Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Mar 25.
Published in final edited form as: Curr Opin Rheumatol. 2013 Sep;25(5):597–605. doi: 10.1097/BOR.0b013e328363eba3

Cardiovascular disease in lupus: insights and updates

Jason S Knight 1, Mariana J Kaplan 1
PMCID: PMC3964608  NIHMSID: NIHMS535872  PMID: 23846339

Abstract

Purpose of review

With improved management of the classical disease manifestations of systemic lupus erythematosus (SLE), cardiovascular disease (CVD) has emerged as one of the most important causes of morbidity and mortality. This review in particular focuses on progress over the past year in clinical and basic aspects of SLE-driven accelerated atherosclerosis.

Recent findings

Both subclinical CVD and CV events continue to be recognized at increased frequency in previously unstudied lupus cohorts and populations. Novel associations have been identified between lupus CVD and cognitive impairment, depression, and low income status. In terms of pathogenesis, there is an ever-increasing focus on the innate immune system and, in particular, type I interferons (IFNs). Recent studies have drawn connections in both human and murine models between neutrophils, plasmacytoid dendritic cells, type I IFNs, and endothelial dysfunction. Whether treatments such as mycophenolate mofetil or statins have a role in prevention of lupus CVD is an area of intensive study.

Summary

CVD is a major complication of lupus and is now a leading cause of death among people living with this disease. As such, additional studies are needed in order to identify the most effective preventive strategies and most predictive vascular risk biomarkers. Type I IFNs may play a critical role in lupus CVD pathogenesis, and it is recommended that vascular outcomes be included in ongoing trials testing the efficacy of anti-IFN biologics.

Keywords: systemic lupus erythematosus, cardiovascular disease, atherosclerosis, type I interferons, neutrophil extracellular traps

INTRODUCTION

The prevalence of premature CVD, especially in young women with SLE, is striking, and may be as high as 50-fold depending on the study and outcome measure [1,2]. A significant percentage of patients with SLE have evidence of subclinical vascular disease, such as increased carotid intima-media thickness (CIMT) [1,3,4] and myocardial perfusion abnormalities [5], findings which are not explained by traditional risk factors [4,6]. While all-cause mortality in SLE has improved significantly with improved monitoring and immunosuppressive treatments, CVD remains a leading cause of death [7]. The interplay between SLE and CVD is a rapidly expanding area of study and has recently been comprehensively reviewed [8,9]. Here, we will focus on those areas that have seen substantial progress over the past year, primarily focusing on the clinical and basic science of CVD attributable to accelerated atherosclerosis.

EPIDEMIOLOGY, RISK FACTORS, AND BIOMARKERS

Investigators of lupus CVD have characterized novel populations, correlations, and biomarkers within the past year.

Epidemiology of CVD in SLE

A nationwide study from Sweden considered whether hospitalization for immune-mediated diseases such as SLE and rheumatoid arthritis was predictive of a subsequent hospitalization for coronary artery disease (CAD). Indeed, the standardized incidence ratio (SIR) for a CAD-related hospitalization in lupus patients was 4.94, making SLE (albeit the subset of patients with enough disease activity to require hospitalization) the second highest-risk condition of 32 immune-mediated diseases considered [10]*. A recent population survey considered death certificates for lupus patients in São Paulo, Brazil, and found that renal failure and infectious diseases were still the most frequent causes of death [11]; the authors contrasted this to the higher proportion of deaths attributable to CVD in North American and European countries. Another interesting study assessed patients with at least two years of serologically active but clinically quiescent (SACQ) SLE [12]**. Treatment with antimalarials was permitted, but corticosteroid and immunosuppressive medications were not. SACQ patients acquired less lupus-related damage after three and 10 years, including renal damage in just 3.6% of SACQ patients versus 23.6% of clinically-active patients at 10 years [12]**. There was also a strong trend toward a reduction in new coronary events (1.8% vs. 7.3%; p=0.06) after 10 years [12]**.

A final group of epidemiologic studies in 2012 drew our attention to patient-centric associations that we might less commonly consider in lupus CVD. One longitudinal study asked whether CV risk factors and outcomes were associated with cognitive impairment [13]. Indeed, the prevalence of cognitive impairment was 15% in lupus patients with CVD, correlating with history of stroke, hypertension, and antiphospholipid (aPL) Abs [13]. A separate cross-sectional analysis considered the association between depression and subclinical CVD in lupus patients, and found a significant odds ratio of 3.85 in a multivariable model regarding the ability of depression to predict CVD [14]. Finally, in white, but not African American, patients with SLE, low income increased the risk of myocardial infarction (MI) and stroke, with significant odds ratios of 3.24 and 2.85, respectively [15]*.

Risk factors

Historically, lupus activity, disease duration, and corticosteroid use have emerged as the most reproducible nontraditional risk factors for CVD in lupus patients; these observations have been supported by recent analyses [1618]. Lupus patients also have a higher burden of traditional risk factors [19,20] as compared to patients without lupus, with one study suggesting a particular role for smoking as a predictor of CV mortality [21].

The genetics of CVD in lupus patients has yet to be extensively assessed, although a recent study asked whether single nucleotide polymorphisms (SNPs) associated with thrombosis in the general population might also predict thrombosis in lupus [22]. In a large lupus cohort, 23% experienced a thrombotic event, and predictive SNPs were identified in genes that included factor V and methylenetetrahydrofolate reductase [22]. Circulating levels of both matrix metalloproteinase (MMP)-2 and MMP-9 have previously been reported as elevated in lupus patients [23]. Given the role of MMP-2 in the breakdown of the endothelial basement membrane, along with its recognized role in CVD [24], a recent study asked whether a MMP-2 promoter polymorphism that affects circulating protein levels would predict risk of CVD in an Iranian cohort of lupus patients [25]. Indeed, the promoter genotype associated with higher MMP-2 activity was associated with both lupus CVD and a higher-risk cholesterol profile [25]. Such genetic analyses will almost surely be applied with increasing frequency to lupus CVD in the future.

Biomarkers

The role of autoAbs in the accelerated atherosclerosis of SLE is a complex topic, without a clear consensus [26]. Antiphospholipid (aPL) Abs are frequently found in patients with SLE and have been considered as possible predictors of atherosclerosis, albeit with mixed results [2731]; a recent review of the topic did suggest a significant increase in the subclinical marker CIMT in patients with primary aPL Ab syndrome as compared to controls [32]. Other autoAbs with suggested roles in lupus CVD are directed against apolipoprotein A1 [33,34], high-density lipoprotein (HDL) [34], and heat-shock protein 60 [35].

Some updates from 2012 include the description of aPL Abs as stimulators of tissue factor (TF) expression on the surface of lupus peripheral blood mononuclear cells, correlating with risk of peripheral vascular disease [36]*. Given the postulated role for TF in atherogenesis [37], as well as in aPL Ab-induced thrombosis [38,39], the authors speculate that TF may play a mechanistic role in lupus CVD [36]*.

HDL normally serves a protective role in atherosclerosis, mediating reverse cholesterol transport and protecting LDL from oxidation. However, in SLE, so-called proinflammatory HDL (piHDL) can be detected in as many as 45% of patients, correlating with increased oxLDL formation, carotid plaque, and CIMT [40,41]. Further, a piHDL phenotype, including the detection of oxidized forms of HDL, has been appreciated in murine models of SLE [42,43]**, where treatment with apoA1 mimetics has shown promise [44]. In general, interplay between oxidative stress and lupus CVD will surely receive increased attention in the coming years [4547].

In addition to autoAb and lipid profiles, some novel biomarkers of CVD in SLE have recently been considered. In a prospective cohort study, both high-sensitivity C-reactive protein (hsCRP) and higher 10-year Framingham risk scores were predictive of future CV events (MI or angina) in lupus patients, with hsCRP levels greater than 1.6 mg/liter giving a statistically-significant hazard ratio of 3.37 [48]. The authors argue for measuring hsCRP to predict CVD risk, as one might do for selected patients in the general population [48]. Defensins activate monocyte-lineage cells to release proinflammatory cytokines, while also serving as chemokines and regulating activation of the complement cascade [49,50]. Both defensin-specific autoAbs [51] and elevated circulating defensin levels [52,53] have been described in SLE. A recent report showed that levels of human beta defensin 2 (hBD2) and human neutrophil peptide (HNP) were predictive of future CV events, as well as progression of subclinical disease [54]*. This result is especially interesting given the presence of defensins in neutrophil extracellular traps (NETs), which will be discussed in more detail below, and which we have speculated on previously with regards to CVD [55]. In general, none of the biomarkers described to date clearly differentiate CVD risk from lupus disease activity, and this will surely be a focus of future clinical studies

PATHOGENESIS: THE CRITICAL ROLE OF INNATE IMMUNITY

Although T and B cells are indispensable for lupus pathogenesis, they have not been definitively linked to lupus CVD. In contrast, links between innate immunity, SLE, and CVD are myriad. In our opinion, the most compelling of these suggests a critical role for type I interferons (IFNs) (Table 1). Recently described roles for cellular mediators of innate immunity such as neutrophils and platelets also exist.

