Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Feb 1.
Published in final edited form as: Arterioscler Thromb Vasc Biol. 2018 Feb;38(2):e10–e16. doi: 10.1161/ATVBAHA.117.310222

HDL Function in Cardiovascular Disease and Diabetes

Yi He 1,*, Vishal Kothari 1,*, Karin E Bornfeldt 1,2
PMCID: PMC5804739  NIHMSID: NIHMS929162  PMID: 29367232

Cardiovascular disease (CVD) is the most common cause of death world-wide 1. Major CVD risk factors are hypertension, smoking, physical inactivity, abnormal glucose levels/diabetes and dyslipidemia. Among these, dyslipidemia characterized by low levels of high-density lipoprotein cholesterol (HDL-C) is strongly and inversely correlated with CVD risk 2-4, and low HDL-C levels is part of the atherogenic dyslipidemia complex associated with diabetes 5. These observations triggered intense interest in increasing HDL-C levels for therapeutic intervention of CVD 6, 7. However, several recent lines of evidence now suggest that the association between HDL-C levels and CVD status may be indirect or more complicated than previously recognized 7. Thus, human genetic studies demonstrate that genetically altered HDL-C levels do not necessarily translate to an altered risk of CVD 8, 9,10,11, 12. Phase III clinical trials of drugs that elevate HDL-C, such as niacin and cholesteryl ester transfer protein (CETP) inhibitors, also have largely failed to reduce CVD events in statin-treated subjects with established CVD 13-15. For several CETP inhibitors, CVD outcomes were unsuccessful because of off-target effects (torcetrapib) or lack of efficacy (dalcetrapib and evacetrapib) 14, 16-18.The exception is the recent REVEAL trial, which included more subjects and was carried out for longer than previous CETP inhibitor trials. The REVEAL demonstrated that the CETP inhibitor anacetrapib exhibited beneficial effects on cardiovascular outcomes on top of those of statin therapy, although the risk reduction was moderate 19, 20 and might have been due, at least in part, to a reduction in non-HDL cholesterol rather than elevated HDL-C 21. Considering these cumulative observations, it remains uncertain whether increased HDL-C directly impacts atherosclerosis and the risk of cardiovascular events. It has become quite apparent that other metrices of HDL, and specifically of HDL anti-atherogenic functions, must be considered.

HDL is a heterogeneous population of particles with sizes ranging from 7 nm to 14 nm diameter. The largest HDL particles contain the most cholesterol 22. Therefore, HDL-C levels do not always correlate with HDL particle concentration, and furthermore, does not provide information on differences in HDL subpopulations. For example, in a recent study published in ATVB, the effects of two different bariatric surgery procedures on HDL metrics were compared. Sleeve gastrectomy increased HDL-C levels more than did Roux-en-Y gastric bypass surgery, calculated as differences from baseline, but reduced HDL particle concentrations 23. The increase in HDL-C was most likely a reflection of a relative increase in HDL particle size 23. Scientists are now therefore increasingly considering HDL-associated metrics other than HDL-C, such as HDL particle concentration, HDL composition, and HDL function as more likely determinants of CVD risk 24. Thus, a large body of recent research has addressed changes in HDL function and particle subpopulations in pathophysiological conditions, including CVD and diabetes, with the goal of discovering novel and reliable biomarkers and targets for the treatment or prevention of CVD. In this review, we highlight research published in ATVB within the past two years, focusing on studies addressing functions of HDL subpopulations in CVD and diabetes.

HDL classification and quantification methods

HDL particles differ in surface charge, size, and lipid and protein composition, and the ensemble of the HDL particles is determined both by its biogenesis and dynamic remodeling by a range of enzymes, including CETP, lecithin–cholesterol acyltransferase, phospholipid transfer protein, platelet-activating factor acetylhydrolase, and hepatic lipase. Thus, one possible explanation for the discrepancies between the epidemiological, genetic, and clinical trial data is that the measurement of cholesterol mass within HDL fails to capture the complexity of this highly dynamic system. While precipitation techniques are commonly used for rapid measurements of HDL-C mass for clinical purposes, the classic method for measurements and separation of lipoprotein sub-fractions, including HDL, is by density gradient ultracentrifugation 25. HDL is classified according to the analytical method used to isolate it. Based on density gradient ultracentrifugation, HDL is classified into different sub-fractions known as HDL2 (large buoyant particles) and HDL3 (smaller dense particles) 26. HDL sub-fractions can further be assessed and classified based on size into HDL2a and 2b (HDL2b larger than HDL2a), and 3a, 3b, and 3c (smallest) by non-denaturing gel electrophoresis, or by charge and size with 2D gel electrophoresis into pre-β1 and pre-β2 (larger than pre-β1), pre-α1, pre-α2, pre-α3 and pre-α4 (larger to smaller) and α1 and α2 (α1 larger than α2) HDL 26, 27.

A key denominator for assessing HDL function may be the molar concentration of HDL particles in blood as it reflects the quantity of HDL particles independent of its composition (i.e. a small cholesterol-poor HDL particle is equivalent to a large, cholesterol-rich particle). Indeed, clinical studies suggest that HDL particle concentration may provide information on CVD status independent of HDL-C 28, 29. Two methods have been described for quantifying HDL particles in human plasma, one based on nuclear magnetic resonance (NMR) 30, 31 and the other based on ion mobility analysis (IMA) 32, classifying HDL into large, medium, and small sub-fractions. One potential limitation of the NMR method is that it is based on measurements of distinctive NMR signals arising from the HDL particle lipids with assumptions made about the physical properties of HDL lipids and HDL volume. The HDL particle concentration is derived from deconvolution of a complex overlapping NMR signal of all lipoprotein classes (including VLDL and LDL) and has not yet been formally biochemically validated 33. To quantify HDL by IMA, HDL particles are separated on the basis of their differential mobility in a carrier gas and directly counted using laser scattering. 32, 34. The IMA method is further calibrated using both nanoparticles and recombinant HDL, yielding molar concentrations of HDL in line with estimated stoichiometry of apolipoprotein A-I (apoA-I) on HDL in contrast to NMR 34. Size exclusion fast protein liquid chromatography (FPLC) or high-performance liquid chromatography systems (HPLC) can then be used separate lipoproteins according to particle size in solution. Furthermore, structure and composition analyses of HDL particles (proteomic/lipidomic methods and immunoaffinity chromatography), provide additional assessment of HDL particles.

Due to the use of different methods to classify, isolate, measure, and normalize HDL, it is difficult to compare studies and experimental results centered on analyzing HDL functions. Thus, to assess the issue of HDL function as a potential therapeutic target, standardized robust, simple and universal analytical methods will be required in the future.

