Introduction
Atherosclerosis is initiated and propagated over decades with no overt clinical symptoms. The complexity and chronicity of its pathophysiological process have been a major challenge to fully define molecular mechanisms and explore more effective therapeutic targets.1,2 This Recent Highlights series summarizes ATVB articles published in 2017 and 2018 on the traditional animal model systems and new approaches that have become powerful tools for studying pathogenesis and mechanisms of atherosclerosis.
Animal Models
Many animal models have been used to study atherosclerosis, as summarized in the recent AHA Scientific Statement,3 which provide suggestions on selecting appropriate animal models for a specific atherosclerosis study. Given its value and feasibility as a proof-of-principle model, mouse models have continued to be the most common species to study atherosclerosis.
Two Classic Mouse Models
The two most commonly used mouse models to study atherosclerosis are apolipoprotein E deficient (Apoe−/−) mouse4 and low-density lipoprotein (LDL) receptor deficient (Ldlr−/−) mouse.5 In 61 recent ATVB articles (Figure 1), studies in 41 articles used Apoe−/− mice,6–46 13 articles reported Ldlr−/− mice,47–59 and 7 articles reported both Apoe−/− and Ldlr−/− mice.60–66 The basis for the more common use of Apoe−/− mice, instead of Ldlr−/− mouse model, is unclear. However, it is worth noting that although both Apoe−/− and Ldlr−/− mice have profound hypercholesterolemia when fed a saturated fat-enriched diet, the mechanisms of lipoprotein metabolism dysfunction and development of atherosclerosis in these two mouse models may be different.3,67,68
Figure 1. Summary of 61 publications in ATVB between 2017 and 2018 that have reported Apoe−/− mice and/or Ldlr−/− mice.
Among the 61 articles, Apoe−/− mice were studied in 41 articles, Ldlr−/− mice were studied in 13 articles, and 7 articles reported both mouse strains. Among articles that studied Apoe−/− mice, 17 only reported male mice, 8 only reported female mice, and 16 reported both male and female mice. Among articles that studied Ldlr−/− mice, 5 only reported male mice, 1 only reported female mice, and 7 reported both male and female mice. Among articles that reported both mouse strains, 4 only reported male, 2 reported both male and female mice, and 1 did not provide the sex information.
Recently Developed Mouse Models
Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates LDL receptor activity.30,69 A D374Y gain-of-function mutation in human leads to hypercholesterolemia.70 Benefitting from the recently advances of using adeno-associated viral vectors (AAV), infection of AAVs carrying this human mutation or its mouse equivalent mutation D377Y in C57BL/6 mice has been used by many researchers to study atherosclerosis.71–81 Since sex difference is an important influence on development of atherosclerosis,82,83 some researchers have also studied potential sex difference of this method in inducing hypercholesterolemia and their contributions to atherosclerosis.80,84 Jarrett and colleagues compared an AAV containing human PCSK9 D374Y mutation (PCSK9D374Y.AAV) to AAV vectors expressing Staphylococcus aureus Cas9 and a guide RNA targeting the Ldlr gene (AAV-CRISPR) in both male and female C57BL/6 mice.80 Their study found that the AAV-CRISPR approach to target LDL receptors led to more profound hypercholesterolemia and increases of atherosclerosis than infection with PCSK9 D374Y.AAV in male mice, whereas PCSK9 D374Y.AAV method had more significant effects on plasma cholesterol concentrations and atherosclerosis than the AAV-CRISPR approach for LDL receptor deletion in female mice.80 The mechanism for sex dimorphism between these two methods is unclear.
Diet-related Effects
For the mouse models described above, it is common to feed these mice a diet that represents the dietary habits in Western countries.3,85–88 The currently available Apoe−/− mice have modest hypercholesterolemia that develop minimal atherosclerosis spontaneously. However, feeding a Western diet profoundly increases plasma cholesterol concentrations and accelerates atherosclerosis. Some laboratories also use a similar fat-enriched diet supplemented with cholesterol (1.25% wt/wt) and cholate, as discussed in the recent AHA statement.3 Some recent studies have provided evidence that gut microbiota influence the development of atherosclerosis,89–91 although findings from one study do not support that microbiota and choline-supplemented diet affect atherosclerotic lesion size in a hypercholesterolemic mouse model.9 Irrespectively, it is important to provide detailed information including mouse housing conditions and whether diets are supplemented with choline when report atherosclerosis studies in animal models.9,92
Large Animal Models
Despite the much higher expense and difficulty in genetic manipulations, large animal models have potential benefits over mice in providing more relevant pathological and mechanistic insights into the disease in human. Three recent articles in ATVB have reported manipulations of lipoprotein metabolism and the consequences on atherosclerosis in rabbits.93–95 In one study, an apoA-I-expressing helper-dependent adenoviral vector delivered to carotid arteries led to reduced atherosclerosis in fat diet-fed rabbits.93 In another study, Wang and colleagues generated transgenic rabbits expressing the human endothelial lipase in liver.94 These rabbits had lower plasma cholesterol concentrations and less atherosclerotic lesions in both the aorta and coronary arteries, compared to their non-transgenic littermates. These investigators also developed a cholesterol ester transfer protein deficient rabbit model.95 These rabbits fed a cholesterol diet for 18 weeks exhibited lower plasma total cholesterol concentrations and atherosclerotic lesions in both the aorta and coronary arteries, compared to their wild type controls.95 It was noted that these rabbit models develop atherosclerotic lesions in coronary arteries, which is common in humans, but not commonly observed in mouse models.
Pigs are also an optimal animal model to study atherosclerosis due to their similarities to human anatomy and comparable patterns of atherosclerotic lesions in coronary arteries.3 Recently, a panel of genes that regulate endothelial cell apoptosis induced by shear stress have been identified in aortas of healthy pigs, implicating potential mechanisms in atherosclerosis development.96 Burke and colleagues successfully depleted LDL receptor in Yucatan miniature pigs.97 A high-fat diet feeding on these pigs led to profound increases of plasma LDL-cholesterol concentrations and large lesions in the aorta and coronary arteries. Bempedoic acid, a drug targeting hepatic ATP-citrate lyase, reduced both plasma cholesterol concentrations and atherosclerosis in this pig model.97
Established Genetic Approaches
This section introduces two well-established genetic manipulations in mouse models that have provided remarkable insights into understanding mechanisms of atherosclerosis. One approach is bone marrow transplantation.98 The other approach is the LoxP-Cre conditional genetic manipulation first reported by Sauer more than 30 years ago.99
Bone Marrow Transplantation
Leukocytes including macrophages, lymphocytes, and neutrophils are present in atherosclerosis, with macrophages being the predominant cell type during development of atherosclerotic lesions.100 One approach for studying effects of a gene-of-interest in leukocytes is bone marrow transplantation. This approach, since it was first reported by Linton and Fazio in 1995101 and by the Curtiss group102 in mouse models for atherosclerosis, it has been used widely to study atherosclerosis. In many recent publications of ATVB, with this approach, it was demonstrated that many components involving from transcription to protein functions contribute to either development or prevention of atherosclerosis. Some studies also report mixed chimeric mouse models using this bone marrow transplantation method. For example, mixed chimeric recipient mice were developed by transplanting bone marrow cells from μMT chain-deficient mice (80%) and bone marrow cells from mice deficient in CD40 (20%) into irradiated Ldlr−/− mice. These chimeric mice had low abundance of CD40 in B cells, which provided evidence that B cell-selective CD40 deficiency reduced atherosclerosis in mice.61 The recent studies published in ATVB that applied this approach in mouse atherosclerosis models are summarized in Table 1 (Ldlr−/− mice as recipient) and Table 2 (Apoe−/− mice as recipient). From these two tables, it is clear that Ldlr−/− mice are more frequently used as recipient mice for bone marrow transplantation since donor mice do not need to be in an Ldlr−/− background, as noted previously.102,103
Table 1.
Bone Marrow Transplantation in Ldlr−/− Recipient Mice
| Manipulation of Gene | Full Name of Gene | Other Manipulations | Effects on Atherosclerosis* | Reference |
|---|---|---|---|---|
| Depletion of Zhx2 | Zinc fingers and homeoboxes 2 | Western diet x 18 or 24 weeks | aortic root ↓; en face ↓ | 47 |
| Depletion of Dab2 | Disabled homolog 2 | Western diet x 20 weeks | aortic root ↔; en face ↓ | 50 |
| Depletion of IL-19 | Interleukin-19 | Western diet x 14 weeks | en face ↑ | 49 |
| Depletion of ACAT1 | Acyl-CoA cholesterol acyltransferase | Western diet x 8 weeks | aortic root ↑ | 52 |
| Depletion of NLRP3 | NACHT, LRR, and PYD domain-containing protein 3 | aortic root ↓ | ||
| Human iNAMPT transgene | Intracellular nicotinamide phosphoribosyltransferase | Western diet x 12 weeks | aortic root ↓ | 57 |
| human mCAT transgene | Mitochondrial catalase | Western diet x 16 weeks | aortic root ↓ | 55 |
| Depletion of myeloid EGFR | Epidermal growth factor receptor | High cholesterol diet x 4, 7, or 12 weeks | aortic root ↓ | 53 |
| Depletion of miR146a | microRNA 146a | High cholesterol diet x 12 weeks | N/A | 63 |
| E2-def | Lack of Ly6C− monocytes | High cholesterol diet x 15 weeks | en face ↑ | 64 |
| Depletion of myeloid MED1 | Mediator 1 | Western diet x 12 weeks | aortic root ↑; en face ↑ | 65 |
| Depletion of MHCII (in follicular B cells) | Major histocompatibility II | Western diet x 8 weeks | aortic root ↓ | 61 |
| Depletion of CD40 (in follicular B cells) | Cluster of differentiation 40 | Western diet x 8 weeks | aortic root ↓ | |
| Depletion of Blimp-1 (in follicular B cells) | B-lymphocyte-induced maturation protein | Western diet x 8 weeks | aortic root ↓ |
Effects on atherosclerosis, compared to their wild type controls. ↑: increase; ↓: decrease; ↔: no change
Table 2.