Table 1.

Impact of type I IFNs on the vasculature and atherosclerotic lesions

Endothelial cells (ECs)
  Increased EC apoptosis [3]
  Decreased levels and function of reparative EC progenitors [43,5661]
  Inhibition of proangiogenic pathways, including VEGF and IL-1β [58]
  Upregulation of IL-18 which interferes with normal EC progenitor differentiation [62]
Macrophages
  Increased macrophage recruitment into atherosclerotic lesions [43,63,64]
  Upregulation of TLR4, with increased TNF-α, IL-12, and MMP-9 production [65]
  Upregulation of scavenger receptor A, thereby priming for foam cell formation [66]
T cells
  Increased T cell recruitment into atherosclerotic lesions [43]
  Promotion of T cell-mediated smooth muscle cell death and plaque instability [67]

Type I IFNs

Type I IFNs (including 15 subtypes of IFN-α, one IFN-β, and several other less-studied forms) share a single receptor, and represent an important group of cytokines in host defense, especially against viral infection [68]. Type I IFNs have received considerable attention as mediators of lupus pathogenesis in human and murine systems [43,6973], and drugs targeting type I IFNs are currently in clinical trials for management of SLE in humans [74,75]. Plasmacytoid dendritic cells (pDCs) are well-recognized secretors of IFN-α in SLE [76], while myeloid-lineage cells may also play a role in type I IFN production [77]. The antiangiogenic properties of type I IFNs have been recognized for decades in the cancer literature [78], but investigators have only more recently considered their role in lupus CVD (Table 1), where endothelial injury, and failed repair, during lupus disease flares could lead to initiation and expansion of vascular lesions, ultimately challenging endothelial integrity and predisposing to atheroma formation.

Lessons from animal models

With the exception of the hypercholesterolemia-inducing knockout models of apolipoprotein E [apoE(−/−)] and the LDL receptor [LDLr(−/−)], mice do not develop atherosclerotic lesions a priori [79]. Therefore, in the absence of spontaneous atherosclerosis in lupus models, investigators have either crossed lupus-prone genotypes with the aforementioned proatherogenic backgrounds, or have focused on subclinical markers of endothelial dysfunction. The combined lupus/atherosclerosis models have generally shown acceleration of both the lupus and atherosclerosis phenotypes [8084], although the role of type I IFNs in these lupus/atherosclerosis models has not been examined.

In an interesting 2012 study, LDLr(−/−) mice were crossed with the lupus-prone strain Sle16 to create a new lupus/atherosclerosis model [85]**. Atherosclerosis was again accelerated by the lupus phenotype, while hypercholesterolemia enhanced both the nephritis score and C3 deposition in kidneys [85]**. Intriguingly, despite more C3 in the kidneys, there was less deposition in atherosclerotic lesions [85]**. Further, lesional C3 deposition was inversely correlated with the number of apoptotic cells, arguing that complement depletion by active lupus may prevent complement-mediated repair of the damaged endothelium and developing plaque [85]**. These observations are reminiscent of some of the aforementioned lupus/atherosclerosis models that also demonstrated accumulation of apoptotic debris [80,82], albeit without considering the role of complement.

In another paper from 2012, the role of type I IFNs in the subclinical vascular dysfunction of lupus-prone mice was assessed [43]**. Knockout of the type I IFN receptor in the lupus-prone model NZM2328 improved endothelium-dependent vasorelaxation, EPC numbers and function, and in vivo neoangiogenesis; these effects were independent of sex (male mice do not typically develop nephritis or high-titer anti-dsDNA Abs in this model) arguing against a global impact of disease activity [43]**. Further, acute administration of IFN-α worsened both endothelium-dependent vasorelaxation and EPC function in lupus-prone, as well as non-lupus-prone, mice [43]**. In the same 2012 study, the impact of type I IFNs on apoE(−/−) mice (without lupus predisposition) was also considered [43]**. Knockout of the type I IFN receptor resulted in decreased atherosclerosis severity, with decreased recruitment of F4/80-positive macrophages and CD3-positive T cells into lesions [43]**. These observations are substantiated by another recent study in which IFN-β administration to apoE(−/−) and LDLr(−/−) mice resulted in accelerated atherosclerosis as well as increased macrophage recruitment into lesions [63]. Further, specific blockade of type I IFN signaling in myeloid cells inhibited atherosclerosis and protected against accumulation of macrophages [63].

Additionally, several recent studies have considered the role of type I IFN-producing pDCs in the atherosclerotic lesions of non-lupus-prone apoE(−/−) mice. Although splenic and aortic pDC numbers did not differ between control and apoE(−/−) mice, pDCs from apoE(−/−) mice had a more activated phenotype [64]*. Further, when pDCs were depleted by monoclonal Ab treatment, atherosclerotic plaque area was significantly reduced, as was macrophage recruitment [64]*. In terms of pDC stimulation, another study considered the possibility that protein-DNA complexes derived from neutrophil extracellular traps (NETs) might activate pDCs and thereby accelerate atherosclerosis [86]**. As will be discussed in more detail below, NETs—consisting of chromatin, the cathelicidin-LL37 peptide, and other proteins derived from azurophilic granules—are potent stimulators of pDCs in various lupus models [8789]. Indeed, both NETs and the murine orthologue of cathelicidin-LL37, CRAMP, were detected at the luminal surface of atherosclerotic arteries, while depletion of either pDCs or CRAMP protected against atherosclerosis and reduced anti-double-stranded DNA (dsDNA) titers [86]**. A final relevant study in 2012 considered the role of CRAMP in apoE(−/−) atherogenesis by focusing in particular on early lesions [90]. CRAMP derived from neutrophils was found especially along the luminal surface in these early lesions, where it was sufficient to enhance monocyte adhesion [90]. Although all these studies were performed in models without a predisposition toward the lupus phenotype, the intriguing link between CRAMP/NETs, pDC activation, and vascular damage seem highly relevant to lupus vasculopathy.

Neutrophils

Although historically receiving relatively little attention in lupus pathogenesis, neutrophils have emerged in the past five years as potentially indispensable players [91]. In 2004, a new form of programmed cell death, NET formation, was described [92]. As above, NETs represent extracellular chromatin decorated with neutrophil-derived proteins, and presumably function to trap and kill pathogens [93]. A subset of lupus patients have an impaired ability to degrade NETs [94,95]. Lupus neutrophils are also predisposed to enhanced NET release, with stimulatory roles for both type I IFNs and autoAbs such as anti-cathelicidin-LL37 and anti-RNP [8789]. Further, our group has suggested that a proinflammatory subset of lupus neutrophils termed low density granulocytes (LDGs) are especially primed for NET release, with LDG-derived NETs capable of endothelial damage in vitro [89]. Our group has also shown that NETs, and especially cathelicin-LL37, can activate the inflammasome in macrophages, resulting in an activated, and potentially pro-atherogenic, phenotype [96].

In other disease models [97,98], including atherosclerosis [90,99], NETs have emerged as mediators of vascular damage, and are also being increasingly recognized as an integral and stimulatory component of deep vein thrombi [100]. Our group has recently shown that these potentially damaging pathways may also be active in lupus models. Lupus-prone NZM2328 mice demonstrate enhanced NET formation and high titers of anti-NET autoAbs as compared to nonautoimmune controls [101]. Further, inhibition of NET formation with an inhibitor of histone deimination, mitigates the endothelial dysfunction and accelerated thrombosis characteristic of these mice [101]. These findings remain to be confirmed in other models and with other NET inhibitors.

Platelets

Activated platelets have a recognized role in atherothrombosis [102], and as mediators of immunity and inflammation. They not only release cytokines, but also express CD40 ligand (CD40L/CD154), FcγRIIa, and TLRs—among other immunologically relevant molecules—on their surface [103]. Patients with SLE have increased platelet activation compared to controls [104,105], and, recently, interplay between type I IFNs, platelets, and vascular damage has been suggested. A type I-IFN signature has been described in the transcriptosome of lupus platelets, extending to the protein level for selected type I IFN-regulated genes [106]. This type I-IFN signature, as well as platelet activation, were more likely to be found in lupus patients with a history of vascular events, compared to lupus patients without [106]. Platelets may also play a role in the vicious cycle of type I IFN production, given that they upregulate CD40L on their surface in response to circulating ICs, and activate pDCs to release more type I IFNs via a CD40L-CD40 interaction [107]. Further, either platelet depletion or inhibition of platelets with clopidogrel can improve nephritis and disease activity in murine models [107]. Whether, antiplatelet agents have a unique role in protecting against lupus CVD in experimental models remains to be determined. Potential interplay between NETs and platelets in lupus CVD, as has been seen in other models of vascular damage [97,108], also remains to be defined.