HDL subpopulations and cholesterol efflux capacity

The cardio-protective nature of HDL has been primarily attributed to its role in promoting reverse cholesterol transport, the net movement of cholesterol from peripheral tissues to the liver for excretion through the bile, in mouse models and human studies. Cholesterol efflux from peripheral cells, such as macrophages in lesions of atherosclerosis, involves the cholesterol transporters ATP-binding cassette transporter A1 (ABCA1) and ABCG1, which are highly induced when macrophages accumulate excess cholesterol. Cholesterol export to lipid-poor apolipoproteins 35 and to small dense HDL 36 by ABCA1 is a key first step in cholesterol efflux from macrophages. ABCG1, which appears to act at least in part intracellularly by redistributing sterols away from the endoplasmic reticulum membrane 37, then mediates the transport of cellular cholesterol to larger more mature lipidated HDL particles 38, 39. A recent study published in ATVB demonstrates that a similar transporter, ABCA8, can also mediate cholesterol efflux to apoA-I 40. However, ABCA8 does not appear to mediate reverse cholesterol transport from macrophages. Instead, ABCA8 acts in the liver, perhaps by interacting with ABCA140 to stimulate HDL biogenesis. Here, the serine/threonine-protein kinase Pim-1 protects ABCA1 from lysosomal degradation, thereby maintaining plasma HDL-C levels 41.

In two large studies by the groups of Rader, Rothblat and colleagues, impaired HDL cholesterol efflux capacity was found to be a stronger predictor of prevalent and incident coronary artery disease than was HDL-C 42, 43. HDL efflux capacity was also found to be decreased in subjects with type 2 diabetes 43. Furthermore, in a separate ten-year follow up study, HDL cholesterol efflux capacity, quantified using a fluorescent substrate assay, was strongly and inversely associated with future cardiovascular events in a large population free of CVD at baseline, whereas HDL-C levels exhibited no such association 44. Recently, Ogura et al 45 came to a similar conclusion by studying patients with familial hypercholesterolemia, who are at high risk for premature atherosclerotic CVD even after statin therapy. They found that cholesterol efflux capacity was independently and inversely associated with the presence of atherosclerotic CVD in heterozygous familial hypercholesterolemia even after adjusting for HDL-C levels 45. In another recent study published in ATVB, Lin and colleagues demonstrated that endogenous cholesterol excretion, as a measure of reverse cholesterol transport, was negatively associated with carotid intima-media thickness in 86 non-diabetic adults, an association that remained significant after adjusting for HDL-C levels 46. Taken together, these observations suggested that the ability of serum HDL to remove cholesterol from peripheral tissues is a promising metric for assessing the cardioprotective effects of HDL, and that HDL’s functionality may be more important than HDL-C levels in predicting CVD risk.

In contrast, in the JUPITER trial (Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin), serum HDL cholesterol efflux capacity was not associated with incident CVD in individuals at baseline 47. Instead, HDL particle number, measured by NMR, was the strongest of four HDL-related biomarkers (cholesterol efflux capacity, HDL-C, apoA-I, and HDL particle number) as an inverse predictor of incident events and biomarker of residual risk. Therefore, the question remains whether serum HDL cholesterol efflux capacity is a better clinically relevant measure of HDL functions than other biomarkers. Recently, Koekemoer and colleagues quantified serum HDL cholesterol efflux capacity of 1988 individuals from the GRAPHIC (Genetic Regulation of Arterial Pressure of Humans in the Community) cohort, comprising individuals from 2 generations 48. They found that cholesterol efflux capacity is a stable trait over time in an individual, but a trait that is only modestly heritable between generations 48. This study also indicated that cholesterol efflux capacity can be determined by multiple specific clinical, serum, and HDL parameters beyond HDL-C, for example, age, systolic blood pressure, triglycerides, phospholipids, lipoprotein(a), HDL particle number, HDL particle size, and apolipoprotein A-II levels 48. To date, it is unknown if HDL’s cholesterol efflux capacity is a universally better predictor than HDL-C for CVD events 49. To understand this issue, we need to better define HDL and its metabolism.

HDL is a heterogeneous population of particles that may collectively contain >95 different proteins 50, 51. Each HDL subspecies can have distinct functions and may be metabolized through different pathways in humans. For example, Du and colleagues used reconstructed HDL and endogenous human HDL isolated by ultracentrifugation to demonstrate that small dense HDLs (HDL3b and HDL3c), not medium and large HDLs (HDL2), are the most efficient lipidated mediators of cholesterol efflux by the ABCA1 pathway, similar to that of lipid-free apoA-I, while ABCG1 has a negligible role in mediating cholesterol efflux to HDL3b and HDL3c subfractions 36. Furthermore, a recent ATVB study showed that HDL subspecies may be metabolized within discrete and stable sizes in humans, suggesting that stepwise size expansion and contraction as major pathways for HDL metabolism may be less important than previously believed22. Four standard HDL size subfractions were each secreted into plasma and circulated mainly within the secreted size for 1–4 days before they were removed from circulation. Thus, subjects with high HDL-C (larger HDL particles) had an increased secretion of large HDL and faster clearance of very small HDL, and subjects with lower HDL-C had a faster clearance of large and very large HDL. In this context, Xu et al showed that the cholesterol and phospholipids of HDL and the structural apoA-I protein are cleared by independent mechanisms that do not involve holo-particle uptake 52. Interesting questions relate to how different HDL therapies affect these HDL subspecies and whether they provide clues to understanding mechanisms of CVD.

Niacin and CETP inhibitors raise HDL-C levels through different mechanisms. Do they also have different effects on HDL’s serum efflux capacity? Niacin raises plasma HDL-C by decreasing the fractional clearance rate of HDL-apoA-I without affecting its synthetic rate53-55. Rosein et al recently analyzed the impact of niacin and statin therapies on HDL related biomarkers56. These researchers reported that niacin markedly increased HDL-C levels, as expected, but failed to improve ABCA1-specific cholesterol efflux 56. Accordingly, HDL particle analysis revealed that niacin selectively raised levels of large HDL instead of boosting levels of small HDL particles, which are superior to large HDL in promoting ABCA1-specific cholesterol efflux. This study and others 57 provided a possible mechanistic explanation for why niacin failed to improve HDL’s atheroprotective properties in humans.