Bone Marrow Transplantation in Apoe−/− Recipient Mice
| Manipulation of Gene | Full Name of Gene | Recipient Mouse Strain | Other Manipulations | Effects on Atherosclerosis* | Reference |
|---|---|---|---|---|---|
| Human p27kip transgene | Cyclin-dependent kinase inhibitor 1B | Apoe−/− | Unknown | aortic root ↓ | 12 |
| Depletion of VWF | von Willebrand factor | Apoe−/− | Western diet x 14 weeks | aortic root ↔; en face ↔ | 19 |
| Depletion of Sema7A | Semaphorin 7A | Apoe−/− | partial carotid artery ligation and Western diet x 2 weeks | carotid artery ↔ | 24 |
Effects on atherosclerosis, compared to their wild type controls. ↓: decrease; ↔: no change
LoxP-Cre Genetic Manipulations
Cross-breeding mice carrying LoxP franked gene with mice carrying Cre transgene driven by a specific promoter is a well-recognized approach for inducing tissue or cell-specific genetic manipulations.104,105
LysM-Cre transgenic mice,106 which generate conditional gene targeting macrophages and granulocytes, have been used frequently to study experimental atherosclerosis. Using mice expressing Cre under the control of the LysM-Cre promoter, attenuation of atherosclerotic development occurred in mice with (1) deletion of myeloid β-catenin, a critical component regulating many cell functions and behaviors differentiation,48 (2) depletion of lipin-1, an important enzyme in the glycerolipid synthesis pathway,81 (3) depletion of ADK (adenosine kinase), an essential enzyme regulating intracellular adenosine levels,46 and (4) depletion of EGFR (Epidermal Growth Factor Receptor), a prominent member of the receptor tyrosine kinase family.53 Mice with hypercholesterolemia, such as Apoe−/− or Ldlr−/−, were used for these studies. Reduced atherosclerosis through manipulation of any of these genes did not affect plasma cholesterol concentrations, supporting the notion that hypercholesterolemia is crucial in the initiation and development of atherosclerosis, but prevention of atherosclerosis can be achieved even in the presence of profound hypercholesterolemia. This concept was also supported by studies of HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase, the rate-limiting enzyme in cholesterol biosynthesis. Inhibition of HMG-CoA by statins has profound effects on reducing plasma cholesterol concentrations in human, but not in the commonly used hypercholesterolemic mouse models.1,107–112 Despite its lack of effects on plasma cholesterol concentrations, statins reduce atherosclerosis in mouse models.111–115 Depletion of HMG-CoA reductase in myeloid cells, as achieved by LysM-Cre activation, attenuates atherosclerosis in Ldlr−/− mice.51
Endothelial cells play critical roles in the initiation and development of atherosclerosis.116 Nucleotide P2Y2 receptor (P2Y2R) regulates vascular cell adhesion molecule-1 (VCAM-1). P2Y2 floxed mice were bred with cadherin 5-Cre mice to generate endothelial cell specific P2Y2 deficiency, which reduced atherosclerosis in Apoe−/− mice.43 In another study, inducible and endothelial cell-specific alpha5 deficient mice were generated by a cadherin promoter driven Cre-ERT2 with tamoxifen injections. Atherosclerotic lesions were reduced by alpha5 endothelial-specific depletion.15 It is worth noting that inducible Cre is not only used for cell-specific studies, but also used to induce Cre activity to deplete a gene globally in adult mice, which could avoid potential impact during embryonic development.117 For example, depletion of ApoE was achieved in Apoe floxed mice with Rosa26 Cre-ERT2 at 10–12 weeks of age, with an oral dose of tamoxifen that increased plasma cholesterol concentrations and atherosclerosis.10
The SM22 promoter has been well-accepted for driving smooth muscle cell-specific genetic manipulation.118 Two recent studies reported smooth muscle cells-specific effects, as achieved by SM22-Cre recombinase activity, on atherosclerosis: Deficiency of IGF1 (insulin-like growth factor) receptor in smooth muscle cells, augmented atherosclerosis;16 and smooth muscle cell-specific overexpression of USP20 (ubiquitin-specific protease 20, a deubiquitinase) reduced atherosclerosis.62
In addition to the commonly used myeloid, endothelial cell, and smooth muscle cell-specific Cre, several other tissue- or cell-specific Cre recombinase studies were reported to have an effect on mouse atherosclerosis. Brown adipocyte-specific PPARγ (peroxisome proliferator-activated receptor gamma) deficient mice were developed by crossbreeding PPARγ floxed mice with UCP-1 (uncoupling protein 1)-driven Cre mice.119 Deficiency of PPARγ in brown adipocytes led to augmented atherosclerosis.8 Adipocyte-specific LRP1 (low-density lipoprotein receptor-related protein 1) deficient mice were generated by mating LRP1 floxed mice with aP2-Cre transgenic mice,120 which exhibited accelerated atherosclerosis in carotid arteries.56 Epithelial cell-specific androgen receptor deficient mice were generated by K5-Cre recombinase activity in androgen receptor floxed mice, which showed increased atherosclerosis.6
Three recent seminal publications have provided comprehensive discussion on adventitial fibroblast,121 endothelial cell,122 and smooth muscle cell-specific123 Cre-recombinase mice. The strengths and weaknesses of multiple Cre-recombinase mouse lines were discussed in these three articles. An important synopsis of these publications is that it is important to validate the location and abundance of the genetic manipulation in Cre transgenic conditional mice.
Emerging Applications
Imaging systems including ultrasound, CT (Computed Tomography), PET (Positron Emission Tomography), and MRI (Magnetic Resonance Imaging) are used for diagnosing, localizing, and characterizing atherosclerotic lesions in humans.124–129 These imaging systems, along with cytometric analysis and microscopy, have provided insights into understanding mechanisms and pathogenesis of atherosclerosis.129 Many of these sophisticated techniques have been applied to animal models to track macrophage biology and behavior because macrophage dynamics are a salient component in the development and progression of atherosclerosis.64,79,130–133
Mass Cytometry
Flow cytometry is traditionally used to quantify a limited number of cell surface markers. Mass cytometry quantifies protein abundance at a single-cell resolution using heavy-metal-conjugated antibodies in tandem with time-of-flight mass spectrometry.134 Thomas and colleagues profiled human monocytes with a panel of 36 cell surface markers using this mass cytometry in combination with a computational analysis method,135 which defined monocyte classifications by incorporating all cell surface markers simultaneously. Using this method, a small but specific panel of surface markers on human blood monocytes were identified, which improved monocyte subset identification with high accuracy and reproducibility.135 This method is expected to facilitate identification of monocyte subsets and enhance consistency in defining monocyte functions not only in atherosclerosis research field but also many other research fields.
Two-photon Microscopy
Microscopy is an essential tool to visualize, characterize, and quantify atherosclerotic lesions. The state-of-the-art intravital two-photon imaging enables visualization of macrophage dynamics in a real-time manner in live animals. In a recent study, Li and colleagues136 transplanted aortic arch grafts from Apoe−/− mice to the right carotid arteries of C57BL/6 CX3CR1 GFP reporter mice. Recipient monocyte-derived macrophages recruited to atherosclerotic lesions was detectable one month after transplantation, but reduced profoundly 4 months after transplantation, which was visualized using a two-photon microscope on the same animal at two time points.136 Two-photon microscopy was also used for intricate characterization of macrophage phenotypes. Williams and colleagues found that in Ldlr−/− mice fed a fat-enriched diet for 12 – 16 weeks, lipid-laden macrophages accumulated in the intima of carotid artery had different morphology and locations: Large asymmetrical cells were static, but smaller round cells near lesional margins were mobile.60 During the progression of atherosclerotic lesions in Apoe−/− mice, macrophages, as detected by nondegradable phagocytic particles, appeared to move to deeper location within atherosclerotic lesions, whereas during regression of atherosclerotic lesions in Apoe−/− mice achieved by application of AAV8 vector encoding ApoE, motile macrophages within atherosclerotic lesions were closer to the surface of lesions.60 These two recent studies provide insights into understanding complex dynamics of macrophages during the development of atherosclerosis in animal models.
Positron Emission Tomography (PET)
Fluorodeoxyglucose (18F-FDG) is a marker for cellular uptake of glucose. Since macrophages have high glycolytic capacity, 18F-FDG-PET has been used to monitor the abundance of macrophage accumulation within atherosclerotic lesions.137–141 Macrophage behavior and functions are highly heterogeneous.131 The classic PET using 18F-FDG alone is not able to identify the heterogeneity of macrophages. To overcome this shortcoming, Tavakoli and colleagues applied PET by combining measurement of glucose (18F-FDG) and 14C-glutamine accumulation.142 This approach identified different patterns of 18F-FDG and 14C-glutamine accumulation in macrophages, further demonstrating heterogeneity of macrophages accumulated in atherosclerotic lesions. In addition to 18F-FDG labeling, a recent study assessed accumulation of monocytes and monocyte-derived macrophages during the regression of atherosclerotic plaques using serial-based scanning with a CCR2 (C-C chemokine receptor 2)-specific imaging probe, which specifically detect CCR2-related responses.136 It is envisioned that advancement of different labeling for PET imaging, combined with histological analysis, will enhance accuracy of atherosclerotic lesion characterization.
Near-infrared calcium imaging
Calcification is a critical condition of atherosclerotic diseases.33,128,143–149 In addition to histology to visualize calcification in serial sections of atherosclerotic lesions, near-infrared calcium imaging was used for ex vivo molecular imaging and quantification of the calcification process, based on the principle that conjugated bisphosphonate compounds could bind to hydroxyapatite deposited by osteoblast-like cells during the mineralization of calcium.150,151 Using this ex vivo near-infrared calcium imaging, Ceneri and colleagues found that calcification signal became gradually enhanced in Apoe−/− mice fed a high fat and high cholesterol diet for 7, 14, or 21 weeks.42 The authors also demonstrated that deletion of Rac2, an important signal transducer in bone marrow-derived cells, resulted in increased calcification in atherosclerotic lesions,42 confirming versatile effects of macrophages in the development of atherosclerosis.
Molecular Ultrasound and Molecular Tomography
Molecular ultrasound imaging using targeted microbubbles visualizes atherosclerotic components at the molecular level beyond classic ultrasonography.152 Curaj and colleagues153 using JAM-A, a junctional adhesion molecule, targeted microbubbles to detected flow-dependent endothelial dysfunction induced by carotid partial ligation in Apoe−/− mice. The authors found JAM-A-targeted microbubbles rapidly bound the ligated side, and reached a peak after two weeks of the ligation procedure. This study provides evidence that JAM-A is a useful imaging biomarker to assess endothelial activation and dysfunction at early stages of atherosclerotic plaque formation.153
In addition to endothelial dysfunction, neutrophil accumulation is present in early stages of atherosclerosis.154–156 In Ldlr−/− mice fed Western diet were imaged at 4, 8, and 12 weeks by fluorescence molecular tomography/x-ray CT after injections of an elastase-targeted fluorescent agent.157 The fluorescent signal in the area of the aortic arch was highest after 4 weeks of Western diet feeding, but decreased after 8 and 12 weeks of Western diet feeding,157 implicating elastase-targeted fluorescence molecular tomography is feasible for detecting early development of atherosclerosis.
Summary
The concept that atherosclerosis is a lipid-driven inflammatory disease has been supported by diverse evidence from basic and clinical research.131,158–163 Cell types that are involved in atherosclerosis development include leukocytes and resident cell types such as endothelial cells, smooth muscle cells, and cells in the adventitia. Although it is hard to conclude which cell type is more important than the other cell types, or one specific molecule is more important than the other multiple molecules in a single cell type, the recent studies have provided more elaborate insights into many components in the development of atherosclerosis. In addition to animal studies, human studies have also continuously supported the importance of lipoprotein metabolisms and inflammation in the development of atherosclerosis.127,148,164–176 We hope that insights from these animal and human studies will enhance our understanding of this complex disease with an ultimate goal to prevent the development of atherosclerosis and improve related adverse outcomes.
Acknowledgments
Sources of Funding
The authors’ research work was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award numbers R01HL133723 and R01HL139748 and the American Heart Association SFRN in Vascular Disease (18SFRN33960001). 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.Rader DJ, Daugherty A. Translating molecular discoveries into new therapies for atherosclerosis. Nature. 2008;451:904–913. [DOI] [PubMed] [Google Scholar]
- 2.Newman CB, Preiss D, Tobert JA et al. Statin Safety and Associated Adverse Events: A Scientific Statement From the American Heart Association. Arterioscler Thromb Vasc Biol. 2019;39:e38–81. [DOI] [PubMed] [Google Scholar]
- 3.Daugherty A, Tall AR, Daemen MJAP, Falk E, Fisher EA, Garcia-Cardena G, Lusis AJ, Owens AP 3rd, Rosenfeld ME, Virmani R. Recommendation on design, execution, and reporting of animal atherosclerosis studies: A Scientific Statement from the American Heart Association. Arterioscler Thromb Vasc Biol. 2017;37:e131–e157. [DOI] [PubMed] [Google Scholar]
- 4.Zhang SH, Reddick RL, Piedrahita JA, Maeda N. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science. 1992;258:468–471. [DOI] [PubMed] [Google Scholar]
- 5.Ishibashi S, Goldstein JL, Brown MS, Herz J, Burns DK. Massive xanthomatosis and atherosclerosis in cholesterol-fed low density lipoprotein receptor-negative mice. J Clin Invest. 1994;93:1885–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wilhelmson AS, Lantero Rodriguez M, Svedlund Eriksson E, Johansson I, Fogelstrand P, Stubelius A, Lindgren S, Fagman JB, Hansson GK, Carlsten H, Karlsson MCI, Ekwall O, Tivesten A. Testosterone Protects Against Atherosclerosis in Male Mice by Targeting Thymic Epithelial Cells-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:1519–1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lallemand T, Rouahi M, Swiader A, Grazide MH, Geoffre N, Alayrac P, Recazens E, Coste A, Salvayre R, Negre-Salvayre A, Auge N. nSMase2 (Type 2-Neutral Sphingomyelinase) Deficiency or Inhibition by GW4869 Reduces Inflammation and Atherosclerosis in Apoe(−/−) Mice. Arterioscler Thromb Vasc Biol. 2018;38:1479–1492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Xiong W, Zhao X, Villacorta L, Rom O, Garcia-Barrio MT, Guo Y, Fan Y, Zhu T, Zhang J, Zeng R, Chen YE, Jiang Z, Chang L. Brown Adipocyte-Specific PPARgamma (Peroxisome Proliferator-Activated Receptor gamma) Deletion Impairs Perivascular Adipose Tissue Development and Enhances Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol. 2018;38:1738–1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lindskog Jonsson A, Caesar R, Akrami R, Reinhardt C, Fak Hallenius F, Boren J, Backhed F. Impact of gut microbiota and diet on the development of atherosclerosis in Apoe(−/−) mice. Arterioscler Thromb Vasc Biol. 2018;38:2318–2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Centa M, Prokopec KE, Garimella MG et al. Acute Loss of Apolipoprotein E Triggers an Autoimmune Response That Accelerates Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:e145–e158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Richardson GD, Sage A, Bennaceur K, Al Zhrany N, Coelho-Lima J, Dookun E, Draganova L, Saretzki G, Breault DT, Mallat Z, Spyridopoulos I. Telomerase Mediates Lymphocyte Proliferation but Not the Atherosclerosis-Suppressive Potential of Regulatory T-Cells. Arterioscler Thromb Vasc Biol. 2018;38:1283–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yamada S, Senokuchi T, Matsumura T et al. Inhibition of Local Macrophage Growth Ameliorates Focal Inflammation and Suppresses Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:994–1006. [DOI] [PubMed] [Google Scholar]
- 13.Cui XB, Luan JN, Dong K, Chen S, Wang Y, Watford WT, Chen SY. RGC-32 (Response Gene to Complement 32) Deficiency Protects Endothelial Cells From Inflammation and Attenuates Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:e36–e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Rinne P, Guillamat-Prats R, Rami M, Bindila L, Ring L, Lyytikainen LP, Raitoharju E, Oksala N, Lehtimaki T, Weber C, van der Vorst EPC, Steffens S. Palmitoylethanolamide Promotes a Proresolving Macrophage Phenotype and Attenuates Atherosclerotic Plaque Formation. Arterioscler Thromb Vasc Biol. 2018;38:2562–2575. [DOI] [PubMed] [Google Scholar]
- 15.Al-Yafeai Z, Yurdagul A Jr, Peretik JM, Alfaidi M, Murphy PA, Orr AW. Endothelial FN (Fibronectin) Deposition by alpha5beta1 Integrins Drives Atherogenic Inflammation. Arterioscler Thromb Vasc Biol. 2018;38:2601–2614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sukhanov S, Higashi Y, Shai SY, Snarski P, Danchuk S, D’Ambra V, Tabony M, Woods TC, Hou X, Li Z, Ozoe A, Chandrasekar B, Takahashi SI, Delafontaine P. SM22alpha (Smooth Muscle Protein 22-alpha) Promoter-Driven IGF1R (Insulin-Like Growth Factor 1 Receptor) Deficiency Promotes Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:2306–2317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nishino T, Horie T, Baba O et al. SREBF1/MicroRNA-33b Axis Exhibits Potent Effect on Unstable Atherosclerotic Plaque Formation In Vivo. Arterioscler Thromb Vasc Biol. 2018;38:2460–2473. [DOI] [PubMed] [Google Scholar]
- 18.Li X, Shao Y, Sha X, Fang P, Kuo YM, Andrews AJ, Li Y, Yang WY, Maddaloni M, Pascual DW, Luo JJ, Jiang X, Wang H, Yang X. IL-35 (Interleukin-35) Suppresses Endothelial Cell Activation by Inhibiting Mitochondrial Reactive Oxygen Species-Mediated Site-Specific Acetylation of H3K14 (Histone 3 Lysine 14). Arterioscler Thromb Vasc Biol. 2018;38:599–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Doddapattar P, Dhanesha N, Chorawala MR, Tinsman C, Jain M, Nayak MK, Staber JM, Chauhan AK. Endothelial Cell-Derived Von Willebrand Factor, But Not Platelet-Derived, Promotes Atherosclerosis in Apolipoprotein E-Deficient Mice. Arterioscler Thromb Vasc Biol. 2018;38:520–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Doddapattar P, Jain M, Dhanesha N, Lentz SR, Chauhan AK. Fibronectin Containing Extra Domain A Induces Plaque Destabilization in the Innominate Artery of Aged Apolipoprotein E-Deficient Mice. Arterioscler Thromb Vasc Biol. 2018;38:500–508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yu H, Fellows A, Foote K, Yang Z, Figg N, Littlewood T, Bennett M. FOXO3a (Forkhead Transcription Factor O Subfamily Member 3a) Links Vascular Smooth Muscle Cell Apoptosis, Matrix Breakdown, Atherosclerosis, and Vascular Remodeling Through a Novel Pathway Involving MMP13 (Matrix Metalloproteinase 13). Arterioscler Thromb Vasc Biol. 2018;38:555–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Karamariti E, Zhai C, Yu B et al. DKK3 (Dickkopf 3) Alters Atherosclerotic Plaque Phenotype Involving Vascular Progenitor and Fibroblast Differentiation Into Smooth Muscle Cells. Arterioscler Thromb Vasc Biol. 2018;38:425–437. [DOI] [PubMed] [Google Scholar]
- 23.Rinne P, Kadiri JJ, Velasco-Delgado M, Nuutinen S, Viitala M, Hollmen M, Rami M, Savontaus E, Steffens S. Melanocortin 1 Receptor Deficiency Promotes Atherosclerosis in Apolipoprotein E(−/−) Mice. Arterioscler Thromb Vasc Biol. 2018;38:313–323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hu S, Liu Y, You T et al. Vascular Semaphorin 7A Upregulation by Disturbed Flow Promotes Atherosclerosis Through Endothelial beta1 Integrin. Arterioscler Thromb Vasc Biol. 2018;38:335–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Park K, Li Q, Evcimen ND et al. Exogenous Insulin Infusion Can Decrease Atherosclerosis in Diabetic Rodents by Improving Lipids, Inflammation, and Endothelial Function. Arterioscler Thromb Vasc Biol. 2018;38:92–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yu P, Xiong T, Tenedero CB, Lebeau P, Ni R, MacDonald ME, Gross PL, Austin RC, Trigatti BL. Rosuvastatin Reduces Aortic Sinus and Coronary Artery Atherosclerosis in SR-B1 (Scavenger Receptor Class B Type 1)/ApoE (Apolipoprotein E) Double Knockout Mice Independently of Plasma Cholesterol Lowering. Arterioscler Thromb Vasc Biol. 2018;38:26–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Yu EPK, Reinhold J, Yu H, Starks L, Uryga AK, Foote K, Finigan A, Figg N, Pung YF, Logan A, Murphy MP, Bennett M. Mitochondrial Respiration Is Reduced in Atherosclerosis, Promoting Necrotic Core Formation and Reducing Relative Fibrous Cap Thickness. Arterioscler Thromb Vasc Biol. 2017;37:2322–2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Trenteseaux C, Gaston AT, Aguesse A, Poupeau G, de Coppet P, Andriantsitohaina R, Laschet J, Amarger V, Krempf M, Nobecourt-Dupuy E, Ouguerram K. Perinatal Hypercholesterolemia Exacerbates Atherosclerosis Lesions in Offspring by Altering Metabolism of Trimethylamine-N-Oxide and Bile Acids. Arterioscler Thromb Vasc Biol. 2017;37:2053–2063. [DOI] [PubMed] [Google Scholar]
- 29.van der Heijden T, Kritikou E, Venema W, van Duijn J, van Santbrink PJ, Slutter B, Foks AC, Bot I, Kuiper J. NLRP3 Inflammasome Inhibition by MCC950 Reduces Atherosclerotic Lesion Development in Apolipoprotein E-Deficient Mice-Brief Report. Arterioscler Thromb Vasc Biol. 2017;37:1457–1461. [DOI] [PubMed] [Google Scholar]
- 30.Sun L, Yang X, Li Q et al. Activation of Adiponectin Receptor Regulates Proprotein Convertase Subtilisin/Kexin Type 9 Expression and Inhibits Lesions in ApoE-Deficient Mice. Arterioscler Thromb Vasc Biol. 2017;37:1290–1300. [DOI] [PubMed] [Google Scholar]
- 31.Ditiatkovski M, Palsson J, Chin-Dusting J, Remaley AT, Sviridov D. Apolipoprotein A-I mimetic peptides: Discordance between in vitro and in vivo properties-Brief Report. Arterioscler Thromb Vasc Biol. 2017;37: 7:1301–1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kayashima Y, Makhanova N, Maeda N. DBA/2J haplotype on distal chromosome 2 reduces Mertk expression, restricts efferocytosis, and increases susceptibility to atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:e82–e91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.McRobb LS, McGrath KCY, Tsatralis T, Liong EC, Tan JTM, Hughes G, Handelsman DJ, Heather AK. Estrogen receptor control of atherosclerotic calcification and smooth muscle cell osteogenic differentiation. Arterioscler Thromb Vasc Biol. 2017;37:1127–1137. [DOI] [PubMed] [Google Scholar]
- 34.Desjarlais M, Dussault S, Dhahri W, Mathieu R, Rivard A. MicroRNA-150 Modulates Ischemia-Induced Neovascularization in Atherosclerotic Conditions. Arterioscler Thromb Vasc Biol. 2017;37:900–908. [DOI] [PubMed] [Google Scholar]
- 35.Hasanov Z, Ruckdeschel T, Konig C, Mogler C, Kapel SS, Korn C, Spegg C, Eichwald V, Wieland M, Appak S, Augustin HG. Endosialin Promotes Atherosclerosis Through Phenotypic Remodeling of Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol. 2017;37:495–505. [DOI] [PubMed] [Google Scholar]
- 36.Krishna SM, Seto SW, Jose RJ, Li J, Morton SK, Biros E, Wang Y, Nsengiyumva V, Lindeman JH, Loots GG, Rush CM, Craig JM, Golledge J. Wnt Signaling Pathway Inhibitor Sclerostin Inhibits Angiotensin II-Induced Aortic Aneurysm and Atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:553–566. [DOI] [PubMed] [Google Scholar]
- 37.Niu X, Pi SL, Baral S, Xia YP, He QW, Li YN, Jin HJ, Li M, Wang MD, Mao L, Hu B. P2Y12 Promotes Migration of Vascular Smooth Muscle Cells Through Cofilin Dephosphorylation During Atherogenesis. Arterioscler Thromb Vasc Biol. 2017;37:515–524. [DOI] [PubMed] [Google Scholar]
- 38.de Jong RJ, Paulin N, Lemnitzer P, Viola JR, Winter C, Ferraro B, Grommes J, Weber C, Reutelingsperger C, Drechsler M, Soehnlein O. Protective Aptitude of Annexin A1 in Arterial Neointima Formation in Atherosclerosis-Prone Mice-Brief Report. Arterioscler Thromb Vasc Biol. 2017;37:312–315. [DOI] [PubMed] [Google Scholar]
- 39.Xu R, Li C, Wu Y, Shen L, Ma J, Qian J, Ge J. Role of KCa3.1 Channels in Macrophage Polarization and Its Relevance in Atherosclerotic Plaque Instability. Arterioscler Thromb Vasc Biol. 2017;37:226–236. [DOI] [PubMed] [Google Scholar]
- 40.McNeill E, Iqbal AJ, Jones D, Patel J, Coutinho P, Taylor L, Greaves DR, Channon KM. Tracking Monocyte Recruitment and Macrophage Accumulation in Atherosclerotic Plaque Progression Using a Novel hCD68GFP/ApoE−/− Reporter Mouse-Brief Report. Arterioscler Thromb Vasc Biol. 2017;37:258–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sanz-Garcia C, Sanchez A, Contreras-Jurado C, Cales C, Barranquero C, Munoz M, Merino R, Escudero P, Sanz MJ, Osada J, Aranda A, Alemany S. Map3k8 Modulates Monocyte State and Atherogenesis in ApoE−/− Mice. Arterioscler Thromb Vasc Biol. 2017;37:237–246. [DOI] [PubMed] [Google Scholar]
- 42.Ceneri N, Zhao L, Young BD et al. Rac2 Modulates Atherosclerotic Calcification by Regulating Macrophage Interleukin-1beta Production. Arterioscler Thromb Vasc Biol. 2017;37:328–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chen X, Qian S, Hoggatt A, Tang H, Hacker TA, Obukhov AG, Herring PB, Seye CI. Endothelial Cell-Specific Deletion of P2Y2 Receptor Promotes Plaque Stability in Atherosclerosis-Susceptible ApoE-Null Mice. Arterioscler Thromb Vasc Biol. 2017;37:75–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wang C, Chen H, Zhu W et al. Nicotine Accelerates Atherosclerosis in Apolipoprotein E-Deficient Mice by Activating alpha7 Nicotinic Acetylcholine Receptor on Mast Cells. Arterioscler Thromb Vasc Biol. 2017;37:53–65. [DOI] [PubMed] [Google Scholar]
- 45.Sasaki N, Yamashita T, Kasahara K et al. UVB Exposure Prevents Atherosclerosis by Regulating Immunoinflammatory Responses. Arterioscler Thromb Vasc Biol. 2017;37:66–74. [DOI] [PubMed] [Google Scholar]
- 46.Zhang M, Zeng X, Yang Q et al. Ablation of myeloid ADK (adenosine kinase) epigenetically suppresses atherosclerosis in ApoE−/− (Apolipoprotein E deficient) mice. Arterioscler Thromb Vasc Biol. 2018;38:2780–2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Erbilgin A, Seldin MM, Wu X, Mehrabian M, Zhou Z, Qi H, Dabirian KS, Sevag Packard RR, Hsieh W, Bensinger SJ, Sinha S, Lusis AJ. Transcription Factor Zhx2 Deficiency Reduces Atherosclerosis and Promotes Macrophage Apoptosis in Mice. Arterioscler Thromb Vasc Biol. 2018;38:2016–2027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wang F, Liu Z, Park SH, Gwag T, Lu W, Ma M, Sui Y, Zhou C. Myeloid beta-Catenin Deficiency Exacerbates Atherosclerosis in Low-Density Lipoprotein Receptor-Deficient Mice. Arterioscler Thromb Vasc Biol. 2018;38:1468–1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ray M, Gabunia K, Vrakas CN, Herman AB, Kako F, Kelemen SE, Grisanti LA, Autieri MV. Genetic Deletion of IL-19 (Interleukin-19) Exacerbates Atherogenesis in Il19(−/−)xLdlr(−/−) Double Knockout Mice by Dysregulation of mRNA Stability Protein HuR (Human Antigen R). Arterioscler Thromb Vasc Biol. 2018;38:1297–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Adamson SE, Polanowska-Grabowska R, Marqueen K, Griffiths R, Angdisen J, Breevoort SR, Schulman IG, Leitinger N. Deficiency of Dab2 (Disabled Homolog 2) in Myeloid Cells Exacerbates Inflammation in Liver and Atherosclerotic Plaques in LDLR (Low-Density Lipoprotein Receptor)-Null Mice-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:1020–1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sakai K, Nagashima S, Wakabayashi T et al. Myeloid HMG-CoA (3-Hydroxy-3-Methylglutaryl-Coenzyme A) Reductase Determines Atherosclerosis by Modulating Migration of Macrophages. Arterioscler Thromb Vasc Biol. 2018;38:2590–2600. [DOI] [PubMed] [Google Scholar]
- 52.Wakabayashi T, Takahashi M, Yamamuro D, Karasawa T, Takei A, Takei S, Yamazaki H, Nagashima S, Ebihara K, Takahashi M, Ishibashi S. Inflammasome Activation Aggravates Cutaneous Xanthomatosis and Atherosclerosis in ACAT1 (Acyl-CoA Cholesterol Acyltransferase 1) Deficiency in Bone Marrow. Arterioscler Thromb Vasc Biol. 2018;38:2576–2589. [DOI] [PubMed] [Google Scholar]
- 53.Zeboudj L, Giraud A, Guyonnet L, Zhang Y, Laurans L, Esposito B, Vilar J, Chipont A, Papac-Milicevic N, Binder CJ, Tedgui A, Mallat Z, Tharaux PL, Ait-Oufella H. Selective EGFR (Epidermal Growth Factor Receptor) Deletion in Myeloid Cells Limits Atherosclerosis-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:114–119. [DOI] [PubMed] [Google Scholar]
- 54.Gromovsky AD, Schugar RC, Brown AL, Helsley RN, Burrows AC, Ferguson D, Zhang R, Sansbury BE, Lee RG, Morton RE, Allende DS, Parks JS, Spite M, Brown JM. Delta-5 Fatty Acid Desaturase FADS1 Impacts Metabolic Disease by Balancing Proinflammatory and Proresolving Lipid Mediators. Arterioscler Thromb Vasc Biol. 2018;38:218–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wang Y, Wang W, Wang N, Tall AR, Tabas I. Mitochondrial Oxidative Stress Promotes Atherosclerosis and Neutrophil Extracellular Traps in Aged Mice. Arterioscler Thromb Vasc Biol. 2017;37:e99–e107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Konaniah ES, Kuhel DG, Basford JE, Weintraub NL, Hui DY. Deficiency of LRP1 in Mature Adipocytes Promotes Diet-Induced Inflammation and Atherosclerosis-Brief Report. Arterioscler Thromb Vasc Biol. 2017;37:1046–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bermudez B, Dahl TB, Medina I et al. Leukocyte overexpression of intracellular NAMPT attenuates atherosclerosis by regulating PPARƒÁ-dependent monocyte differentiation and function. Arterioscler Thromb Vasc Biol. 2017;37:1157–1167. [DOI] [PubMed] [Google Scholar]
- 58.Rossignoli A, Shang MM, Gladh H, Moessinger C, Foroughi Asl H, Talukdar HA, Franzen O, Mueller S, Bjorkegren JL, Folestad E, Skogsberg J. Poliovirus Receptor-Related 2: A Cholesterol-Responsive Gene Affecting Atherosclerosis Development by Modulating Leukocyte Migration. Arterioscler Thromb Vasc Biol. 2017;37:534–542. [DOI] [PubMed] [Google Scholar]
- 59.Nicolaou A, Zhao Z, Northoff BH, Sass K, Herbst A, Kohlmaier A, Chalaris A, Wolfrum C, Weber C, Steffens S, Rose-John S, Teupser D, Holdt LM. Adam17 Deficiency Promotes Atherosclerosis by Enhanced TNFR2 Signaling in Mice. Arterioscler Thromb Vasc Biol. 2017;37:247–257. [DOI] [PubMed] [Google Scholar]
- 60.Williams JW, Martel C, Potteaux S, Esaulova E, Ingersoll MA, Elvington A, Saunders BT, Huang LH, Habenicht AJ, Zinselmeyer BH, Randolph GJ. Limited Macrophage Positional Dynamics in Progressing or Regressing Murine Atherosclerotic Plaques-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:1702–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tay C, Liu YH, Kanellakis P, Kallies A, Li Y, Cao A, Hosseini H, Tipping P, Toh BH, Bobik A, Kyaw T. Follicular B Cells Promote Atherosclerosis via T Cell-Mediated Differentiation Into Plasma Cells and Secreting Pathogenic Immunoglobulin G. Arterioscler Thromb Vasc Biol. 2018;38:e71–e84. [DOI] [PubMed] [Google Scholar]
- 62.Jean-Charles PY, Wu JH, Zhang L, Kaur S, Nepliouev I, Stiber JA, Brian L, Qi R, Wertman V, Shenoy SK, Freedman NJ. USP20 (Ubiquitin-Specific Protease 20) Inhibits TNF (Tumor Necrosis Factor)-Triggered Smooth Muscle Cell Inflammation and Attenuates Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:2295–2305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Nguyen MA, Karunakaran D, Geoffrion M, Cheng HS, Tandoc K, Perisic Matic L, Hedin U, Maegdefessel L, Fish JE, Rayner KJ. Extracellular Vesicles Secreted by Atherogenic Macrophages Transfer MicroRNA to Inhibit Cell Migration. Arterioscler Thromb Vasc Biol. 2018;38:49–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Marcovecchio PM, Thomas GD, Mikulski Z, Ehinger E, Mueller KAL, Blatchley A, Wu R, Miller YI, Nguyen AT, Taylor AM, McNamara CA, Ley K, Hedrick CC. Scavenger Receptor CD36 Directs Nonclassical Monocyte Patrolling Along the Endothelium During Early Atherogenesis. Arterioscler Thromb Vasc Biol. 2017;37:2043–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bai L, Li Z, Li Q, Guan H, Zhao S, Liu R, Wang R, Zhang J, Jia Y, Fan J, Wang N, Reddy JK, Shyy JY, Liu E. Mediator 1 Is Atherosclerosis Protective by Regulating Macrophage Polarization. Arterioscler Thromb Vasc Biol. 2017;37:1470–1481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ouimet M, Ediriweera H, Afonso MS, Ramkhelawon B, Singaravelu R, Liao X, Bandler RC, Rahman K, Fisher EA, Rayner KJ, Pezacki JP, Tabas I, Moore KJ. microRNA-33 Regulates Macrophage Autophagy in Atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:1058–1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Getz GS, Reardon CA. Apoprotein E as a lipid transport and signaling protein in the blood, liver, and artery wall. J Lipid Res. 2009;50 Suppl:S156–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Getz GS, Reardon CA. Do the Apoe−/− and Ldlr−/− mice yield the same insight on atherogenesis? Arterioscler Thromb Vasc Biol. 2016;36:1734–1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Horton JD, Cohen JC, Hobbs HH. Molecular biology of PCSK9: its role in LDL metabolism. Trends Biochem Sci. 2007;32:71–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Cunningham D, Danley DE, Geoghegan KF et al. Structural and biophysical studies of PCSK9 and its mutants linked to familial hypercholesterolemia. Nat Struct Mol Biol. 2007;14:413–419. [DOI] [PubMed] [Google Scholar]
- 71.Daugherty A, Tabas I, Rader DJ. Accelerating the pace of atherosclerosis research. Arterioscler Thromb Vasc Biol. 2015;35:11–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.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] [PubMed] [Google Scholar]
- 73.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] [PubMed] [Google Scholar]
- 74.Goettsch C, Hutcheson JD, Hagita S, Rogers MA, Creager MD, Pham T, Choi J, Mlynarchik AK, Pieper B, Kjolby M, Aikawa M, Aikawa E. A single injection of gain-of-function mutant PCSK9 adeno-associated virus vector induces cardiovascular calcification in mice with no genetic modification. Atherosclerosis. 2016;251:109–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lu H, Howatt DA, Balakrishnan A, Graham MJ, Mullick AE, Daugherty A. Hypercholesterolemia induced by a PCSK9 gain-of-function mutation augments angiotensin II-induced abdominal aortic aneurysms in C57BL/6 mice. Arterioscler Thromb Vasc Biol. 2016;36:1753–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Peled M, Nishi H, Weinstock A, Barrett TJ, Zhou F, Quezada A, Fisher EA. A wild-type mouse-based model for the regression of inflammation in atherosclerosis. PLoS One. 2017;12:e0173975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cansby E, Magnusson E, Nunez-Duran E, Amrutkar M, Pedrelli M, Parini P, Hoffmann J, Stahlman M, Howell BW, Marschall HU, Boren J, Mahlapuu M. STK25 regulates cardiovascular disease progression in a mouse model of hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2018;38:1723–1737. [DOI] [PubMed] [Google Scholar]
- 78.Lutshumba J, Liu S, Zhong Y, Hou T, Daugherty A, Lu H, Guo Z, Gong MC. Deletion of BMAL1 in Smooth Muscle Cells Protects Mice From Abdominal Aortic Aneurysms. Arterioscler Thromb Vasc Biol. 2018;38:1063–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wu CH, Mohammadmoradi S, Chen JZ, Sawada H, Daugherty A, Lu HS. Renin-angiotensin system and cardiovascular functions. Arterioscler Thromb Vasc Biol. 2018;38:e108–e116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Jarrett KE, Lee C, De Giorgi M, Hurley A, Gillard BK, Doerfler AM, Li A, Pownall HJ, Bao G, Lagor WR. Somatic editing of Ldlr with adeno-associated viral-CRISPR is an efficient tool for atherosclerosis research. Arterioscler Thromb Vasc Biol. 2018;38:1997–2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Vozenilek AE, Navratil AR, Green JM, Coleman DT, Blackburn CMR, Finney AC, Pearson BH, Chrast R, Finck BN, Klein RL, Orr AW, Woolard MD. Macrophage-Associated Lipin-1 Enzymatic Activity Contributes to Modified Low-Density Lipoprotein-Induced Proinflammatory Signaling and Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:324–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Robinet P, Milewicz DM, Cassis LA, Leeper NJ, Lu HS, Smith JD. Consideration of sex differences in design and reporting of experimental arterial pathology studies-Statement From ATVB Council. Arterioscler Thromb Vasc Biol. 2018;38:292–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Arnold AP, Cassis LA, Eghbali M, Reue K, Sandberg K. Sex hormones and sex chromosomes cause sex differences in the development of cardiovascular diseases. Arterioscler Thromb Vasc Biol. 2017;37:746–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Vozenilek AE, Blackburn CMR, Schilke RM, Chandran S, Castore R, Klein RL, Woolard MD. AAV8-mediated overexpression of mPCSK9 in liver differs between male and female mice. Atherosclerosis. 2018;278:66–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Plump AS, Smith JD, Hayek T, Aaltosetala K, Walsh A, Verstuyft JG, Rubin EM, Breslow JL. Severe hypercholesterolemia and atherosclerosis in apolipoprotein-E-deficient mice created by homologous recombination in ES cells. Cell. 1992;71:343–353. [DOI] [PubMed] [Google Scholar]
- 86.Nakashima Y, Plump AS, Raines EW, Breslow JL, Ross R. ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler Thromb. 1994;14:133–140. [DOI] [PubMed] [Google Scholar]
- 87.Lu H, Wu C, Howatt DA, Balakrishnan A, Moorleghen JJ, Chen X, Zhao M, Graham MJ, Mullick AE, Crooke RM, Feldman DL, Cassis LA, Vander Kooi CW, Daugherty A. Angiotensinogen exerts effects independent of angiotensin II. Arterioscler Thromb Vasc Biol. 2016;36:256–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Samsoondar JP, Burke AC, Sutherland BG, Telford DE, Sawyez CG, Edwards JY, Pinkosky SL, Newton RS, Huff MW. Prevention of Diet-Induced Metabolic Dysregulation, Inflammation, and Atherosclerosis in Ldlr(−/−) Mice by Treatment With the ATP-Citrate Lyase Inhibitor Bempedoic Acid. Arterioscler Thromb Vasc Biol. 2017;37:647–656. [DOI] [PubMed] [Google Scholar]
- 89.Koren O, Spor A, Felin J, Fak F, Stombaugh J, Tremaroli V, Behre CJ, Knight R, Fagerberg B, Ley RE, Backhed F. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci U S A. 2011;108 Suppl 1:4592–4598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Jie Z, Xia H, Zhong SL et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun. 2017;8:845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang Z, Klipfell E, Bennett BJ et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Getz GS, Reardon CA. Diet, Microbes, and Murine Atherosclerosis. Arterioscler Thromb Vasc Biol. 2018;38:2269–2271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Wacker BK, Dronadula N, Bi L, Stamatikos A, Dichek DA. Apo A-I (Apolipoprotein A-I) Vascular Gene Therapy Provides Durable Protection Against Atherosclerosis in Hyperlipidemic Rabbits. Arterioscler Thromb Vasc Biol. 2018;38:206–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Wang C, Nishijima K, Kitajima S, Niimi M, Yan H, Chen Y, Ning B, Matsuhisa F, Liu E, Zhang J, Chen YE, Fan J. Increased Hepatic Expression of Endothelial Lipase Inhibits Cholesterol Diet-Induced Hypercholesterolemia and Atherosclerosis in Transgenic Rabbits. Arterioscler Thromb Vasc Biol. 2017;37:1282–1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Zhang J, Niimi M, Yang D et al. Deficiency of Cholesteryl Ester Transfer Protein Protects Against Atherosclerosis in Rabbits. Arterioscler Thromb Vasc Biol. 2017;37:1068–1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Serbanovic-Canic J, de Luca A, Warboys C et al. Zebrafish Model for Functional Screening of Flow-Responsive Genes. Arterioscler Thromb Vasc Biol. 2017;37:130–143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Burke AC, Telford DE, Sutherland BG, Edwards JY, Sawyez CG, Barrett PHR, Newton RS, Pickering JG, Huff MW. Bempedoic Acid Lowers Low-Density Lipoprotein Cholesterol and Attenuates Atherosclerosis in Low-Density Lipoprotein Receptor-Deficient (LDLR(+/−) and LDLR(−/−)) Yucatan Miniature Pigs. Arterioscler Thromb Vasc Biol. 2018;38:1178–1190. [DOI] [PubMed] [Google Scholar]
- 98.Henig I, Zuckerman T. Hematopoietic stem cell transplantation-50 years of evolution and future perspectives. Rambam Maimonides Med J. 2014;5:e0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Sauer B Functional expression of the cre-lox site-specific recombination system in the yeast Saccharomyces cerevisiae. Mol Cell Biol. 1987;7:2087–2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lu H, Rateri DL, Feldman DL, Charnigo RJ Jr, Fukamizu A, Ishida J, Oesterling EG, Cassis LA, Daugherty A. Renin inhibition reduces hypercholesterolemia-induced atherosclerosis in mice. J Clin Invest. 2008;118:984–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Linton MF, Atkinson JB, Fazio S. Prevention of atherosclerosis in apolipoprotein E-deficient mice by bone marrow transplantation. Science. 1995;267:1034–1037. [DOI] [PubMed] [Google Scholar]
- 102.Boisvert WA, Spangenberg J, Curtiss LK. Role of leukocyte-specific LDL receptors on plasma lipoprotein cholesterol and atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 1997;17:340–347. [DOI] [PubMed] [Google Scholar]
- 103.Fazio S, Babaev VR, Burleigh ME, Major AS, Hasty AH, Linton MF. Physiological expression of macrophage apoE in the artery wall reduces atherosclerosis in severely hyperlipidemic mice. J Lipid Res. 2002;43:1602–1609. [DOI] [PubMed] [Google Scholar]
- 104.Abremski K, Wierzbicki A, Frommer B, Hoess RH. Bacteriophage P1 Cre-loxP site-specific recombination. Site-specific DNA topoisomerase activity of the Cre recombination protein. J Biol Chem. 1986;261:391–396. [PubMed] [Google Scholar]
- 105.Orban PC, Chui D, Marth JD. Tissue- and site-specific DNA recombination in transgenic mice. Proc. Natl. Acad. Sci. USA. 1992;89:6861–6865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Clausen BE, Burkhardt C, Reith W, Renkawitz R, Forster I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 1999;8:265–277. [DOI] [PubMed] [Google Scholar]
- 107.Baigent C, Keech A, Kearney PM, Blackwell L, Buck G, Pollicino C, Kirby A, Sourjina T, Peto R, Collins R, Simes R. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366:1267–1278. [DOI] [PubMed] [Google Scholar]
- 108.Chan DC, Watts GF, Somaratne R, Wasserman SM, Scott R, Barrett PHR. Comparative Effects of PCSK9 (Proprotein Convertase Subtilisin/Kexin Type 9) Inhibition and Statins on Postprandial Triglyceride-Rich Lipoprotein Metabolism. Arterioscler Thromb Vasc Biol. 2018;38:1644–1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Labos C, Brophy JM, Smith GD, Sniderman AD, Thanassoulis G. Evaluation of the Pleiotropic Effects of Statins: A Reanalysis of the Randomized Trial Evidence Using Egger Regression-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:262–265. [DOI] [PubMed] [Google Scholar]
- 110.Ouweneel AB, van der Sluis RJ, Nahon JE, Van Eck M, Hoekstra M. Simvastatin treatment aggravates the glucocorticoid insufficiency associated with hypocholesterolemia in mice. Atherosclerosis. 2017;261:99–104. [DOI] [PubMed] [Google Scholar]
- 111.Fukuda D, Enomoto S, Nagai R, Sata M. Inhibition of renin-angiotensin system attenuates periadventitial inflammation and reduces atherosclerotic lesion formation. Biomed Pharmacother. 2009; 63:754–761. [DOI] [PubMed] [Google Scholar]
- 112.Wang JA, Chen WA, Wang Y, Zhang S, Bi H, Hong B, Luo Y, Daugherty A, Xie X. Statins exert differential effects on angiotensin II-induced atherosclerosis, but no benefit for abdominal aortic aneurysms. Atherosclerosis. 2011;217:90–6. [DOI] [PubMed] [Google Scholar]
- 113.Werner N, Priller J, Laufs U, Endres M, Bohm M, Dirnagl U, Nickenig G. Bone marrow-derived progenitor cells modulate vascular reendothelialization and neointimal formation - Effect of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition. Arterioscler Thromb Vasc Biol. 2002;22:1567–1572. [DOI] [PubMed] [Google Scholar]
- 114.Aprahamian T, Bonegio R, Rizzo J, Perlman H, Lefer DJ, Rifkin IR, Walsh K. Simvastatin treatment ameliorates autoimmune disease associated with accelerated atherosclerosis in a murine lupus model. J Immunol. 2006;177:3028–3034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Calkin AC, Giunti S, Sheehy KJ, Chew C, Boolell V, Rajaram YS, Cooper ME, Jandeleit-Dahm KA. The HMG-CoA reductase inhibitor rosuvastatin and the angiotensin receptor antagonist candesartan attenuate atherosclerosis in an apolipoprotein E-deficient mouse model of diabetes via effects on advanced glycation, oxidative stress and inflammation. Diabetologia. 2008;51:1731–1740. [DOI] [PubMed] [Google Scholar]
- 116.Boulanger CM. Highlight on Endothelial Activation and Beyond. Arterioscler Thromb Vasc Biol. 2018;38:e198–e201. [DOI] [PubMed] [Google Scholar]
- 117.Ventura A, Kirsch DG, McLaughlin ME, Tuveson DA, Grimm J, Lintault L, Newman J, Reczek EE, Weissleder R, Jacks T. Restoration of p53 function leads to tumour regression in vivo. Nature. 2007;445:661–665. [DOI] [PubMed] [Google Scholar]
- 118.Zhang J, Zhong W, Cui T, Yang M, Hu X, Xu K, Xie C, Xue C, Gibbons GH, Liu C, Li L, Chen YE. Generation of an adult smooth muscle cell-targeted Cre recombinase mouse model. Arterioscler Thromb Vasc Biol. 2006;26:e23–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Kong X, Banks A, Liu T et al. IRF4 is a key thermogenic transcriptional partner of PGC-1alpha. Cell. 2014;158:69–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hofmann SM, Zhou L, Perez-Tilve D, Greer T, Grant E, Wancata L, Thomas A, Pfluger PT, Basford JE, Gilham D, Herz J, Tschop MH, Hui DY. Adipocyte LDL receptor-related protein-1 expression modulates postprandial lipid transport and glucose homeostasis in mice. J Clin Invest. 2007;117:3271–3282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kuwabara JT, Tallquist MD. Tracking Adventitial Fibroblast Contribution to Disease: A Review of Current Methods to Identify Resident Fibroblasts. Arterioscler Thromb Vasc Biol. 2017;37:1598–1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Payne S, De Val S, Neal A. Endothelial-Specific Cre Mouse Models. Arterioscler Thromb Vasc Biol. 2018;38:2550–2561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Chakraborty R, Saddouk FZ, Carrao AC, Krause DS, Greif DM, Martin KA. Promoters to study vascular smooth muscle. Arterioscler Thromb Vasc Biol. 2019; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Xie Y, Jin H, Zeng M, Li D. Coronary Artery Plaque Imaging. Curr Atheroscler Rep. 2017;19:37. [DOI] [PubMed] [Google Scholar]
- 125.Ripa RS, Pedersen SF, Kjaer A. PET/MR Imaging in Vascular Disease: Atherosclerosis and Inflammation. PET Clin. 2016;11:479–488. [DOI] [PubMed] [Google Scholar]
- 126.Watase H, Sun J, Hippe DS, Balu N, Li F, Zhao X, Mani V, Fayad ZA, Fuster V, Hatsukami TS, Yuan C. Carotid Artery Remodeling Is Segment Specific: An In Vivo Study by Vessel Wall Magnetic Resonance Imaging. Arterioscler Thromb Vasc Biol. 2018;38:927–934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Chai JT, Ruparelia N, Goel A, Kyriakou T, Biasiolli L, Edgar L, Handa A, Farrall M, Watkins H, Choudhury RP. Differential Gene Expression in Macrophages From Human Atherosclerotic Plaques Shows Convergence on Pathways Implicated by Genome-Wide Association Study Risk Variants. Arterioscler Thromb Vasc Biol. 2018;38:2718–2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lin R, Chen S, Liu G, Xue Y, Zhao X. Association Between Carotid Atherosclerotic Plaque Calcification and Intraplaque Hemorrhage: A Magnetic Resonance Imaging Study. Arterioscler Thromb Vasc Biol. 2017;37:1228–1233. [DOI] [PubMed] [Google Scholar]
- 129.Wang X, Peter K. Molecular Imaging of Atherothrombotic Diseases: Seeing Is Believing. Arterioscler Thromb Vasc Biol. 2017;37:1029–1040. [DOI] [PubMed] [Google Scholar]
- 130.Kratofil RM, Kubes P, Deniset JF. Monocyte Conversion During Inflammation and Injury. Arterioscler Thromb Vasc Biol. 2017;37:35–42. [DOI] [PubMed] [Google Scholar]
- 131.Mallat Z. Macrophages. Arterioscler Thromb Vasc Biol. 2017;37:e92–98. [DOI] [PubMed] [Google Scholar]
- 132.Guo L, Harari E, Virmani R, Finn AV. Linking Hemorrhage, Angiogenesis, Macrophages, and Iron Metabolism in Atherosclerotic Vascular Diseases. Arterioscler Thromb Vasc Biol. 2017;37:e33–e39. [DOI] [PubMed] [Google Scholar]
- 133.Ye F, Wang JA, Daugherty A, Lu H. Macrophage-mediated mechanisms in atherosclerosis: still tangled. Curr Opin Lipidol. 2017;28:286–287. [DOI] [PubMed] [Google Scholar]
- 134.Bendall SC, Nolan GP, Roederer M, Chattopadhyay PK. A deep profiler’s guide to cytometry. Trends Immunol. 2012;33:323–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Thomas GD, Hamers AAJ, Nakao C, Marcovecchio P, Taylor AM, McSkimming C, Nguyen AT, McNamara CA, Hedrick CC. Human Blood Monocyte Subsets: A New Gating Strategy Defined Using Cell Surface Markers Identified by Mass Cytometry. Arterioscler Thromb Vasc Biol. 2017;37:1548–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Li W, Luehmann HP, Hsiao HM, Tanaka S, Higashikubo R, Gauthier JM, Sultan D, Lavine KJ, Brody SL, Gelman AE, Gropler RJ, Liu Y, Kreisel D. Visualization of monocytic cells in regressing atherosclerotic plaques by intravital 2-Photon and positron emission tomography-based imaging-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:1030–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Keliher EJ, Ye YX, Wojtkiewicz GR et al. Polyglucose nanoparticles with renal elimination and macrophage avidity facilitate PET imaging in ischaemic heart disease. Nat Commun. 2017;8:14064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Fayad ZA, Mani V, Woodward M, Kallend D, Abt M, Burgess T, Fuster V, Ballantyne CM, Stein EA, Tardif JC, Rudd JH, Farkouh ME, Tawakol A. Safety and efficacy of dalcetrapib on atherosclerotic disease using novel non-invasive multimodality imaging (dal-PLAQUE): a randomised clinical trial. Lancet. 2011;378:1547–1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Dweck MR, Aikawa E, Newby DE, Tarkin JM, Rudd JH, Narula J, Fayad ZA. Noninvasive Molecular Imaging of Disease Activity in Atherosclerosis. Circ Res. 2016;119:330–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Tawakol A, Singh P, Mojena M et al. HIF-1alpha and PFKFB3 Mediate a Tight Relationship Between Proinflammatory Activation and Anerobic Metabolism in Atherosclerotic Macrophages. Arterioscler Thromb Vasc Biol. 2015;35:1463–1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Tavakoli S, Vashist A, Sadeghi MM. Molecular imaging of plaque vulnerability. J Nucl Cardiol. 2014;21:1112–1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Tavakoli S, Downs K, Short JD, Nguyen HN, Lai Y, Jerabek PA, Goins B, Toczek J, Sadeghi MM, Asmis R. Characterization of Macrophage Polarization States Using Combined Measurement of 2-Deoxyglucose and Glutamine Accumulation: Implications for Imaging of Atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:1840–1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Stabley JN, Towler DA. Arterial Calcification in Diabetes Mellitus: Preclinical Models and Translational Implications. Arterioscler Thromb Vasc Biol. 2017;37:205–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Goettsch C, Kjolby M, Aikawa E. Sortilin and Its Multiple Roles in Cardiovascular and Metabolic Diseases. Arterioscler Thromb Vasc Biol. 2018;38:19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Yahagi K, Kolodgie FD, Lutter C, Mori H, Romero ME, Finn AV, Virmani R. Pathology of Human Coronary and Carotid Artery Atherosclerosis and Vascular Calcification in Diabetes Mellitus. Arterioscler Thromb Vasc Biol. 2017;37:191–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Hodroge A, Trécherel E, Cornu M et al. Oligogalacturonic Acid Inhibits Vascular Calcification by Two Mechanisms: Inhibition of Vascular Smooth Muscle Cell Osteogenic Conversion and Interaction With Collagen. Arterioscler Thromb Vasc Biol. 2017;37:1391–1401. [DOI] [PubMed] [Google Scholar]
- 147.Lino M, Wan MH, Rocca AS, Ngai D, Shobeiri N, Hou G, Ge C, Franceschi RT, Bendeck MP. Diabetic Vascular Calcification Mediated by the Collagen Receptor Discoidin Domain Receptor 1 via the Phosphoinositide 3-Kinase/Akt/Runt-Related Transcription Factor 2 Signaling Axis. Arterioscler Thromb Vasc Biol. 2018;38:1878–1889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Thomas IC, Thompson CA, Yang M, Allison MA, Forbang NI, Michos ED, McClelland RL, Budoff MJ, Criqui MH. Thoracic Aorta Calcification and Noncardiovascular Disease-Related Mortality. Arterioscler Thromb Vasc Biol. 2018;38:1926–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Liu L, Zeng P, Yang X et al. Inhibition of Vascular Calcification. Arterioscler Thromb Vasc Biol. 2018;38:2382–2395. [DOI] [PubMed] [Google Scholar]
- 150.Zaheer A, Lenkinski RE, Mahmood A, Jones AG, Cantley LC, Frangioni JV. In vivo near-infrared fluorescence imaging of osteoblastic activity. Nat Biotechnol. 2001;19:1148–1154. [DOI] [PubMed] [Google Scholar]
- 151.Zaheer A, Murshed M, De Grand AM, Morgan TG, Karsenty G, Frangioni JV. Optical imaging of hydroxyapatite in the calcified vasculature of transgenic animals. Arterioscler Thromb Vasc Biol. 2006;26:1132–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Curaj A, Wu Z, Fokong S, Liehn EA, Weber C, Burlacu A, Lammers T, van Zandvoort M, Kiessling F. Noninvasive molecular ultrasound monitoring of vessel healing after intravascular surgical procedures in a preclinical setup. Arterioscler Thromb Vasc Biol. 2015;35:1366–1373. [DOI] [PubMed] [Google Scholar]
- 153.Curaj A, Wu Z, Rix A, Gresch O, Sternkopf M, Alampour-Rajabi S, Lammers T, van Zandvoort M, Weber C, Koenen RR, Liehn EA, Kiessling F. Molecular Ultrasound Imaging of Junctional Adhesion Molecule A Depicts Acute Alterations in Blood Flow and Early Endothelial Dysregulation. Arterioscler Thromb Vasc Biol. 2018;38:40–48. [DOI] [PubMed] [Google Scholar]
- 154.van Leeuwen M, Gijbels MJ, Duijvestijn A, Smook M, van de Gaar MJ, Heeringa P, de Winther MP, Tervaert JW. Accumulation of myeloperoxidase-positive neutrophils in atherosclerotic lesions in LDLR−/− mice. Arterioscler Thromb Vasc Biol. 2008;28:84–89. [DOI] [PubMed] [Google Scholar]
- 155.Naruko T, Ueda M, Haze K et al. Neutrophil infiltration of culprit lesions in acute coronary syndromes. Circulation. 2002;106:2894–2900. [DOI] [PubMed] [Google Scholar]
- 156.Rotzius P, Thams S, Soehnlein O, Kenne E, Tseng CN, Bjorkstrom NK, Malmberg KJ, Lindbom L, Eriksson EE. Distinct infiltration of neutrophils in lesion shoulders in ApoE−/− mice. Am J Pathol. 2010;177:493–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Glinzer A, Ma X, Prakash J et al. Targeting Elastase for Molecular Imaging of Early Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol. 2017;37:525–533. [DOI] [PubMed] [Google Scholar]
- 158.Rouwet E, Lutgens E. 2016 Jeffrey M. Hoeg Award Lecture. Arterioscler Thromb Vasc Biol. 2018;38:1678–1688. [DOI] [PubMed] [Google Scholar]
- 159.Ley K, Gerdes N, Winkels H. ATVB Distinguished Scientist Award: How Costimulatory and Coinhibitory Pathways Shape Atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:764–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Tabas I 2016 Russell Ross Memorial Lecture in Vascular Biology: Molecular-Cellular Mechanisms in the Progression of Atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:183–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Di Bartolo BA, Psaltis PJ, Bursill CA, Nicholls SJ. Translating Evidence of HDL and Plaque Regression. Arterioscler Thromb Vasc Biol. 2018;38:1961–1968. [DOI] [PubMed] [Google Scholar]
- 162.Sacks FM, Jensen MK. From High-Density Lipoprotein Cholesterol to Measurements of Function: Prospects for the Development of Tests for High-Density Lipoprotein Functionality in Cardiovascular Disease. Arterioscler Thromb Vasc Biol. 2018;38:487–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Dron JS, Lazarte J, Hegele RA. Recent Highlights of ATVB: Lipoproteins. Arterioscler Thromb Vasc Biol. 2018;38:e185–e197. [DOI] [PubMed] [Google Scholar]
- 164.El Khoudary SR, Ceponiene I, Samargandy S, Stein JH, Li D, Tattersall MC, Budoff MJ. HDL (High-Density Lipoprotein) Metrics and Atherosclerotic Risk in Women. Arterioscler Thromb Vasc Biol. 2018;38:2236–2244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wilson JM, Nguyen AT, Schuyler AJ, Commins SP, Taylor AM, Platts-Mills TAE, McNamara CA. IgE to the Mammalian Oligosaccharide Galactose-alpha-1,3-Galactose Is Associated With Increased Atheroma Volume and Plaques With Unstable Characteristics-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:1665–1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Zylla S, Dorr M, Volzke H, Schminke U, Felix SB, Nauck M, Friedrich N. Association of Circulating Chemerin With Subclinical Parameters of Atherosclerosis: Results of a Population-Based Study. Arterioscler Thromb Vasc Biol. 2018;38:1656–1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Akishima-Fukasawa Y, Ishikawa Y, Mikami T, Akasaka Y, Ishii T. Settlement of Stenotic Site and Enhancement of Risk Factor Load for Atherosclerosis in Left Anterior Descending Coronary Artery by Myocardial Bridge. Arterioscler Thromb Vasc Biol. 2018;38:1407–1414. [DOI] [PubMed] [Google Scholar]
- 168.Hertle E, Arts ICW, van der Kallen CJH, Feskens EJM, Schalkwijk CG, Stehouwer CDA, van Greevenbroek MMJ. Classical Pathway of Complement Activation: Longitudinal Associations of C1q and C1-INH With Cardiovascular Outcomes: The CODAM Study (Cohort on Diabetes and Atherosclerosis Maastricht)-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:1242–1244. [DOI] [PubMed] [Google Scholar]
- 169.Li Y, Wang DW, Chen Y, Chen C, Guo J, Zhang Seal. Genome-wide association and functional studies identify SCML4 and THSD7A as novel susceptibility genes for coronary artery disease. Arterioscler Thromb Vasc Biol. 2018;38:964–975. [DOI] [PubMed] [Google Scholar]
- 170.Gille A, D’Andrea D, Tortorici MA, Hartel G, Wright SD. CSL112 (Apolipoprotein A-I. Arterioscler Thromb Vasc Biol. 2018;38:953–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Steffen BT, Duprez D, Bertoni AG, Guan W, Tsai MY. Lp(a). Arterioscler Thromb Vasc Biol. 2018;38:2498–2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Hippe DS, Phan BAP, Sun J, Isquith DA, O’Brien KD, Crouse JR, Anderson T, Huston J, Marcovina SM, Hatsukami TS, Yuan C, Zhao XQ. Lp(a) (Lipoprotein(a)) Levels Predict Progression of Carotid Atherosclerosis in Subjects With Atherosclerotic Cardiovascular Disease on Intensive Lipid Therapy: An Analysis of the AIM-HIGH (Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides: Impact on Global Health Outcomes) Carotid Magnetic Resonance Imaging Substudy-Brief Report. Arterioscler Thromb Vasc Biol. 2018;38:673–678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Amor AJ, Ortega E, Perea V, Cofan M, Sala-Vila A, Nunez I, Gilabert R, Ros E. Relationship Between Total Serum Bilirubin Levels and Carotid and Femoral Atherosclerosis in Familial Dyslipidemia. Arterioscler Thromb Vasc Biol. 2017;37:2356–2363. [DOI] [PubMed] [Google Scholar]
- 174.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. Arterioscler Thromb Vasc Biol. 2017;37:2364–2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Safouris A, Krogias C, Sharma VK et al. Statin Pretreatment and Microembolic Signals in Large Artery Atherosclerosis. Arterioscler Thromb Vasc Biol. 2017;37:1415–1422. [DOI] [PubMed] [Google Scholar]
- 176.Prasad A, Clopton P, Ayers C, Khera A, de Lemos JA, Witztum JL, Tsimikas S. Relationship of Autoantibodies to MDA-LDL and ApoB-Immune Complexes to Sex, Ethnicity, Subclinical Atherosclerosis, and Cardiovascular Events. Arterioscler Thromb Vasc Biol. 2017;37:1213–1221. [DOI] [PMC free article] [PubMed] [Google Scholar]