LUPUS THERAPEUTICS

Corticosteroids, widely used as adjuvant treatment in SLE, likely have mixed effects on CVD. While there has long been evidence that duration of prednisone use can independently predict risk of CVD events [109], there is also evidence that, over time, more aggressive treatment with drugs like cyclophosphamide and corticosteroids, will correlate with reduced CVD burden [4]. In terms of drugs with a specific role in the treatment of the disease manifestations of SLE, two—the antimalarials and mycophenolate mofetil—have received the most attention for their potential cardioprotective benefits. In recent years, there has also been increasing interest in whether drugs with defined roles in diseases like hypercholesterolemia and diabetes might have particular benefit in patients with SLE.

Antimalarials

In general, antimalarials are probably vasculoprotective, and, indeed, antimalarial use in SLE correlates with reduced vascular stiffness [110], decreased prevalence of carotid plaque [4], and lower total cholesterol levels, especially in patients taking corticosteroids [111]. Further, in a nested case-control study, antimalarials were the only protective factor against thrombovascular events in a multivariate analysis [112], consistent with the antithrombotic effect of these drugs in relevant murine models [113]. One recent study did suggest a counter to the generally vasculoprotective data for antimalarials, showing that chloroquine impairs cholesterol efflux from macrophage foam cells [114], a phenomenon that will require further investigation in the context of SLE.

Mycophenolate mofetil

In patients without SLE, a short course of mycophenolate mofetil (MMF) decreases T cell activation and increases regulatory T cells in carotid artery plaques [115], and may also protect against cardiovascular mortality in renal transplant patients with pretransplant diabetes [116]. Further, recent murine data showed a promising effect of MMF on atherogenesis in a combined model of lupus and atherosclerosis [117]*. When LDLr(−/−) mice were transplanted with bone marrow from a lupus-prone strain, MMF, but not atorvastatin, attenuated atherosclerosis and the recruitment of CD4-positive T cells into lesions; this was without an effect on markers of lupus activity, such as kidney pathology [117]*. Despite this interesting experimental data, in another 2012 study, MMF did not improve either CIMT or CAC over a two-year period in a prospectively-followed lupus cohort [118]**. However, an important caveat of this study is that a relatively small number of patients, 25 out of 187, were treated with MMF at any point during the two years, and the question will therefore need to be reconsidered in larger cohorts.

Statins

In lupus-prone NZB/W F1 mice, atorvastatin treatment reduced anti-dsDNA Ab levels and also improved both proteinuria and kidney histology; the clinical changes were accompanied by reduced expression of MHC class II and CD80/86 on B cells [119]. Further, in a combined lupus/atherosclerosis model, simvastatin therapy not only reduced atherosclerotic lesion area, but also reduced lymphadenopathy and nephritis, while shifting the inflammatory pattern from TH1- to TH2-predominant [120]. In patients, anecdotal reports suggest that statins may reduce proteinuria in lupus nephritis, at least in the short term [121], while an eight-week course of atorvastatin improved flow-mediated dilation in young, female lupus patients [122]. However, a two-year trial of atorvastatin in adult patients with SLE, but without clinical CVD, showed no difference in CAC, CIMT, or disease activity [123]. In addition, the recently published APPLE trial, which administered atorvastatin to pediatric lupus patients over 36 months, reported no difference in CIMT, although hsCRP and lipid profiles did improve [124]*. An important, although understandable, caveat to the clinical trials published to date is their relatively small size and power [125]. Nevertheless, despite interesting experimental and shortterm clinical data, there is no evidence at present to support the use of statins in lupus patients who do not meet criteria for their use based on standard CVD guidelines.

Thiazolidinediones

Thiazolidinediones (TZDs), such as rosiglitazone and pioglitazone, modify gene expression as agonists of peroxisome proliferator-activated receptor gamma (PPARγ), a regulator of adipocyte differentiation, lipid and carbohydrate metabolism, and inflammation. Given the anti-inflammatory properties of TZDs, there has been interest in their impact on both SLE in general and lupus CVD. In a combined lupus/atherosclerosis model, rosiglitazone mitigated autoAb production, renal disease, and atherosclerosis [126], with the beneficial effect mediated by the anti-inflammatory adipocytokine adiponectin. Both rosiglitazone and pioglitazone are also renal-protective in NZB/W F1 mice [127,128], while pioglitazone additionally was shown to have a positive impact on endothelial regeneration and function [128]. Recently, for the first time, the impact of pioglitazone on the CV risk profile of lupus patients was considered [129]. In a small group of young, female lupus patients, pioglitazone administration over three months improved HDL levels, insulin resistance, and HDL size, while decreasing markers of inflammation such as CRP and serum amyloid A; specific markers of endothelial function were not considered [129]. It also remains to be determined whether TZDs (or other non-TZD PPARγ agonists) might improve disease activity in lupus patients.

CONCLUSIONS

Evidence indicates that the rheumatologist can help mitigate CV risk by effectively controlling lupus disease activity. The treatment of traditional CV risk factors should also be attempted, and further education of rheumatologists regarding the need to identify, monitor, and treat these factors is required. We propose an important role for type I IFNs in lupus CVD, a hypothesis that has gained support in the field of CVD in general. Going forward, there should be continued focus on understanding the extent to which specific therapeutic regimens—whether already available, like antimalarials and MMF, or novel, like anti-IFN—might be particularly effective against this significant cause of morbidity and mortality in patients with SLE.

KEY POINTS.

  • SLE—and especially active SLE—is a risk factor for subclinical endothelial dysfunction and CV events, an observation which continues to be supported by the most recent epidemiologic studies.

  • Type I IFNs likely play a critical role in both endothelial dysfunction and atherogenesis in lupus patients, patients with other autoimmune diseases, and, potentially, in the general population.

  • NET formation—a process by which neutrophils externalize chromatin and granule-derived proteins—is a likely cause of endothelial damage in SLE, which may be one of the first steps in atherogenesis.

  • Further study is needed to better understand the impact of lupus-specific medications such as antimalarials and mycophenolate mofetil, as well as cholesterol-lowering and anti-platelet agents, on lupus CVD.

ACKNOWLEDGEMENTS

This work was supported by the Alliance for Lupus Research, the Lupus Research Institute, and the National Institutes of Health through PHS grant HL088419 (to MJK). JSK was supported by a Scientist Development Award from the Rheumatology Research Foundation. We apologize to the many authors whose interesting and important studies could not be cited due to space constraints.

Footnotes

The authors have no relevant conflicts of interest to disclose.

REFERENCE SECTION

  • 1.Manzi S, Selzer F, Sutton-Tyrrell K, Fitzgerald SG, Rairie JE, Tracy RP, Kuller LH. Prevalence and risk factors of carotid plaque in women with systemic lupus erythematosus. Arthritis Rheum. 1999;42(1):51–60. doi: 10.1002/1529-0131(199901)42:1<51::AID-ANR7>3.0.CO;2-D. [DOI] [PubMed] [Google Scholar]
  • 2.Hak AE, Karlson EW, Feskanich D, Stampfer MJ, Costenbader KH. Systemic lupus erythematosus and the risk of cardiovascular disease: results from the nurses' health study. Arthritis Rheum. 2009;61(10):1396–1402. doi: 10.1002/art.24537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rajagopalan S, Somers EC, Brook RD, Kehrer C, Pfenninger D, Lewis E, Chakrabarti A, Richardson BC, Shelden E, McCune WJ, Kaplan MJ. Endothelial cell apoptosis in systemic lupus erythematosus: a common pathway for abnormal vascular function and thrombosis propensity. Blood. 2004;103(10):3677–3683. doi: 10.1182/blood-2003-09-3198. [DOI] [PubMed] [Google Scholar]
  • 4.Roman MJ, Shanker BA, Davis A, Lockshin MD, Sammaritano L, Simantov R, Crow MK, Schwartz JE, Paget SA, Devereux RB, Salmon JE. Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus. N Engl J Med. 2003;349(25):2399–2406. doi: 10.1056/NEJMoa035471. [DOI] [PubMed] [Google Scholar]
  • 5.Bruce IN, Burns RJ, Gladman DD, Urowitz MB. Single photon emission computed tomography dual isotope myocardial perfusion imaging in women with systemic lupus erythematosus. I. Prevalence and distribution of abnormalities. J Rheumatol. 2000;27(10):2372–2377. [PubMed] [Google Scholar]
  • 6.Esdaile JM, Abrahamowicz M, Grodzicky T, Li Y, Panaritis C, du Berger R, Cote R, Grover SA, Fortin PR, Clarke AE, Senecal JL. Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus. Arthritis Rheum. 2001;44(10):2331–2337. doi: 10.1002/1529-0131(200110)44:10<2331::aid-art395>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
  • 7.Nossent J, Cikes N, Kiss E, Marchesoni A, Nassonova V, Mosca M, Olesinska M, Pokorny G, Rozman B, Schneider M, Vlachoyiannopoulos PG, et al. Current causes of death in systemic lupus erythematosus in Europe, 2000–2004: relation to disease activity and damage accrual. Lupus. 2007;16(5):309–317. doi: 10.1177/0961203307077987. [DOI] [PubMed] [Google Scholar]
  • 8.Kahlenberg JM, Kaplan MJ. Mechanisms of premature atherosclerosis in rheumatoid arthritis and lupus. Annu Rev Med. 2013;64:249–263. doi: 10.1146/annurev-med-060911-090007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Skaggs BJ, Hahn BH, McMahon M. Accelerated atherosclerosis in patients with SLE--mechanisms and management. Nat Rev Rheumatol. 2012;8(4):214–223. doi: 10.1038/nrrheum.2012.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Zoller B, Li X, Sundquist J, Sundquist K. Risk of subsequent coronary heart disease in patients hospitalized for immune-mediated diseases: a nationwide follow-up study from Sweden. PLoS One. 2012;7(3):e33442. doi: 10.1371/journal.pone.0033442. Patients hospitalized with a main diagnosis of an immune-mediated disease, and with no past history of CVD, were followed for first hospitalization for coronary heart disease. A diagnosis of SLE carried more risk than all diagnoses except chorea minor.
  • 11.Souza DC, Santo AH, Sato EI. Mortality profile related to systemic lupus erythematosus: a multiple cause-of-death analysis. J Rheumatol. 2012;39(3):496–503. doi: 10.3899/jrheum.110241. [DOI] [PubMed] [Google Scholar]
  • 12. Steiman AJ, Gladman DD, Ibanez D, Urowitz MB. Outcomes in patients with systemic lupus erythematosus with and without a prolonged serologically active clinically quiescent period. Arthritis Care Res (Hoboken) 2012;64(4):511–518. doi: 10.1002/acr.21568. Serologically active but clinically quiescent lupus patients are at less risk for renal and CV damage than their counterparts with active clinical disease. More studies in these patients who often present a management quandary for the rheumatologist are definitely warranted.
  • 13.Murray SG, Yazdany J, Kaiser R, Criswell LA, Trupin L, Yelin EH, Katz PP, Julian LJ. Cardiovascular disease and cognitive dysfunction in systemic lupus erythematosus. Arthritis Care Res (Hoboken) 2012;64(9):1328–1333. doi: 10.1002/acr.21691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Greco CM, Li T, Sattar A, Kao AH, Danchenko N, Edmundowicz D, Sutton-Tyrrell K, Tracy RP, Kuller LH, Manzi S. Association between depression and vascular disease in systemic lupus erythematosus. J Rheumatol. 2012;39(2):262–268. doi: 10.3899/jrheum.110327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Maynard JW, Fang H, Petri M. Low socioeconomic status is associated with cardiovascular risk factors and outcomes in systemic lupus erythematosus. J Rheumatol. 2012;39(4):777–783. doi: 10.3899/jrheum.110724. Low socioeconomic status has previously been linked to bad outcomes in lupus nephritis. This association may also be true for CVD.
  • 16.Magder LS, Petri M. Incidence of and risk factors for adverse cardiovascular events among patients with systemic lupus erythematosus. Am J Epidemiol. 2012;176(8):708–719. doi: 10.1093/aje/kws130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang XY, Tang XQ, Huang YJ, Chen WY, Yu XQ. Frequency of established cardiovascular disease and its risk factors in Chinese patients with systemic lupus erythematosus. Clin Rheumatol. 2012;31(4):669–675. doi: 10.1007/s10067-011-1910-3. [DOI] [PubMed] [Google Scholar]
  • 18.Romero-Diaz J, Vargas-Vorackova F, Kimura-Hayama E, Cortazar-Benitez LF, Gijon-Mitre R, Criales S, Cabiedes-Contreras J, Iniguez-Rodriguez Mdel R, Lara-Garcia EA, Nunez-Alvarez C, Llorente L, et al. Systemic lupus erythematosus risk factors for coronary artery calcifications. Rheumatology (Oxford) 2012;51(1):110–119. doi: 10.1093/rheumatology/ker307. [DOI] [PubMed] [Google Scholar]
  • 19.Karp I, Abrahamowicz M, Fortin PR, Pilote L, Neville C, Pineau CA, Esdaile JM. Longitudinal evolution of risk of coronary heart disease in systemic lupus erythematosus. J Rheumatol. 2012;39(5):968–973. doi: 10.3899/jrheum.111127. [DOI] [PubMed] [Google Scholar]
  • 20.Yang L, Tao J, Tang X, Wang Y, He X, Xu G, Ren Y, Tu Y. Prevalence and correlation of conventional and lupus-specific risk factors for cardiovascular disease in Chinese systemic lupus erythematosus patients. J Eur Acad Dermatol Venereol. 2012;26(1):95–101. doi: 10.1111/j.1468-3083.2011.04211.x. [DOI] [PubMed] [Google Scholar]
  • 21.Gustafsson JT, Simard JF, Gunnarsson I, Elvin K, Lundberg IE, Hansson LO, Larsson A, Svenungsson E. Risk factors for cardiovascular mortality in patients with systemic lupus erythematosus, a prospective cohort study. Arthritis Res Ther. 2012;14(2):R46. doi: 10.1186/ar3759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kaiser R, Li Y, Chang M, Catanese J, Begovich AB, Brown EE, Edberg JC, McGwin G, Jr, Alarcon GS, Ramsey-Goldman R, Reveille JD, et al. Genetic risk factors for thrombosis in systemic lupus erythematosus. J Rheumatol. 2012;39(8):1603–1610. doi: 10.3899/jrheum.111451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chang YH, Lin IL, Tsay GJ, Yang SC, Yang TP, Ho KT, Hsu TC, Shiau MY. Elevated circulatory MMP-2 and MMP-9 levels and activities in patients with rheumatoid arthritis and systemic lupus erythematosus. Clin Biochem. 2008;41(12):955–959. doi: 10.1016/j.clinbiochem.2008.04.012. [DOI] [PubMed] [Google Scholar]
  • 24.Galis ZS, Khatri JJ. Matrix metalloproteinases in vascular remodeling and atherogenesis: the good, the bad, and the ugly. Circ Res. 2002;90(3):251–262. [PubMed] [Google Scholar]
  • 25.Bahrehmand F, Vaisi-Raygani A, Kiani A, Rahimi Z, Tavilani H, Navabi SJ, Shakiba E, Hassanzadeh N, Pourmotabbed T. Matrix metalloproteinase-2 functional promoter polymorphism G1575A is associated with elevated circulatory MMP-2 levels and increased risk of cardiovascular disease in systemic lupus erythematosus patients. Lupus. 2012;21(6):616–624. doi: 10.1177/0961203312436857. [DOI] [PubMed] [Google Scholar]
  • 26.Narshi CB, Giles IP, Rahman A. The endothelium: an interface between autoimmunity and atherosclerosis in systemic lupus erythematosus? Lupus. 2011;20(1):5–13. doi: 10.1177/0961203310382429. [DOI] [PubMed] [Google Scholar]
  • 27.Baron MA, Khamashta MA, Hughes GR, D'Cruz DP. Prevalence of an abnormal ankle-brachial index in patients with primary antiphospholipid syndrome: preliminary data. Ann Rheum Dis. 2005;64(1):144–146. doi: 10.1136/ard.2003.016204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ames PR, Margarita A, Delgado Alves J, Tommasino C, Iannaccone L, Brancaccio V. Anticardiolipin antibody titre and plasma homocysteine level independently predict intima media thickness of carotid arteries in subjects with idiopathic antiphospholipid antibodies. Lupus. 2002;11(4):208–214. doi: 10.1191/0961203302lu165oa. [DOI] [PubMed] [Google Scholar]
  • 29.Gresele P, Migliacci R, Vedovati MC, Ruffatti A, Becattini C, Facco M, Guglielmini G, Boscaro E, Mezzasoma AM, Momi S, Pengo V. Patients with primary antiphospholipid antibody syndrome and without associated vascular risk factors present a normal endothelial function. Thromb Res. 2009;123(3):444–451. doi: 10.1016/j.thromres.2008.05.015. [DOI] [PubMed] [Google Scholar]
  • 30.Ames PR, Margarita A, Sokoll KB, Weston M, Brancaccio V. Premature atherosclerosis in primary antiphospholipid syndrome: preliminary data. Ann Rheum Dis. 2005;64(2):315–317. doi: 10.1136/ard.2004.023952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Petri M. Update on anti-phospholipid antibodies in SLE: the Hopkins' Lupus Cohort. Lupus. 2010;19(4):419–423. doi: 10.1177/0961203309360541. [DOI] [PubMed] [Google Scholar]
  • 32.Ames PR, Margarita A, Alves JD. Antiphospholipid antibodies and atherosclerosis: insights from systemic lupus erythematosus and primary antiphospholipid syndrome. Clin Rev Allergy Immunol. 2009;37(1):29–35. doi: 10.1007/s12016-008-8099-5. [DOI] [PubMed] [Google Scholar]
  • 33.Dinu AR, Merrill JT, Shen C, Antonov IV, Myones BL, Lahita RG. Frequency of antibodies to the cholesterol transport protein apolipoprotein A1 in patients with SLE. Lupus. 1998;7(5):355–360. doi: 10.1191/096120398678920262. [DOI] [PubMed] [Google Scholar]
  • 34.O'Neill SG, Giles I, Lambrianides A, Manson J, D'Cruz D, Schrieber L, March LM, Latchman DS, Isenberg DA, Rahman A. Antibodies to apolipoprotein A-I, high-density lipoprotein, and C-reactive protein are associated with disease activity in patients with systemic lupus erythematosus. Arthritis Rheum. 2010;62(3):845–854. doi: 10.1002/art.27286. [DOI] [PubMed] [Google Scholar]
  • 35.Mandal K, Foteinos G, Jahangiri M, Xu Q. Role of antiheat shock protein 60 autoantibodies in atherosclerosis. Lupus. 2005;14(9):742–746. doi: 10.1191/0961203305lu2212oa. [DOI] [PubMed] [Google Scholar]
  • 36. Motoki Y, Nojima J, Yanagihara M, Tsuneoka H, Matsui T, Yamamoto M, Ichihara K. Anti-phospholipid antibodies contribute to arteriosclerosis in patients with systemic lupus erythematosus through induction of tissue factor expression and cytokine production from peripheral blood mononuclear cells. Thromb Res. 2012;130(4):667–673. doi: 10.1016/j.thromres.2011.11.048. aPL Abs bring TF to the surface of peripheral blood mononuclear cells, both in vivo and in vitro. This interestingly correlated with risk of peripheral vascular disease in lupus patients, pointing toward the need for further study of dysregulated TF in the accelerated atherosclerosis of SLE.
  • 37.Loeffen R, Spronk HM, ten Cate H. The impact of blood coagulability on atherosclerosis and cardiovascular disease. J Thromb Haemost. 2012;10(7):1207–1216. doi: 10.1111/j.1538-7836.2012.04782.x. [DOI] [PubMed] [Google Scholar]
  • 38.Boles J, Mackman N. Role of tissue factor in thrombosis in antiphospholipid antibody syndrome. Lupus. 2010;19(4):370–378. doi: 10.1177/0961203309360810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kinev AV, Roubey RA. Tissue factor in the antiphospholipid syndrome. Lupus. 2008;17(10):952–958. doi: 10.1177/0961203308096662. [DOI] [PubMed] [Google Scholar]
  • 40.McMahon M, Grossman J, FitzGerald J, Dahlin-Lee E, Wallace DJ, Thong BY, Badsha H, Kalunian K, Charles C, Navab M, Fogelman AM, et al. Proinflammatory highdensity lipoprotein as a biomarker for atherosclerosis in patients with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum. 2006;54(8):2541–2549. doi: 10.1002/art.21976. [DOI] [PubMed] [Google Scholar]
  • 41.McMahon M, Grossman J, Skaggs B, Fitzgerald J, Sahakian L, Ragavendra N, Charles-Schoeman C, Watson K, Wong WK, Volkmann E, Chen W, et al. Dysfunctional proinflammatory high-density lipoproteins confer increased risk of atherosclerosis in women with systemic lupus erythematosus. Arthritis Rheum. 2009;60(8):2428–2437. doi: 10.1002/art.24677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Srivastava R, Yu S, Parks BW, Black LL, Kabarowski JH. Autoimmune-mediated reduction of high-density lipoprotein-cholesterol and paraoxonase 1 activity in systemic lupus erythematosus-prone gld mice. Arthritis Rheum. 2011;63(1):201–211. doi: 10.1002/art.27764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Thacker SG, Zhao W, Smith CK, Luo W, Wang H, Vivekanandan-Giri A, Rabquer BJ, Koch AE, Pennathur S, Davidson A, Eitzman DT, et al. Type I interferons modulate vascular function, repair, thrombosis, and plaque progression in murine models of lupus and atherosclerosis. Arthritis Rheum. 2012;64(9):2975–2985. doi: 10.1002/art.34504. Using a variety of murine models including exogenous type I IFN administration and knockout of the type I IFN receptor, the authors show that subclinical vascular dysfunction in lupus-prone strains, as well as atherosclerosis in apoE(−/−) mice, are at least partially dependent on type I IFNs.
  • 44.Woo JM, Lin Z, Navab M, Van Dyck C, Trejo-Lopez Y, Woo KM, Li H, Castellani LW, Wang X, Iikuni N, Rullo OJ, et al. Treatment with apolipoprotein A-1 mimetic peptide reduces lupus-like manifestations in a murine lupus model of accelerated atherosclerosis. Arthritis Res Ther. 2010;12(3):R93. doi: 10.1186/ar3020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dhaun N, Kluth DC. Oxidative stress promotes hypertension and albuminuria during the autoimmune disease systemic lupus erythematosus. Hypertension. 2012;59(5):e47. doi: 10.1161/HYPERTENSIONAHA.112.193276. author reply e48. [DOI] [PubMed] [Google Scholar]
  • 46.Hanasand M, Omdal R, Norheim KB, Goransson LG, Brede C, Jonsson G. Improved detection of advanced oxidation protein products in plasma. Clin Chim Acta. 2012;413(9–10):901–906. doi: 10.1016/j.cca.2012.01.038. [DOI] [PubMed] [Google Scholar]
  • 47.Li KJ, Wu CH, Hsieh SC, Lu MC, Tsai CY, Yu CL. Deranged bioenergetics and defective redox capacity in T lymphocytes and neutrophils are related to cellular dysfunction and increased oxidative stress in patients with active systemic lupus erythematosus. Clin Dev Immunol. 2012;2012:548516. doi: 10.1155/2012/548516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nikpour M, Harvey PJ, Ibanez D, Gladman DD, Urowitz MB. High-sensitivity C-reactive protein as a marker of cardiovascular risk in systemic lupus erythematosus. Arthritis Rheum. 2012;64(9):3052–3053. doi: 10.1002/art.34541. [DOI] [PubMed] [Google Scholar]
  • 49.Lehrer RI, Ganz T. Defensins of vertebrate animals. Curr Opin Immunol. 2002;14(1):96–102. doi: 10.1016/s0952-7915(01)00303-x. [DOI] [PubMed] [Google Scholar]
  • 50.Lehrer RI, Lichtenstein AK, Ganz T. Defensins: antimicrobial and cytotoxic peptides of mammalian cells. Annu Rev Immunol. 1993;11:105–128. doi: 10.1146/annurev.iy.11.040193.000541. [DOI] [PubMed] [Google Scholar]
  • 51.Tamiya H, Tani K, Miyata J, Sato K, Urata T, Lkhagvaa B, Otsuka S, Shigekiyo S, Sone S. Defensins- and cathepsin G-ANCA in systemic lupus erythematosus. Rheumatol Int. 2006;27(2):147–152. doi: 10.1007/s00296-006-0173-9. [DOI] [PubMed] [Google Scholar]
  • 52.Sthoeger ZM, Bezalel S, Chapnik N, Asher I, Froy O. High alpha-defensin levels in patients with systemic lupus erythematosus. Immunology. 2009;127(1):116–122. doi: 10.1111/j.1365-2567.2008.02997.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Vordenbaumen S, Fischer-Betz R, Timm D, Sander O, Chehab G, Richter J, Bleck E, Schneider M. Elevated levels of human beta-defensin 2 and human neutrophil peptides in systemic lupus erythematosus. Lupus. 2010;19(14):1648–1653. doi: 10.1177/0961203310377089. [DOI] [PubMed] [Google Scholar]
  • 54. Vordenbaumen S, Sander O, Bleck E, Schneider M, Fischer-Betz R. Cardiovascular disease and serum defensin levels in systemic lupus erythematosus. Clin Exp Rheumatol. 2012;30(3):364–370. Circulating defensin levels, which can be derived from NETs, predict progression of subclinical lupus CVD.
  • 55.Knight JS, Carmona-Rivera C, Kaplan MJ. Proteins derived from neutrophil extracellular traps may serve as self-antigens and mediate organ damage in autoimmune diseases. Front Immunol. 2012;3:380. doi: 10.3389/fimmu.2012.00380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Westerweel PE, Luijten RK, Hoefer IE, Koomans HA, Derksen RH, Verhaar MC. Haematopoietic and endothelial progenitor cells are deficient in quiescent systemic lupus erythematosus. Ann Rheum Dis. 2007;66(7):865–870. doi: 10.1136/ard.2006.065631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Denny MF, Thacker S, Mehta H, Somers EC, Dodick T, Barrat FJ, McCune WJ, Kaplan MJ. Interferon-alpha promotes abnormal vasculogenesis in lupus: a potential pathway for premature atherosclerosis. Blood. 2007;110(8):2907–2915. doi: 10.1182/blood-2007-05-089086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Thacker SG, Berthier CC, Mattinzoli D, Rastaldi MP, Kretzler M, Kaplan MJ. The detrimental effects of IFN-alpha on vasculogenesis in lupus are mediated by repression of IL-1 pathways: potential role in atherogenesis and renal vascular rarefaction. J Immunol. 2010;185(7):4457–4469. doi: 10.4049/jimmunol.1001782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Schmidt-Lucke C, Rossig L, Fichtlscherer S, Vasa M, Britten M, Kamper U, Dimmeler S, Zeiher AM. Reduced number of circulating endothelial progenitor cells predicts future cardiovascular events: proof of concept for the clinical importance of endogenous vascular repair. Circulation. 2005;111(22):2981–2987. doi: 10.1161/CIRCULATIONAHA.104.504340. [DOI] [PubMed] [Google Scholar]
  • 60.Lee PY, Li Y, Richards HB, Chan FS, Zhuang H, Narain S, Butfiloski EJ, Sobel ES, Reeves WH, Segal MS. Type I interferon as a novel risk factor for endothelial progenitor cell depletion and endothelial dysfunction in systemic lupus erythematosus. Arthritis Rheum. 2007;56(11):3759–3769. doi: 10.1002/art.23035. [DOI] [PubMed] [Google Scholar]
  • 61.Somers EC, Zhao W, Lewis EE, Wang L, Wing JJ, Sundaram B, Kazerooni EA, McCune WJ, Kaplan MJ. Type I interferons are associated with subclinical markers of cardiovascular disease in a cohort of systemic lupus erythematosus patients. PLoS One. 2012;7(5):e37000. doi: 10.1371/journal.pone.0037000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Kahlenberg JM, Thacker SG, Berthier CC, Cohen CD, Kretzler M, Kaplan MJ. Inflammasome activation of IL-18 results in endothelial progenitor cell dysfunction in systemic lupus erythematosus. J Immunol. 2011;187(11):6143–6156. doi: 10.4049/jimmunol.1101284. Although IL-1 appears to promote healthy endothelial progenitor differentiation, the process is impaired by another cytokine dependent on caspase-1 for posttranslational activation, IL-18. This dichotomy needs further study, especially in animal models.
  • 63.Goossens P, Gijbels MJ, Zernecke A, Eijgelaar W, Vergouwe MN, van der Made I, Vanderlocht J, Beckers L, Buurman WA, Daemen MJ, Kalinke U, et al. Myeloid type I interferon signaling promotes atherosclerosis by stimulating macrophage recruitment to lesions. Cell Metab. 2010;12(2):142–153. doi: 10.1016/j.cmet.2010.06.008. [DOI] [PubMed] [Google Scholar]
  • 64. Macritchie N, Grassia G, Sabir SR, Maddaluno M, Welsh P, Sattar N, Ialenti A, Kurowska-Stolarska M, McInnes IB, Brewer JM, Garside P, et al. Plasmacytoid dendritic cells play a key role in promoting atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2012;32(11):2569–2579. doi: 10.1161/ATVBAHA.112.251314. Although pDCs have previously been identified in human atherosclerotic lesions, the authors here tested the concept experimentally and showed that pDC depletion mitigates atherosclerosis.
  • 65.Niessner A, Shin MS, Pryshchep O, Goronzy JJ, Chaikof EL, Weyand CM. Synergistic proinflammatory effects of the antiviral cytokine interferon-alpha and Toll-like receptor 4 ligands in the atherosclerotic plaque. Circulation. 2007;116(18):2043–2052. doi: 10.1161/CIRCULATIONAHA.107.697789. [DOI] [PubMed] [Google Scholar]
  • 66.Li J, Fu Q, Cui H, Qu B, Pan W, Shen N, Bao C. Interferon-alpha priming promotes lipid uptake and macrophage-derived foam cell formation: a novel link between interferon-alpha and atherosclerosis in lupus. Arthritis Rheum. 2011;63(2):492–502. doi: 10.1002/art.30165. [DOI] [PubMed] [Google Scholar]
  • 67.Niessner A, Sato K, Chaikof EL, Colmegna I, Goronzy JJ, Weyand CM. Pathogensensing plasmacytoid dendritic cells stimulate cytotoxic T-cell function in the atherosclerotic plaque through interferon-alpha. Circulation. 2006;114(23):2482–2489. doi: 10.1161/CIRCULATIONAHA.106.642801. [DOI] [PubMed] [Google Scholar]
  • 68.Trinchieri G. Type I interferon: friend or foe? J Exp Med. 2010;207(10):2053–2063. doi: 10.1084/jem.20101664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Agrawal H, Jacob N, Carreras E, Bajana S, Putterman C, Turner S, Neas B, Mathian A, Koss MN, Stohl W, Kovats S, et al. Deficiency of type I IFN receptor in lupus-prone New Zealand mixed 2328 mice decreases dendritic cell numbers and activation and protects from disease. J Immunol. 2009;183(9):6021–6029. doi: 10.4049/jimmunol.0803872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Thacker SG, Duquaine D, Park J, Kaplan MJ. Lupus-prone New Zealand Black/New Zealand White F1 mice display endothelial dysfunction and abnormal phenotype and function of endothelial progenitor cells. Lupus. 2010;19(3):288–299. doi: 10.1177/0961203309353773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mathian A, Weinberg A, Gallegos M, Banchereau J, Koutouzov S. IFN-alpha induces early lethal lupus in preautoimmune (New Zealand Black × New Zealand White) F1 but not in BALB/c mice. J Immunol. 2005;174(5):2499–2506. doi: 10.4049/jimmunol.174.5.2499. [DOI] [PubMed] [Google Scholar]
  • 72.Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A. 2003;100(5):2610–2615. doi: 10.1073/pnas.0337679100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kim T, Kanayama Y, Negoro N, Okamura M, Takeda T, Inoue T. Serum levels of interferons in patients with systemic lupus erythematosus. Clin Exp Immunol. 1987;70(3):562–569. [PMC free article] [PubMed] [Google Scholar]
  • 74.McBride JM, Jiang J, Abbas AR, Morimoto A, Li J, Maciuca R, Townsend M, Wallace DJ, Kennedy WP, Drappa J. Safety and pharmacodynamics of rontalizumab in patients with systemic lupus erythematosus: results of a phase I, placebo-controlled, double-blind, dose-escalation study. Arthritis Rheum. 2012;64(11):3666–3676. doi: 10.1002/art.34632. [DOI] [PubMed] [Google Scholar]
  • 75.Merrill JT, Wallace DJ, Petri M, Kirou KA, Yao Y, White WI, Robbie G, Levin R, Berney SM, Chindalore V, Olsen N, et al. Safety profile and clinical activity of sifalimumab, a fully human anti-interferon alpha monoclonal antibody, in systemic lupus erythematosus: a phase I, multicentre, double-blind randomised study. Ann Rheum Dis. 2011;70(11):1905–1913. doi: 10.1136/ard.2010.144485. [DOI] [PubMed] [Google Scholar]
  • 76.Elkon KB, Wiedeman A. Type I IFN system in the development and manifestations of SLE. Curr Opin Rheumatol. 2012;24(5):499–505. doi: 10.1097/BOR.0b013e3283562c3e. [DOI] [PubMed] [Google Scholar]
  • 77.Denny MF, Yalavarthi S, Zhao W, Thacker SG, Anderson M, Sandy AR, McCune WJ, Kaplan MJ. A distinct subset of proinflammatory neutrophils isolated from patients with systemic lupus erythematosus induces vascular damage and synthesizes type I IFNs. J Immunol. 2010;184(6):3284–3297. doi: 10.4049/jimmunol.0902199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Folkman J, Ingber D. Inhibition of angiogenesis. Semin Cancer Biol. 1992;3(2):89–96. [PubMed] [Google Scholar]
  • 79.Hansson GK, Hermansson A. The immune system in atherosclerosis. Nat Immunol. 2011;12(3):204–212. doi: 10.1038/ni.2001. [DOI] [PubMed] [Google Scholar]
  • 80.Aprahamian T, Rifkin I, Bonegio R, Hugel B, Freyssinet JM, Sato K, Castellot JJ, Jr, Walsh K. Impaired clearance of apoptotic cells promotes synergy between atherogenesis and autoimmune disease. J Exp Med. 2004;199(8):1121–1131. doi: 10.1084/jem.20031557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Braun N, Wade NS, Wakeland EK, Major AS. Accelerated atherosclerosis is independent of feeding high fat diet in systemic lupus erythematosus-susceptible LDLr(−/−) mice. Lupus. 2008;17(12):1070–1078. doi: 10.1177/0961203308093551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Feng X, Li H, Rumbin AA, Wang X, La Cava A, Brechtelsbauer K, Castellani LW, Witztum JL, Lusis AJ, Tsao BP. ApoE−/−Fas−/− C57BL/6 mice: a novel murine model simultaneously exhibits lupus nephritis, atherosclerosis, and osteopenia. J Lipid Res. 2007;48(4):794–805. doi: 10.1194/jlr.M600512-JLR200. [DOI] [PubMed] [Google Scholar]
  • 83.Ma Z, Choudhury A, Kang SA, Monestier M, Cohen PL, Eisenberg RA. Accelerated atherosclerosis in ApoE deficient lupus mouse models. Clin Immunol. 2008;127(2):168–175. doi: 10.1016/j.clim.2008.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Stanic AK, Stein CM, Morgan AC, Fazio S, Linton MF, Wakeland EK, Olsen NJ, Major AS. Immune dysregulation accelerates atherosclerosis and modulates plaque composition in systemic lupus erythematosus. Proc Natl Acad Sci U S A. 2006;103(18):7018–7023. doi: 10.1073/pnas.0602311103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Lewis MJ, Malik TH, Fossati-Jimack L, Carassiti D, Cook HT, Haskard DO, Botto M. Distinct roles for complement in glomerulonephritis and atherosclerosis revealed in mice with a combination of lupus and hyperlipidemia. Arthritis Rheum. 2012;64(8):2707–2718. doi: 10.1002/art.34451. This study carefully examined complement deposition in a combined lupus/atherosclerosis model. The authors make the case that complement depletion by the lupus phenotype leads to accumulation of debris in the developing atherosclerotic plaque.
  • 86. Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, Busch M, Manca M, Koenen RR, Pelisek J, Daemen MJ, et al. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promote atherosclerosis. Circulation. 2012;125(13):1673–1683. doi: 10.1161/CIRCULATIONAHA.111.046755. Using a number of strategies, including knockout of the critical NET protein CRAMP and administration of NET-like complexes, the authors convincingly show that NETs exacerbate atherosclerosis in vivo, probably through pDC stimulation and type I IFN production. Intringuigly, anti-double-stranded-DNA titers also correlated with atherosclerosis, both in the animal model, and in patients with atherosclerosis, but without SLE.
  • 87.Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, Meller S, Chamilos G, Sebasigari R, Riccieri V, Bassett R, et al. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med. 2011;3(73):73ra19. doi: 10.1126/scitranslmed.3001180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Garcia-Romo GS, Caielli S, Vega B, Connolly J, Allantaz F, Xu Z, Punaro M, Baisch J, Guiducci C, Coffman RL, Barrat FJ, et al. Netting neutrophils are major inducers of type I IFN production in pediatric systemic lupus erythematosus. Sci Transl Med. 2011;3(73):73ra20. doi: 10.1126/scitranslmed.3001201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Villanueva E, Yalavarthi S, Berthier CC, Hodgin JB, Khandpur R, Lin AM, Rubin CJ, Zhao W, Olsen SH, Klinker M, Shealy D, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011;187(1):538–552. doi: 10.4049/jimmunol.1100450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.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(8):1052–1056. doi: 10.1161/CIRCRESAHA.112.265868. [DOI] [PubMed] [Google Scholar]
  • 91.Knight JS, Kaplan MJ. Lupus neutrophils: 'NET' gain in understanding lupus pathogenesis. Curr Opin Rheumatol. 2012;24(5):441–450. doi: 10.1097/BOR.0b013e3283546703. [DOI] [PubMed] [Google Scholar]
  • 92.Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–1535. doi: 10.1126/science.1092385. [DOI] [PubMed] [Google Scholar]
  • 93.Brinkmann V, Zychlinsky A. Neutrophil extracellular traps: is immunity the second function of chromatin? J Cell Biol. 2012;198(5):773–783. doi: 10.1083/jcb.201203170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hakkim A, Furnrohr BG, Amann K, Laube B, Abed UA, Brinkmann V, Herrmann M, Voll RE, Zychlinsky A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc Natl Acad Sci U S A. 2010;107(21):9813–9818. doi: 10.1073/pnas.0909927107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Leffler J, Martin M, Gullstrand B, Tyden H, Lood C, Truedsson L, Bengtsson AA, Blom AM. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J Immunol. 2012;188(7):3522–3531. doi: 10.4049/jimmunol.1102404. [DOI] [PubMed] [Google Scholar]
  • 96.Kahlenberg JM, Carmona-Rivera C, Smith CK, Kaplan MJ. Neutrophil extracellular trap-associated protein activation of the NLRP3 inflammasome is enhanced in lupus macrophages. J Immunol. 2013;190(3):1217–1226. doi: 10.4049/jimmunol.1202388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Caudrillier A, Kessenbrock K, Gilliss BM, Nguyen JX, Marques MB, Monestier M, Toy P, Werb Z, Looney MR. Platelets induce neutrophil extracellular traps in transfusion-related acute lung injury. J Clin Invest. 2012;122(7):2661–2671. doi: 10.1172/JCI61303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gupta AK, Joshi MB, Philippova M, Erne P, Hasler P, Hahn S, Resink TJ. Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis-mediated cell death. FEBS Lett. 2010;584(14):3193–3197. doi: 10.1016/j.febslet.2010.06.006. [DOI] [PubMed] [Google Scholar]
  • 99.Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, Weber C, Soehnlein O. Presence of luminal neutrophil extracellular traps in atherosclerosis. Thromb Haemost. 2012;107(3):597–598. doi: 10.1160/TH11-09-0650. [DOI] [PubMed] [Google Scholar]
  • 100.Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol. 2012;32(8):1777–1783. doi: 10.1161/ATVBAHA.111.242859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Knight JS, Zhao W, Luo W, Subramanian V, O’Dell AA, Yalavarthi S, Hodgin JB, Eitzman DT, Thompson PR, Kaplan MJ. Peptidylarginine deiminase inhibition is immunomodulatory and vasculoprotective in murine lupus. Journal of Clinical Investigation. 2013 doi: 10.1172/JCI67390. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med. 2007;357(24):2482–2494. doi: 10.1056/NEJMra071014. [DOI] [PubMed] [Google Scholar]
  • 103.Vieira-de-Abreu A, Campbell RA, Weyrich AS, Zimmerman GA. Platelets: versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin Immunopathol. 2012;34(1):5–30. doi: 10.1007/s00281-011-0286-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Nagahama M, Nomura S, Ozaki Y, Yoshimura C, Kagawa H, Fukuhara S. Platelet activation markers and soluble adhesion molecules in patients with systemic lupus erythematosus. Autoimmunity. 2001;33(2):85–94. doi: 10.3109/08916930108995993. [DOI] [PubMed] [Google Scholar]
  • 105.Tam LS, Fan B, Li EK, Thomas GN, Yim SF, Haines CJ, Tomlinson B. Patients with systemic lupus erythematosus show increased platelet activation and endothelial dysfunction induced by acute hyperhomocysteinemia. J Rheumatol. 2003;30(7):1479–1484. [PubMed] [Google Scholar]
  • 106.Lood C, Amisten S, Gullstrand B, Jonsen A, Allhorn M, Truedsson L, Sturfelt G, Erlinge D, Bengtsson AA. Platelet transcriptional profile and protein expression in patients with systemic lupus erythematosus: up-regulation of the type I interferon system is strongly associated with vascular disease. Blood. 2010;116(11):1951–1957. doi: 10.1182/blood-2010-03-274605. [DOI] [PubMed] [Google Scholar]
  • 107.Duffau P, Seneschal J, Nicco C, Richez C, Lazaro E, Douchet I, Bordes C, Viallard JF, Goulvestre C, Pellegrin JL, Weil B, et al. Platelet CD154 potentiates interferon-alpha secretion by plasmacytoid dendritic cells in systemic lupus erythematosus. Sci Transl Med. 2010;2(47):47ra63. doi: 10.1126/scitranslmed.3001001. [DOI] [PubMed] [Google Scholar]
  • 108.Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13(4):463–469. doi: 10.1038/nm1565. [DOI] [PubMed] [Google Scholar]
  • 109.Petri M, Perez-Gutthann S, Spence D, Hochberg MC. Risk factors for coronary artery disease in patients with systemic lupus erythematosus. Am J Med. 1992;93(5):513–519. doi: 10.1016/0002-9343(92)90578-y. [DOI] [PubMed] [Google Scholar]
  • 110.Selzer F, Sutton-Tyrrell K, Fitzgerald S, Tracy R, Kuller L, Manzi S. Vascular stiffness in women with systemic lupus erythematosus. Hypertension. 2001;37(4):1075–1082. doi: 10.1161/01.hyp.37.4.1075. [DOI] [PubMed] [Google Scholar]
  • 111.Rahman P, Gladman DD, Urowitz MB, Yuen K, Hallett D, Bruce IN. The cholesterol lowering effect of antimalarial drugs is enhanced in patients with lupus taking corticosteroid drugs. J Rheumatol. 1999;26(2):325–330. [PubMed] [Google Scholar]
  • 112.Jung H, Bobba R, Su J, Shariati-Sarabi Z, Gladman DD, Urowitz M, Lou W, Fortin PR. The protective effect of antimalarial drugs on thrombovascular events in systemic lupus erythematosus. Arthritis Rheum. 2010;62(3):863–868. doi: 10.1002/art.27289. [DOI] [PubMed] [Google Scholar]
  • 113.Edwards MH, Pierangeli S, Liu X, Barker JH, Anderson G, Harris EN. Hydroxychloroquine reverses thrombogenic properties of antiphospholipid antibodies in mice. Circulation. 1997;96(12):4380–4384. doi: 10.1161/01.cir.96.12.4380. [DOI] [PubMed] [Google Scholar]
  • 114.Ouimet M, Franklin V, Mak E, Liao X, Tabas I, Marcel YL. Autophagy regulates cholesterol efflux from macrophage foam cells via lysosomal acid lipase. Cell Metab. 2011;13(6):655–667. doi: 10.1016/j.cmet.2011.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.van Leuven SI, van Wijk DF, Volger OL, de Vries JP, van der Loos CM, de Kleijn DV, Horrevoets AJ, Tak PP, van der Wal AC, de Boer OJ, Pasterkamp G, et al. Mycophenolate mofetil attenuates plaque inflammation in patients with symptomatic carotid artery stenosis. Atherosclerosis. 2010;211(1):231–236. doi: 10.1016/j.atherosclerosis.2010.01.043. [DOI] [PubMed] [Google Scholar]
  • 116.David KM, Morris JA, Steffen BJ, Chi-Burris KS, Gotz VP, Gordon RD. Mycophenolate mofetil vs. azathioprine is associated with decreased acute rejection, late acute rejection, and risk for cardiovascular death in renal transplant recipients with pretransplant diabetes. Clin Transplant. 2005;19(2):279–285. doi: 10.1111/j.1399-0012.2005.00338.x. [DOI] [PubMed] [Google Scholar]
  • 117. van Leuven SI, Mendez-Fernandez YV, Wilhelm AJ, Wade NS, Gabriel CL, Kastelein JJ, Stroes ES, Tak PP, Major AS. Mycophenolate mofetil but not atorvastatin attenuates atherosclerosis in lupus-prone LDLr(−/−) mice. Ann Rheum Dis. 2012;71(3):408–414. doi: 10.1136/annrheumdis-2011-200071. In a combined lupus/atherosclerosis model, MMF, but surprisingly not atorvastatin, improved atherosclerosis. Also perhaps surprisingly, MMF did not protect against nephritis.
  • 118. Kiani AN, Magder LS, Petri M. Mycophenolate mofetil (MMF) does not slow the progression of subclinical atherosclerosis in SLE over 2 years. Rheumatol Int. 2012;32(9):2701–2705. doi: 10.1007/s00296-011-2048-y. Although of relatively small size, this study is the first to prospectively assess the impact of MMF on lupus CVD. Despite no benefit here, the vasculoprotective effects of MMF in the context of renal transplatation certainly warrant its further study in lupus CVD.
  • 119.Lawman S, Mauri C, Jury EC, Cook HT, Ehrenstein MR. Atorvastatin inhibits autoreactive B cell activation and delays lupus development in New Zealand black/white F1 mice. J Immunol. 2004;173(12):7641–7646. doi: 10.4049/jimmunol.173.12.7641. [DOI] [PubMed] [Google Scholar]
  • 120.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(5):3028–3034. doi: 10.4049/jimmunol.177.5.3028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Abud-Mendoza C, de la Fuente H, Cuevas-Orta E, Baranda L, Cruz-Rizo J, Gonzalez-Amaro R. Therapy with statins in patients with refractory rheumatic diseases: a preliminary study. Lupus. 2003;12(8):607–611. doi: 10.1191/0961203303lu429oa. [DOI] [PubMed] [Google Scholar]
  • 122.Ferreira GA, Navarro TP, Telles RW, Andrade LE, Sato EI. Atorvastatin therapy improves endothelial-dependent vasodilation in patients with systemic lupus erythematosus: an 8 weeks controlled trial. Rheumatology (Oxford) 2007;46(10):1560–1565. doi: 10.1093/rheumatology/kem186. [DOI] [PubMed] [Google Scholar]
  • 123.Petri MA, Kiani AN, Post W, Christopher-Stine L, Magder LS. Lupus Atherosclerosis Prevention Study (LAPS) Ann Rheum Dis. 2011;70(5):760–765. doi: 10.1136/ard.2010.136762. [DOI] [PubMed] [Google Scholar]
  • 124. Schanberg LE, Sandborg C, Barnhart HX, Ardoin SP, Yow E, Evans GW, Mieszkalski KL, Ilowite NT, Eberhard A, Imundo LF, Kimura Y, et al. Use of atorvastatin in systemic lupus erythematosus in children and adolescents. Arthritis Rheum. 2012;64(1):285–296. doi: 10.1002/art.30645. The ambitious APPLE study prospectively assessed the effects of atorvastatin on preclinical markers of CVD in pediatric patients with SLE. Some commentors have pointed out that more patients may be necessary to see a beneficial effect, a challenge that will clearly require a significant collaborative effort going forward.
  • 125.Steichen O. A clinically relevant benefit of statins in young patients with systemic lupus erythematosus cannot be excluded: comment on the article by Schanberg et al. Arthritis Rheum. 2012;64(8):2805. doi: 10.1002/art.34521. author reply 2805–2806. [DOI] [PubMed] [Google Scholar]
  • 126.Aprahamian T, Bonegio RG, Richez C, Yasuda K, Chiang LK, Sato K, Walsh K, Rifkin IR. The peroxisome proliferator-activated receptor gamma agonist rosiglitazone ameliorates murine lupus by induction of adiponectin. J Immunol. 2009;182(1):340–346. doi: 10.4049/jimmunol.182.1.340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Venegas-Pont M, Sartori-Valinotti JC, Maric C, Racusen LC, Glover PH, McLemore GR, Jr, Jones AV, Reckelhoff JF, Ryan MJ. Rosiglitazone decreases blood pressure and renal injury in a female mouse model of systemic lupus erythematosus. Am J Physiol Regul Integr Comp Physiol. 2009;296(4):R1282–R1289. doi: 10.1152/ajpregu.90992.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Zhao W, Thacker SG, Hodgin JB, Zhang H, Wang JH, Park JL, Randolph A, Somers EC, Pennathur S, Kretzler M, Brosius FC, 3rd, et al. The peroxisome proliferatoractivated receptor gamma agonist pioglitazone improves cardiometabolic risk and renal inflammation in murine lupus. J Immunol. 2009;183(4):2729–2740. doi: 10.4049/jimmunol.0804341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Juarez-Rojas JG, Medina-Urrutia AX, Jorge-Galarza E, Caracas-Portilla NA, Posadas-Sanchez R, Cardoso-Saldana GC, Goycochea-Robles MV, Silveira LH, Lino-Perez L, Mas-Oliva J, Perez-Mendez O, et al. Pioglitazone improves the cardiovascular profile in patients with uncomplicated systemic lupus erythematosus: a double-blind randomized clinical trial. Lupus. 2012;21(1):27–35. doi: 10.1177/0961203311422096. [DOI] [PubMed] [Google Scholar]

RESOURCES