CETP inhibitors increase HDL-C by preventing transfer of cholesteryl esters from HDL to other lipoprotein particles, but it is unclear how they affect HDL functions. Recently, Zhang and colleagues developed a CETP knockout rabbit by zinc finger nuclease-mediated gene targeting to clarify the pathophysiological functions of CETP in atherosclerosis 58. This study supported a beneficial role of CETP inhibition, because deletion of CETP protected against cholesterol diet–induced atherosclerosis concomitant with an increase in HDL-C and an increase in cholesterol efflux capacity, as compared with the wildtype controls 58. However, like in the REVEAL trial, plasma levels of total cholesterol were reduced by CETP inhibition, making it difficult to conclude that improved HDL function was responsible for the anti-atherosclerotic effects. Reyes-Soffer et al recently investigated the effects of anacetrapib on HDL subpopulations in human subjects in the presence or absence of statin therapy 59. Similar to effects of niacin, this group observed an increase in HDL-C and apoA-I levels due to a reduction in the fractional clearance rate of HDL apoA-I, and a concomitant shift in HDL subspecies from smaller to larger HDL particles 59. However, unlike niacin therapy, the absolute mass of apoA-I in pre-β particles increased 59, which supports the idea that inhibition of CETP results in improved cholesterol efflux capacity 57, 60.

The concept that diabetes might impair HDL’s cholesterol efflux capacity was addressed by Apro and colleagues 61. They showed that HDL isolated by ultracentrifugation from plasma and interstitial fluid from suction blisters from subjects with diabetes exhibited reduced cholesterol efflux capacity when normalized to HDL cholesterol content, as compared with HDL from subjects without diabetes, and that the defect in cholesterol efflux was more pronounced in interstitial fluid than in plasma. The results also demonstrated that HDL from subjects with diabetes on average was smaller (determined by gel filtration FPLC) than HDL from controls, and that these subjects had reduced levels of apoA-I in both plasma and interstitial fluid. The measurements in interstitial fluid are important because cholesterol efflux occurs in peripheral tissues, such as lesions of atherosclerosis, rather than in circulation. A recent study by Mehta and colleagues supports the concept that reactive oxygen species, which are believed to be increased in diabetes, can impact HDL function. They demonstrated that hemopexin, a heme scavenger, results in reduced reactive oxygen species, and with reduced pro-inflammatory actions and increased cholesterol efflux capacity of HDL, concomitant with increased atherosclerosis 62.

These studies demonstrate that HDL is a heterogeneous population of particles that can be divided into multiple subspecies, and that HDL measures other than HDL-C need to be considered. Each of the HDL subspecies may have distinct functions and may be metabolized partly through distinct pathways. Further studies are needed to elucidate the role of each HDL subspecies and possibilities to take advantage of this new knowledge for development of CVD therapeutics.

Direct endothelial cell effects of HDL in CVD and diabetes

In addition to promoting cholesterol efflux from macrophages, HDL can exert anti-atherosclerotic effects in endothelial cells, such as provide direct stimulation of endothelial nitric oxide (NO) production (a vasoprotective molecule), anti-inflammatory effects, and anti-oxidant effects 63. Several different mechanisms have been proposed to account for the endothelial NO-stimulating capacity of HDL. Early studies suggested that HDL acts by preventing the detrimental effects of oxidized LDL on endothelial NO-synthase (eNOS), while a subsequent study by Yuhanna et al suggested that HDL can bind to endothelial scavenger receptor class B type 1 (SR-BI) and thereby directly stimulate eNOS-mediated NO production through a process that involves eNOS stimulation in plasmalemmal caveolae 64. Mechanistically, binding of HDL to SR-BI initially leads to tyrosine kinase Src-mediated activation of phosphoinositide (PI) 3-kinase (PI3K), which in turn activates Akt and Erk pathways 65. The activation of endothelial Akt has been shown to stimulate phosphorylation of eNOS at serine residue 1177, known to be an important regulatory mechanism leading to eNOS activation 65. HDL also can preserve endothelial function by improving ABCG1-mediated efflux of oxysterols, thereby preventing inhibitory interactions of eNOS with caveolin-1, and restoring eNOS activity in cholesterol-loaded endothelial cells 66.

HDL must be transported through endothelial cell barriers to reach atherosclerotic lesion macrophages and other peripheral tissues. Velagapudi et al recently used microscopy-based high-content screening of 141 kinase inhibiting drugs to demonstrate that vascular endothelial growth factor receptor 2 (VEGFR2) is required for transendothelial transport of HDL, but not LDL 67. Furthermore, VEGF-A was found to be required for the localization of SR-BI in the plasma membrane of endothelial cells. These results indicate that VEGF-A may play an important regulatory role in the vascular protective effects of HDL 67.

Stimulation of eNOS-mediated NO production is also induced by binding of HDL-associated sphingosine-1-phosphate (S1P) to the lysophospholipid receptor S1P3, which is expressed in endothelial cells and may partially mediate HDL- and lysophospholipid-induced vasodilation 68. Studies have revealed that apolipoprotein M (apoM), a minor apolipoprotein on HDL, is a carrier and a modulator of S1P 68. A recent report defines apoM-bound S1P as a key component of HDL, which is partially responsible for several HDL-associated protective functions in the endothelium, including anti-inflammatory effects, regulation of adhesion molecule expression, leukocyte-endothelial adhesion, and endothelial barrier function 68. However, it is important to note that not all S1P in HDL is associated with apoM, as HDL from apoM-deficient mice still carries appreciable amounts of S1P and exerts similar biological activities as HDL from control mice 69.

Notably, current evidence suggests that vascular effects of HDL can be highly heterogeneous and may be altered in patients with CVD and diabetes. Thus, when normalized based on HDL protein content, HDL preparations from subjects with diabetes failed to stimulate endothelial cell NO production and to promote endothelial repair when compared with HDL from subjects without diabetes 70. Accordingly, subjects with type 2 diabetes were shown to have reduced levels of S1P in HDL 71, and HDL from normoglycemic patients with metabolic syndrome, before onset of diabetes, exhibited a reduced ability to activate eNOS; an defect explained by S1P depletion of HDL 72.

To understand the functional disparity of different HDL subfractions, Frej et al carried out an interesting study on HDL isolated from women with type 1 diabetes and matched control subjects. They showed that buoyant HDL is less anti-inflammatory in endothelial cells, as compared with dense HDL 73. They also demonstrated that type 1 diabetic subjects have similar levels of S1P and apoM in total HDL as compared with controls, but that the HDL-associated apoM/S1P complex shifts from dense to buoyant HDL particles in type 1 diabetes, where the complex is less able to suppress expression of the adhesion molecule VCAM-1 73. These reduced anti-inflammatory effects of apoM/S1P-containing buoyant HDL were explained by differential activation of the S1P1 receptor, leading to differential Akt and Erk activation depending on the density and size of the HDL particle 73. Recently, it has been shown that CETP can shift the distribution of S1P and apoM from HDL to apoB-containing lipoproteins, on which S1P might exert more potent bioactivities 74. In this regard, Wu et al showed that increasing HDL-C levels by inhibiting CETP activity with the anacetrapib analog des-fluoro-anacetrapib reduced intimal thickening and regenerated functional endothelium in New Zealand White rabbits after balloon injury in an SR-B1-dependent and PI3K/Akt-dependent manner 75, 76. It will be interesting to determine the effects of CETP inhibitors on the S1P/apoM complex in different lipoproteins, and its functional repercussions.

Together, these studies suggest that the biological functions of HDL in the endothelium are heterogeneous, depend on signal transduction mediated by different receptors and alterations in cellular membranes, and might be altered in patients with CVD and diabetes. Recent and future mechanistic and clinical observations may have profound implications for the understanding of direct vasoprotective effects of HDL and how these relate to HDL’s cholesterol efflux capacities.

HDL regulation by epigenetic mechanisms, microRNAs, and transcription factors

Several studies demonstrate that genetic variations that associate with altered HDL-C levels do not strongly associate with altered CVD risk 77, 78. However, alterations in epigenetic mechanisms, microRNAs and transcription factors have recently been shown to impact HDL function. Thus, epigenetic DNA methylation at different loci associates with HDL-C levels, but its role in HDL functions was not clear 79. Recently, Elosua et al designed an epigenome-wide association study with individuals from the REGICOR study (Registre Gironi del Cor) to investigate the association between DNA methylation, HDL cholesterol efflux capacity and HDL’s “inflammatory index” 80. They identified five methylation sites in HOXA3, PEX5, PER3, and CMIP (two sites) associated with HDL cholesterol efflux capacity and one methylation site in GABRR1 associated with HDL inflammatory index 80. Further work is needed to validate these associations and their functional significance in humans.

Work over the past several years has identified microRNAs (miRNAs) as important regulators of HDL-C metabolism 81, 82. MicroRNAs, small non-coding RNAs, control most of the steps of the reverse cholesterol transport pathway, including HDL biogenesis, cellular cholesterol efflux, and hepatic HDL-C uptake 83. In particular, miR-33a/b and miR-27b have been found to act as regulatory hubs that repress genes involved in lipid metabolism 84-86. MicroRNA-33a/b targets genes involved in cholesterol metabolism and insulin signaling, including ABCA1 and ABCG1, the endolysosomal transport protein Niemann-Pick C1 (NPC1) and insulin receptor substrate 2 (IRS2) 86, 87. MicroRNA-27b regulates the expression of several key lipid metabolism genes, including angiopoietin-like 3 (ANGPTL3), which increases triglycerides and HDL-C 84. Interestingly, HDL isolated from miRNA-33-deficient mice exhibits increased ability to suppress expression of matrix metalloproteinase 9 in macrophages and monocyte chemotactic protein-1 in vascular smooth muscle cells, suggesting that HDL function is altered by miR-33 88. In recent work by Ouimet et al, oxysterol-binding protein-like 6 (OSBPL6) was identified as a novel target gene in the miR-33/miR27b network that contributes to cholesterol homeostasis and HDL cholesterol efflux capacity 89. Knockdown of OSBPL6 in mice resulted in aberrant clustering of endosomes and accumulation of free cholesterol in these structures, which resulted in reduced cholesterol esterification in the endoplasmic reticulum. In contrast, overexpression of OSBPL6 enhanced cholesterol trafficking and cholesterol efflux in macrophages and hepatocytes 89. Hepatic expression of OSBPL6 was positively correlated with plasma levels of HDL-C in humans, although the correlation was relatively weak 89.

Lastly, recent work in mice demonstrates that the transcription factors cAMP-responsive element-binding protein H (CREBH) 90 and iodothyronine deiodinase 1 (DIO1) 91 can regulate apoA-I expression. In particular, Liu et al found that hepatic insulin signaling plays a significant role in the regulation of Apoa1 gene expression through the regulation of Dio191. This work suggested a new pathway linking hepatic insulin signaling, Dio1 and apoA-I, which may be relevant to the increased risk of CVD in subjects with metabolic syndrome or type 2 diabetes.

Thus, HDL and HDL functions are modulated by a large number of mechanisms. Future studies are needed to explore these mechanisms as possible treatment strategies.

Summary

Recent articles published in ATVB and elsewhere have highlighted the heterogeneity of HDL and the effects of different HDL subpopulations in cells as they relate to CVD risk. A relatively new focus of research is to understand the links among HDL composition and HDL functions, including cholesterol efflux capacity, endothelial anti-inflammatory effects, and effects in other cells. The molecular mechanisms leading to HDL with optimal cardioprotective effects, and the possibility of the presence of dysfunctional HDL, need to be further studied and validated in large clinical studies in subjects with cardiovascular disease and diabetes.

Acknowledgments

Sources of Funding

Research in the authors’ laboratories is funded in part by the National Institutes of Health, grants DP3DK108209, P01HL092969, P30 DK017047 and P01HL128203.

Footnotes

Disclosures

None

References

  • 1.Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart disease and stroke statistics-2017 update: A report from the american heart association. Circulation. 2017;135:e146–e603. doi: 10.1161/CIR.0000000000000485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Gordon DJ, Rifkind BM. High-density lipoprotein–the clinical implications of recent studies. The New England journal of medicine. 1989;321:1311–1316. doi: 10.1056/NEJM198911093211907. [DOI] [PubMed] [Google Scholar]
  • 3.Mahdy Ali K, Wonnerth A, Huber K, Wojta J. Cardiovascular disease risk reduction by raising hdl cholesterol–current therapies and future opportunities. British journal of pharmacology. 2012;167:1177–1194. doi: 10.1111/j.1476-5381.2012.02081.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wilson PW, Abbott RD, Castelli WP. High density lipoprotein cholesterol and mortality. The framingham heart study. Arteriosclerosis. 1988;8:737–741. doi: 10.1161/01.atv.8.6.737. [DOI] [PubMed] [Google Scholar]
  • 5.Xiao C, Dash S, Morgantini C, Hegele RA, Lewis GF. Pharmacological targeting of the atherogenic dyslipidemia complex: The next frontier in cvd prevention beyond lowering ldl cholesterol. Diabetes. 2016;65:1767–1778. doi: 10.2337/db16-0046. [DOI] [PubMed] [Google Scholar]
  • 6.Kempen HJ, Asztalos BF, Moerland M, Jeyarajah E, Otvos J, Kallend DG, Bellibas SE, Wijngaard PL. High-density lipoprotein subfractions and cholesterol efflux capacities after infusion of mdco-216 (apolipoprotein a-imilano/palmitoyl-oleoyl-phosphatidylcholine) in healthy volunteers and stable coronary artery disease patients. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:736–742. doi: 10.1161/ATVBAHA.115.307052. [DOI] [PubMed] [Google Scholar]
  • 7.Hoekstra M, Van Berkel TJ. Functionality of high-density lipoprotein as antiatherosclerotic therapeutic target. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:e87–e94. doi: 10.1161/ATVBAHA.116.308262. [DOI] [PubMed] [Google Scholar]
  • 8.Rader DJ, Hovingh GK. Hdl and cardiovascular disease. Lancet. 2014;384:618–625. doi: 10.1016/S0140-6736(14)61217-4. [DOI] [PubMed] [Google Scholar]
  • 9.Rosenson RS. The high-density lipoprotein puzzle: Why classic epidemiology, genetic epidemiology, and clinical trials conflict? Arteriosclerosis, thrombosis, and vascular biology. 2016;36:777–782. doi: 10.1161/ATVBAHA.116.307024. [DOI] [PubMed] [Google Scholar]
  • 10.Mank-Seymour AR, Durham KL, Thompson JF, Seymour AB, Milos PM. Association between single-nucleotide polymorphisms in the endothelial lipase (lipg) gene and high-density lipoprotein cholesterol levels. Biochimica et biophysica acta. 2004;1636:40–46. doi: 10.1016/j.bbalip.2003.12.001. [DOI] [PubMed] [Google Scholar]
  • 11.Zanoni P, Khetarpal SA, Larach DB, et al. Rare variant in scavenger receptor bi raises hdl cholesterol and increases risk of coronary heart disease. Science. 2016;351:1166–1171. doi: 10.1126/science.aad3517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Trigatti BL, Hegele RA. Rare genetic variants and high-density lipoprotein: Marching to a different drum. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:e53–55. doi: 10.1161/ATVBAHA.116.307688. [DOI] [PubMed] [Google Scholar]
  • 13.Rader DJ, Tall AR. The not-so-simple hdl story: Is it time to revise the hdl cholesterol hypothesis? Nature medicine. 2012;18:1344–1346. doi: 10.1038/nm.2937. [DOI] [PubMed] [Google Scholar]
  • 14.Schwartz GG, Olsson AG, Abt M, et al. Effects of dalcetrapib in patients with a recent acute coronary syndrome. The New England journal of medicine. 2012;367:2089–2099. doi: 10.1056/NEJMoa1206797. [DOI] [PubMed] [Google Scholar]
  • 15.Investigators A-H. 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. The New England journal of medicine. 2011;365:2255–2267. doi: 10.1056/NEJMoa1107579. [DOI] [PubMed] [Google Scholar]
  • 16.Barter PJ, Caulfield M, Eriksson M, et al. Effects of torcetrapib in patients at high risk for coronary events. The New England journal of medicine. 2007;357:2109–2122. doi: 10.1056/NEJMoa0706628. [DOI] [PubMed] [Google Scholar]
  • 17.Nissen SE, Tardif JC, Nicholls SJ, Revkin JH, Shear CL, Duggan WT, Ruzyllo W, Bachinsky WB, Lasala GP, Tuzcu EM, Investigators I Effect of torcetrapib on the progression of coronary atherosclerosis. The New England journal of medicine. 2007;356:1304–1316. doi: 10.1056/NEJMoa070635. [DOI] [PubMed] [Google Scholar]
  • 18.McLain JH, Alsterda AJ, Arora RR. Cholesteryl ester transfer protein inhibitors: Trials and tribulations. Journal of cardiovascular pharmacology and therapeutics. 2016 doi: 10.1177/1074248416662349. In press. [DOI] [PubMed] [Google Scholar]
  • 19.Group HTRC. 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. The New England journal of medicine. 2017;377:1217–1227. doi: 10.1056/NEJMoa1706444. [DOI] [PubMed] [Google Scholar]
  • 20.Barter PJ, Rye KA. Cholesteryl ester transfer protein inhibition is not yet dead–pro. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:439–441. doi: 10.1161/ATVBAHA.115.306879. [DOI] [PubMed] [Google Scholar]
  • 21.Tall AR, Rader DJ. The trials and tribulations of cetp inhibitors. Circulation research. 2017 doi: 10.1161/CIRCRESAHA.117.311978. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Mendivil CO, Furtado J, Morton AM, Wang L, Sacks FM. Novel pathways of apolipoprotein a-i metabolism in high-density lipoprotein of different sizes in humans. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:156–165. doi: 10.1161/ATVBAHA.115.306138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Heffron SP, Lin B, Parikh M, Scolaro B, Adelman SJ, Collins HL, Berger JS, Fisher EA. Changes in high-density lipoprotein cholesterol efflux capacity after bariatric surgery are procedure dependent. Arteriosclerosis, thrombosis, and vascular biology. 2017 doi: 10.1161/ATVBAHA.117.310102. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Heinecke JW, Bornfeldt KE. A long road ahead for discovering new hdl metrics that reflect cardiovascular disease risk. Journal of the American College of Cardiology. 2017;70:179–181. doi: 10.1016/j.jacc.2017.05.038. [DOI] [PubMed] [Google Scholar]
  • 25.Hafiane A, Genest J. High density lipoproteins: Measurement techniques and potential biomarkers of cardiovascular risk. BBA clinical. 2015;3:175–188. doi: 10.1016/j.bbacli.2015.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Pirillo A, Norata GD, Catapano AL. High-density lipoprotein subfractions–what the clinicians need to know. Cardiology. 2013;124:116–125. doi: 10.1159/000346463. [DOI] [PubMed] [Google Scholar]
  • 27.Asztalos BF, Tani M, Schaefer EJ. Metabolic and functional relevance of hdl subspecies. Curr Opin Lipidol. 2011;22:176–185. doi: 10.1097/MOL.0b013e3283468061. [DOI] [PubMed] [Google Scholar]
  • 28.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) Journal of the American College of Cardiology. 2012;60:508–516. doi: 10.1016/j.jacc.2012.03.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Mora S, Caulfield MP, Wohlgemuth J, Chen Z, Superko HR, Rowland CM, Glynn RJ, Ridker PM, Krauss RM. Atherogenic lipoprotein subfractions determined by ion mobility and first cardiovascular events after random allocation to high-intensity statin or placebo: The justification for the use of statins in prevention: An intervention trial evaluating rosuvastatin (jupiter) trial. Circulation. 2015;132:2220–2229. doi: 10.1161/CIRCULATIONAHA.115.016857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Jeyarajah EJ, Cromwell WC, Otvos JD. Lipoprotein particle analysis by nuclear magnetic resonance spectroscopy. Clinics in laboratory medicine. 2006;26:847–870. doi: 10.1016/j.cll.2006.07.006. [DOI] [PubMed] [Google Scholar]
  • 31.Bell G, Mora S, Greenland P, Tsai M, Gill E, Kaufman JD. Association of air pollution exposures with high-density lipoprotein cholesterol and particle number: The multi-ethnic study of atherosclerosis. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:976–982. doi: 10.1161/ATVBAHA.116.308193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Caulfield MP, Li S, Lee G, Blanche PJ, Salameh WA, Benner WH, Reitz RE, Krauss RM. Direct determination of lipoprotein particle sizes and concentrations by ion mobility analysis. Clinical chemistry. 2008;54:1307–1316. doi: 10.1373/clinchem.2007.100586. [DOI] [PubMed] [Google Scholar]
  • 33.Davidson WS. Hdl-c vs hdl-p: How changing one letter could make a difference in understanding the role of high-density lipoprotein in disease. Clinical chemistry. 2014;60:e1–3. doi: 10.1373/clinchem.2014.232769. [DOI] [PubMed] [Google Scholar]
  • 34.Hutchins PM, Ronsein GE, Monette JS, Pamir N, Wimberger J, He Y, Anantharamaiah GM, Kim DS, Ranchalis JE, Jarvik GP, Vaisar T, Heinecke JW. Quantification of hdl particle concentration by calibrated ion mobility analysis. Clinical chemistry. 2014;60:1393–1401. doi: 10.1373/clinchem.2014.228114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Oram JF, Heinecke JW. Atp-binding cassette transporter a1: A cell cholesterol exporter that protects against cardiovascular disease. Physiological reviews. 2005;85:1343–1372. doi: 10.1152/physrev.00005.2005. [DOI] [PubMed] [Google Scholar]
  • 36.Du XM, Kim MJ, Hou L, et al. Hdl particle size is a critical determinant of abca1-mediated macrophage cellular cholesterol export. Circulation research. 2015;116:1133–1142. doi: 10.1161/CIRCRESAHA.116.305485. [DOI] [PubMed] [Google Scholar]
  • 37.Tarling EJ, Edwards PA. Intracellular localization of endogenous mouse abcg1 is mimicked by both abcg1-l550 and abcg1-p550-brief report. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1323–1327. doi: 10.1161/ATVBAHA.116.307414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kennedy MA, Barrera GC, Nakamura K, Baldan A, Tarr P, Fishbein MC, Frank J, Francone OL, Edwards PA. Abcg1 has a critical role in mediating cholesterol efflux to hdl and preventing cellular lipid accumulation. Cell metabolism. 2005;1:121–131. doi: 10.1016/j.cmet.2005.01.002. [DOI] [PubMed] [Google Scholar]
  • 39.Yvan-Charvet L, Wang N, Tall AR. Role of hdl, abca1, and abcg1 transporters in cholesterol efflux and immune responses. Arteriosclerosis, thrombosis, and vascular biology. 2010;30:139–143. doi: 10.1161/ATVBAHA.108.179283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Trigueros-Motos L, van Capelleveen JC, Torta F, et al. Abca8 regulates cholesterol efflux and high-density lipoprotein cholesterol levels. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:2147–2155. doi: 10.1161/ATVBAHA.117.309574. [DOI] [PubMed] [Google Scholar]
  • 41.Katsube A, Hayashi H, Kusuhara H. Pim-1l protects cell surface-resident abca1 from lysosomal degradation in hepatocytes and thereby regulates plasma high-density lipoprotein level. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:2304–2314. doi: 10.1161/ATVBAHA.116.308472. [DOI] [PubMed] [Google Scholar]
  • 42.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. The New England journal of medicine. 2011;364:127–135. doi: 10.1056/NEJMoa1001689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Saleheen D, Scott R, Javad S, et al. Association of hdl cholesterol efflux capacity with incident coronary heart disease events: A prospective case-control study. The lancet. Diabetes & endocrinology. 2015;3:507–513. doi: 10.1016/S2213-8587(15)00126-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.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. The New England journal of medicine. 2014;371:2383–2393. doi: 10.1056/NEJMoa1409065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ogura M, Hori M, Harada-Shiba M. Association between cholesterol efflux capacity and atherosclerotic cardiovascular disease in patients with familial hypercholesterolemia. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:181–188. doi: 10.1161/ATVBAHA.115.306665. [DOI] [PubMed] [Google Scholar]
  • 46.Lin X, Racette SB, Ma L, Wallendorf M, Davila-Roman VG, Ostlund RE., Jr Endogenous cholesterol excretion is negatively associated with carotid intima-media thickness in humans. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:2364–2369. doi: 10.1161/ATVBAHA.117.310081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Khera AV, Demler OV, Adelman SJ, Collins HL, Glynn RJ, Ridker PM, Rader DJ, Mora S. Cholesterol efflux capacity, high-density lipoprotein particle number, and incident cardiovascular events: An analysis from the jupiter trial (justification for the use of statins in prevention: An intervention trial evaluating rosuvastatin) Circulation. 2017;135:2494–2504. doi: 10.1161/CIRCULATIONAHA.116.025678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Koekemoer AL, Codd V, Masca NGD, Nelson CP, Musameh MD, Kaess BM, Hengstenberg C, Rader DJ, Samani NJ. Large-scale analysis of determinants, stability, and heritability of high-density lipoprotein cholesterol efflux capacity. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:1956–1962. doi: 10.1161/ATVBAHA.117.309201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Li XM, Tang WH, Mosior MK, Huang Y, Wu Y, Matter W, Gao V, Schmitt D, Didonato JA, Fisher EA, Smith JD, Hazen SL. Paradoxical association of enhanced cholesterol efflux with increased incident cardiovascular risks. Arteriosclerosis, thrombosis, and vascular biology. 2013;33:1696–1705. doi: 10.1161/ATVBAHA.113.301373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Vaisar T, Pennathur S, Green PS, et al. Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of hdl. The Journal of clinical investigation. 2007;117:746–756. doi: 10.1172/JCI26206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Shah AS, Tan L, Long JL, Davidson WS. Proteomic diversity of high density lipoproteins: Our emerging understanding of its importance in lipid transport and beyond. Journal of lipid research. 2013;54:2575–2585. doi: 10.1194/jlr.R035725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Xu B, Gillard BK, Gotto AM, Jr, Rosales C, Pownall HJ. Abca1-derived nascent high-density lipoprotein-apolipoprotein ai and lipids metabolically segregate. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:2260–2270. doi: 10.1161/ATVBAHA.117.310290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Blum CB, Levy RI, Eisenberg S, Hall M, 3rd, Goebel RH, Berman M. High density lipoprotein metabolism in man. The Journal of clinical investigation. 1977;60:795–807. doi: 10.1172/JCI108833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Shepherd J, Packard CJ, Patsch JR, Gotto AM, Jr, Taunton OD. Effects of nicotinic acid therapy on plasma high density lipoprotein subfraction distribution and composition and on apolipoprotein a metabolism. The Journal of clinical investigation. 1979;63:858–867. doi: 10.1172/JCI109385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Jin FY, Kamanna VS, Kashyap ML. Niacin decreases removal of high-density lipoprotein apolipoprotein a-i but not cholesterol ester by hep g2 cells. Implication for reverse cholesterol transport. Arteriosclerosis, thrombosis, and vascular biology. 1997;17:2020–2028. doi: 10.1161/01.atv.17.10.2020. [DOI] [PubMed] [Google Scholar]
  • 56.Ronsein GE, Hutchins PM, Isquith D, Vaisar T, Zhao XQ, Heinecke JW. Niacin therapy increases high-density lipoprotein particles and total cholesterol efflux capacity but not abca1-specific cholesterol efflux in statin-treated subjects. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:404–411. doi: 10.1161/ATVBAHA.115.306268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yvan-Charvet L, Kling J, Pagler T, Li H, Hubbard B, Fisher T, Sparrow CP, Taggart AK, Tall AR. Cholesterol efflux potential and antiinflammatory properties of high-density lipoprotein after treatment with niacin or anacetrapib. Arteriosclerosis, thrombosis, and vascular biology. 2010;30:1430–1438. doi: 10.1161/ATVBAHA.110.207142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhang J, Niimi M, Yang D, et al. Deficiency of cholesteryl ester transfer protein protects against atherosclerosis in rabbits. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:1068–1075. doi: 10.1161/ATVBAHA.117.309114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Reyes-Soffer G, Millar JS, Ngai C, et al. Cholesteryl ester transfer protein inhibition with anacetrapib decreases fractional clearance rates of high-density lipoprotein apolipoprotein a-i and plasma cholesteryl ester transfer protein. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:994–1002. doi: 10.1161/ATVBAHA.115.306680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Nicholls SJ, Ruotolo G, Brewer HB, Kane JP, Wang MD, Krueger KA, Adelman SJ, Nissen SE, Rader DJ. Cholesterol efflux capacity and pre-beta-1 hdl concentrations are increased in dyslipidemic patients treated with evacetrapib. Journal of the American College of Cardiology. 2015;66:2201–2210. doi: 10.1016/j.jacc.2015.09.013. [DOI] [PubMed] [Google Scholar]
  • 61.Apro J, Tietge UJ, Dikkers A, Parini P, Angelin B, Rudling M. Impaired cholesterol efflux capacity of high-density lipoprotein isolated from interstitial fluid in type 2 diabetes mellitus-brief report. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:787–791. doi: 10.1161/ATVBAHA.116.307385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mehta NU, Grijalva V, Hama S, Wagner A, Navab M, Fogelman AM, Reddy ST. Apolipoprotein e-/- mice lacking hemopexin develop increased atherosclerosis via mechanisms that include oxidative stress and altered macrophage function. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1152–1163. doi: 10.1161/ATVBAHA.115.306991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Besler C, Luscher TF, Landmesser U. Molecular mechanisms of vascular effects of high-density lipoprotein: Alterations in cardiovascular disease. EMBO molecular medicine. 2012;4:251–268. doi: 10.1002/emmm.201200224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yuhanna IS, Zhu Y, Cox BE, Hahner LD, Osborne-Lawrence S, Lu P, Marcel YL, Anderson RG, Mendelsohn ME, Hobbs HH, Shaul PW. High-density lipoprotein binding to scavenger receptor-bi activates endothelial nitric oxide synthase. Nature medicine. 2001;7:853–857. doi: 10.1038/89986. [DOI] [PubMed] [Google Scholar]
  • 65.Mineo C, Yuhanna IS, Quon MJ, Shaul PW. High density lipoprotein-induced endothelial nitric-oxide synthase activation is mediated by akt and map kinases. The Journal of biological chemistry. 2003;278:9142–9149. doi: 10.1074/jbc.M211394200. [DOI] [PubMed] [Google Scholar]
  • 66.Terasaka N, Westerterp M, Koetsveld J, Fernandez-Hernando C, Yvan-Charvet L, Wang N, Sessa WC, Tall AR. Atp-binding cassette transporter g1 and high-density lipoprotein promote endothelial no synthesis through a decrease in the interaction of caveolin-1 and endothelial no synthase. Arteriosclerosis, thrombosis, and vascular biology. 2010;30:2219–2225. doi: 10.1161/ATVBAHA.110.213215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Velagapudi S, Yalcinkaya M, Piemontese A, Meier R, Norrelykke SF, Perisa D, Rzepiela A, Stebler M, Stoma S, Zanoni P, Rohrer L, von Eckardstein A. Vegf-a regulates cellular localization of sr-bi as well as transendothelial transport of hdl but not ldl. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:794–803. doi: 10.1161/ATVBAHA.117.309284. [DOI] [PubMed] [Google Scholar]
  • 68.Ruiz M, Frej C, Holmer A, Guo LJ, Tran S, Dahlback B. High-density lipoprotein-associated apolipoprotein m limits endothelial inflammation by delivering sphingosine-1-phosphate to the sphingosine-1-phosphate receptor 1. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:118–129. doi: 10.1161/ATVBAHA.116.308435. [DOI] [PubMed] [Google Scholar]
  • 69.Sutter I, Park R, Othman A, Rohrer L, Hornemann T, Stoffel M, Devuyst O, von Eckardstein A. Apolipoprotein m modulates erythrocyte efflux and tubular reabsorption of sphingosine-1-phosphate. Journal of lipid research. 2014;55:1730–1737. doi: 10.1194/jlr.M050021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sorrentino SA, Besler C, Rohrer L, et al. Endothelial-vasoprotective effects of high-density lipoprotein are impaired in patients with type 2 diabetes mellitus but are improved after extended-release niacin therapy. Circulation. 2010;121:110–122. doi: 10.1161/CIRCULATIONAHA.108.836346. [DOI] [PubMed] [Google Scholar]
  • 71.Brinck JW, Thomas A, Lauer E, et al. Diabetes mellitus is associated with reduced high-density lipoprotein sphingosine-1-phosphate content and impaired high-density lipoprotein cardiac cell protection. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:817–824. doi: 10.1161/ATVBAHA.115.307049. [DOI] [PubMed] [Google Scholar]
  • 72.Denimal D, Monier S, Brindisi MC, Petit JM, Bouillet B, Nguyen A, Demizieux L, Simoneau I, Pais de Barros JP, Verges B, Duvillard L. Impairment of the ability of hdl from patients with metabolic syndrome but without diabetes mellitus to activate enos: Correction by s1p enrichment. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:804–811. doi: 10.1161/ATVBAHA.117.309287. [DOI] [PubMed] [Google Scholar]
  • 73.Frej C, Mendez AJ, Ruiz M, Castillo M, Hughes TA, Dahlback B, Goldberg RB. A shift in apom/s1p between hdl-particles in women with type 1 diabetes mellitus is associated with impaired anti-inflammatory effects of the apom/s1p complex. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:1194–1205. doi: 10.1161/ATVBAHA.117.309275. [DOI] [PubMed] [Google Scholar]
  • 74.Kurano M, Hara M, Ikeda H, Tsukamoto K, Yatomi Y. Involvement of cetp (cholesteryl ester transfer protein) in the shift of sphingosine-1-phosphate among lipoproteins and in the modulation of its functions. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:506–514. doi: 10.1161/ATVBAHA.116.308692. [DOI] [PubMed] [Google Scholar]
  • 75.Wu BJ, Li Y, Ong KL, Sun Y, Shrestha S, Hou L, Johns D, Barter PJ, Rye KA. Reduction of in-stent restenosis by cholesteryl ester transfer protein inhibition. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:2333–2341. doi: 10.1161/ATVBAHA.117.310051. [DOI] [PubMed] [Google Scholar]
  • 76.Wu BJ, Shrestha S, Ong KL, Johns D, Hou L, Barter PJ, Rye KA. Cholesteryl ester transfer protein inhibition enhances endothelial repair and improves endothelial function in the rabbit. Arteriosclerosis, thrombosis, and vascular biology. 2015;35:628–636. doi: 10.1161/ATVBAHA.114.304747. [DOI] [PubMed] [Google Scholar]
  • 77.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;299:2524–2532. doi: 10.1001/jama.299.21.2524. [DOI] [PubMed] [Google Scholar]
  • 78.Voight BF, Peloso GM, Orho-Melander M, et al. Plasma hdl cholesterol and risk of myocardial infarction: A mendelian randomisation study. Lancet. 2012;380:572–580. doi: 10.1016/S0140-6736(12)60312-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sayols-Baixeras S, Subirana I, Lluis-Ganella C, et al. Identification and validation of seven new loci showing differential DNA methylation related to serum lipid profile: An epigenome-wide approach. The regicor study. Human molecular genetics. 2016;25:4556–4565. doi: 10.1093/hmg/ddw285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sayols-Baixeras S, Hernaez A, Subirana I, Lluis-Ganella C, Munoz D, Fito M, Marrugat J, Elosua R. DNA methylation and high-density lipoprotein functionality-brief report: The regicor study (registre gironi del cor) Arteriosclerosis, thrombosis, and vascular biology. 2017;37:567–569. doi: 10.1161/ATVBAHA.116.308831. [DOI] [PubMed] [Google Scholar]
  • 81.Ouimet M, Moore KJ. A big role for small rnas in hdl homeostasis. Journal of lipid research. 2013;54:1161–1167. doi: 10.1194/jlr.R036327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sun X, Feinberg MW. Microrna-management of lipoprotein homeostasis. Circulation research. 2014;115:2–6. doi: 10.1161/CIRCRESAHA.114.304228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Canfran-Duque A, Lin CS, Goedeke L, Suarez Y, Fernandez-Hernando C. Micro-rnas and high-density lipoprotein metabolism. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1076–1084. doi: 10.1161/ATVBAHA.116.307028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Vickers KC, Shoucri BM, Levin MG, Wu H, Pearson DS, Osei-Hwedieh D, Collins FS, Remaley AT, Sethupathy P. Microrna-27b is a regulatory hub in lipid metabolism and is altered in dyslipidemia. Hepatology. 2013;57:533–542. doi: 10.1002/hep.25846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Allen RM, Marquart TJ, Albert CJ, Suchy FJ, Wang DQ, Ananthanarayanan M, Ford DA, Baldan A. Mir-33 controls the expression of biliary transporters, and mediates statin- and diet-induced hepatotoxicity. EMBO molecular medicine. 2012;4:882–895. doi: 10.1002/emmm.201201228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Davalos A, Goedeke L, Smibert P, et al. Mir-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:9232–9237. doi: 10.1073/pnas.1102281108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Rayner KJ, Suarez Y, Davalos A, Parathath S, Fitzgerald ML, Tamehiro N, Fisher EA, Moore KJ, Fernandez-Hernando C. Mir-33 contributes to the regulation of cholesterol homeostasis. Science. 2010;328:1570–1573. doi: 10.1126/science.1189862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Nakao T, Horie T, Baba O, et al. Genetic ablation of microrna-33 attenuates inflammation and abdominal aortic aneurysm formation via several anti-inflammatory pathways. Arteriosclerosis, thrombosis, and vascular biology. 2017;37:2161–2170. doi: 10.1161/ATVBAHA.117.309768. [DOI] [PubMed] [Google Scholar]
  • 89.Ouimet M, Hennessy EJ, van Solingen C, et al. Mirna targeting of oxysterol-binding protein-like 6 regulates cholesterol trafficking and efflux. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:942–951. doi: 10.1161/ATVBAHA.116.307282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Park JG, Xu X, Cho S, Lee AH. Loss of transcription factor crebh accelerates diet-induced atherosclerosis in ldlr-/- mice. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1772–1781. doi: 10.1161/ATVBAHA.116.307790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Liu J, Hernandez-Ono A, Graham MJ, Galton VA, Ginsberg HN. Type 1 deiodinase regulates apoa-i gene expression and apoa-i synthesis independent of thyroid hormone signaling. Arteriosclerosis, thrombosis, and vascular biology. 2016;36:1356–1366. doi: 10.1161/ATVBAHA.116.307330. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES