Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Mar 1.
Published in final edited form as: Arterioscler Thromb Vasc Biol. 2015 Mar;35(3):485–491. doi: 10.1161/ATVBAHA.115.305380

Recent Highlights of ATVB Atherosclerosis

Hong Lu 1, Alan Daugherty 1
PMCID: PMC4511379  NIHMSID: NIHMS658929  PMID: 25717174

Summary

Mechanistic studies over the past decades using in vitro systems, animal models, and human tissues have highlighted the complexity of pathophysiological processes of atherosclerosis. Hypercholesterolemia, as one of the major risk factors for the development and progression of atherosclerosis, is still the focus of many mechanistic studies and the major therapeutic target of atherosclerosis. Although there is a dire need to validate many experimental findings in humans, there is a large number of approaches that have been showing promise for contributing to future therapeutic strategies.

Keywords: atherosclerosis, Editorials, mechanisms, inflammation, leukocytes, endothelial cells, lipoproteins, microRNAs, neuronal guidance cues

Introduction

Atherosclerosis contributes to major mortality and modality of cardiovascular diseases in western countries. Lipid-laden macrophage accumulation in the subendothelial area of the arterial wall is a hallmark of atherosclerosis. These lipid-laden cells promote inflammatory responses in the arterial wall, and lead to multiple fatal pathological consequences such as hemorrhage, rupture, and calcification.1-3 Mechanisms of atherosclerosis relating to disruption of lipoprotein metabolism as well as inflammation have been the major focuses of atherosclerosis research.4,5 Animal models are still the major tools to determine mechanisms and discover potential targets for therapeutic purpose. Although hypercholesterolemic mouse models such as low density lipoprotein (LDL) receptor deficient and apolipoprotein (apo)E deficient mice are used in most experimental studies,6 other animal models such as rabbits7-9 and pigs10,11 have also been used frequently to study atherosclerosis. This article highlights some recent ATVB publications that have either extended the traditional concepts or provided new insights into understanding mechanisms of atherosclerosis.

Lipid and Lipoprotein Metabolisms in Atherosclerosis

Low density lipoproteins (LDL) and high density lipoproteins (HDL)

There is convincing evidence that high plasma LDL cholesterol concentrations contribute to the initiation and progression of atherosclerosis, and lowering this lipoprotein reduces atherosclerosis-related cardiovascular events.5,12-14 In contrast, plasma HDL cholesterol concentrations are negatively associated with atherosclerosis.5,15 Although the major clinical use of statins is to reduce plasma LDL cholesterol concentrations, this class of drug may also increase plasma HDL cholesterol concentrations.16,17

Both LDL and HDL particles are highly heterogenous.18 Recent advance in research of lipoproteins have provided new insights at least from two aspects. On one hand, using techniques to detect and characterize subclasses of LDL particles,19-22 small dense LDL particles have been demonstrated in recent human studies to be positively associated with coronary heart disease.21,23,24 On the other hand, raising plasma HDL cholesterol concentrations had no apparent beneficial effects on atherosclerosis.25 One lesson learned from the failure of the latter study is that HDL function may play a more critical role in preventing and protecting against atherosclerosis.26,27 Studies focusing on exploring mechanisms of HDL dysfunction showed that myeloperoxidase impaired effects of apoA-I on reverse cholesterol transport,28,29 scavenger receptor type BI played a crucial role in HDL regulation of hematopoietic stem/progenitor cell proliferation and differentiation,30 and anti-inflammatory effects of HDL in macrophages were mediated by activating transcription factor 3 (ATF3), a protein involved in toll-like receptor signaling pathway.31,32 Using lipid chromatography-mass spectrometry technique, small, dense HDL3 particles were found to be associated with multiple protective effects in atherosclerosis such as cholesterol efflux, anti-inflammation, and anti-oxidation.33

ATP-binding cassette subfamily A member 1 (ABCA1)

ABCA1 in macrophages facilitates cellular cholesterol efflux. Previous studies determining effects of ABCA1 on atherosclerosis in mouse models have been consistent. Deficiency of ABCA1 alone, or in combination with deficiency of ABCG1, in leukocytes, as demonstrated by bone marrow transplantation, augmented atherosclerosis in mice.34-38 Conversely, overexpression of ABCA1 in macrophages reduced atherosclerosis.39 However, conflicting findings were reported recently in studies using a genetic conditional deletion approach rather than bone marrow transplantation.40 Cell-specific deficiency of ABCA1 was created using Cre-Lox recombination technique.41 ABCA1 floxed mice expressed Cre transgene under the control of either the LysM or albumin promoter to develop myeloid or hepatocyte specific ABCA1 deficiency. In one study, depletion of ABCA1 in hepatocytes augmented atherosclerosis in aortic roots of apoE −/− mice, whereas macrophage deficiency of ABCA1 did not influence atherosclerotic development in LDL receptor −/− mice.42 A subsequent study confirmed that myeloid cell-specific deficiency of ABCA1 had no significant effects on atherosclerosis development, although it resulted in profound cellular cholesterol accumulation in resident peritoneal macrophages.43 In contrast to its deficiency in apoE −/− mice, hepatocyte-specific deficiency of ABCA1 in LDL receptor −/− mice attenuated atherosclerosis in aortic roots, but had no effect on atherosclerotic lesion size of the entire aorta.44 In LDL receptor −/− mice with hepatocyte-specific deficiency of ABCA1 fed an atherogenic diet, both apoB-containing lipoproteins and HDL were reduced. In vitro experiment inferred that HDL concentrations per se were not the primary contributor to plasma efflux capacity.44 Liver X receptors (LXR) regulates both ABCA1 and ABCG1, and contributes to cholesterol efflux. A recent study reported that activation of LXR attenuated atherosclerosis, in the absence of both ABCA1 and ABCG1 in bone marrow-derived cells or myeloid cells, providing evidence that LXR has ABCA1 and ABCG1 independent effects on the development of atherosclerosis.45,46

Proprotein convertase subtilisin/kexin type 9 (PCSK9)

Since its discovery, studies have identified functional mutations of PCSK9 that are either related to hypercholesterolemia (gain-of-function mutations) or hypocholesterolemia (loss-of-function mutations) in humans, which are associated with increased and reduced cardiovascular risks, respectively.47-51 Profiling of carotid atherosclerosis from a human biobank using microarray technique found increased PCSK9 mRNA abundance in fibrous caps of symptomatic carotid atherosclerotic lesions.52 Findings in human studies have led to development of PCSK9 inhibitors to reduce plasma cholesterol concentrations and risks for cardiovascular events.53

In the past several decades, LDL receptor −/− and apoE −/− mice have been the most commonly used hypercholesterolemic mouse models to study mechanisms of atherosclerosis. Therefore, manipulations of genes of interest in mice have also been bred to generate LDL receptor −/− or apoE −/− background. Recently, two research groups have reported that delivery of either a human (D377Y) or a mouse (D374Y) mutation of PCSK9 expressed in adeno-associated virus (AAV) by a single injection led to rapid and profound increases of plasma cholesterol concentrations in several mouse strains, and promoted atherosclerosis that was comparable to lesions developed in LDL receptor −/− or apoE −/− mice.54-57 In addition to providing a new hypercholesterolemic mouse model for atherosclerosis study, these two studies have also enhanced our understanding of PCSK9 mediated LDL receptor regulation, atherosclerosis, and related potential mechanisms.

Inflammation in Atherosclerosis

Inflammation is a critical contributor to the development of atherosclerosis. Accumulation of leukocytes, predominantly macrophages, is a prominent feature of atherosclerosis from initiation to advanced stages of evolution. Lipid accumulation in macrophages induces inflammation, and inflammation promotes and augments atherosclerotic development. Therefore, inflammation and atherosclerotic lesion development form a positive feedback loop. Benefitted from recent enhancements to techniques, mechanisms by which inflammation contributes to atherosclerosis is continuously providing new insights. These include determining the processes of macrophages in invading the arterial wall, trapping, polarization, and triggering a spectrum of inflammatory signals within atherosclerotic lesions.58-60

Leukocyte specific effects

Since the initial experiment using bone marrow transplantation approach to studying atherosclerosis in mice,61 this method has been used frequently by researchers to determine effects of genes of interest in leukocytes in the development of atherosclerosis in mouse models. Recently, using this technique, it has been demonstrated that cathepsin C deficiency62, CD43 (an integral membrane glycoprotein),63 CC chemokine ligand 3,64 or angiotensin-converting enzyme (a critical enzyme to generate angiotensin II)65 in leukocytes led to reductions of atherosclerosis in hypercholesterolemic mice at a single time point. In mice studied at multiple durations of Western diet feeding, deficiency of the common β subunit of the granulocyte macrophage colony-stimulating factor/interleukin-3 receptor in leukocytes only reduced lesion size transiently.66 In contrast to these components that reduced atherosclerosis, acyl-CoA: cholesterol acyltransferase (ACAT) 1 deficiency in leukocytes augmented atherosclerosis.67

Another commonly used approach to determining macrophage-specific effects of genes of interest is to use conditional knockout mice developed with LysM Cre-Lox recombination technique.68 Using this approach with mice expressing LysM Cre recombinase, Basu et al.69 found that genetic depletion of ribosomal protein L13a in macrophages augmented atherosclerosis in mice.

Neutrophils

Neutrophils are the largest population of leukocytes in blood. Although neutrophils play a rapid and critical role in acute inflammatory response, the contribution of this cell type to atherosclerosis has not been drawn much attention until recently.70-72 Hypercholesterolemia induced neutrophilia, which was associated with the initiation of atherosclerosis in apoE −/− mice.70 Cathelicidin is a neutrophil granule protein. Genetic deficiency of cathelicidin led to reductions of atherosclerosis in apoE −/− mice.73 Activation of neutrophils leads to cell death and releasing neutrophil extracellular traps. A recent cross-sectional prospective clinical study reported that plasma neutrophil extracellular traps were positively associated with coronary artery disease.74 Neutrophil gelatinase-associated lipocalin is a neutrophil granular glycoprotein. A human prospectively investigation found that this glycoprotein was positively associated with cardiovascular events including atherosclerotic disease.75 These two recent human studies provide clinical evidence that neutrophils may play an important role in the development of atherosclerosis.76

Chemokines

The presence and activation of multiple chemokines in the development of atherosclerosis is a commonly described feature of atherosclerosis.77 Many chemokine-chemokine receptor interactions such as CCL2-CCR2, CX3C-chemokine ligand 1 (CX3CL1)-CX3C-chemokine receptor 1(CX3CR1), CCL5–CCR5, and CCL19/CCL21-CCR7 contribute to atherosclerotic development.78 A pharmacological inhibitor of CX3CR1 reduced atherosclerosis in both LDL receptor −/− and apoE −/− mice, implicating this chemokine receptor might be a potential target for treatment of atherosclerosis-related inflammation.79 Important roles of chemokine/chemokine receptor interaction were also noticed in human studies. For example, CCL19/CCL21-CCR7 axis were associated with risk for coronary artery disease in a Chinese population.80

Resident Cell Types in Atherosclerosis

Endothelial cell specific effects

Endothelial dysfunction of the arterial wall is an initial step to provoke monocyte adhesion. This is followed by macrophage intruding the subendothelial area to form foam cells. Hypercholesterolemia is a critical contributor to endothelial dysfunction. As determined in hypercholesterolemic mice with several genetic manipulations, endothelial dysfunction after prolonged hypercholesterolemia was the result of both impairment of sensitivity to nitric oxide and reduced nitric oxide synthase cofactor bioavailability.81 In a gene expression analysis of mouse aortic endothelium in response to hypercholesterolemia, inflammation, or aging, 14 genes were significantly different between these different atherogenic stimulations and normal condition.82 Angiopoietin-like protein 2 (Angptl2), a proinflammatory protein, is abundant in endothelial cells. Findings using whole body genetic deficiency of Angptl2, bone marrow transplantation, and endothelial specific overexpression of Angptl2, demonstrated that endothelial cell-derived Angptl2 contributed to atherosclerotic lesion formation, attributed to endothelial dysfunction.83 Krüppel-like factor 2 (KLF2), a member in zinc finger transcription factors, plays an important role in regulating endothelial function.84 LDL inhibited endothelial KLF2 through regulation of DNA and histone methylation. This was proposed as one mechanism by which LDL induced endothelial dysfunction.85 Type 1 insulin-like growth factor-1 receptor (IGF1R) is a negative regulator of nitric oxide bioavailability in endothelium.86 Incomplete deletion of IGF1R (IGF1R+/−) led to accelerated endothelium regeneration in mice, implicating potentially enhanced endothelium repair.87 Bone morphogenic protein (BMP)4 interacts with its receptors BMPR1 or BMPR2 for its bioactive actions. Stimulations such as hypercholesterolemia, disturbed blood flow or inflammatory responses, suppressed BMPRII in endothelium. Partial genetic deletion of BMPRII (BMPRII +/−) led to augmentation of atherosclerosis in apoE −/− mice.88 In addition to evidence from experimental studies, endothelial dysfunction was associated with macrophage infiltration and neovascularization in patients with early stage coronary atherosclerosis.89

Smooth muscle cell specific effects

Vascular smooth muscle cells (SMCs) are the predominant cell type in the arterial wall. This cell type also plays a critical role in atherosclerosis development through multiple mechanisms. Recent studies explored mechanisms by which survival or apoptosis of this cell type influenced atherosclerosis. Wnt/β-catenin responsive gene, Wnt1-inducible secreted protein-1 (WISP-1), was associated with suppression of mouse aortic SMC apoptosis induced by oxidative stress, and WISP-1 was lower in unstable, compared to stable, human atherosclerotic lesions.90 Inhibition of Akt1 (the serine-threonine kinase) interaction with forkhead class O transcription factor 3a (FoxO3a) contributed to survival of vascular SMCs in vitro.91 Transgenic mice with SMC-specific overexpression of Akt1 inhibited SMC apoptosis in atherosclerotic lesions without changing lesion size.92 Small proline-rich repeat protein 3 (SPRR3) promoted cell survival and was more abundant in SMCs of human atherosclerotic lesions than normal arterial tissues.93 Genetic deficiency of SPRR3 augmented atherosclerosis in apoE −/− mice, which were attributed to regulation of Akt signaling in SMCs.94 Therefore, roles of SMC survival and apoptosis in the development of atherosclerosis through Akt signaling pathway have not been consistent in the literature.

New Emerging Mechanisms

MicroRNAs

Since the discovery of microRNAs (miRs) in 1993,95 this class of small non-coding RNAs has been recognized as comprehensive regulators to many physiological and pathophysiological conditions. Their contributions to atherosclerosis have also been studied in the past few years.96 Since its initial study in regulation of cholesterol homeostasis and subsequently in atherosclerosis development,97-109 miR-33 has become a most attractive target in miRs for drug development to treat dyslipidemia and atherosclerosis. Although initial studies showed that inhibition of miR-33 reduced atherosclerosis in LDL receptor −/− or apoE −/− mice,99,108 recent studies have reported that inhibition of this miR did not attenuate the progression of atherosclerosis in LDL receptor −/− mice,107,109 or its deficiency in bone marrow-derived cells did not reduce atherosclerotic lesions.108 In addition to miR-33, many other miRs contribute to the development of atherosclerosis through different mechanisms.110,111 For example, miR-24 regulates macrophage behavior,112 miR-126-5p promotes endothelial proliferation, 113,114 miR-145 controls smooth muscle cell differentiation,115 miR-155 contributes to inflammation,116-118 and miR-302a modulates cholesterol homeostasis.119

Neuronal Guidance Cues

Neuronal guidance cues regulate neuronal migration and vascular patterning and development. Recent studies have identified several neuronal guidance cues as a potential mechanism of atherosclerosis, which added another exciting layer to the complexity of atherosclerosis mechanisms.120,121 Netrin-1 is a neuronal guidance cue that mediates chemorepulsion and chemoattraction of axons. van Gils et al. reported that netrin-1 and its chemorepulsive receptor UNC5b were present in macrophages of human atherosclerotic lesions.120 This molecule inhibited macrophage migration and was a chemoattractant for SMCs. Netrin-1 deficiency in hematopoietic stem cells isolated from fetal liver reduced atherosclerosis in mice through multiple mechanisms including regulating inflammation.120 A subsequent study found that netrin-1 and UNC5b were increased in macrophages in vitro by incubation with oxidized LDL or inducers of oxidative stress.122 These effects were inhibited by hypoxia-inducible transcription factor-1α, implicating that hypoxia is a potential contributor to activation of netrin-1 - UNC5b axis.122

Semaphorin 3E (Sema3E), another neuronal guidance molecule, has also been implicated in atherosclerosis development.123 This molecule was abundant in macrophages of mouse atherosclerotic lesions, and was increased by stimulation with oxidized LDL or hypoxia.123 In addition to important roles of netril-1 and Sema3E on macrophage function, these two guidance cues and another molecule, ephrinB2, were expressed in arterial endothelial cells. Netrin-1 and Sema3E inhibited chemokine-mediated monocyte migration and leukocyte adhesion, whereas ephrinB2 was a potent monocyte chemoattractant.124

ACKNOWLEDGMENTS

None.

SOURCES OF FUNDING

The authors’ research work were supported by a grant (HL107319 to Alan Daugherty) from the National Institutes of Health of the United States of America. The content in this manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Footnotes

DISCLOSURES

None.

References

  • 1.Arbab-Zadeh A, Fuster V. The Myth of the "Vulnerable Plaque": Transitioning From a Focus on Individual Lesions to Atherosclerotic Disease Burden for Coronary Artery Disease Risk Assessment. J Am Coll Cardiol. 2015 doi: 10.1016/j.jacc.2014.11.041. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Otsuka F, Sakakura K, Yahagi K, Joner M, Virmani R. Has our understanding of calcification in human coronary atherosclerosis progressed? Arterioscler Thromb Vasc Biol. 2014;34:724–736. doi: 10.1161/ATVBAHA.113.302642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Virmani R, Joner M, Sakakura K. Recent highlights of ATVB: calcification. Arterioscler Thromb Vasc Biol. 2014;34:1329–1332. doi: 10.1161/ATVBAHA.114.304000. [DOI] [PubMed] [Google Scholar]
  • 4.Lusis AJ. Atherosclerosis. Nature. 2000;407:233–241. doi: 10.1038/35025203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rader DJ, Daugherty A. Translating molecular discoveries into new therapies for atherosclerosis. Nature. 2008;451:904–913. doi: 10.1038/nature06796. [DOI] [PubMed] [Google Scholar]
  • 6.Daugherty A, Rateri DL. Development of experimental designs for atherosclerosis studies in mice. Methods. 2005;36:129–138. doi: 10.1016/j.ymeth.2004.11.008. [DOI] [PubMed] [Google Scholar]
  • 7.Wang Y, Niimi M, Nishijima K, et al. Human apolipoprotein A-II protects against diet-induced atherosclerosis in transgenic rabbits. Arterioscler Thromb Vasc Biol. 2013;33:224–231. doi: 10.1161/ATVBAHA.112.300445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shiomi M, Ishida T, Kobayashi T, Nitta N, Sonoda A, Yamada S, Koike T, Kuniyoshi N, Murata K, Hirata K, Ito T, Libby P. Vasospasm of atherosclerotic coronary arteries precipitates acute ischemic myocardial damage in myocardial infarction-prone strain of the Watanabe heritable hyperlipidemic rabbits. Arterioscler Thromb Vasc Biol. 2013;33:2518–2523. doi: 10.1161/ATVBAHA.113.301303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Zhang J, Yang S, Ji X, Zhou Q, Xing D. Characterization of lipid-rich aortic plaques by intravascular photoacoustic tomography: ex vivo and in vivo validation in a rabbit atherosclerosis model with histologic correlation. J Am Coll Cardiol. 2014;64:385–390. doi: 10.1016/j.jacc.2014.04.053. [DOI] [PubMed] [Google Scholar]
  • 10.Koskinas KC, Sukhova GK, Baker AB, Papafaklis MI, Chatzizisis YS, Coskun AU, Quillard T, Jonas M, Maynard C, Antoniadis AP, Shi GP, Libby P, Edelman ER, Feldman CL, Stone PH. Thin-capped atheromata with reduced collagen content in pigs develop in coronary arterial regions exposed to persistently low endothelial shear stress. Arterioscler Thromb Vasc Biol. 2013;33:1494–1504. doi: 10.1161/ATVBAHA.112.300827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Deuse T, Hua X, Wang D, et al. Dichloroacetate prevents restenosis in preclinical animal models of vessel injury. Nature. 2014;509:641–644. doi: 10.1038/nature13232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Herder M, Arntzen KA, Johnsen SH, Eggen AE, Mathiesen EB. Long-term use of lipid-lowering drugs slows progression of carotid atherosclerosis: the Tromso study 1994 to 2008. Arterioscler Thromb Vasc Biol. 2013;33:858–862. doi: 10.1161/ATVBAHA.112.300767. [DOI] [PubMed] [Google Scholar]
  • 13.Bedi US, Singh M, Singh PP, Bhuriya R, Bahekar A, Molnar J, Khosla S, Arora R. Effects of statins on progression of carotid atherosclerosis as measured by carotid intimal--medial thickness: a meta-analysis of randomized controlled trials. J Cardiovasc Pharmacol Ther. 2010;15:268–273. doi: 10.1177/1074248410369110. [DOI] [PubMed] [Google Scholar]
  • 14.Puri R, Nissen SE, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nicholls SJ. Antiatherosclerotic effects of long-term maximally intensive statin therapy after acute coronary syndrome: insights from Study of Coronary Atheroma by Intravascular Ultrasound: Effect of Rosuvastatin Versus Atorvastatin. Arterioscler Thromb Vasc Biol. 2014;34:2465–2472. doi: 10.1161/ATVBAHA.114.303932. [DOI] [PubMed] [Google Scholar]
  • 15.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]
  • 16.Schaefer JR, Schweer H, Ikewaki K, Stracke H, Seyberth HJ, Kaffarnik H, Maisch B, Steinmetz A. Metabolic basis of high density lipoproteins and apolipoprotein A-I increase by HMG-CoA reductase inhibition in healthy subjects and a patient with coronary artery disease. Atherosclerosis. 1999;144:177–184. doi: 10.1016/s0021-9150(99)00053-2. [DOI] [PubMed] [Google Scholar]
  • 17.Asztalos BF, Horvath KV, McNamara JR, Roheim PS, Rubinstein JJ, Schaefer EJ. Comparing the effects of five different statins on the HDL subpopulation profiles of coronary heart disease patients. Atherosclerosis. 2002;164:361–369. doi: 10.1016/s0021-9150(02)00149-1. [DOI] [PubMed] [Google Scholar]
  • 18.Berneis KK, Krauss RM. Metabolic origins and clinical significance of LDL heterogeneity. J Lipid Res. 2002;43:1363–1379. doi: 10.1194/jlr.r200004-jlr200. [DOI] [PubMed] [Google Scholar]
  • 19.Hammond MG, Fisher WR. The characterization of a discrete series of low density lipoproteins in the disease, hyper-pre-beta-lipoproteinemia. Implications relating to the structure of plasma lipoproteins. J Biol Chem. 1971;246:5454–5465. [PubMed] [Google Scholar]
  • 20.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. Clin Chem. 2008;54:1307–1316. doi: 10.1373/clinchem.2007.100586. [DOI] [PubMed] [Google Scholar]
  • 21.Tsai MY, Steffen BT, Guan W, McClelland RL, Warnick R, McConnell J, Hoefner DM, Remaley AT. New automated assay of small dense low-density lipoprotein cholesterol identifies risk of coronary heart disease: the Multi-ethnic Study of Atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34:196–201. doi: 10.1161/ATVBAHA.113.302401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ito Y, Fujimura M, Ohta M, Hirano T. Development of a homogeneous assay for measurement of small dense LDL cholesterol. Clin Chem. 2011;57:57–65. doi: 10.1373/clinchem.2010.149559. [DOI] [PubMed] [Google Scholar]
  • 23.Hoogeveen RC, Gaubatz JW, Sun W, Dodge RC, Crosby JR, Jiang J, Couper D, Virani SS, Kathiresan S, Boerwinkle E, Ballantyne CM. Small dense low-density lipoprotein-cholesterol concentrations predict risk for coronary heart disease: the Atherosclerosis Risk In Communities (ARIC) study. Arterioscler Thromb Vasc Biol. 2014;34:1069–1077. doi: 10.1161/ATVBAHA.114.303284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Parish S, Offer A, Clarke R, Hopewell JC, Hill MR, Otvos JD, Armitage J, Collins R. Lipids and lipoproteins and risk of different vascular events in the MRC/BHF Heart Protection Study. Circulation. 2012;125:2469–2478. doi: 10.1161/CIRCULATIONAHA.111.073684. [DOI] [PubMed] [Google Scholar]
  • 25.Barter PJ, Caulfield M, Eriksson M, et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007;357:2109–2122. doi: 10.1056/NEJMoa0706628. [DOI] [PubMed] [Google Scholar]
  • 26.Toth PP, Barter PJ, Rosenson RS, Boden WE, Chapman MJ, Cuchel M, D'Agostino RB, Sr, Davidson MH, Davidson WS, Heinecke JW, Karas RH, Kontush A, Krauss RM, Miller M, Rader DJ. High-density lipoproteins: a consensus statement from the National Lipid Association. J Clin Lipidol. 2013;7:484–525. doi: 10.1016/j.jacl.2013.08.001. [DOI] [PubMed] [Google Scholar]
  • 27.Degoma EM, Rader DJ. Novel HDL-directed pharmacotherapeutic strategies. Nat Rev Cardiol. 2011;8:266–277. doi: 10.1038/nrcardio.2010.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hewing B, Parathath S, Barrett T, et al. Effects of native and myeloperoxidase-modified apolipoprotein a-I on reverse cholesterol transport and atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2014;34:779–789. doi: 10.1161/ATVBAHA.113.303044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Getz GS, Reardon CA. Myeloperoxidase-mediated dysfunctional high-density lipoprotein. Arterioscler Thromb Vasc Biol. 2014;34:695–696. doi: 10.1161/ATVBAHA.114.303282. [DOI] [PubMed] [Google Scholar]
  • 30.Gao M, Zhao D, Schouteden S, Sorci-Thomas MG, Van Veldhoven PP, Eggermont K, Liu G, Verfaillie CM, Feng Y. Regulation of high-density lipoprotein on hematopoietic stem/progenitor cells in atherosclerosis requires scavenger receptor type BI expression. Arterioscler Thromb Vasc Biol. 2014;34:1900–1909. doi: 10.1161/ATVBAHA.114.304006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.De Nardo D, Labzin LI, Kono H, et al. High-density lipoprotein mediates anti-inflammatory reprogramming of macrophages via the transcriptional regulator ATF3. Nat Immunol. 2014;15:152–160. doi: 10.1038/ni.2784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhang H, Reilly MP. Anti-inflammatory effects of high-density lipoprotein through activating transcription factor 3: benefit beyond cholesterol transport-dependent processes. Arterioscler Thromb Vasc Biol. 2014;34:e11–12. doi: 10.1161/ATVBAHA.114.303553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Camont L, Lhomme M, Rached F, Le Goff W, Negre-Salvayre A, Salvayre R, Calzada C, Lagarde M, Chapman MJ, Kontush A. Small, dense high-density lipoprotein-3 particles are enriched in negatively charged phospholipids: relevance to cellular cholesterol efflux, antioxidative, antithrombotic, anti-inflammatory, and antiapoptotic functionalities. Arterioscler Thromb Vasc Biol. 2013;33:2715–2723. doi: 10.1161/ATVBAHA.113.301468. [DOI] [PubMed] [Google Scholar]
  • 34.Aiello RJ, Brees D, Bourassa PA, Royer L, Lindsey S, Coskran T, Haghpassand M, Francone OL. Increased atherosclerosis in hyperlipidemic mice with inactivation of ABCA1 in macrophages. Arterioscler Thromb Vasc Biol. 2002;22:630–637. doi: 10.1161/01.atv.0000014804.35824.da. [DOI] [PubMed] [Google Scholar]
  • 35.van Eck M, Bos IS, Kaminski WE, Orso E, Rothe G, Twisk J, Bottcher A, Van Amersfoort ES, Christiansen-Weber TA, Fung-Leung WP, Van Berkel TJ, Schmitz G. Leukocyte ABCA1 controls susceptibility to atherosclerosis and macrophage recruitment into tissues. Proc Natl Acad Sci USA. 2002;99:6298–6303. doi: 10.1073/pnas.092327399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yvan-Charvet L, Ranalletta M, Wang N, Han S, Terasaka N, Li R, Welch C, Tall AR. Combined deficiency of ABCA1 and ABCG1 promotes foam cell accumulation and accelerates atherosclerosis in mice. J Clin Invest. 2007;117:3900–3908. doi: 10.1172/JCI33372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Out R, Hoekstra M, Habets K, Meurs I, de Waard V, Hildebrand RB, Wang Y, Chimini G, Kuiper J, Van Berkel TJ, Van Eck M. Combined deletion of macrophage ABCA1 and ABCG1 leads to massive lipid accumulation in tissue macrophages and distinct atherosclerosis at relatively low plasma cholesterol levels. Arterioscler Thromb Vasc Biol. 2008;28:258–264. doi: 10.1161/ATVBAHA.107.156935. [DOI] [PubMed] [Google Scholar]
  • 38.Zhao Y, Pennings M, Hildebrand RB, Ye D, Calpe-Berdiel L, Out R, Kjerrulf M, Hurt-Camejo E, Groen AK, Hoekstra M, Jessup W, Chimini G, Van Berkel TJ, Van Eck M. Enhanced foam cell formation, atherosclerotic lesion development, and inflammation by combined deletion of ABCA1 and SR-BI in Bone marrow-derived cells in LDL receptor knockout mice on western-type diet. Circ Res. 2010;107:e20–31. doi: 10.1161/CIRCRESAHA.110.226282. [DOI] [PubMed] [Google Scholar]
  • 39.van Eck M, Singaraja RR, Ye D, Hildebrand RB, James ER, Hayden MR, van Berkel TJ. Macrophage ATP-Binding Cassette Transporter 1 Overexpression Inhibits Atherosclerotic Lesion Progression in Low-Density Lipoprotein Receptor Knockout Mice. Arterioscler Thromb Vasc Biol. 2006;26:929–934. doi: 10.1161/01.ATV.0000208364.22732.16. [DOI] [PubMed] [Google Scholar]
  • 40.Van Eck M, Van Berkel TJ. ATP-binding cassette transporter A1 in lipoprotein metabolism and atherosclerosis: a new piece of the complex puzzle. Arterioscler Thromb Vasc Biol. 2013;33:2281–2283. doi: 10.1161/ATVBAHA.113.301719. [DOI] [PubMed] [Google Scholar]
  • 41.Timmins JM, Lee JY, Boudyguina E, Kluckman KD, Brunham LR, Mulya A, Gebre AK, Coutinho JM, Colvin PL, Smith TL, Hayden MR, Maeda N, Parks JS. Targeted inactivation of hepatic Abca1 causes profound hypoalphalipoproteinemia and kidney hypercatabolism of apoA-I. J Clin Invest. 2005;115:1333–1342. doi: 10.1172/JCI23915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Brunham LR, Singaraja RR, Duong M, Timmins JM, Fievet C, Bissada N, Kang MH, Samra A, Fruchart JC, McManus B, Staels B, Parks JS, Hayden MR. Tissue-specific roles of ABCA1 influence susceptibility to atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29:548–554. doi: 10.1161/ATVBAHA.108.182303. [DOI] [PubMed] [Google Scholar]
  • 43.Bi X, Zhu X, Gao C, Shewale S, Cao Q, Liu M, Boudyguina E, Gebre AK, Wilson MD, Brown AL, Parks JS. Myeloid cell-specific ATP-binding cassette transporter A1 deletion has minimal impact on atherogenesis in atherogenic diet-fed low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol. 2014;34:1888–1899. doi: 10.1161/ATVBAHA.114.303791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bi X, Zhu X, Duong M, Boudyguina E, Wilson MD, Gebre AK, Parks JS. Liver ABCA1 deletion in LDLrKO mice does not impair macrophage reverse cholesterol transport of exacerbate atherogenesis. Arterioscler Thromb Vasc Biol. 2013;33:2288–2296. doi: 10.1161/ATVBAHA.112.301110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kappus MS, Murphy AJ, Abramowicz S, Ntonga V, Welch CL, Tall AR, Westerterp M. Activation of liver X receptor decreases atherosclerosis in Ldlr(−)/(−) mice in the absence of ATP-binding cassette transporters A1 and G1 in myeloid cells. Arterioscler Thromb Vasc Biol. 2014;34:279–284. doi: 10.1161/ATVBAHA.113.302781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ignatova ID, Schulman IG. Liver X receptors and atherosclerosis: it is not all cholesterol. Arterioscler Thromb Vasc Biol. 2014;34:242–243. doi: 10.1161/ATVBAHA.113.302987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Abifadel M, Varret M, Rabes JP, et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet. 2003;34:154–156. doi: 10.1038/ng1161. [DOI] [PubMed] [Google Scholar]
  • 48.Maxwell KN, Breslow JL. Proprotein convertase subtilisin kexin 9: the third locus implicated in autosomal dominant hypercholesterolemia. Curr Opin Lipidol. 2005;16:167–172. doi: 10.1097/01.mol.0000162321.31925.a3. [DOI] [PubMed] [Google Scholar]
  • 49.Allard D, Amsellem S, Abifadel M, Trillard M, Devillers M, Luc G, Krempf M, Reznik Y, Girardet JP, Fredenrich A, Junien C, Varret M, Boileau C, Benlian P, Rabes JP. Novel mutations of the PCSK9 gene cause variable phenotype of autosomal dominant hypercholesterolemia. Hum Mutat. 2005;26:497. doi: 10.1002/humu.9383. [DOI] [PubMed] [Google Scholar]
  • 50.Hallman DM, Srinivasan SR, Chen W, Boerwinkle E, Berenson GS. Relation of PCSK9 mutations to serum low-density lipoprotein cholesterol in childhood and adulthood (from The Bogalusa Heart Study) Am J Cardiol. 2007;100:69–72. doi: 10.1016/j.amjcard.2007.02.057. [DOI] [PubMed] [Google Scholar]
  • 51.Saavedra YG, Dufour R, Davignon J, Baass A. PCSK9 R46L, lower LDL, and cardiovascular disease risk in familial hypercholesterolemia: a cross-sectional cohort study. Arterioscler Thromb Vasc Biol. 2014;34:2700–2705. doi: 10.1161/ATVBAHA.114.304406. [DOI] [PubMed] [Google Scholar]
  • 52.Perisic L, Hedin E, Razuvaev A, Lengquist M, Osterholm C, Folkersen L, Gillgren P, Paulsson-Berne G, Ponten F, Odeberg J, Hedin U. Profiling of atherosclerotic lesions by gene and tissue microarrays reveals PCSK6 as a novel protease in unstable carotid atherosclerosis. Arterioscler Thromb Vasc Biol. 2013;33:2432–2443. doi: 10.1161/ATVBAHA.113.301743. [DOI] [PubMed] [Google Scholar]
  • 53.Verma DR, Brinton EA. Management of hypercholesterolemia for prevention of atherosclerotic cardiovascular disease: focus on the potential role of recombinant anti-PCSK9 monoclonal antibodies. Rev Cardiovasc Med. 2014;15:86–101. doi: 10.3909/ricm0741. [DOI] [PubMed] [Google Scholar]
  • 54.Roche-Molina M, Sanz-Rosa D, Cruz FM, Garcia-Prieto J, Lopez S, Abia R, Muriana FJ, Fuster V, Ibanez B, Bernal JA. Induction of Sustained Hypercholesterolemia by Single Adeno-Associated Virus-Mediated Gene Transfer of Mutant hPCSK9. Arterioscler Thromb Vasc Biol. 2015;35:50–59. doi: 10.1161/ATVBAHA.114.303617. [DOI] [PubMed] [Google Scholar]
  • 55.Bjorklund MM, Hollensen AK, Hagensen MK, Dagnaes-Hansen F, Christoffersen C, Mikkelsen JG, Bentzon JF. Induction of atherosclerosis in mice and hamsters without germline genetic engineering. Circ Res. 2014;114:1684–1689. doi: 10.1161/CIRCRESAHA.114.302937. [DOI] [PubMed] [Google Scholar]
  • 56.Daugherty A, Tabas I, Rader DJ. Accelerating the pace of atherosclerosis research. Arterioscler Thromb Vasc Biol. 2015;35:11–12. doi: 10.1161/ATVBAHA.114.304833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lu H, Daugherty A. Atherosclerosis: cell biology and lipoproteins. Curr Opin Lipidol. 2015 doi: 10.1097/MOL.0000000000000166. In press. [DOI] [PubMed] [Google Scholar]
  • 58.Hilgendorf I, Swirski FK, Robbins CS. Monocyte fate in atherosclerosis. Arterioscler Thromb Vasc Biol. 2015;35:272–279. doi: 10.1161/ATVBAHA.114.303565. [DOI] [PubMed] [Google Scholar]
  • 59.Rosenfeld ME. Macrophage proliferation in atherosclerosis: an historical perspective. Arterioscler Thromb Vasc Biol. 2014;34:e21–22. doi: 10.1161/ATVBAHA.114.303379. [DOI] [PubMed] [Google Scholar]
  • 60.Leitinger N, Schulman IG. Phenotypic polarization of macrophages in atherosclerosis. Arterioscler Thromb Vasc Biol. 2013;33:1120–1126. doi: 10.1161/ATVBAHA.112.300173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Linton MF, Atkinson JB, Fazio S. Prevention of atherosclerosis in apolipoprotein E-deficient mice by bone marrow transplantation. Science. 1995;267:1034–1037. doi: 10.1126/science.7863332. [DOI] [PubMed] [Google Scholar]
  • 62.Herias V, Biessen EA, Beckers C, Delsing D, Liao M, Daemen MJ, Pham CC, Heeneman S. Leukocyte cathepsin C deficiency attenuates atherosclerotic lesion progression by selective tuning of innate and adaptive immune responses. Arterioscler Thromb Vasc Biol. 2015;35:79–86. doi: 10.1161/ATVBAHA.114.304292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Meiler S, Baumer Y, McCurdy S, Lee B, Kitamoto S, Boisvert WA. Cluster of differentiation 43 deficiency in leukocytes leads to reduced atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;35:323–331. doi: 10.1161/ATVBAHA.114.304513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.De Jager SC, Bot I, Kraaijeveld AO, Korporaal SJ, Bot M, van Santbrink PJ, van Berkel TJ, Kuiper J, Biessen EA. Leukocyte-specific CCL3 deficiency inhibits atherosclerotic lesion development by affecting neutrophil accumulation. Arterioscler Thromb Vasc Biol. 2013;33:e75–83. doi: 10.1161/ATVBAHA.112.300857. [DOI] [PubMed] [Google Scholar]
  • 65.Chen XC, Lu H, Zhao M, Tashiro K, Cassis LA, Daugherty A. Angiotensin-converting enzyme promotes atherosclerosis through an angiotensin I to angiotensin II pathway involving leukocytes. Arterioscler Thromb Vasc Biol. 2013;33:2075–2080. doi: 10.1161/ATVBAHA.113.301777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang M, Subramanian M, Abramowicz S, Murphy AJ, Gonen A, Witztum J, Welch C, Tabas I, Westerterp M, Tall AR. Interleukin-3/granulocyte macrophage colony-stimulating factor receptor promotes stem cell expansion, monocytosis, and atheroma macrophage burden in mice with hematopoietic ApoE deficiency. Arterioscler Thromb Vasc Biol. 2014;34:976–984. doi: 10.1161/ATVBAHA.113.303097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Huang LH, Gui J, Artinger E, Craig R, Berwin BL, Ernst PA, Chang CC, Chang TY. Acat1 gene ablation in mice increases hematopoietic progenitor cell proliferation in bone marrow and causes leukocytosis. Arterioscler Thromb Vasc Biol. 2013;33:2081–2087. doi: 10.1161/ATVBAHA.112.301080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Nagy A. Cre recombinase: the universal reagent for genome tailoring. Genesis. 2000;26:99–109. [PubMed] [Google Scholar]
  • 69.Basu A, Poddar D, Robinet P, Smith JD, Febbraio M, Baldwin WM, 3rd, Mazumder B. Ribosomal protein L13a deficiency in macrophages promotes atherosclerosis by limiting translation control-dependent retardation of inflammation. Arterioscler Thromb Vasc Biol. 2014;34:533–542. doi: 10.1161/ATVBAHA.113.302573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Drechsler M, Megens RT, van Zandvoort M, Weber C, Soehnlein O. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation. 2010;122:1837–1845. doi: 10.1161/CIRCULATIONAHA.110.961714. [DOI] [PubMed] [Google Scholar]
  • 71.Soehnlein O. Multiple roles for neutrophils in atherosclerosis. Circ Res. 2012;110:875–888. doi: 10.1161/CIRCRESAHA.111.257535. [DOI] [PubMed] [Google Scholar]
  • 72.Doring Y, Drechsler M, Soehnlein O, Weber C. Neutrophils in Atherosclerosis: From Mice to Man. Arterioscler Thromb Vasc Biol. 2015;35:288–295. doi: 10.1161/ATVBAHA.114.303564. [DOI] [PubMed] [Google Scholar]
  • 73.Doring Y, Drechsler M, Wantha S, Kemmerich K, Lievens D, Vijayan S, Gallo RL, Weber C, Soehnlein O. Lack of neutrophil-derived CRAMP reduces atherosclerosis in mice. Circ Res. 2012;110:1052–1056. doi: 10.1161/CIRCRESAHA.112.265868. [DOI] [PubMed] [Google Scholar]
  • 74.Borissoff JI, Joosen IA, Versteylen MO, Brill A, Fuchs TA, Savchenko AS, Gallant M, Martinod K, Ten Cate H, Hofstra L, Crijns HJ, Wagner DD, Kietselaer BL. Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arterioscler Thromb Vasc Biol. 2013;33:2032–2040. doi: 10.1161/ATVBAHA.113.301627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lindberg S, Jensen JS, Mogelvang R, Pedersen SH, Galatius S, Flyvbjerg A, Magnusson NE. Plasma neutrophil gelatinase-associated lipocalinin in the general population: association with inflammation and prognosis. Arterioscler Thromb Vasc Biol. 2014;34:2135–2142. doi: 10.1161/ATVBAHA.114.303950. [DOI] [PubMed] [Google Scholar]
  • 76.Doring Y, Weber C, Soehnlein O. Footprints of neutrophil extracellular traps as predictors of cardiovascular risk. Arterioscler Thromb Vasc Biol. 2013;33:1735–1736. doi: 10.1161/ATVBAHA.113.301889. [DOI] [PubMed] [Google Scholar]
  • 77.Zernecke A, Weber C. Chemokines in atherosclerosis: proceedings resumed. Arterioscler Thromb Vasc Biol. 2014;34:742–750. doi: 10.1161/ATVBAHA.113.301655. [DOI] [PubMed] [Google Scholar]
  • 78.Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. 2013;13:709–721. doi: 10.1038/nri3520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Poupel L, Boissonnas A, Hermand P, Dorgham K, Guyon E, Auvynet C, Charles FS, Lesnik P, Deterre P, Combadiere C. Pharmacological inhibition of the chemokine receptor, CX3CR1, reduces atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2013;33:2297–2305. doi: 10.1161/ATVBAHA.112.300930. [DOI] [PubMed] [Google Scholar]
  • 80.Cai W, Tao J, Zhang X, Tian X, Liu T, Feng X, Bai J, Yan C, Han Y. Contribution of homeostatic chemokines CCL19 and CCL21 and their receptor CCR7 to coronary artery disease. Arterioscler Thromb Vasc Biol. 2014;34:1933–1941. doi: 10.1161/ATVBAHA.113.303081. [DOI] [PubMed] [Google Scholar]
  • 81.Miller JD, Chu Y, Castaneda LE, Serrano KM, Brooks RM, Heistad DD. Vascular function during prolonged progression and regression of atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2013;33:459–465. doi: 10.1161/ATVBAHA.112.252700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Erbilgin A, Siemers N, Kayne P, Yang WP, Berliner J, Lusis AJ. Gene expression analyses of mouse aortic endothelium in response to atherogenic stimuli. Arterioscler Thromb Vasc Biol. 2013;33:2509–2517. doi: 10.1161/ATVBAHA.113.301989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Horio E, Kadomatsu T, Miyata K, Arai Y, Hosokawa K, Doi Y, Ninomiya T, Horiguchi H, Endo M, Tabata M, Tazume H, Tian Z, Takahashi O, Terada K, Takeya M, et al. Role of endothelial cell-derived angptl2 in vascular inflammation leading to endothelial dysfunction and atherosclerosis progression. Arterioscler Thromb Vasc Biol. 2014;34:790–800. doi: 10.1161/ATVBAHA.113.303116. [DOI] [PubMed] [Google Scholar]
  • 84.Jain MK, Sangwung P, Hamik A. Regulation of an inflammatory disease: Kruppel-like factors and atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34:499–508. doi: 10.1161/ATVBAHA.113.301925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kumar A, Kumar S, Vikram A, Hoffman TA, Naqvi A, Lewarchik CM, Kim YR, Irani K. Histone and DNA methylation-mediated epigenetic downregulation of endothelial Kruppel-like factor 2 by low-density lipoprotein cholesterol. Arterioscler Thromb Vasc Biol. 2013;33:1936–1942. doi: 10.1161/ATVBAHA.113.301765. [DOI] [PubMed] [Google Scholar]
  • 86.Abbas A, Imrie H, Viswambharan H, et al. The insulin-like growth factor-1 receptor is a negative regulator of nitric oxide bioavailability and insulin sensitivity in the endothelium. Diabetes. 2011;60:2169–2178. doi: 10.2337/db11-0197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yuldasheva NY, Rashid ST, Haywood NJ, et al. Haploinsufficiency of the insulin-like growth factor-1 receptor enhances endothelial repair and favorably modifies angiogenic progenitor cell phenotype. Arterioscler Thromb Vasc Biol. 2014;34:2051–2058. doi: 10.1161/ATVBAHA.114.304121. [DOI] [PubMed] [Google Scholar]
  • 88.Kim CW, Song H, Kumar S, Nam D, Kwon HS, Chang KH, Son DJ, Kang DW, Brodie SA, Weiss D, Vega JD, Alberts-Grill N, Griendling K, Taylor WR, Jo H. Anti-inflammatory and antiatherogenic role of BMP receptor II in endothelial cells. Arterioscler Thromb Vasc Biol. 2013;33:1350–1359. doi: 10.1161/ATVBAHA.112.300287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Choi BJ, Matsuo Y, Aoki T, Kwon TG, Prasad A, Gulati R, Lennon RJ, Lerman LO, Lerman A. Coronary endothelial dysfunction is associated with inflammation and vasa vasorum proliferation in patients with early atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34:2473–2477. doi: 10.1161/ATVBAHA.114.304445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Mill C, Monk BA, Williams H, Simmonds SJ, Jeremy JY, Johnson JL, George SJ. Wnt5a-induced Wnt1-inducible secreted protein-1 suppresses vascular smooth muscle cell apoptosis induced by oxidative stress. Arterioscler Thromb Vasc Biol. 2014;34:2449–2456. doi: 10.1161/ATVBAHA.114.303922. [DOI] [PubMed] [Google Scholar]
  • 91.Allard D, Figg N, Bennett MR, Littlewood TD. Akt regulates the survival of vascular smooth muscle cells via inhibition of FoxO3a and GSK3. J Biol Chem. 2008;283:19739–19747. doi: 10.1074/jbc.M710098200. [DOI] [PubMed] [Google Scholar]
  • 92.Tucka J, Yu H, Gray K, Figg N, Maguire J, Lam B, Bennett M, Littlewood T. Akt1 regulates vascular smooth muscle cell apoptosis through FoxO3a and Apaf1 and protects against arterial remodeling and atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34:2421–2428. doi: 10.1161/ATVBAHA.114.304284. [DOI] [PubMed] [Google Scholar]
  • 93.Pyle AL, Atkinson JB, Pozzi A, Reese J, Eckes B, Davidson JM, Crimmins DL, Young PP. Regulation of the atheroma-enriched protein, SPRR3, in vascular smooth muscle cells through cyclic strain is dependent on integrin alpha1beta1/collagen interaction. Am J Pathol. 2008;173:1577–1588. doi: 10.2353/ajpath.2008.080042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Segedy AK, Pyle AL, Li B, Zhang Y, Babaev VR, Jat P, Fazio S, Atkinson JB, Linton MF, Young PP. Identification of small proline-rich repeat protein 3 as a novel atheroprotective factor that promotes adaptive Akt signaling in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2014;34:2527–2536. doi: 10.1161/ATVBAHA.114.303644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lee R, Feinbaum R, Ambros V. A short history of a short RNA. Cell. 2004;116:S89–92. doi: 10.1016/s0092-8674(04)00035-2. [DOI] [PubMed] [Google Scholar]
  • 96.Rayner KJ, Fernandez-Hernando C, Moore KJ. MicroRNAs regulating lipid metabolism in atherogenesis. Thromb Haemost. 2012;107:642–647. doi: 10.1160/TH11-10-0694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.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]
  • 98.Rayner KJ, Esau CC, Hussain FN, et al. Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature. 2011;478:404–407. doi: 10.1038/nature10486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, Parathath S, van Gils JM, Rayner AJ, Chang AN, Suarez Y, Fernandez-Hernando C, Fisher EA, Moore KJ. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J Clin Invest. 2011;121:2921–2931. doi: 10.1172/JCI57275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, Gerszten RE, Naar AM. MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science. 2010;328:1566–1569. doi: 10.1126/science.1189123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Gerin I, Clerbaux LA, Haumont O, Lanthier N, Das AK, Burant CF, Leclercq IA, MacDougald OA, Bommer GT. Expression of miR-33 from an SREBP2 intron inhibits cholesterol export and fatty acid oxidation. J Biol Chem. 2010;285:33652–33661. doi: 10.1074/jbc.M110.152090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Marquart TJ, Allen RM, Ory DS, Baldan A. miR-33 links SREBP-2 induction to repression of sterol transporters. Proc Natl Acad Sci U S A. 2010;107:12228–12232. doi: 10.1073/pnas.1005191107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Goedeke L, Vales-Lara FM, Fenstermaker M, Cirera-Salinas D, Chamorro-Jorganes A, Ramirez CM, Mattison JA, de Cabo R, Suarez Y, Fernandez-Hernando C. A regulatory role for microRNA 33* in controlling lipid metabolism gene expression. Mol Cell Biol. 2013;33:2339–2352. doi: 10.1128/MCB.01714-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Davalos A, Goedeke L, Smibert P, et al. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc Natl Acad Sci U S A. 2011;108:9232–9237. doi: 10.1073/pnas.1102281108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Goedeke L, Salerno A, Ramirez CM, Guo L, Allen RM, Yin X, Langley SR, Esau C, Wanschel A, Fisher EA, Suarez Y, Baldan A, Mayr M, Fernandez-Hernando C. Long-term therapeutic silencing of miR-33 increases circulating triglyceride levels and hepatic lipid accumulation in mice. EMBO Mol Med. 2014;6:1133–1141. doi: 10.15252/emmm.201404046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Rotllan N, Ramirez CM, Aryal B, Esau CC, Fernandez-Hernando C. Therapeutic silencing of microRNA-33 inhibits the progression of atherosclerosis in Ldlr−/− mice--brief report. Arterioscler Thromb Vasc Biol. 2013;33:1973–1977. doi: 10.1161/ATVBAHA.113.301732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Marquart TJ, Wu J, Lusis AJ, Baldan A. Anti-miR-33 therapy does not alter the progression of atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol. 2013;33:455–458. doi: 10.1161/ATVBAHA.112.300639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Horie T, Baba O, Kuwabara Y, et al. MicroRNA-33 deficiency reduces the progression of atherosclerotic plaque in ApoE−/− mice. J Am Heart Assoc. 2012;1:e003376. doi: 10.1161/JAHA.112.003376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Naar AM. Anti-atherosclerosis or No Anti-atherosclerosis: That is the miR-33 question. Arterioscler Thromb Vasc Biol. 2013;33:447–448. doi: 10.1161/ATVBAHA.112.301021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Wei Y, Nazari-Jahantigh M, Neth P, Weber C, Schober A. MicroRNA-126, -145, and -155: a therapeutic triad in atherosclerosis? Arterioscler Thromb Vasc Biol. 2013;33:449–454. doi: 10.1161/ATVBAHA.112.300279. [DOI] [PubMed] [Google Scholar]
  • 111.Kumar S, Kim CW, Simmons RD, Jo H. Role of flow-sensitive microRNAs in endothelial dysfunction and atherosclerosis: mechanosensitive athero-miRs. Arterioscler Thromb Vasc Biol. 2014;34:2206–2216. doi: 10.1161/ATVBAHA.114.303425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Di Gregoli K, Jenkins N, Salter R, White S, Newby AC, Johnson JL. MicroRNA-24 regulates macrophage behavior and retards atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34:1990–2000. doi: 10.1161/ATVBAHA.114.304088. [DOI] [PubMed] [Google Scholar]
  • 113.Schober A, Nazari-Jahantigh M, Wei Y, Bidzhekov K, Gremse F, Grommes J, Megens RT, Heyll K, Noels H, Hristov M, Wang S, Kiessling F, Olson EN, Weber C. MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat Med. 2014;20:368–376. doi: 10.1038/nm.3487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Boon RA, Dimmeler S. MicroRNA-126 in atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;34:e15–16. doi: 10.1161/ATVBAHA.114.303572. [DOI] [PubMed] [Google Scholar]
  • 115.Lovren F, Pan Y, Quan A, Singh KK, Shukla PC, Gupta N, Steer BM, Ingram AJ, Gupta M, Al-Omran M, Teoh H, Marsden PA, Verma S. MicroRNA-145 targeted therapy reduces atherosclerosis. Circulation. 2012;126:S81–90. doi: 10.1161/CIRCULATIONAHA.111.084186. [DOI] [PubMed] [Google Scholar]
  • 116.Du F, Yu F, Wang Y, Hui Y, Carnevale K, Fu M, Lu H, Fan D. MicroRNA-155 deficiency results in decreased macrophage inflammation and attenuated atherogenesis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2014;34:759–767. doi: 10.1161/ATVBAHA.113.302701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Donners MM, Wolfs IM, Stoger LJ, van der Vorst EP, Pottgens CC, Heymans S, Schroen B, Gijbels MJ, de Winther MP. Hematopoietic miR155 deficiency enhances atherosclerosis and decreases plaque stability in hyperlipidemic mice. PLoS One. 2012;7:e35877. doi: 10.1371/journal.pone.0035877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Nazari-Jahantigh M, Wei Y, Noels H, Akhtar S, Zhou Z, Koenen RR, Heyll K, Gremse F, Kiessling F, Grommes J, Weber C, Schober A. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J Clin Invest. 2012;122:4190–4202. doi: 10.1172/JCI61716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Meiler S, Baumer Y, Toulmin E, Seng K, Boisvert WA. MicroRNA 302a Is a Novel Modulator of Cholesterol Homeostasis and Atherosclerosis. Arterioscler Thromb Vasc Biol. 2014;35:323–331. doi: 10.1161/ATVBAHA.114.304878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.van Gils JM, Derby MC, Fernandes LR, et al. The neuroimmune guidance cue netrin-1 promotes atherosclerosis by inhibiting the emigration of macrophages from plaques. Nat Immunol. 2012;13:136–143. doi: 10.1038/ni.2205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Randolph GJ, Gautier EL. Emerging roles of neural guidance molecules in atherosclerosis: sorting out the complexity. Arterioscler Thromb Vasc Biol. 2013;33:882–883. doi: 10.1161/ATVBAHA.113.301346. [DOI] [PubMed] [Google Scholar]
  • 122.Ramkhelawon B, Yang Y, van Gils JM, Hewing B, Rayner KJ, Parathath S, Guo L, Oldebeken S, Feig JL, Fisher EA, Moore KJ. Hypoxia induces netrin-1 and Unc5b in atherosclerotic plaques: mechanism for macrophage retention and survival. Arterioscler Thromb Vasc Biol. 2013;33:1180–1188. doi: 10.1161/ATVBAHA.112.301008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Wanschel A, Seibert T, Hewing B, Ramkhelawon B, Ray TD, van Gils JM, Rayner KJ, Feig JE, O'Brien ER, Fisher EA, Moore KJ. Neuroimmune guidance cue Semaphorin 3E is expressed in atherosclerotic plaques and regulates macrophage retention. Arterioscler Thromb Vasc Biol. 2013;33:886–893. doi: 10.1161/ATVBAHA.112.300941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.van Gils JM, Ramkhelawon B, Fernandes L, Stewart MC, Guo L, Seibert T, Menezes GB, Cara DC, Chow C, Kinane TB, Fisher EA, Balcells M, Alvarez-Leite J, Lacy-Hulbert A, Moore KJ. Endothelial expression of guidance cues in vessel wall homeostasis dysregulation under proatherosclerotic conditions. Arterioscler Thromb Vasc Biol. 2013;33:911–919. doi: 10.1161/ATVBAHA.112.301155. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES