+ All Categories
Home > Documents > Inflammatory biomarkers for predicting cardiovascular disease

Inflammatory biomarkers for predicting cardiovascular disease

Date post: 13-Nov-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
52
Inflammatory biomarkers for predicting cardiovascular disease Lee Stoner, Adam A. Lucero, Barry R. Palmer, Lynnette M. Jones, Joanna M. Young, James Faulkner PII: S0009-9120(13)00275-0 DOI: doi: 10.1016/j.clinbiochem.2013.05.070 Reference: CLB 8412 To appear in: Clinical Biochemistry Received date: 26 December 2012 Revised date: 27 May 2013 Accepted date: 30 May 2013 Please cite this article as: Stoner Lee, Lucero Adam A., Palmer Barry R., Jones Lynnette M., Young Joanna M., Faulkner James, Inflammatory biomarkers for predicting cardio- vascular disease, Clinical Biochemistry (2013), doi: 10.1016/j.clinbiochem.2013.05.070 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Transcript

�������� ����� ��

Inflammatory biomarkers for predicting cardiovascular disease

Lee Stoner, Adam A. Lucero, Barry R. Palmer, Lynnette M. Jones, Joanna M.Young, James Faulkner

PII: S0009-9120(13)00275-0DOI: doi: 10.1016/j.clinbiochem.2013.05.070Reference: CLB 8412

To appear in: Clinical Biochemistry

Received date: 26 December 2012Revised date: 27 May 2013Accepted date: 30 May 2013

Please cite this article as: Stoner Lee, Lucero Adam A., Palmer Barry R., Jones LynnetteM., Young Joanna M., Faulkner James, Inflammatory biomarkers for predicting cardio-vascular disease, Clinical Biochemistry (2013), doi: 10.1016/j.clinbiochem.2013.05.070

This is a PDF file of an unedited manuscript that has been accepted for publication.As a service to our customers we are providing this early version of the manuscript.The manuscript will undergo copyediting, typesetting, and review of the resulting proofbefore it is published in its final form. Please note that during the production processerrors may be discovered which could affect the content, and all legal disclaimers thatapply to the journal pertain.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 1

INFLAMMATORY BIOMARKERS FOR PREDICTING CARDIOVASCULAR DISEASE

Running title: Inflammatory Biomarkers

Lee Stoner1*

, Adam A. Lucero1, Barry R. Palmer

2, Lynnette M. Jones

3, Joanna M. Young

4, James Faulkner

1

1School of Sport and Exercise, Massey University, Wellington, New Zealand.

2Institute of Food, Nutrition and Human Health, Massey University, New Zealand.

3School of Physical Education, Sport and Exercise Sciences, University of Otago, Dunedin, New Zealand.

4Lipid and Diabetes Research Group, Diabetes Research Institute, Christchurch, New Zealand.

*Corresponding Author: Lee Stoner, Ph.D.

Email: [email protected]

Telephone: +64.4.801.5799 ext 6240

Fax: +64.4.801.4994

Address: School of Sport and Exercise, Private Bag 756, Massey University, Wellington 6140, New Zealand.

Word Count: 9,488

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 2

ABSTRACT

The pathology of cardiovascular disease (CVD) is complex; multiple biological pathways have been implicated,

including, but not limited to, inflammation and oxidative stress. Biomarkers of inflammation and oxidative stress

may serve to help identify patients at risk for CVD, to monitor the efficacy of treatments, and to develop new

pharmacological tools. However, due to the complexities of CVD pathogenesis there is no single biomarker

available to estimate absolute risk of future cardiovascular events. Furthermore, not all biomarkers are equal; the

functions of many biomarkers overlap, some offer better prognostic information than others, and some are better

suited to identify/predict the pathogenesis of particular cardiovascular events. The identification of the most

appropriate set of biomarkers can provide a detailed picture of the specific nature of the cardiovascular event. The

following review provides an overview of existing and emerging inflammatory biomarkers, pro-inflammatory

cytokines, anti-inflammatory cytokines, chemokines, oxidative stress biomarkers, and antioxidant biomarkers. The

functions of each biomarker are discussed, and prognostic data are provided where available.

Keyword: biomarkers, inflammation, oxidative stress, cardiovascular disease, chemokines, cytokines

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 3

INTRODUCTION

Multiple biological pathways have been implicated in the etiology of cardiovascular disease (CVD), including, but

not limited to inflammation and oxidative stress. The identification of biological markers of inflammation and

oxidative stress may assist the physician in monitoring the efficacy of treatments, and facilitate the development of

new pharmacological tools for patients at risk of CVD. However, due to the complexities of CVD pathogenesis there

is no single biomarker available to estimate absolute risk of future cardiovascular events. Furthermore, not all

biomarkers are equal; the functions of many biomarkers overlap, some offer better prognostic information than

others, and some are better suited to identify/predict the pathogenesis of particular cardiovascular events. The

following review provides an overview of existing and emerging biomarkers of inflammation and oxidative stress.

BIOMARKER DEFINITION

The US National Institutes of Health/Food and Drug Administration in 2001 defined a biomarker as a characteristic

that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or

pharmacological responses to a therapeutic intervention [1]. The levels of an ideal biomarker should be measurable

in a sample such as blood, urine, or tissue biopsy and should relate to a clinical phenotype either causally or

indirectly.

INFLAMMATORY BIOMARKERS

Atherosclerosis, the precursor to CVD, is recognized as a chronic inflammatory disease of large arteries [2, 3].

While the process of atheroma formation has been extensively reviewed elsewhere [4], some of the relevant steps in

atheromatous plaque development are briefly outlined here. Pathological studies show the abundant presence of

inflammatory cells, like monocyte-derived macrophages and T-lymphocytes at the site of rupture or superficial

erosion [3, 5]. These morphological characteristics are preceded by dysfunction of activated endothelial cells, which

produce adhesion molecules that interact with inflammatory cells [3, 6]. The ability of monocyte-derived

macrophages to secrete various cytokines, chemokines, growth-factors, and disintegrins, then leads to activation and

proliferation of smooth muscle cells (SMCs), lesion progression, and finally to the weakening of a vulnerable plaque

by matrix degradation of its fibrous cap [5]. Once inflammation has been triggered and cytokine release is initiated

at the onset of atherosclerotic lesion development, a number of factors that are found in the atherosclerotic plaque

can participate in maintaining and amplifying cytokine production, including: adipokines, angiotensin II (ANG II),

heat shock proteins (HSPs), immune complexes, reactive oxygen species (ROS), and pro-inflammatory cytokines

(Table 1).

ADVANCED GLYCATION END PRODUCTS (AGEs)

AGEs, the products of non-enzymatic glycation and oxidation of proteins and lipids, accumulate in the vessel wall

following oxidative stress [7]. AGEs may exert their pathogenic effects by engaging cellular binding sites/receptors.

The interaction of AGEs with macrophages activates macrophages in an NF-κB-dependent fashion, leading to the

induction of platelet-derived growth factor (PDGF), insulin-like growth factor (IGF)-I, and pro-inflammatory

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 4

cytokines, such as interleukin-1β (IL-1β) and tumour necrosis factor- α (TNF-α) [8]. Binding of AGEs to the

endothelial receptor for advanced glycation end products (RAGE) results in the depletion of cellular antioxidant

defence mechanisms (e.g., glutathione, vitamin C) [9] and the generation of reactive oxygen species (ROS) [10]. As

a consequence of the increased cellular oxidative stress, AGE-activated endothelial cells express pro-coagulant

tissue factor and adhesion molecules such as E-selectin, intracellular cell-adhesion molecule-1 (ICAM-1), and

vascular cell-adhesion molecule-1 (VCAM-1) [9, 11].

Increased levels of AGEs have been positively associated with increased arterial stiffness in relatively

healthy adults [12], and have been shown to predict all-cause [13] and cardiovascular mortality in older adults [13-

15]. Semba et al [13] reported a hazard ratio (HR) of 2.11 (95% CI: 1.27–3.49, P=0.003) for 1,013 adults aged 65 or

above with the highest tertile for adjusted plasma carboxymethyl-lysine (CML, a dominant AGE). Notably, the HR

was lower for all-cause mortality (HR: 1.84, 95%CI: 1.30-2.50, P=0.006), suggesting that high plasma CML may be

more specifically involved in CVD.

ANGIOTENSIN II (ANG II)

ANG II is the main effector of the renin-angiotensin-aldosterone system [16]. Vasoconstrictor action and anti-

natriuretic properties allow ANG II to regulate blood pressure and salt and fluid volume homeostasis [16]. ANG II

is also pro-inflammatory, inducing the production of ROS, pro-inflammatory cytokines, and adhesion molecules.

ANG II stimulates ICAM-1 and VCAM-1 expression in endothelial cells and SMCs [17-20], as well as E- and P-

selectin expression in endothelial cells [21, 22]. ANG II also enhances the functional adhesion of monocytes to

endothelial cells [23, 24] and stimulates MCP-1 production in SMCs and monocytes [25, 26]. In addition, ANG II

enhances the vascular expression of TNF-α, IL-6, and IL-1β as well as chemokines and chemokine receptors [27-

29]. These effects are primarily mediated by angiotensin II type 1 receptors (AT1R) and opposed by AT2R receptors

(AT2R); the latter receptors are also associated with pro-apoptotic and neuroprotective effects [16, 27, 30].

Increased ANG II levels have been reported in patients with hypertension [31], chronic heart failure (CHF)

[32] and unstable angina (UA) [33]. Conversely, both angiotensin-converting enzyme (ACE) inhibitors

and angiotensin II receptor blockers (ARBs) have been shown to favorably reduce left ventricular mass [34], intima-

media thickness [35] and arterial stiffness [36]. ACE inhibitors have also been shown to improve survival following

myocardial infarction (MI) [37], and to reduce the risk of cardiovascular events in patients with stable CAD [38].

E-SELECTIN

E-selectin is a member of the cellular adhesion molecule family, which also includes VCAM and ICAM-1. Each has

a plasma soluble form, which can serve as a surrogate marker for increased expression of CAM’s on vascular

endothelial cells, and reflect inflammation and activation of endothelial cells [39]. The selectin family has three

members: L-, P- and E-selectin, of which the latter is of particular interest, as it is found only on the endothelium

[40, 41]. E-selectin promotes endothelial-leukocyte interactions [42]. The adherence of certain leukocytes to the

intimal surface seems to be a subsequent event in atherogenesis, and this phenomenon is mediated in part by E-

selectin [40].

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 5

Increased levels of E-selectin have been observed in patients with acute coronary syndromes (ACS) [43],

CAD [44, 45], and UA [46, 47]. However, other studies have failed to show elevated levels of E-selectin in patients

with ACS [48] or UA [49]. An epidemiological study reported raised E-selectin levels in subjects with CAD,

although this was lost in multivariate analysis with included traditional risk factors [50]. In the British Regional

Heart Study of 643 men [51], Malik et al. reported an odds ratio (OR) of 1.13 (95% CI: 0.78-1.62) for adjusted E-

selectin, concluding that measurement of this molecule is unlikely to add much predictive information to that

provided by more established risk factors. Taken together, these findings indicate that the prognostic value of E-

selectin remains unclear [52].

HEAT SHOCK PROTEIN (HSPs)

HSPs are a group of highly conserved proteins found in the cells of all organisms, from the simplest of prokaryotes

to the most complex mammals, including man [53]. HSPs are grouped into various families according to their

molecular weight: namely the 110, 90, 70, 60, 40 kDa and low molecular weight families. HSP synthesis is

increased in response to many environmental stresses (stress-inducible), including raised temperature, oxidative

stress, nutritional deficiency, ultraviolet radiation, chemicals, viruses and ischemia-reperfusion injury [54]. Although

stress-inducible, HSPs constitute 5–10% of the total protein content in healthy growth conditions [55, 56]. HSPs

function as molecular chaperones, guiding newly formed polypeptides through folding/unfolding steps to achieve

correct functional configuration [57]. They are also involved in protein transport across intracellular membranes and

the repair of denatured proteins.

The inflammatory component of atherosclerosis might, at least in part, involve immune reactivity to HSPs

[58, 59]. HSP60 and HSP70 families have been most widely investigated. Studies have shown that HSP60 localizes

selectively in atherosclerotic lesions as opposed to non-atherosclerotic regions of the arterial wall [60]. Animal

models of atherosclerosis have shown a very early role for HSP60 in the development of the disease [61]. HSP60

might be an important auto-antigen in atherosclerosis and may play a role similar to that of oxLDL in triggering an

autoreactive T-cell response. Elevated HSP60 levels predict the progression of atherosclerosis in hypertensive

patients [62] and are associated with early atherosclerosis in clinically normal subjects [63, 64]. HSPs also have

prognostic significance in predicting morbidity and mortality due to atherosclerosis. In a cohort of 79 men with

documented CAD, significantly higher levels of anti-HSP65 antibodies were found in those who went on to have

cardiovascular events [65]. Similarly, anti-HSP65 antibodies predicted cardiovascular events (OR: 2.1, 95% CI: 1.2

to 3.9) in 386 age- and sex-matched, high risk patients, independent of conventional cardiovascular risk factors and

other inflammatory markers [66].

MATRIX METALLOPROTEINASES (MMPs)

MMPs are a family of endopeptidases, secreted by a variety of inflammatory or tumour cells as zymogens (pro-

MMPs), subsequently activated by proteinases [67]. Expression of pro-MMPs is strictly regulated [68], and their

action is inhibited by specific tissue inhibitors of metalloproteinases (TIMPs) [69]. MMPs are expressed

predominantly in macrophages, and also in vascular SMCs, lymphocytes, and endothelial cells [70]. MMPs play an

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 6

important role in diverse physiological processes such as organ development, angiogenesis, and tissue repair, and

contribute to pathology including inflammation and cancer [71]. In vascular pathology, MMPs play a role in

vascular remodelling, aneurysm formation, post-angioplastic restenosis, progression of atherosclerosis, and plaque

destabilization [72]. Several studies have indicated that MMPs can directly or indirectly affect the activity of various

cytokines that participate in inflammation and repair processes, including IFN-β, vascular endothelial growth factor

(VEGF), epidermal growth factor (EGF), and fibroblast growth factor (FGF) [73]. Of particular interest in the

context of atherosclerosis are the effects of MMPs on TGF- β, IL-1 β, and TNF-α. By activating TGF-β in vivo,

MMPs would restrain, rather than augment, inflammation. This might, at least in part, account for reduced

macrophage infiltration in atherosclerotic lesions of MMP-3-deficient apoE-/-

mice [74].

MMPs have been reported to be elevated in patients following MI [75], UA [76] and sudden cardiac death

(SCD) [77], and are considered particularly important in ACS [76, 78-80]. MMPs weaken coronary plaque caps by

degrading the extracellular matrix (ECM) at certain locations, often in the shoulder areas of the plaque [81]. Focal

destruction of the ECM renders the plaque less resistant to mechanical stresses imposed during systole, thereby

making the plaque vulnerable to rupture. Disruption of an atherosclerotic plaque accounts for more than two-thirds

of acute cardiovascular events [82, 83].

MYELOPEROXIDASE (MPO)

MPO is a heme protein that is secreted by phagocytic white blood cells when they become activated. The principal

sources of MPO are activated neutrophils and monocytes. MPO is proposed to be an active mediator of

atherogenesis [84]. In support of a pro-atherogenic role of MPO in vivo, expression of human MPO in macrophages

of LDL receptor-deficient mice led to a 2-fold increase in atherosclerotic lesion size [85]. MPO has also been

identified in human plaques [86] and has been found to exert potent pro-atherogenic effects. These include oxidation

of LDL, rendering it atherogenic [87], as well as oxidative modification of apolipoprotein AI (ApoAI), attenuating

its capacity to promote cholesterol efflux [88, 89]. MPO activity also diminishes nitric oxide (NO) bioavailability,

which leads to endothelial dysfunction [90, 91] – the precursor to atherosclerosis [92, 93].

MPO levels were independently predictive of mortality in acute MI patients [94, 95]. MPO levels are also

higher in patients with CAD and can predict future cardiovascular events in this cohort, even after correction for

traditional risk factors and CRP [94, 96, 97]. Elevated MPO levels also predicted future risk of CAD in apparently

healthy individuals [98]; however, the association of MPO with future CAD was weaker than that of traditional

cardiovascular risk factors and CRP.

PLATELET ENDOTHELIAL CELL ADHESION MOLECULE-1 (PECAM-1)

Also known as CD31, PECAM-1is a member of the immunoglobulin gene superfamily [99]. PECAM-1 is expressed

at high density at the lateral borders of endothelial cells and at lower density on the surface of hematopoietic and

immune cells [100]. PECAM-1 is involved in angiogenesis [101], integrin regulation [102], apoptosis [103] and

more importantly, transendothelial migration of monocytes [104], and also plays an important role in plaque

formation and thrombosis [105].

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 7

PECAM-1 gene polymorphisms and elevated PECAM-1 levels are associated with atherosclerosis, CAD

and MI [106-109]. PECAM-1 may contribute to the pathogenesis of atherosclerosis through its ability to mediate

leukocyte infiltration. Oxidized LDL has been shown to promote monocyte migration through cytokine-stimulated

endothelial cells in vitro by a mechanism involving up-regulation of endothelial cell PECAM-1 and down-regulation

of VE-cadherin [110]. Furthermore, in light of the fact that diabetic patients exhibit a higher incidence of

atherosclerosis, it is of relevance that high glucose and insulin levels can promote increased monocyte migration

through cultured endothelial cells in a PECAM-1–dependent manner [111, 112].

INTRACELLULAR CELL-ADHESION MOLECULE-1 (ICAM-1)

Soluble ICAM-1 is expressed on the surface of endothelial cells, leukocytes and SMCs in reaction to stimuli such as

shear stress, bacterial toxins, pro-inflammatory cytokines and oxidants. ICAM-1 can be induced by IL-1 and TNF-α

and is expressed by the vascular endothelium, macrophages and lymphocytes. ICAM-1 mediates attachment of

circulating leukocytes to the endothelium and their subsequent transmigration and accumulation in the arterial

intima [113, 114], processes critical to the development and progression of atherosclerosis [3]. The circulating

soluble form of ICAM-1 (sICAM-1) released from endothelial cell membranes may therefore reflect on-going

atherosclerosis.

ICAM-1 has been implicated in mature atherosclerotic lesions [115]; however, its role in initiation and

formation of lesions is overshadowed by the contribution of VCAM-1 [116, 117]. Nonetheless, in several cross-

sectional studies, ICAM-1 levels were reported to be higher in patients with CAD [118-121], and have been

significantly associated with death due to cardiovascular events in patients with CAD [45]. Furthermore, higher

levels of ICAM-1 have been shown to predict future cardiovascular events, including SCD in those free of clinical

CVD at baseline [77, 122-124].

VASCULAR CELL ADHESION MOLECULE-1 (VCAM-1)

VCAM-1 is expressed on both large and small vessels after the endothelial cells are stimulated by cytokines [125].

These molecules facilitate the rolling, adhesion and migration of leukocytes across the endothelial barrier. Up-

regulation of VCAM-1 in endothelial cells by cytokines occurs as a result of increased gene transcription (e.g., in

response to TNF-α and IL-1) and through stabilization of mRNA (e.g., IL-4).

In large prospective studies of healthy individuals, sICAM-l, but not sVCAM-1, appears consistently

related to incident CAD [50, 51, 122, 126, 127]. sICAM-l correlates with acute phase reactants like CRP, and

provides similar predictive information to CRP in settings of primary prevention [126]. sICAM-l, therefore, appears

as a general marker of a pro-inflammatory status [128]. By contrast, VCAM-1 is not expressed in baseline

conditions, but is rapidly induced by pro-atherosclerotic conditions in animal models and in humans [129].

Therefore, it seems that sVCAM-1 does not appear as a risk factor in healthy individuals, but emerges as a strong

risk predictor in patients suffering from pre-existing disease. Support for a different role of ICAM-1 and VCAM-1

according to the type of population comes from studies on peripheral arterial disease (PAD); in healthy individuals,

ICAM-1 predicts symptomatic disease [123], whereas in patients with established disease, VCAM-1 is a better

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 8

marker of the extent and severity of atherosclerosis [130, 131]. In a prospective cohort of 1,246 CAD patients,

VCAM, ICAM, and E-selectin were independently higher in patients with future death from cardiovascular causes

[45]. However, sVCAM-1 levels revealed the strongest association with future death from cardiovascular causes,

and in a Cox regression model that simultaneously controlled for all inflammatory and soluble adhesion markers,

only VCAM-1 remained independently significant for future cardiovascular events (HRR: 2.8, 95% CI: 1.4-5.4, P =

0.003). VCAM-1 has also been reported to predict future cardiovascular events patients with CAD [45, 51], type 2

diabetes [132, 133] or UA [47].

SUMMARY

MMPs play a role in plaque destabilization [72] and have been reported to be elevated in patients following the

development of CVD [75-77], in particular ACS [76, 78-80]. MMPs may be useful for prognosis in patients with

ACS. Conversely, a number of the biomarkers may be particularly useful for illuminating the early stages of

atherosclerosis. MPO may exert important oxidative effects [87-89], including diminished NO bioavailability [90,

91], which may lead to lead to endothelial dysfunction, the precursor to atherosclerosis [92, 93]. However, while

elevated MPO levels have been reported to predict future risk of CAD in apparently healthy individuals [98], this

association with CAD was weaker than that of traditional cardiovascular risk factors. Likewise, AGE has been found

to induce pro-inflammatory cytokines [8], and following binding to RAGE, results in the production of ROS [10],

which would also lead to endothelial dysfunction. Not surprisingly, AGE has been associated with raised central

arterial stiffness, an early marker of CVD [134, 135], as well as CVD mortality [13], though further longitudinal,

predictive studies are warranted. ANG II has also been found to induce the production of ROS, as well as pro-

inflammatory cytokines, and adhesion molecules. High levels of ANG II have been found in hypertensive patients,

and ARBs have been shown to reduce intima-media thickness [35] and arterial stiffness [36], suggesting the ANG II

may be involved in the early development of atherosclerosis. However, the predictive capacity of this molecule

remains to be determined. Lastly, it has been postulated that the inflammatory component of atherosclerosis might,

at least in part, involve immune reactivity to HSPs [57]. HSPs represent an emerging, potentially exciting

biomarker, but longitudinal predictive studies are required. HSPs have been associated with early atherosclerosis in

clinically normal subjects [63, 64], but the majority of the literature has looked at individuals with pre-existing

disease [62, 65, 66],

The cellular adhesion molecule family, including E-selectin, VCAM and ICAM-1, serve has soluble

markers for the increased expression of CAM’s on vascular endothelial cells, and reflect inflammation and

activation of endothelial cells [39]. However, while these markers may represent the early onset of inflammation,

their prognostic value remains unclear [52]. For example, it has been reported that adjusted E-selectin levels are

unlikely to add much predictive information to that provided by more established risk factors [51]. Likewise,

VCAM-1 does not appear as a risk factor in healthy individuals, but does emerge as a strong risk predictor in

patients suffering from pre-existing disease [129].

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 9

CYTOKINES

Most cytokines are glycoproteins that are secreted by cells using classical secretory pathways. Cytokines consist of

more than 50 secreted factors involved in intercellular communication, which regulate fundamental biological

processes including body growth, lactation, adiposity, and haematopoiesis [73, 136]. They are especially important

for regulating inflammatory and immune responses and have crucial functions in controlling both innate and

adaptive immunity. Indeed, the pathogenesis of atherosclerosis involves a complex interplay between cytokines,

chemokines and adhesion molecules, leading to monocyte infiltration and multiple other leukocyte responses within

the arterial wall [2, 73].

Once produced, cytokines are rapidly trapped by neighbouring cells via their high-affinity receptors.

Accordingly, measuring the levels of circulating cytokines is not necessarily a perfect surrogate end point.

Nevertheless, a variety of plasma inflammatory markers have been shown to predict future cardiovascular risk. They

may be useful for risk stratification and also to identify patients who might benefit from targeted therapy. Cytokines

are often classified according to their pro- or anti-inflammatory activities (see Table 2 and Table 3). The balance

between pro- and anti-inflammatory cytokines has emerged as a major determinant of plaque stability [73].

PRO-INFLAMMATORY CYTOKINES

CD40/CD40L

CD40 ligand (CD40L) is a trimeric, transmembrane protein of the TNF family, and together with its receptor CD40

is an important contributor to the inflammatory processes that lead to atherosclerosis, plaque destabilization, and

thrombosis [137-139]. A large variety of immunological and vascular cells express CD40 and/or CD40L [140].

CD40L directly regulates platelet-dependent inflammatory and thrombotic responses that contribute to the

pathogenesis of atherothrombosis [141]. Both CD40 and CD40L are present in human atherosclerotic plaques.

Interestingly, only membrane CD40L is on the surface of platelets [142], but non-platelet derived CD40L [143] can

activate endothelial cells and upregulate adhesion molecules, pro-inflammatory cytokines and chemokines in vitro.

This might limit vascular inflammation following the cleavage of CD40L from the surface of activated platelets

[143].

It has been reported that apparently healthy women with elevated levels of CD40L have an increased risk

of MI, stroke, or cardiovascular death, a finding that remained after adjustment for traditional cardiovascular risk

factors [144]. Furthermore, among patients with carotid atheroma, sCD40L levels predicted the presence of an

intraplaque lipid pool on high resolution carotid magnetic resonance imaging [145]. Platelet stimulation is the major

source of circulating CD40L, suggesting that the CD40L levels may be of greatest predictive value among those

with ACS. Consistent with this hypothesis, sCD40L levels identify patients at risk of having recurrent ischemic

events [146]. The CAPTURE study demonstrated that elevated levels of CD40L identified the subgroup of patients

with ACS who were at highest risk of death or nonfatal MI over a 6-month follow-up [146].

C-REACTIVE PROTEIN (CRP)

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 10

C-reactive protein (CRP), an acute-phase protein, is a potentially important etiological factor in inflammation and

atherosclerosis [147]. Production occurs primarily in the liver, by the hepatocytes as part of the acute phase response

upon stimulation by IL-6, and to a lesser degree by TNF-α and IL-1β, originating at the site of inflammation. Among

other effects, recombinant CRP has been shown to enhance the expression of ICAM-1, VCAM-1, E-selectin, and

MCP-1 in endothelial cells [148, 149]. CRP has been extensively studied, and there is now robust evidence from

primary prevention cohorts and among patients presenting with ACS that elevated CRP levels predict future

cardiovascular events [150]. The lack of correlation between LDL-cholesterol and CRP observed in the Women’s

Health Study allowed the identification of a subgroup of subjects with increased incidence of CAD, but normal lipid

profiles [151]. CRP levels also were found to correlate only marginally to Lp-PLA2 [152, 153] and MCP-1 [154],

and were unrelated to CD40L in patients with UA.

Several papers have demonstrated that most of the in vitro effects of recombinant CRP previously reported

in the literature were most likely artifactual and due to the presence of sodium azide [155] or contamination by

bacterial products [149] in the commercial CRP preparation used in the experiments. Moreover, in vivo experiments

assessing the direct role of CRP on atherosclerosis in CRP transgenic apoE-/-

mice failed to observe any effect [156],

or reported a very small effect in male, but not female mice [157]. There is even some evidence that CRP might be

protective against atherosclerosis [158, 159] and has a clear anti-inflammatory activity that protects mice from

lethality due to LPS challenge [160]. It is therefore unlikely that CRP is a mediator of atherosclerosis and its

complications, even though it appears to be a strong independent predictor of cardiovascular events.

INTERFERON-GAMMA (IFN- γ)

IFN-γ is a Th1 cytokine that is produced by T and NK cells following synergistic activation by IL-12 and IL-18.

Previous studies have shown that IFN-γ plays a major role in atherosclerosis. IFN-γ receptor deficiency was

associated with a reduction in atherosclerotic lesion size in apoE-/-

mice [161], and cholesterol diet-induced

atherosclerosis in LDLr-/-

mice was significantly reduced in the absence of IFN-γ [162]. Moreover, IFN-γ

administered intraperitoneally promoted atherosclerosis in apoE-/-

mice [163]. It has been suggested that IFN-γ may

affect atherosclerosis in a gender-specific manner, IFN-γ being pro-atherogenic only in males [164]. However, two

studies show conflicting effects of IFN-γ on atherosclerosis in female mice [161, 165]. In humans, IFN-γ levels

significantly correlate with CRP (r = 0.782, P<0.001, [166]) and TNF-α (r = 0.702, P value not reported, [167]),

have been shown to be raised in patients with MI [166, 168], PAD [169], CAD [120, 170], and CHF [171], and

predict death following MI (OR 1.119, 95% CI: 1.005-1.246, P=0.040) [167].

INTERLEUKIN-1 (IL-1)

Interleukins are an important group of inflammatory mediators, released from various cells into the blood and

interstitium where they bind to interleukin receptors on cell surfaces and can induce cellular activation. Some

interleukins are well-documented as aggravating (e.g., IL-6) the inflammatory response, while others are

documented to reduce (e.g, IL-10) inflammation. The pathogenic role of IL-1 has been investigated in apoE-/-

mice

fed a cholesterol-rich diet receiving subcutaneous administration of recombinant human IL-1Ra [172], and in LDLr-

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 11

/- mice [173] or apoE

-/- mice [174] crossed with transgenic mice expressing high levels of IL-1R. Over-expression of

IL-1Ra increased total cholesterol levels by ~50%, and in spite of this, decreased the size of atherosclerotic lesions

by 50–70%. In contrast, IL-1ra knockout C57BL/6J mice fed a cholesterol/cholate diet had a threefold decrease in

non-HDL cholesterol and a trend toward increased foam-cell lesion area compared with wild-type littermate controls

[173]. Taken together, these results clearly indicate that IL-1 contributes to atherosclerosis in mice.

Increased levels of IL-1 have been reported in patients with CAD [121], UA [175], acute MI [176], and

have been associated with adverse events following coronary stenting [177]. Patti et al [176] investigated IL-1

receptor antagonist (IL-1Ra) values in patients (N = 44) who reported to the emergency department with acute MI.

IL-1Ra values were elevated (>230 pg/ml) in 82% of patients, compared to 57% with raised CRP, 57% with raised

troponin I, 48% with raised myoglobin, and 41% with raised creatine kinase. These findings suggest that IL-1 may

be a sensitive marker of clinical instability in patients with CAD.

INTERLEUKIN (IL-6)

IL-6 is a pleiotropic cytokine produced by a variety of cells including fibroblasts, endothelial cells, mononuclear

phagocytes, neutrophils, hepatocytes, lymphocytes (T and B) and neural tissue - neurons, astrocytes and glial cells

[178]. IL-6 acts on a wide range of tissues influencing cell growth and differentiation, including angiogenesis, re-

vascularisation, and healing. In healthy individuals, IL-6 is expressed at low levels, kept in check by a complex

network that comprises glucocorticoids, catecholamines and secondary sex steroids [179].While the specific role of

IL-6 in the progression of atherosclerosis remains unclear, IL-6 has been shown to enhance fatty lesion development

in mice [180], and positively correlates with the degree and number of atherosclerotic plaques in ApoE knockout

mice [181]. IL-6 also mediates the synthesis and secretion of CRP [182], and is involved in the induction of MMP

[183], therefore playing an important role in the instability of vulnerable plaque. However, IL-6 also induces the

synthesis of IL-1Ra and release of soluble TNF receptor leading to reduced activity of pro-inflammatory cytokines

[184-186]. Taken together, these findings suggest that IL-6 levels in a physiological range are necessary to keep

inflammatory responses in check [179].

IL-6 levels appear to be predictive of death due to CVD [187], and are elevated in patients with stable CAD

[120], UA [76, 188] acute MI [76, 189, 190] and CHF [191], and are associated with adverse outcomes in patients

with ACS [190, 192, 193]. Patients with persistently elevated IL-6 levels demonstrate a worse in-hospital outcome

following admission with UA [194]. In the FRISC II trial, IL-6 was an independent predictor of mortality among

patients presenting with ACS, even when high sensitivity CRP was included in the analysis [195]. Interestingly,

elevated IL-6 levels appeared to have utility in terms of directing subsequent care. Early invasive strategies in

patients with elevated IL-6 levels led to a 65% relative reduction in mortality at 1 year [195]. In contrast, among

patients with lower levels of IL-6, randomization to an early invasive strategy did not confer any benefit over a

conservative strategy.

TUMOR NECROSIS FACTOR-α (TNF-α)

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 12

TNF-α is a cytokine with a wide range of pro-inflammatory activities [196, 197]. It is primarily produced by

macrophages, endothelial cells, and SMCs of atherosclerotic arteries [198]. TNF-α may influence the atherosclerotic

process both by causing metabolic perturbations and by increasing the expression of cellular adhesion molecules.

TNF-α induces the expression of surface leukocyte adhesion molecules [199, 200], chemokines [201] and enhances

the production of cytokines and growth factors [202]. TNF-α also stimulates new vessel formation and induces

features characteristic of developing atheroma.

Disturbances in the TNF-α metabolism have been implicated in metabolic disorders, such as obesity and

insulin resistance [203], indicating that perturbations of TNF-α metabolism may affect the onset of non-insulin-

dependent diabetes mellitus and play a role in the development of cardiovascular disorders. Indeed, increased

plasma concentrations of TNF-α have been found in patients with premature CAD [121, 204, 205], acute MI [206],

PAD [169], and CHF [191].

ANTI-INFLAMMATORY CYTOKINES

ADIPONECTIN

Adiponectin is a protein produced by adipocytes and present at high concentration in human peripheral circulation

[207-209]. Adiponectin exerts important cardiovascular functions, including anti-inflammatory, anti-apoptotic and

anti-hypertrophic effects [210]. Adiponectin also controls monocyte adhesion to the vascular endothelium [211] and

is an important regulator of endothelial nitric oxide synthase (eNOS) [212]. In humans, adiponectin levels appear to

be closely correlated with forearm reactive hyperaemia (endothelial function)[213], flow-mediated-dilation

(endothelial function) [214] and carotid intima-media thickness [214, 215]. Anti-inflammatory actions include

reduced TNFα-stimulated expression of E-selectin, nuclear factor-κB, VCAM-1 and IL-8 [211, 216, 217]. These

anti-inflammatory and anti-atherogenic properties appear to have important protective effects at the cardiovascular

level [207-209].

Adiponectin is reduced in the serum of both diabetic and obese individuals, and is further decreased in

patients with CAD [218, 219]. Improvement of glycaemic control with diet, weight loss and hypoglycaemic agents,

has resulted in normalization of adiponectin levels [220], suggesting an intimate connection between this adipokine

and metabolic control. In patients with CHF, levels of adiponectin were inversely related to mortality independent of

other risk markers [221]. High levels of adiponectin have also been associated with lower risk for MI [222], even

after correction for HDL- and LDL-cholesterol and BMI. Furthermore, elevated circulating concentrations of

adiponectin are associated with a lower risk for CAD independently of other well-known risk factors [223].

INTERLEUKIN-4 (IL-4)

IL-4 is a pleiotropic cytokine produced by Th2 lymphocytes, eosinophils, basophils, and mast cells. Since IL-4

inhibits the synthesis of pro-inflammatory cytokines it is generally considered anti-inflammatory [224-228].

However, a body of evidence indicates that IL-4 may also play a role in atherosclerosis through induction of

inflammatory responses, such as upregulation of VCAM-1 [229] and MCP-1 [230]. Consistent with this hypothesis,

transplantation of bone marrow stem cells from IL-4-deficient LDLr-/-

mice decreased atherosclerotic lesion

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 13

formation in a site-specific manner [231]. In contrast, IL-4 deficiency in C57BL/6 mice fed an atherogenic diet did

not affect the development of early lesions [232]. Findings are more consistent in human studies. Szkodzinski et al

[233] investigated 53 patients with acute MI undergoing percutaneous coronary intervention; higher IL-4

concentrations significantly characterized left ventricular dysfunction (ejection fraction <30%) when measured prior

to (specificity = 79%, P <0.001) or immediately post (specificity = 67%, P <0.001) surgery. IL-4 has also been

shown to be elevated in patients with CAD [120]. Furthermore, in type 2 diabetic subjects, mixed modality physical

exercise upregulates IL-4 and IL-10 and down-regulates inflammatory markers (hs-CRP, IL-1β, IL-6, TNF-α and

IFN- γ) [234, 235].

INTERLEUKIN-10 (IL-10)

IL-10 is a pleiotropic cytokine produced by Th2 cells, B cells, monocytes, and macrophages that inhibits a broad

array of immune parameters including the expression Th1 cytokines, antigen presentation, and antigen-specific T-

cell proliferation [236]. IL-10 also has potent anti-inflammatory properties on macrophages [237] and plays an

active role in limiting the inflammatory response in the vessel wall [238]. The role of endogenous IL-10 has been

clearly established in mouse models of atherosclerosis. IL-10 deficiency in C57BL/6 mice fed an atherogenic

cholate-containing diet promotes early atherosclerotic lesion formation, characterized by increased infiltration of

inflammatory cells and by increased production of pro-inflammatory cytokines [239]. Furthermore, in IL-10

deficient LDLr -/-

mice, it was found that IL-10 is instrumental in the prevention of atherosclerotic lesion

development [240].

Decreased IL-10 levels have been reported in patients with acute MI [168], UA [241] and other ACS [242].

Heeschen et al [242] assessed the prognostic impact of CRP and IL-10 in patients (n = 547) presenting with ACS;

IL-10 was inversely correlated with CRP (r = -0.31, P = 0.49), and patients with elevated IL-10 (>3.5 pg/mL) had

decreased risk for MI and death at 24 hours (HR: 0.19, 95% CI: 0.04-0.88) and 6 months (HR: 0.43, 95% CI: 0.25-

74). Notably, the predictive value of IL-10 was restricted to patients with elevated CRP levels, indicating the

complex interplay between inflammatory and anti-inflammatory molecules.

TRANSFORMING GROWTH FACTOR (TGF-β)

TGF-β is a potent anti-inflammatory, immunosuppressive and pro-fibrotic cytokine, with major effects on the

biology of SMCs. Three TGF-β isoforms (TGF-β1, TGF-β2, and TGF-β3) are expressed by cells in the vessel wall

and are capable of modulating vascular development and remodelling by altering cell differentiation, proliferation,

migration, extracellular matrix production, and the activities of immune cells [243] [244]. TGF-β appears to

determine the extent to which developing atherosclerotic lesions are stabilized by a collagen-rich fibrous cap.

Indeed, SMCs in stable lesions express greater amounts of TGF- β than unstable lesions [245]. In mice, specific

inhibition of TGF-β signalling in T cells leads to the development of atherosclerotic plaques with a phenotype that

may potentially increase plaque vulnerability to rupture [246-248]. There is also evidence that TGF-β1 down-

regulates inflammatory cytokine-induced expression of VCAM-1[249], leukocyte adhesion to the endothelium

[250], decreases macrophage activity by releasing IL-1 Ra [251], and is inversely correlated (-0.27, P=0.05) to CRP

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 14

[252]. Taken together, these findings suggest that TGF-β plays a pivotal role in the maintenance of normal blood

vessel wall architecture, and protects against vulnerability to atherosclerosis

In humans, genetic polymorphisms and defective TGF-β signalling have been linked to the development of

a number of cardiovascular diseases, including CAD [253], CHF [254], aortic aneurysm [255, 256], hypertrophic

cardiomyopathy [257], hypertension [258, 259], rheumatic heart disease [260], stroke [261, 262], as well as acute

MI [253, 263, 264] and SCD [77, 265]. Decreased levels of TGF-β1 have been reported in patients with CAD, CVD

and PAD compared to controls [252], and in patients receiving regular haemodialysis, a 1 ng/mL reduction in TGF-

β1 concentration was associated with a 9% increase in the relative risk of a cardiovascular events [252].

SUMMARY

The balance between pro- and anti-inflammatory cytokines may be important for risk stratification and also to

identify patients who might benefit from targeted therapy. The pro-inflammatory cytokines, CD40/CD40L, CRP,

and IFN-γ, appear to play an important role in atherosclerosis, has been shown to be elevated in patients with several

CVDs [120, 166, 168-171], and to predict death following MI [167]. TNF-α may play a particularly important role

in metabolic disorders such as obesity and insulin resistance [203], and increased plasma concentrations of TNF-α

have been found in patients with a range of CVD [121, 169, 191, 204-206]. CD40L levels identify patients at risk of

having recurrent ischemic events [146], and elevated levels of CD40L identified the subgroup of patients with ACS

who were at highest risk of death or nonfatal MI [146], suggesting that CD40L may be of greatest predictive value

among those with ACS. Even though CRP appears to be a strong independent predictor of cardiovascular events

[150], the validity of this biomarker remains unclear; there is evidence that CRP might be protective against

atherosclerosis [158-160], and commercial CRP preparation may be contaminated by the presence of sodium azide

[155] or bacterial products [149].

Two promising anti-inflammatory cytokines include adiponectin and TGF-β. Adiponectin appears to play a

role in regulating endothelial function [211-213] and has been inversely correlated with flow-mediated dilation

(endothelial function) and carotid intima-media thickness [214, 215], two widely used and early markers of

atherosclerosis. High levels of adiponectin have been associated with a lower risk for CAD [222] and MI [223].

TGF-β appears to play a pivotal role in the maintenance of normal blood vessel wall architecture and decreased

levels of TGF-β1 have been reported in patients with several CVD [77, 191, 253, 254, 261-265].

Interleukins are an important group of inflammatory mediators, some of which aggravate the inflammatory

response, while others reduce inflammation. IL-1 and IL-6 are thought to be pro-inflammatory. Increased levels of

IL-1 have been documented in patients with a range of CVD [121, 175-177], and may serve as a sensitive marker of

clinical instability in CAD patients [176]. Raised levels of IL-6 levels have also been reported in patients with a

range of CVD [76, 120, 188-193], and have been associated with worse in-hospital outcomes following admission

with UA [194]. Conversely, IL-4 and IL-10 are considered anti-inflammatory. While findings from IL-4 studies are

inconstant, the role of endogenous IL-10 has been clearly established in mouse models of atherosclerosis, and

decreased IL-10 have been reported in patients with ACS [168, 241, 242]. Measurement of IL-1, IL-6 and IL10 may

be useful for indicating the complex interplay between inflammatory and anti-inflammatory processes.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 15

CHEMOKINES

Chemokines belong to a large group of structurally related and secretable, largely basic, chemotactic cytokines [266,

267]. Chemokines are pro-inflammatory, characterized by their ability to cause directed migration of leukocytes into

inflamed tissue. These chemotactic cytokines seem not only to be raised in the circulation, but also within

atherosclerotic lesions [73]. Furthermore, several other leukocyte responses such as cell proliferation, enzyme

secretion and induction of ROS, have been observed in vitro after chemokine stimulation [73]. Chemokines may

also interfere with SMC migration and growth, as well as platelet activation [268, 269].

INTERLUKIN-8 (IL-8)

While a wide variety of cell types are potential sources of IL-8, the principal cellular sources are typically

monocytes and macrophages [270, 271]. IL-8 induces angiogenesis [272, 273] and functions as a chemoattractant

for neutrophils and T lymphocytes, which also produce IL-8 [274]. Injection of an IL-8 inhibitor in rabbits and rats

significantly reduced neutrophil migration towards inflammatory foci [275]. In irradiated mice reconstituted with

bone marrow cells lacking the murine homologue of the IL-8 receptor CXCR2, the lesion size, as well as the amount

of macrophages within the plaques, was reduced [276].

IL-8 has been associated with acute MI [168, 189], UA [277, 278], CHF [279], and CAD [280-282]. Inoue

et al [280] evaluated the serum levels of 10 cytokines as potential markers of long-term outcomes in patients with

angiographically identified stable CAD; IL-8 was the only cytokine to predict cardiovascular events, doing so

independently of the other nine cytokines. More recently, Pronzinsky et al [168] investigated the prognostic value

of IL-1β, IL-6, IL-7, IL-8, and IL-10 in 40 patients presenting with acute MI complicated by cardiogenic shock . IL-

8 showed the highest diagnostic accuracy for infarct-related mortality over the subsequent 96 hours, with an area

under the curve (AUC) of 0.80 ± 0.08, followed by IL-6 (AUC 0.79 ± 0.08), IL-10 (AUC 0.76 ± 0.08) and IL-7

(AUC 0.69 ± 0.08). Moreover, physical exercise has reported to decrease IL-8 and MCP-1 levels in patients with

metabolic syndrome [283], suggesting that the protective effect of physical exercise might be due, in part, to the

suppression inflammatory molecules.

MONOCYTE CHEMOATTRACTANT-1 (MCP-1)

MCP-1, also known chemokine ligand 2 (CCL2), is produced by a variety of cell types, either constitutively or after

induction by oxidative stress, cytokines, or growth factors [284]. MCP-1 is one of the key chemokines that regulate

migration and infiltration of monocytes and macrophages, and appears to play a principal role in atherosclerotic

lesions. In mice over-expressing apolipoprotein B, deletion of the MCP-1 gene protected against monocyte

recruitment [285]. Similarly, in MCP-1-deficient LDLr-/-

mice, macrophage recruitment is diminished, suggesting

that MCP-1 attracts monocytes into the vessel wall [286].

MCP-1 has been associated with subclinical atherosclerosis (coronary artery calcium score) [287], CAD

[154, 281, 282, 288], PAD [154], ACS [288-290], UA [291], MI [206, 290], SCD[290] and UA [291]. de Lemos

[290] et al investigated MCP-1 levels in 2,279 patients presenting with ACS; MCP-1 levels above the 75th

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 16

percentile (238 pg/ml) were associated with an increased risk of MI or death (HR: 1.53, 95% CI: 1.09-2.14) during

follow-up (10 months) after adjustment for traditional risk factors, including CRP. Martinovic et al [288] assessed

serum MCP-1 levels in 263 patients admitted to hospital for non-invasive diagnostic procedures; elevated MCP-1

levels were found in patients with CAD or increased coronary risk factors. In addition, endothelial activation

markers such as soluble adhesion molecules, soluble intercellular adhesion molecule and soluble E-selectin were

also increased in these patients. This suggests that elevated MCP-1 levels might serve as a direct marker of

inflammatory activity for those at risk for CVD [292].

MIGRATION INHIBITORY FACTOR (MIF)

MIF is a pleiotropic inflammatory T cell and macrophage cytokine. Originally discovered and named as an inhibitor

of macrophage migration, MIF has now been shown to be a key player in acute and chronic immune-inflammatory

conditions, including atherosclerosis [293], rheumatoid arthritis [294], sepsis [295], and asthma [296]. There is also

evidence that MIF plays a key role in metabolic and inflammatory processes underlying metabolic disorders [297].

MIF is widely expressed in numerous types of tissue, and can act in an autocrine and paracrine fashion to stimulate

its own synthesis and the synthesis of other pro-inflammatory mediators [298]. An up-regulation of MIF has been

observed in endothelial cells, SMCs, and macrophages during progression of atherosclerosis [299, 300]. Inhibition

of MIF in apoE-/-

mice by treatment with neutralizing MIF antibodies resulted in a shift in the cellular composition

of neointimal plaques toward a more stable phenotype with reduced macrophage and increased SMCs content [301],

as well reduced circulating levels of inflammatory markers (fibrinogen, MIF and IL-6) [299].

In humans, the prognostic capacity of MIF in predicting future cardiac events remains elusive. One study

[302] demonstrated that high MIF levels were a marginal, but significant predictor of MI and SCD, while another

[303] reported a lack of association between serum levels of MIF and incident CAD. However, these data are based

on a group of subjects without pre-existing CAD. A more recent study [304] examined whether high circulating

levels of MIF are related to future cardiovascular events in patients with stable CAD and, after controlling for

conventional risk factors, MIF was a significant predictor of cardiovascular events in patients with type 2 diabetes

mellitus (HR: 3.3, 95% CI: 1.6–8.3), but not in patients without diabetes.

SUMMARY

Several chemokines have been widely investigated, including IL-8, MCP-1 and MIF. Of these, IL-8 and MCP-1

have shown particular promise as cardiac biomarkers. MCP-1 levels might serve as a direct marker of inflammatory

activity for those at risk for CVD [292], and hasve been associated with subclinical atherosclerosis [287] and a range

of CVD [154, 206, 281, 282, 288-291]. IL-8 has been associated with a number of CVD [168, 189, 277-282], and

appears to be a sensitive marker of long-term outcomes in patients with stable CAD [280]. The prognostic capacity

of MIF for predicating future cardiovascular events may be limited to those with pre-existing CVD or diabetes

[304].

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 17

OXIDATIVE STRESS

Oxidative stress results when free radical formation is unbalanced in proportion to protective antioxidants [305].

Free radicals are formed during a variety of biochemical reactions and cellular functions (such as mitochondria

metabolism). Examples of free radicals (oxidizing molecules) are hydrogen peroxide, hydroxyl radical, NO, singlet

oxygen, superoxide anion and peroxyl radical. Free radicals are highly reactive, unstable molecules that have an

unpaired electron in their outer shell. They bind with electrons from other molecules, potentially initiating chain

reactions as successive molecules lose and gain electrons. They oxidize various cellular components including

DNA, proteins, lipids/fatty acids and AGEs [306]. These reactions between cellular components and free radicals

lead to DNA damage, mitochondrial malfunction, cell membrane damage and eventually cell death (apoptosis).

Antioxidants are molecules or compounds that act as free radical scavengers (Table 4). Most antioxidants

are electron donors and react with the free radicals to form innocuous end products such as water. These

antioxidants bind and inactivate the free radicals. Thus, antioxidants protect against oxidative stress and prevent

damage to cells.

OXIDATIVE STRESS BIOMARKERS

ISOPROSTANES

Isoprostanes are cellular non-enzymatic products resulting from free radical–catalyzed peroxidation of arachidonic

acid [307, 308]. In contrast to enzymatically generated prostaglandins, which are generated from free arachidonic

acid, isoprostanes can be generated on intact cholesteryl esters and phospholipids, which are major components of

lipoprotein particles and cell membranes. Following generation, isoprostanes are released by phospholipase activity,

circulate in plasma, and are ultimately excreted in urine. F2 isoprostanes are stable, specific and unique end-products

(up to 64 species can be generated) of lipoprotein metabolism.

F2 isoprostane levels are raised by traditional risk factors, including cigarette smoking, diabetes mellitus,

obesity and hypertension [307], and correlate with unstable carotid plaques [309]. A recent systematic review [310]

assessed the role of F2 isoprostane as a biomarker for CVD. Of the 22 eligible studies retrieved, 20 studies showed

a significant association between F2-isoprostanes and CVD, of which only 4 were population based studies, and

only 2 were prospective. Thus, while F2 isoprostanes may be considered the gold standard in measuring oxidative

stress in vivo [311], prognostic data remain limited. Furthermore, the relative sophistication and expense required to

perform isoprostane assays inhibits widespread use.

LIPOPROTEIN-ASSOCIATED PHOSPHOLIPASE A2 (Lp-PLA2)

Lp-PLA2 is a member of phospholipase A2 superfamily [312]. The secreted isoform was first identified on the basis

of its ability to degrade platelet-activating factor (PAF), hence it is also known as PAF-acetylhydrolase [313]. Lp-

PLA2 is secreted by various inflammatory cells, including monocyte-macrophages, T-cells, and mast cells [314]. It

cleaves the ester bond of oxidized phospholipids (OxPLs) at the sn-2 position to give rise to two potentially pro-

inflammatory products, free oxidized fatty acids and lysophosphatidylcholine [315]. The biological role of Lp-PLA2

has been controversial with seemingly contradictory, with anti- or pro-atherogenic functions being proposed. The

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 18

anti-atherogenic properties of Lp-PLA2 were suggested because of the enzymatic catabolism of biologically active

oxidized phospholipids in LDL and degradation of the unrelated polar phospholipid, PAF [316, 317]. To this end,

Lp-PLA2 was reported to alter biological properties of minimally modified LDL by abrogating the ability of LDL to

promote endothelial cell binding of monocytes [317]. In contrast, Lp-PLA2 may directly promote atherogenesis by

generating potent pro-inflammatory and pro-atherogenic products, such as lysophosphatidylcholine and oxidized

free fatty acids from oxidation of LDL [315, 318, 319], an important step in atherogenesis.

Seemingly conflicting evidence has been found in population and clinical studies. For example, loss-of-

function mutations have been found to be associated with a higher risk for CVD in Japanese populations [320,

321].Three subsequent studies [152, 322, 323] reported that high levels of Lp-PLA2 independently predicted CAD

or cardiovascular events. However, in all studies, Lp-PLA2 levels were strongly correlated with LDL cholesterol

levels, and the predictive value of Lp-PLA2 was attenuated substantially after adjusting for LDL cholesterol and

traditional risk factors. Further study is required to determine the extent to which Lp-PLA2 levels provide

independent information beyond traditional risk assessments such as the Framingham risk model.

NITROTYROSINES

Nitrotyrosine, one of the most important mediators of MPO, plays a key role in the process of oxidation seen early

on in atherosclerosis [324, 325]. Nitrotyrosine accumulation reflects a loss of balance between oxidant formation

and antioxidant defence mechanisms [325]. Nitrotyrosine originates as tyrosine in both a free and protein-bound

form. The protein-bound form that is involved in atherosclerosis is attached to LDL. This molecule is then nitrated

to form the biologically active nitrotyrosine, and occurs in a highly efficient manner in human serum [326]. Once

modified, the nitrated form of LDL is collected and consumed by macrophages via phagocytosis. The end product of

this degradation is the deposition of cholesterol and foam cells that are vital in plaque development.

Several animal studies have linked nitrotyrosine to inflammation. Mice injected with Klebsiella pneumonia

produced an inflammatory fluid rich in nitrotyrosine [326]. Rats genetically engineered to be spontaneously

hypertensive have been shown to have an increased production of nitrotyrosine [327]. A study that links the

association of nitrotyrosine with the oxidative stress of renal failure showed an increase in nitrotyrosine serum levels

in uremic mice [328]. In humans, nitrotyrosine levels are raised in patients afflicted with diseases associated with

high oxidative stress such as diabetes mellitus [329], and multiple studies have reported that raised levels are

detected in atheromatous plaques [329, 330]. Nitrotyrosine has also been found to be elevated in patients with PAD

[331], CAD [332-334], and MI [335]. Systemic levels of protein-bound nitrotyrosine have been reported to be

associated with the presence of CAD even following multivariable adjustments for traditional risk factors and CRP

(OR:4.4, 95%CI: 1.8-10.6, P<0.001)[332].

OXIDIZED LDL (oxLDL)

oxLDL is thought to be a very early trigger of vascular inflammation and a major cause of injury to the endothelium

[336]. LDL accumulation and modification in the sub-endothelium triggers monocyte and lymphocyte recruitment.

Thereafter, activated macrophages and lymphocytes secrete abundant amounts of cytokines that in turn can activate

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 19

endothelial cells, SMCs, and macrophages/lymphocytes to foster cytokine production, leading to a self-perpetuating

inflammatory process that becomes less dependent on the presence of oxLDL [337]. In addition, oxLDL cholesterol

has been demonstrated to induce the production of endothelin-1 (ET-1) in human macrophages and increases ET-1

release from endothelial cells by inducing ET-1 gene expression [338]. oxLDL may therefore indirectly contribute

to endothelial dysfunction in atherosclerosis [339].

Elevated levels of oxLDL have been associated with CVD [340], ACS [341-345], acute MI [346-348], UA

[347, 349], PAD [350], CHF [351, 352], transient ischemic stroke (TIA) [340, 353, 354], CAD [343, 355, 356], and

the severity of CAD [357-359]. In a recent study by Tsimikas et al [340], oxLDL levels were found to be

significantly raised in patients with incident CVD versus controls, and after adjustment for traditional risk factors

were found to be associated with higher risk for CVD (HR: 1.18, 95% CI: 1.02-1.37, P=0.028) and stroke (HR: 1.32,

95% CI: 1.11-1.58, P=0.002) after a 15 year follow up. The time course of change in oxLDL has been measured in

patients following acute MI [360] and TIA [353]; oxLDL levels were high during admission to hospital, but

approached normal levels at the time of discharge. These findings suggest that plasma oxLDL is released into the

circulation following plaque rupture [361, 362].

ANTIOXIDANT BIOMARKERS

COENZYME Q10 (CoQ10)

CoQ10, present in all cellular membranes [363], originates from endogenous synthesis as well as from food intake

and oral supplements [364]. Tissues generally synthesize CoQ10 from farnesyl diphosphate and tyrosine [365]. Iron,

magnesium, and vitamin B6 are cofactors for CoQ10 biosynthesis. CoQ10 can be obtained from dietary sources of

meat, fish, vegetables, and fruits. CoQ10 is an important antioxidant; its reduced form, ubiquinol, protects

membrane phospholipids and mitochondrial membrane proteins, as well as DNA, from free-radical-induced

oxidative damage [366-368]. Not only can ubiquinol act to remove oxygen radicals, but it can also reduce

tocopheryl radicals and semi dehydroascorbate back to tocopherol and ascorbate, respectively [369, 370]. The

ubiquinone formed in this process is reduced back to ubiquinol in the electron transport chain by metabolic supply

of NADH or NADPH [371, 372]. Ubiquinol has also been reported to protect LDL from oxidation [373].

Myocardial tissues of CVD patients are reported to be deficient in CoQ10 [374-376]. Littarru et al [377]

first reported its deficiency in heart disease. These authors reported that 75% of cardiac surgery patients had

decreased CoQ10 in the blood and myocardium. Folkers et al [374] reported low myocardial CoQ10 in patients with

aortic and mitral wall disease, diabetic cardiopathy, and congenital valvular defects. The same group of authors

[375] reported that, compared with people with less-severe CHF (Class I and II), people with more-severe CHF

(class III and IV) had lower plasma and myocardial CoQ10, suggesting an increased risk of CoQ10 deficiency as

heart disease worsens. Decreased CoQ10 levels have also been reported in patients with hyperlipidemia [378] and

diabetes [379].

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 20

GLUTATHIONE (GSH)

GSH is an important intracellular antioxidant required for normal functioning of immune cells [380]. Certain

cellular functions, such as the DNA synthetic response, are very sensitive to ROS and are favoured in high levels of

GSH [381]. A study by Kinscherf et al [382] found that individuals with intermediate level of GSH (20-30 nmol/mg

protein) had, on average, a significantly higher number of CD4+ T-cells than those with either higher or lower

intracellular GSH levels. This indicates that the immune system is very sensitive to changes in GSH levels and that

the mean GSH levels of healthy human subjects are approximately optimal. Low GSH levels have also been

associated with impaired immune function, including impaired T cell proliferation and activation, as well as

decreased IL-2 production, impaired IL-2 responses and a shift to Th2 response as compared to Th1[383].

Decreased levels of GSH have been reported in patients with CHF [384, 385] and acute MI [383, 386]. In patients

reporting with a first acute MI, plasma-reduced GSH was independently associated with future cardiovascular

events (HR: 0.42, 95% CI: 0.18-0.99, P=0.04) [383].

SUPEROXIDE DISMUTASE (SOD)

In mammals, there are 3 isoforms of superoxide dismutase (SOD). Each isoform is the product of a distinct gene but

catalyses the same reaction. The 3 isoforms are: the cytosolic or copper-zinc SOD (CuZn-SOD SOD-1), the

manganese SOD (Mn-SOD or SOD-2) localized in the mitochondria, and the extracellular form of SOD (EC-SOD

or SOD-3). EC-SOD is the only antioxidant enzyme localized on the surface of the vascular lumen [387]. Studies in

nonvascular cells suggest the different isoforms of SOD have distinctive roles [388]. An important function of EC-

SOD in the arterial wall may be the preservation of NO bioactivity [389-391]; NO is considered the most important

molecule governing endothelial function and health [93, 392-395]. NO reacts with superoxide at a rate three times

faster than dismutation of superoxide by SOD [396, 397]. Moreover, EC-SOD degrades superoxide at a rate 4-5

times faster than antioxidants such as vitamin C or E [398]. Studies using rabbit aortas or bovine coronary arteries

have shown that inhibition of vascular SOD activity results in a rapid impairment of NO-mediated vasodilation,

suggesting that SOD levels are critical for the ability of NO to modulate vascular tone [390, 391].

The role of SOD in cardiovascular events was initially the subject of controversy [399]. Arterial expression

and/or activity of CuZn-SOD and Mn-SOD increases in several animal models of hypertension [400, 401], as well

as in the initial phases of atherosclerosis, but is decreased in the later stages of this disease [402]. Knock-out

experiments showed neonatal lethality of mice lacking Mn-SOD and reduced lifespan in mice lacking CuZn-SOD

[403-406]. EC-SOD-deficient mice had higher blood pressures in two hypertension models compared with wild-type

animals [407]. Over-expression studies of SODs strongly suggest a protective role of these enzymes in many

diseases, as well as in aging [404-406]. Indeed, SOD was reported to be increased in the initial stages of CAD to

protect and prevent lipid peroxidation, but it decreased thereafter with the severity of disease [408]. Studies in

patients with stable CAD [409, 410], UA [411] , MI [412] and SCD [413] have reported substantially reduced levels

of vascular EC-SOD.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 21

SUMMARY

A number of biomarkers have emerged that may be used to represent the balance between free radical formation and

protective antioxidants. Oxidative stress biomarkers include F2 isoprostane, LpPLA1, Nitrotyrosine, and oxLDL.

The biological role of Lp-PLA2 has been controversial with seemingly contradictory anti-atherogenic [316, 317] and

pro-atherogenic [315, 318, 319] functions being proposed. F2 isoprostane levels are raised by traditional risk factors

[307], correlate with unstable carotid plaques [309], and a recent systematic review reported a significant

association between F2-isoprostanes and CVD. However, prognostic data remain limited and the expense required

to perform isoprostane assays inhibits widespread use. Alternatively, oxLDL is thought to be an early trigger of

vascular inflammation and has been associated with a range of CVD [340-359]. oxLDL levels have also been

associated with higher risk for CVD, even after adjustment for traditional risk factors [340]. Raised nitrotyrosine

levels may also be used to indicate an imbalance between oxidant formation and antioxidant defence mechanisms

[325], and raised levels have been reported in patients with several CVD [331-335]. Nitrotyrosine levels may play a

particularly prominent role in diseases associated with high oxidative stress, such as diabetes mellitus [329].

Prominent antioxidant biomarkers include coQ10, GSH, and SOD. Decreased levels of coQ10 have been

reported in patients presenting with CVD [375, 376], and have been associated with CVD risk factors, including

hyperlipidemia [378] and diabetes [379]. Levels of GSH and SOD may be indicative of the stage of CVD. Up-

regulation of vascular EC-SOD may indicate the initial stages of CVD [408], whereas substantially decreased levels

may indicate stable CVD [409-413]. Decreased levels of GSH have been reported in patients presenting with CHF

[384, 385] and acute MI [383, 386], and reduced GSH levels were associated with future cardiovascular events in

patients with acute MI [383]. Further investigation is required to elucidate the prognostic capacity of these

potentially useful biomarkers.

CONCLUSIONS

Not surprisingly - due to the complexities of CVD pathogenesis - a single biomarker cannot be used to estimate

absolute risk of future cardiovascular events. Furthermore, particular biomarkers are more suited for the prognosis of

particular cardiovascular events, or for a given stage of a given CVD and it should also be recognized that the

functions of many biomarkers overlap. For example, soluble ICAM-1 reflects in part endothelial function and in part

inflammation, HSP60 might play a role similar to that of oxLDL in triggering an autoreactive T-cell response, and

MMPs can directly or indirectly affect the activity of various cytokines. Therefore, biomarkers should be selected

for a specific stage of a given CVD, and a particular biomarker should not be considered in isolation. Simultaneous

measurements of disease appropriate biomarkers over time can provide a more detailed picture of the specific nature

of the cardiovascular event.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 22

REFERENCES

[1] Atkinson AJ, Colburn WA, DeGruttola VG, DeMets DL, Downing GJ, Hoth DF, et al. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin Pharmacol Ther 2001;69(3):89-95.

[2] Hansson GK. Inflammation, Atherosclerosis, and Coronary Artery Disease. N Engl J Med 2005;352(16):1685-95.

[3] Ross R. Atherosclerosis -- An Inflammatory Disease. N Engl J Med 1999;340(2):115-26. [4] Fleg JL, Stone GW, Fayad ZA, Granada JF, Hatsukami TS, Kolodgie FD, et al. Detection of high-risk

atherosclerotic plaque: report of the NHLBI Working Group on current status and future directions. JACC Cardiovasc Imaging 2012;5(9):941-55.

[5] Shah PK. Mechanisms of plaque vulnerability and rupture. Journal of the American College of Cardiology 2003;41(4, Supplement 1):S15-S22.

[6] Nissen SE, Yock P. Intravascular Ultrasound : Novel Pathophysiological Insights and Current Clinical Applications. Circulation 2001;103(4):604-16.

[7] Basta G, Schmidt AM, De Caterina R. Advanced glycation end products and vascular inflammation: implications for accelerated atherosclerosis in diabetes. Cardiovasc Res 2004;63(4):582-92.

[8] Hofmann MA, Drury S, Fu C, Qu W, Taguchi A, Lu Y, et al. RAGE Mediates a Novel Proinflammatory Axis: A Central Cell Surface Receptor for S100/Calgranulin Polypeptides. Cell 1999;97(7):889-901.

[9] Bierhaus A, Chevion S, Chevion M, Hofmann M, Quehenberger P, Illmer T, et al. Advanced glycation end product-induced activation of NF-kappaB is suppressed by alpha-lipoic acid in cultured endothelial cells. Diabetes 1997;46(9):1481-90.

[10] Yan SF, Ramasamy R, Naka Y, Schmidt AM. Glycation, Inflammation, and RAGE: A Scaffold for the Macrovascular Complications of Diabetes and Beyond. Circ Res 2003;93(12):1159-69.

[11] Basta G, Lazzerini G, Massaro M, Simoncini T, Tanganelli P, Fu C, et al. Advanced Glycation End Products Activate Endothelium Through Signal-Transduction Receptor RAGE: A Mechanism for Amplification of Inflammatory Responses. Circulation 2002;105(7):816-22.

[12] Semba RD, Najjar SS, Sun K, Lakatta EG, Ferrucci L. Serum carboxymethyl-lysine, an advanced glycation end product, is associated with increased aortic pulse wave velocity in adults. Am J Hypertens 2009;22(1):74-9.

[13] Semba RD, Bandinelli S, Sun K, Guralnik JM, Ferrucci L. Plasma carboxymethyl-lysine, an advanced glycation end product, and all-cause and cardiovascular disease mortality in older community-dwelling adults. J Am Geriatr Soc 2009;57(10):1874-80.

[14] Meerwaldt R, Hartog JW, Graaff R, Huisman RJ, Links TP, den Hollander NC, et al. Skin autofluorescence, a measure of cumulative metabolic stress and advanced glycation end products, predicts mortality in hemodialysis patients. J Am Soc Nephrol 2005;16(12):3687-93.

[15] Semba RD, Ferrucci L, Sun K, Beck J, Dalal M, Varadhan R, et al. Advanced glycation end products and their circulating receptors predict cardiovascular disease mortality in older community-dwelling women. Aging Clin Exp Res 2009;21(2):182-90.

[16] Rautureau Y, Paradis P, Schiffrin EL. Cross-talk between aldosterone and angiotensin signaling in vascular smooth muscle cells. Steroids 2011;76(9):834-9.

[17] Costanzo A, Moretti F, Burgio VL, Bravi C, Guido F, Levrero M, et al. Endothelial activation by angiotensin II through NF?B and p38 pathways: Involvement of NF?B-inducible kinase (NIK), free oxygen radicals, and selective inhibition by aspirin. Journal of Cellular Physiology 2003;195(3):402-10.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 23

[18] Pastore L, Tessitore A, Martinotti S, Toniato E, Alesse E, Bravi MC, et al. Angiotensin II Stimulates Intercellular Adhesion Molecule-1 (ICAM-1) Expression by Human Vascular Endothelial Cells and Increases Soluble ICAM-1 Release In Vivo. Circulation 1999;100(15):1646-52.

[19] Pueyo ME, Gonzalez W, Nicoletti A, Savoie F, Arnal J-F, Michel J-B. Angiotensin II Stimulates Endothelial Vascular Cell Adhesion Molecule-1 via Nuclear Factor-{kappa}B Activation Induced by Intracellular Oxidative Stress. Arterioscler Thromb Vasc Biol 2000;20(3):645-51.

[20] Tummala PE, Chen X-L, Sundell CL, Laursen JB, Hammes CP, Alexander RW, et al. Angiotensin II Induces Vascular Cell Adhesion Molecule-1 Expression In Rat Vasculature : A Potential Link Between the Renin-Angiotensin System and Atherosclerosis. Circulation 1999;100(11):1223-9.

[21] Alvarez A, Cerda-Nicolas M, Naim Abu Nabah Y, Mata M, Issekutz AC, Panes J, et al. Direct evidence of leukocyte adhesion in arterioles by angiotensin II. Blood 2004;104(2):402-8.

[22] Grafe M, Auch-Schwelk W, Zakrzewicz A, Regitz-Zagrosek V, Bartsch P, Graf K, et al. Angiotensin II–Induced Leukocyte Adhesion on Human Coronary Endothelial Cells Is Mediated by E-Selectin. Circ Res 1997;81(5):804-11.

[23] AbdAlla S, Lother H, Langer A, el Faramawy Y, Quitterer U. Factor XIIIA Transglutaminase Crosslinks AT1 Receptor Dimers of Monocytes at the Onset of Atherosclerosis. Cell 2004;119(3):343-54.

[24] Kim JA, Berliner JA, Nadler JL. Angiotensin II increases monocyte binding to endothelial cells. Biochem Biophys Res Commun 1996;226(3):862-8.

[25] Chen X-L, Tummala PE, Olbrych MT, Alexander RW, Medford RM. Angiotensin II Induces Monocyte Chemoattractant Protein-1 Gene Expression in Rat Vascular Smooth Muscle Cells. Circ Res 1998;83(9):952-9.

[26] Funakoshi Y, Ichiki T, Shimokawa H, Egashira K, Takeda K, Kaibuchi K, et al. Rho-Kinase Mediates Angiotensin II-Induced Monocyte Chemoattractant Protein-1 Expression in Rat Vascular Smooth Muscle Cells. Hypertension 2001;38(1):100-4.

[27] Schmieder RE, Hilgers KF, Schlaich MP, Schmidt BM. Renin-angiotensin system and cardiovascular risk. Lancet 2007;369(9568):1208-19.

[28] Ni W, Kitamoto S, Ishibashi M, Usui M, Inoue S, Hiasa K-i, et al. Monocyte Chemoattractant Protein-1 Is an Essential Inflammatory Mediator in Angiotensin II-Induced Progression of Established Atherosclerosis in Hypercholesterolemic Mice. Arterioscler Thromb Vasc Biol 2004;24(3):534-9.

[29] Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2-/- mice reveals a role for chemokines in the initiation of atherosclerosis. Nature 1998;394(6696):894-7.

[30] Savoia C, D'Agostino M, Lauri F, Volpe M. Angiotensin type 2 receptor in hypertensive cardiovascular disease. Curr Opin Nephrol Hypertens 2011;20(2):125-32.

[31] Schmieder RE, Langenfeld MR, Friedrich A, Schobel HP, Gatzka CD, Weihprecht H. Angiotensin II related to sodium excretion modulates left ventricular structure in human essential hypertension. Circulation 1996;94(6):1304-9.

[32] Zheng H, Sharma NM, Liu X, Patel KP. Exercise training normalizes enhanced sympathetic activation from the paraventricular nucleus in chronic heart failure: role of angiotensin II. Am J Physiol Regul Integr Comp Physiol 2012;303(4):R387-94.

[33] Neri Serneri GG, Boddi M, Modesti PA, Coppo M, Cecioni I, Toscano T, et al. Cardiac angiotensin II participates in coronary microvessel inflammation of unstable angina and strengthens the immunomediated component. Circ Res 2004;94(12):1630-7.

[34] Klingbeil AU, Schneider M, Martus P, Messerli FH, Schmieder RE. A meta-analysis of the effects of treatment on left ventricular mass in essential hypertension. Am J Med 2003;115(1):41-6.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 24

[35] Ariff B, Zambanini A, Vamadeva S, Barratt D, Xu Y, Sever P, et al. Candesartan- and atenolol-based treatments induce different patterns of carotid artery and left ventricular remodeling in hypertension. Stroke 2006;37(9):2381-4.

[36] Lacourciere Y, Beliveau R, Conter HS, Burgess ED, Lepage S, Pesant Y, et al. Effects of perindopril on elastic and structural properties of large arteries in essential hypertension. Can J Cardiol 2004;20(8):795-9.

[37] Indications for ACE inhibitors in the early treatment of acute myocardial infarction: systematic overview of individual data from 100,000 patients in randomized trials. ACE Inhibitor Myocardial Infarction Collaborative Group. Circulation 1998;97(22):2202-12.

[38] Fox KM, Henderson JR, Bertrand ME, Ferrari R, Remme WJ, Simoons ML. The European trial on reduction of cardiac events with perindopril in stable coronary artery disease (EUROPA). Eur Heart J 1998;19 Suppl J(J52-5.

[39] Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev Immunol 2007;7(10):803-15.

[40] Kansas GS. Selectins and their ligands: current concepts and controversies. Blood 1996;88(9):3259-87.

[41] Eppihimer MJ, Wolitzky B, Anderson DC, Labow MA, Granger DN. Heterogeneity of Expression of E- and P-Selectins In Vivo. Circ Res 1996;79(3):560-9.

[42] Plutzky J. Inflammatory pathways in atherosclerosis and acute coronary syndromes. Am J Cardiol 2001;88(8A):10K-5K.

[43] Kozuka K, Kohriyama T, Nomura E, Ikeda J, Kajikawa H, Nakamura S. Endothelial markers and adhesion molecules in acute ischemic stroke--sequential change and differences in stroke subtype. Atherosclerosis 2002;161(1):161-8.

[44] Blann AD, Amiral J, McCollum CN. Circulating endothelial cell/leucocyte adhesion molecules in ischaemic heart disease. Br J Haematol 1996;95(2):263-5.

[45] Blankenberg S, Rupprecht HJ, Bickel C, Peetz D, Hafner G, Tiret L, et al. Circulating Cell Adhesion Molecules and Death in Patients With Coronary Artery Disease. Circulation 2001;104(12):1336-42.

[46] Miwa K, Igawa A, Inoue H. Soluble E-selectin, ICAM-1 and VCAM-1 levels in systemic and coronary circulation in patients with variant angina. Cardiovasc Res 1997;36(1):37-44.

[47] Mulvihill NT, Foley JB, Murphy RT, Curtin R, Crean PA, Walsh M. Risk stratification in unstable angina and non-Q wave myocardial infarction using soluble cell adhesion molecules. Heart 2001;85(6):623-7.

[48] Blann A, Kumar P, Krupinski J, McCollum C, Beevers DG, Lip GY. Soluble intercelluar adhesion molecule-1, E-selectin, vascular cell adhesion molecule-1 and von Willebrand factor in stroke. Blood Coagul Fibrinolysis 1999;10(5):277-84.

[49] Shyu KG, Chang H, Lin CC, Kuan P. Circulating intercellular adhesion molecule-1 and E-selectin in patients with acute coronary syndrome. Chest 1996;109(6):1627-30.

[50] Hwang S-J, Ballantyne CM, Sharrett AR, Smith LC, Davis CE, Gotto AM, Jr., et al. Circulating Adhesion Molecules VCAM-1, ICAM-1, and E-selectin in Carotid Atherosclerosis and Incident Coronary Heart Disease Cases : The Atherosclerosis Risk In Communities (ARIC) Study. Circulation 1997;96(12):4219-25.

[51] Malik I, Danesh J, Whincup P, Bhatia V, Papacosta O, Walker M, et al. Soluble adhesion molecules and prediction of coronary heart disease: a prospective study and meta-analysis. The Lancet 2001;358(9286):971-6.

[52] Roldan V, Marin F, Lip GY, Blann AD. Soluble E-selectin in cardiovascular disease and its risk factors. A review of the literature. Thromb Haemost 2003;90(6):1007-20.

[53] Lindquist S. The heat-shock response. Annu Rev Biochem 1986;55(1151-91.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 25

[54] Mehta TA, Greenman J, Ettelaie C, Venkatasubramaniam A, Chetter IC, McCollum PT. Heat shock proteins in vascular disease--a review. Eur J Vasc Endovasc Surg 2005;29(4):395-402.

[55] Whitley D, Goldberg SP, Jordan WD. Heat shock proteins: a review of the molecular chaperones. J Vasc Surg 1999;29(4):748-51.

[56] Pockley AG. Heat shock proteins as regulators of the immune response. Lancet 2003;362(9382):469-76.

[57] Beckmann RP, Mizzen LE, Welch WJ. Interaction of Hsp 70 with newly synthesized proteins: implications for protein folding and assembly. Science 1990;248(4957):850-4.

[58] Pockley AG. Heat Shock Proteins, Inflammation, and Cardiovascular Disease. Circulation 2002;105(8):1012-7.

[59] Frostegard J. Immunity, atherosclerosis and cardiovascular disease. BMC Med 2013;11(1):117. [60] Kol A, Sukhova GK, Lichtman AH, Libby P. Chlamydial Heat Shock Protein 60 Localizes in Human

Atheroma and Regulates Macrophage Tumor Necrosis Factor-{alpha} and Matrix Metalloproteinase Expression. 1998;98(4):300-7.

[61] Wick G, Knoflach M, Xu Q. Autoimmune and inflammatory mechanisms in atherosclerosis. Annu Rev Immunol 2004;22(361-403.

[62] Pockley AG, Georgiades A, Thulin T, de Faire U, Frostegard J. Serum Heat Shock Protein 70 Levels Predict the Development of Atherosclerosis in Subjects With Established Hypertension. Hypertension 2003;42(3):235-8.

[63] Xu Q, Schett G, Perschinka H, Mayr M, Egger G, Oberhollenzer F, et al. Serum Soluble Heat Shock Protein 60 Is Elevated in Subjects With Atherosclerosis in a General Population. Circulation 2000;102(1):14-20.

[64] Pockley AG, Wu R, Lemne C, Kiessling R, Ulf de F, Frostegard J. Circulating Heat Shock Protein 60 Is Associated With Early Cardiovascular Disease. Hypertension 2000;36(2):303-7.

[65] Hoppichler F, Koch T, Dzien A, Gschwandtner G, Lechleitner M. Prognostic value of antibody titre to heat-shock protein 65 on cardiovascular events. Cardiology 2000;94(4):220-3.

[66] Veres A, Fust G, Smieja M, McQueen M, Horvath A, Yi Q, et al. Relationship of Anti-60 kDa Heat Shock Protein and Anti-Cholesterol Antibodies to Cardiovascular Events. 2002;106(22):2775-80.

[67] Dollery CM, McEwan JR, Henney AM. Matrix Metalloproteinases and Cardiovascular Disease. Circ Res 1995;77(5):863-8.

[68] Shapiro SD. Matrix metalloproteinase degradation of extracellular matrix: biological consequences. Curr Opin Cell Biol 1998;10(5):602-8.

[69] Shah PK, Falk E, Badimon JJ, Fernandez-Ortiz A, Mailhac A, Villareal-Levy G, et al. Human monocyte-derived macrophages induce collagen breakdown in fibrous caps of atherosclerotic plaques. Potential role of matrix-degrading metalloproteinases and implications for plaque rupture. Circulation 1995;92(6):1565-9.

[70] Newby AC. Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc Res 2006;69(3):614-24.

[71] Brinckerhoff CE, Matrisian LM. Matrix metalloproteinases: a tail of a frog that became a prince. Nat Rev Mol Cell Biol 2002;3(3):207-14.

[72] Lijnen HR. Plasmin and matrix metalloproteinases in vascular remodeling. Thromb Haemost 2001;86(1):324-33.

[73] Tedgui A, Mallat Z. Cytokines in Atherosclerosis: Pathogenic and Regulatory Pathways. Physiol Rev 2006;86(2):515-81.

[74] Silence J, Lupu F, Collen D, Lijnen HR. Persistence of Atherosclerotic Plaque but Reduced Aneurysm Formation in Mice With Stromelysin-1 (MMP-3) Gene Inactivation. Arterioscler Thromb Vasc Biol 2001;21(9):1440-5.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 26

[75] Eckart RE, Uyehara CF, Shry EA, Furgerson JL, Krasuski RA. Matrix metalloproteinases in patients with myocardial infarction and percutaneous revascularization. J Interv Cardiol 2004;17(1):27-31.

[76] Manginas A, Bei E, Chaidaroglou A, Degiannis D, Koniavitou K, Voudris V, et al. Peripheral levels of matrix metalloproteinase-9, interleukin-6, and C-reactive protein are elevated in patients with acute coronary syndromes: correlations with serum troponin I. Clin Cardiol 2005;28(4):182-6.

[77] Ceausu M, Curca C, Dermengiu D, Ardeleanu C. Simultaneous immunophenotypical assessment of troponin and extracellular matrix molecules in myocardium of patients with sudden cardiac death. Rom J Morphol Embryol 2009;50(1):103-6.

[78] Haft JI, Mariano DL, Goldstein J. Comparison of the histopathology of culprit lesions in chronic stable angina, unstable angina, and myocardial infarction. Clin Cardiol 1997;20(7):651-5.

[79] Inokubo Y, Hanada H, Ishizaka H, Fukushi T, Kamada T, Okumura K. Plasma levels of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 are increased in the coronary circulation in patients with acute coronary syndrome. Am Heart J 2001;141(2):211-7.

[80] Kai H, Ikeda H, Yasukawa H, Kai M, Seki Y, Kuwahara F, et al. Peripheral blood levels of matrix metalloproteases-2 and -9 are elevated in patients with acute coronary syndromes. J Am Coll Cardiol 1998;32(2):368-72.

[81] Ikeda U, Shimada K. Matrix metalloproteinases and coronary artery diseases. Clin Cardiol 2003;26(2):55-9.

[82] Burke AP, Farb A, Malcom GT, Liang Y-h, Smialek J, Virmani R. Coronary Risk Factors and Plaque Morphology in Men with Coronary Disease Who Died Suddenly. N Engl J Med 1997;336(18):1276-82.

[83] Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons From Sudden Coronary Death : A Comprehensive Morphological Classification Scheme for Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol 2000;20(5):1262-75.

[84] Nicholls SJ, Hazen SL. Myeloperoxidase and Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2005;25(6):1102-11.

[85] McMillen TS, Heinecke JW, LeBoeuf RC. Expression of human myeloperoxidase by macrophages promotes atherosclerosis in mice. Circulation 2005;111(21):2798-804.

[86] Daugherty A, Dunn JL, Rateri DL, Heinecke JW. Myeloperoxidase, a catalyst for lipoprotein oxidation, is expressed in human atherosclerotic lesions. J Clin Invest 1994;94(1):437-44.

[87] Podrez EA, Schmitt D, Hoff HF, Hazen SL. Myeloperoxidase-generated reactive nitrogen species convert LDL into an atherogenic form in vitro. J Clin Invest 1999;103(11):1547-60.

[88] Zheng L, Nukuna B, Brennan M-L, Sun M, Goormastic M, Settle M, et al. Apolipoprotein A-I is a selective target for myeloperoxidase-catalyzed oxidation and functional impairment in subjects with cardiovascular disease. J Clin Invest 2004;114(4):529-41.

[89] Shao B, Oda MN, Bergt C, Fu X, Green PS, Brot N, et al. Myeloperoxidase Impairs ABCA1-dependent Cholesterol Efflux through Methionine Oxidation and Site-specific Tyrosine Chlorination of Apolipoprotein A-I. J Biol Chem 2006;281(14):9001-4.

[90] Abu-Soud HM, Hazen SL. Nitric Oxide Is a Physiological Substrate for Mammalian Peroxidases. J Biol Chem 2000;275(48):37524-32.

[91] Vita JA, Brennan M-L, Gokce N, Mann SA, Goormastic M, Shishehbor MH, et al. Serum Myeloperoxidase Levels Independently Predict Endothelial Dysfunction in Humans. Circulation 2004;110(9):1134-9.

[92] Luscher TF, Barton M. Biology of the endothelium. Clin Cardiol 1997;20(3-10. [93] Stoner L, Erickson ML, Young JM, Fryer S, Sabatier MJ, Faulkner J, et al. There's more to flow-

mediated dilation than nitric oxide. J Atheroscler Thromb 2012;19(7):589-600.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 27

[94] Brennan M-L, Penn MS, Van Lente F, Nambi V, Shishehbor MH, Aviles RJ, et al. Prognostic Value of Myeloperoxidase in Patients with Chest Pain. N Engl J Med 2003;349(17):1595-604.

[95] Mocatta TJ, Pilbrow AP, Cameron VA, Senthilmohan R, Frampton CM, Richards AM, et al. Plasma concentrations of myeloperoxidase predict mortality after myocardial infarction. J Am Coll Cardiol 2007;49(20):1993-2000.

[96] Baldus S, Heeschen C, Meinertz T, Zeiher AM, Eiserich JP, Munzel T, et al. Myeloperoxidase Serum Levels Predict Risk in Patients With Acute Coronary Syndromes. Circulation 2003;108(12):1440-5.

[97] Zhang R, Brennan M-L, Fu X, Aviles RJ, Pearce GL, Penn MS, et al. Association Between Myeloperoxidase Levels and Risk of Coronary Artery Disease. JAMA 2001;286(17):2136-42.

[98] Meuwese MC, Stroes ES, Hazen SL, van Miert JN, Kuivenhoven JA, Schaub RG, et al. Serum myeloperoxidase levels are associated with the future risk of coronary artery disease in apparently healthy individuals: the EPIC-Norfolk Prospective Population Study. J Am Coll Cardiol 2007;50(2):159-65.

[99] Galkina E, Ley K. Vascular Adhesion Molecules in Atherosclerosis. Arterioscler Thromb Vasc Biol 2007;27(11):2292-301.

[100] Muller WA. Leukocyte-endothelial-cell interactions in leukocyte transmigration and the inflammatory response. Trends Immunol 2003;24(6):327-34.

[101] Matsumura T, Wolff K, Petzelbauer P. Endothelial cell tube formation depends on cadherin 5 and CD31 interactions with filamentous actin. J Immunol 1997;158(7):3408-16.

[102] Piali L, Hammel P, Uherek C, Bachmann F, Gisler RH, Dunon D, et al. CD31/PECAM-1 is a ligand for alpha v beta 3 integrin involved in adhesion of leukocytes to endothelium. J Cell Biol 1995;130(2):451-60.

[103] Gao C, Sun W, Christofidou-Solomidou M, Sawada M, Newman DK, Bergom C, et al. PECAM-1 functions as a specific and potent inhibitor of mitochondrial-dependent apoptosis. Blood 2003;102(1):169-79.

[104] Muller WA, Weigl SA, Deng X, Phillips DM. PECAM-1 is required for transendothelial migration of leukocytes. J Exp Med 1993;178(2):449-60.

[105] Mahooti S, Graesser D, Patil S, Newman P, Duncan G, Mak T, et al. PECAM-1 (CD31) Expression Modulates Bleeding Time in Vivo. Am J Pathol 2000;157(1):75-81.

[106] Elrayess MA, Webb KE, Flavell DM, Syvanne M, Taskinen M-R, Frick MH, et al. A novel functional polymorphism in the PECAM-1 gene (53G>A) is associated with progression of atherosclerosis in the LOCAT and REGRESS studies. Atherosclerosis 2003;168(1):131-8.

[107] Elrayess MA, Webb KE, Bellingan GJ, Whittall RA, Kabir J, Hawe E, et al. R643G polymorphism in PECAM-1 influences transendothelial migration of monocytes and is associated with progression of CHD and CHD events. Atherosclerosis 2004;177(1):127-35.

[108] Fang L, Wei H, Chowdhury SH, Gong N, Song J, Heng CK, et al. Association of Leu125Val polymorphism of platelet endothelial cell adhesion molecule-1 (PECAM-1) gene & soluble level of PECAM-1 with coronary artery disease in Asian Indians. Indian J Med Res 2005;121(2):92-9.

[109] Listi F, Candore G, Lio D, Cavallone L, Colonna-Romano G, Caruso M, et al. Association between platelet endothelial cellular adhesion molecule 1 (PECAM-1/CD31) polymorphisms and acute myocardial infarction: a study in patients from Sicily. Eur J Immunogenet 2004;31(4):175-8.

[110] Hashimoto K, Kataoka N, Nakamura E, Tsujioka K, Kajiya F. Oxidized LDL specifically promotes the initiation of monocyte invasion during transendothelial migration with upregulated PECAM-1 and downregulated VE-cadherin on endothelial junctions. Atherosclerosis 2007;194(2):e9-17.

[111] Rattan V, Shen Y, Sultana C, Kumar D, Kalra VK. Glucose-induced transmigration of monocytes is linked to phosphorylation of PECAM-1 in cultured endothelial cells. 1996;271(4):E711-7.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 28

[112] Okouchi M, Okayama N, Imai S, Omi H, Shimizu M, Fukutomi T, et al. High insulin enhances neutrophil transendothelial migration through increasing surface expression of platelet endothelial cell adhesion molecule-1 via activation of mitogen activated protein kinase. Diabetologia 2002;45(10):1449-56.

[113] Tailor A, Granger DN. Role of adhesion molecules in vascular regulation and damage. Curr Hypertens Rep 2000;2(1):78-83.

[114] Kubes P, Ward PA. Leukocyte recruitment and the acute inflammatory response. Brain Pathol 2000;10(1):127-35.

[115] Collins RG, Velji R, Guevara NV, Hicks MJ, Chan L, Beaudet AL. P-Selectin or Intercellular Adhesion Molecule (ICAM)-1 Deficiency Substantially Protects against Atherosclerosis in Apolipoprotein E-deficient Mice. J Exp Med 2000;191(1):189-94.

[116] Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, et al. A major role for VCAM-1, but not ICAM-1, in early atherosclerosis. J Clin Invest 2001;107(10):1255-62.

[117] Nakashima Y, Raines EW, Plump AS, Breslow JL, Ross R. Upregulation of VCAM-1 and ICAM-1 at Atherosclerosis-Prone Sites on the Endothelium in the ApoE-Deficient Mouse. Arterioscler Thromb Vasc Biol 1998;18(5):842-51.

[118] Haim M, Tanne D, Boyko V, Reshef T, Goldbourt U, Leor J, et al. Soluble intercellular adhesion molecule-1 and long-term risk of acute coronary events in patients with chronic coronary heart disease. Data from the Bezafibrate Infarction Prevention (BIP) Study. J Am Coll Cardiol 2002;39(7):1133-8.

[119] Schumacher A, Seljeflot I, Sommervoll L, Christensen B, Otterstad JE, Arnesen H. Increased levels of markers of vascular inflammation in patients with coronary heart disease. Scand J Clin Lab Invest 2002;62(1):59-68.

[120] Jha HC, Divya A, Prasad J, Mittal A. Plasma circulatory markers in male and female patients with coronary artery disease. Heart Lung 2010;39(4):296-303.

[121] Huang Y, Yin H, Wang J, Ma X, Zhang Y, Chen K. The significant increase of FcγRIIIA (CD16), a sensitive marker, in patients with coronary heart disease. Gene 2012;504(2):284-7.

[122] Luc G, Arveiler D, Evans A, Amouyel P, Ferrieres J, Bard JM, et al. Circulating soluble adhesion molecules ICAM-1 and VCAM-1 and incident coronary heart disease: the PRIME Study. Atherosclerosis 2003;170(1):169-76.

[123] Pradhan AD, Rifai N, Ridker PM. Soluble Intercellular Adhesion Molecule-1, Soluble Vascular Adhesion Molecule-1, and the Development of Symptomatic Peripheral Arterial Disease in Men. Circulation 2002;106(7):820-5.

[124] Jenny NS, Arnold AM, Kuller LH, Sharrett AR, Fried LP, Psaty BM, et al. Soluble intracellular adhesion molecule-1 is associated with cardiovascular disease risk and mortality in older adults. J Thromb Haemost 2006;4(1):107-13.

[125] Carlos TM, Schwartz BR, Kovach NL, Yee E, Rosa M, Osborn L, et al. Vascular cell adhesion molecule-1 mediates lymphocyte adherence to cytokine-activated cultured human endothelial cells. Blood 1990;76(5):965-70.

[126] Ridker PM, Hennekens CH, Roitman-Johnson B, Stampfer MJ, Allen J. Plasma concentration of soluble intercellular adhesion molecule 1 and risks of future myocardial infarction in apparently healthy men. Lancet 1998;351(9096):88-92.

[127] de Lemos JA, Hennekens CH, Ridker PM. Plasma concentration of soluble vascular cell adhesion molecule-1 and subsequent cardiovascular risk. J Am Coll Cardiol 2000;36(2):423-6.

[128] Blankenberg S, Barbaux S, Tiret L. Adhesion molecules and atherosclerosis. Atherosclerosis 2003;170(2):191-203.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 29

[129] O'Brien KD, Allen MD, McDonald TO, Chait A, Harlan JM, Fishbein D, et al. Vascular cell adhesion molecule-1 is expressed in human coronary atherosclerotic plaques. Implications for the mode of progression of advanced coronary atherosclerosis. J Clin Invest 1993;92(2):945-51.

[130] De Caterina R, Basta G, Lazzerini G, Dell'Omo G, Petrucci R, Morale M, et al. Soluble Vascular Cell Adhesion Molecule-1 as a Biohumoral Correlate of Atherosclerosis. Arterioscler Thromb Vasc Biol 1997;17(11):2646-54.

[131] Peter K, Nawroth P, Conradt C, Nordt T, Weiss T, Boehme M, et al. Circulating Vascular Cell Adhesion Molecule-1 Correlates With the Extent of Human Atherosclerosis in Contrast to Circulating Intercellular Adhesion Molecule-1, E-Selectin, P-Selectin, and Thrombomodulin. Arterioscler Thromb Vasc Biol 1997;17(3):505-12.

[132] Jager A, van Hinsbergh VW, Kostense PJ, Emeis JJ, Nijpels G, Dekker JM, et al. Increased levels of soluble vascular cell adhesion molecule 1 are associated with risk of cardiovascular mortality in type 2 diabetes: the Hoorn study. Diabetes 2000;49(3):485-91.

[133] Stehouwer CDA, Gall M-A, Twisk JWR, Knudsen E, Emeis JJ, Parving H-H. Increased Urinary Albumin Excretion, Endothelial Dysfunction, and Chronic Low-Grade Inflammation in Type 2 Diabetes: Progressive, Interrelated, and Independently Associated With Risk of Death. Diabetes 2002;51(4):1157-65.

[134] Stoner L, Young JM, Fryer S. Assessments of arterial stiffness and endothelial function using pulse wave analysis. Int J Vasc Med 2012;2012(903107.

[135] Stoner L, Lambrick DM, Faulkner J, Young J. Guidelines for the Use of Pulse Wave Analysis in Adults and Children. J Atheroscler Thromb 2013.

[136] Boulay J-L, O'Shea JJ, Paul WE. Molecular Phylogeny within Type I Cytokines and Their Cognate Receptors. Immunity 2003;19(2):159-63.

[137] Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature 1998;394(6689):200-3.

[138] Lutgens E, Lievens D, Beckers L, Donners M, Daemen M. CD40 and Its Ligand in Atherosclerosis. Trends in Cardiovascular Medicine 2007;17(4):118-23.

[139] Lutgens E, Daemen MJAP. CD40-CD40L Interactions in Atherosclerosis. Trends in Cardiovascular Medicine 2002;12(1):27-32.

[140] Schonbeck U, Libby P. CD40 Signaling and Plaque Instability. Circ Res 2001;89(12):1092-103. [141] Chakrabarti S, Varghese S, Vitseva O, Tanriverdi K, Freedman JE. CD40 Ligand Influences Platelet

Release of Reactive Oxygen Intermediates. Arterioscler Thromb Vasc Biol 2005;25(11):2428-34. [142] Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G, et al. CD40 ligand

on activated platelets triggers an inflammatory reaction of endothelial cells. Nature 1998;391(6667):591-4.

[143] Henn V, Steinbach S, Buchner K, Presek P, Kroczek RA. The inflammatory action of CD40 ligand (CD154) expressed on activated human platelets is temporally limited by coexpressed CD40. Blood 2001;98(4):1047-54.

[144] Schonbeck U, Varo N, Libby P, Buring J, Ridker PM. Soluble CD40L and Cardiovascular Risk in Women. Circulation 2001;104(19):2266-8.

[145] Blake GJ, Ostfeld RJ, Yucel EK, Varo N, Schonbeck U, Blake MA, et al. Soluble CD40 ligand levels indicate lipid accumulation in carotid atheroma: an in vivo study with high-resolution MRI. Arterioscler Thromb Vasc Biol 2003;23(1):e11-4.

[146] Heeschen C, Dimmeler S, Hamm CW, van den Brand MJ, Boersma E, Zeiher AM, et al. Soluble CD40 ligand in acute coronary syndromes. N Engl J Med 2003;348(12):1104-11.

[147] Jialal I, Devaraj S, Venugopal SK. C-Reactive Protein: Risk Marker or Mediator in Atherothrombosis? Hypertension 2004;44(1):6-11.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 30

[148] Pasceri V, Chang J, Willerson JT, Yeh ETH. Modulation of C-Reactive Protein-Mediated Monocyte Chemoattractant Protein-1 Induction in Human Endothelial Cells by Anti-Atherosclerosis Drugs. Circulation 2001;103(21):2531-4.

[149] Pasceri V, Willerson JT, Yeh ETH. Direct Proinflammatory Effect of C-Reactive Protein on Human Endothelial Cells. Circulation 2000;102(18):2165-8.

[150] Libby P, Ridker PM. Inflammation and atherosclerosis: role of C-Reactive protein in risk assessment. The American Journal of Medicine 2004;116(6, Supplement 1):9-16.

[151] Ridker PM, Rifai N, Rose L, Buring JE, Cook NR. Comparison of C-Reactive Protein and Low-Density Lipoprotein Cholesterol Levels in the Prediction of First Cardiovascular Events. N Engl J Med 2002;347(20):1557-65.

[152] Koenig W, Khuseyinova N, Lowel H, Trischler G, Meisinger C. Lipoprotein-Associated Phospholipase A2 Adds to Risk Prediction of Incident Coronary Events by C-Reactive Protein in Apparently Healthy Middle-Aged Men From the General Population: Results From the 14-Year Follow-Up of a Large Cohort From Southern Germany. Circulation 2004;110(14):1903-8.

[153] Caslake MJ, Packard CJ. Lipoprotein-associated phospholipase A2 as a biomarker for coronary disease and stroke. Nat Clin Pract Cardiovasc Med 2005;2(10):529-35.

[154] Hoogeveen RC, Morrison A, Boerwinkle E, Miles JS, Rhodes CE, Sharrett AR, et al. Plasma MCP-1 level and risk for peripheral arterial disease and incident coronary heart disease: Atherosclerosis Risk in Communities study. Atherosclerosis 2005;183(2):301-7.

[155] Liu C, Wang S, Deb A, Nath KA, Katusic ZS, McConnell JP, et al. Proapoptotic, Antimigratory, Antiproliferative, and Antiangiogenic Effects of Commercial C-Reactive Protein on Various Human Endothelial Cell Types In Vitro: Implications of Contaminating Presence of Sodium Azide in Commercial Preparation. Circ Res 2005;97(2):135-43.

[156] Reifenberg K, Lehr H-A, Baskal D, Wiese E, Schaefer SC, Black S, et al. Role of C-Reactive Protein in Atherogenesis: Can the Apolipoprotein E Knockout Mouse Provide the Answer? Arterioscler Thromb Vasc Biol 2005;25(8):1641-6.

[157] Paul A, Ko KWS, Li L, Yechoor V, McCrory MA, Szalai AJ, et al. C-Reactive Protein Accelerates the Progression of Atherosclerosis in Apolipoprotein E-Deficient Mice. Circulation 2004;109(5):647-55.

[158] Bhakdi S, Torzewski M, Paprotka K, Schmitt S, Barsoom H, Suriyaphol P, et al. Possible Protective Role for C-Reactive Protein in Atherogenesis: Complement Activation by Modified Lipoproteins Halts Before Detrimental Terminal Sequence. Circulation 2004;109(15):1870-6.

[159] Schwedler SB, Amann K, Wernicke K, Krebs A, Nauck M, Wanner C, et al. Native C-Reactive Protein Increases Whereas Modified C-Reactive Protein Reduces Atherosclerosis in Apolipoprotein E-Knockout Mice. Circulation 2005;112(7):1016-23.

[160] Xia D, Samols D. Transgenic mice expressing rabbit C-reactive protein are resistant to endotoxemia. Proceedings of the National Academy of Sciences 1997;94(6):2575-80.

[161] Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C. IFN-gamma potentiates atherosclerosis in ApoE knock-out mice. J Clin Invest 1997;99(11):2752-61.

[162] Buono C, Come CE, Stavrakis G, Maguire GF, Connelly PW, Lichtman AH. Influence of Interferon-{gamma} on the Extent and Phenotype of Diet-Induced Atherosclerosis in the LDLR-Deficient Mouse. Arterioscler Thromb Vasc Biol 2003;23(3):454-60.

[163] Whitman SC, Ravisankar P, Elam H, Daugherty A. Exogenous Interferon-{{gamma}} Enhances Atherosclerosis in Apolipoprotein E-/- Mice. Am J Pathol 2000;157(6):1819-24.

[164] Whitman SC, Ravisankar P, Daugherty A. Interleukin-18 Enhances Atherosclerosis in Apolipoprotein E-/- Mice Through Release of Interferon-{gamma}. Circ Res 2002;90(2):e34-8.

[165] Buono C Fau - Come CE, Come Ce Fau - Stavrakis G, Stavrakis G Fau - Maguire GF, Maguire Gf Fau - Connelly PW, Connelly Pw Fau - Lichtman AH, Lichtman AH. Influence of interferon-gamma

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 31

on the extent and phenotype of diet-induced atherosclerosis in the LDLR-deficient mouse. SO - Arterioscler Thromb Vasc Biol 2003 Mar 1;23(3):454-60 Epub 2003 Jan 30.

[166] Patel KD, Duggan SP, Currid CA, Gallagher WM, McManus R, Kelleher D, et al. High sensitivity cytokine detection in acute coronary syndrome reveals up-regulation of interferon gamma and interleukin-10 post myocardial infarction. Clin Immunol 2009;133(2):251-6.

[167] Prondzinsky R, Unverzagt S, Lemm H, Wegener N, Heinroth K, Buerke U, et al. Acute myocardial infarction and cardiogenic shock: prognostic impact of cytokines: INF-gamma, TNF-alpha, MIP-1beta, G-CSF, and MCP-1beta. Med Klin Intensivmed Notfmed 2012;107(6):476-84.

[168] Prondzinsky R, Unverzagt S, Lemm H, Wegener NA, Schlitt A, Heinroth KM, et al. Interleukin-6, -7, -8 and -10 predict outcome in acute myocardial infarction complicated by cardiogenic shock. Clin Res Cardiol 2012;101(5):375-84.

[169] Botti C, Maione C, Dogliotti G, Russo P, Signoriello G, Molinari A, et al. Circulating cytokines present in the serum of peripheral arterial disease patients induce endothelial dysfunction. Journal of biological regulators and homeostatic agents 2012;26(1):67.

[170] Haider M, Rizvi M, Malik A, Azam M, Rabbani MU. Acute and chronic Chlamydia pneumoniae infection and inflammatory markers in coronary artery disease patients. J Infect Dev Ctries 2011;5(8):580-6.

[171] Cappuzzello C, Di Vito L, Melchionna R, Melillo G, Silvestri L, Cesareo E, et al. Increase of plasma IL-9 and decrease of plasma IL-5, IL-7, and IFN-γ in patients with chronic heart failure. Journal of Translational Medicine 2011;9(9):28.

[172] Elhage R, Maret A, Pieraggi MT, Thiers JC, Arnal JF, Bayard F. Differential Effects of Interleukin-1 Receptor Antagonist and Tumor Necrosis Factor Binding Protein on Fatty-Streak Formation in Apolipoprotein E–Deficient Mice. Circulation 1998;97(3):242-4.

[173] Devlin CM, Kuriakose G, Hirsch E, Tabas I. Genetic alterations of IL-1 receptor antagonist in mice affect plasma cholesterol level and foam cell lesion size. Proceedings of the National Academy of Sciences 2002;99(9):6280-5.

[174] Merhi-Soussi F, Kwak BR, Magne D, Chadjichristos C, Berti M, Pelli G, et al. Interleukin-1 plays a major role in vascular inflammation and atherosclerosis in male apolipoprotein E-knockout mice. Cardiovasc Res 2005;66(3):583-93.

[175] Patti G, D'Ambrosio A, Dobrina A, Dicuonzo G, Giansante C, Fiotti N, et al. Interleukin-1 receptor antagonist: a sensitive marker of instability in patients with coronary artery disease. J Thromb Thrombolysis 2002;14(2):139-43.

[176] Patti G, D'Ambrosio A, Mega S, Giorgi G, Zardi EM, Zardi DM, et al. Early interleukin-1 receptor antagonist elevation in patients with acute myocardial infarction. J Am Coll Cardiol 2004;43(1):35-8.

[177] Patti G, Di Sciascio G, D'Ambrosio A, Dicuonzo G, Abbate A, Dobrina A. Inflammatory markers and coronary interventions: a potentially useful follow-up modality after stenting. Catheter Cardiovasc Interv 2002;56(3):341-5.

[178] Abeywardena MY, Leifert WR, Warnes KE, Varghese JN, Head RJ. Cardiovascular biology of interleukin-6. Curr Pharm Des 2009;15(15):1809-21.

[179] Schuett H, Luchtefeld M, Grothusen C, Grote K, Schieffer B. How much is too much? Interleukin-6 and its signalling in atherosclerosis. Thromb Haemost 2009;102(2):215-22.

[180] Huber SA, Sakkinen P, Conze D, Hardin N, Tracy R. Interleukin-6 Exacerbates Early Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 1999;19(10):2364-7.

[181] Sukovich DA, Kauser K, Shirley FD, DelVecchio V, Halks-Miller M, Rubanyi GM. Expression of interleukin-6 in atherosclerotic lesions of male ApoE-knockout mice: inhibition by 17beta-estradiol. Arterioscler Thromb Vasc Biol 1998;18(9):1498-505.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 32

[182] Casas JP, Shah T, Hingorani AD, Danesh J, Pepys MB. C-reactive protein and coronary heart disease: a critical review. J Intern Med 2008;264(4):295-314.

[183] Suzuki H, Kusuyama T, Sato R, Yokota Y, Tsunoda F, Sato T, et al. Elevation of matrix metalloproteinases and interleukin-6 in the culprit coronary artery of myocardial infarction. Eur J Clin Invest 2008;38(3):166-73.

[184] Barton BE. The biological effects of interleukin 6. Med Res Rev 1996;16(1):87-109. [185] Tilg H, Trehu E, Atkins MB, Dinarello CA, Mier JW. Interleukin-6 (IL-6) as an anti-inflammatory

cytokine: induction of circulating IL-1 receptor antagonist and soluble tumor necrosis factor receptor p55. Blood 1994;83(1):113-8.

[186] Xing Z, Gauldie J, Cox G, Baumann H, Jordana M, Lei XF, et al. IL-6 is an antiinflammatory cytokine required for controlling local or systemic acute inflammatory responses. J Clin Invest 1998;101(2):311-20.

[187] Harris TB, Ferrucci L, Tracy RP, Corti MC, Wacholder S, Ettinger WH, Jr., et al. Associations of elevated interleukin-6 and C-reactive protein levels with mortality in the elderly. Am J Med 1999;106(5):506-12.

[188] Biasucci LM, Vitelli A, Liuzzo G, Altamura S, Caligiuri G, Monaco C, et al. Elevated Levels of Interleukin-6 in Unstable Angina. Circulation 1996;94(5):874-7.

[189] Neumann FJ, Ott I, Gawaz M, Richardt G, Holzapfel H, Jochum M, et al. Cardiac release of cytokines and inflammatory responses in acute myocardial infarction. Circulation 1995;92(4):748-55.

[190] Jaremo P, Nilsson O. Interleukin-6 and neutrophils are associated with long-term survival after acute myocardial infarction. Eur J Intern Med 2008;19(5):330-3.

[191] Kinugawa T, Kato M, Yamamoto K, Hisatome I, Nohara R. Proinflammatory cytokine activation is linked to apoptotic mediator, soluble fas level in patients with chronic heart failure. International Heart Journal 2012;53(3):182-6.

[192] Mielniczuk LM, Pfeffer MA, Lewis EF, Blazing MA, de Lemos JA, Mohanavelu S, et al. Acute decline in renal function, inflammation, and cardiovascular risk after an acute coronary syndrome. Clin J Am Soc Nephrol 2009;4(11):1811-7.

[193] Tan J, Hua Q, Li J, Fan Z. Prognostic value of interleukin-6 during a 3-year follow-up in patients with acute ST-segment elevation myocardial infarction. Heart Vessels 2009;24(5):329-34.

[194] Biasucci LM, Liuzzo G, Fantuzzi G, Caligiuri G, Rebuzzi AG, Ginnetti F, et al. Increasing Levels of Interleukin (IL)-1Ra and IL-6 During the First 2 Days of Hospitalization in Unstable Angina Are Associated With Increased Risk of In-Hospital Coronary Events. Circulation 1999;99(16):2079-84.

[195] Lindmark E, Diderholm E, Wallentin L, Siegbahn A. Relationship Between Interleukin 6 and Mortality in Patients With Unstable Coronary Artery Disease: Effects of an Early Invasive or Noninvasive Strategy. JAMA 2001;286(17):2107-13.

[196] Vassalli P. The pathophysiology of tumor necrosis factors. Annu Rev Immunol 1992;10(411-52. [197] Beutler B. TNF, immunity and inflammatory disease: lessons of the past decade. J Investig Med

1995;43(3):227-35. [198] Beutler B, Cerami A. The biology of cachectin/TNF--a primary mediator of the host response.

Annu Rev Immunol 1989;7(625-55. [199] Meager A. Cytokine regulation of cellular adhesion molecule expression in inflammation.

Cytokine Growth Factor Rev 1999;10(1):27-39. [200] Ohta H, Wada H, Niwa T, Kirii H, Iwamoto N, Fujii H, et al. Disruption of tumor necrosis factor-

[alpha] gene diminishes the development of atherosclerosis in ApoE-deficient mice. Atherosclerosis 2005;180(1):11-7.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 33

[201] Rollins BJ, Yoshimura T, Leonard EJ, Pober JS. Cytokine-activated human endothelial cells synthesize and secrete a monocyte chemoattractant, MCP-1/JE. Am J Pathol 1990;136(6):1229-33.

[202] Libby P, Ordovas JM, Auger KR, Robbins AH, Birinyi LK, Dinarello CA. Endotoxin and tumor necrosis factor induce interleukin-1 gene expression in adult human vascular endothelial cells. Am J Pathol 1986;124(2):179-85.

[203] Hotamisligil GS, Spiegelman BM. Tumor necrosis factor alpha: a key component of the obesity-diabetes link. Diabetes 1994;43(11):1271-8.

[204] Jovinge S, Hamsten A, Tornvall P, Proudler A, Bavenholm P, Ericsson CG, et al. Evidence for a role of tumor necrosis factor alpha in disturbances of triglyceride and glucose metabolism predisposing to coronary heart disease. Metabolism 1998;47(1):113-8.

[205] Skoog T, Dichtl W, Boquist S, Skoglund-Andersson C, Karpe F, Tang R, et al. Plasma tumour necrosis factor-{alpha} and early carotid atherosclerosis in healthy middle-aged men. Eur Heart J 2002;23(5):376-83.

[206] Prondzinsky R, Unverzagt S, Lemm H, Wegener N, Heinroth K, Buerke U, et al. Acute myocardial infarction and cardiogenic shock: Prognostic impact of cytokines: INF-γ, TNF-α, MIP-1β, G-CSF, and MCP-1β.

[207] Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 1995;270(45):26746-9.

[208] Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem 1996;271(18):10697-703.

[209] Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Matsubara K. cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (AdiPose Most abundant Gene transcript 1). Biochem Biophys Res Commun 1996;221(2):286-9.

[210] Ouchi N, Shibata R, Walsh K. Cardioprotection by adiponectin. Trends Cardiovasc Med 2006;16(5):141-6.

[211] Ouchi N, Kihara S, Arita Y, Maeda K, Kuriyama H, Okamoto Y, et al. Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation 1999;100(25):2473-6.

[212] Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ. Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 2003;278(45):45021-6.

[213] Shimabukuro M, Higa N, Asahi T, Oshiro Y, Takasu N, Tagawa T, et al. Hypoadiponectinemia is closely linked to endothelial dysfunction in man. J Clin Endocrinol Metab 2003;88(7):3236-40.

[214] Saarikoski LA, Huupponen RK, Viikari JS, Marniemi J, Juonala M, Kahonen M, et al. Adiponectin is related with carotid artery intima-media thickness and brachial flow-mediated dilatation in young adults--the Cardiovascular Risk in Young Finns Study. Ann Med 2010;42(8):603-11.

[215] Juonala M, Saarikoski LA, Viikari JS, Oikonen M, Lehtimaki T, Lyytikainen LP, et al. A longitudinal analysis on associations of adiponectin levels with metabolic syndrome and carotid artery intima-media thickness. The Cardiovascular Risk in Young Finns Study. Atherosclerosis 2011;217(1):234-9.

[216] Andersson CX, Gustafson B, Hammarstedt A, Hedjazifar S, Smith U. Inflamed adipose tissue, insulin resistance and vascular injury. Diabetes Metab Res Rev 2008;24(8):595-603.

[217] Kobashi C, Urakaze M, Kishida M, Kibayashi E, Kobayashi H, Kihara S, et al. Adiponectin inhibits endothelial synthesis of interleukin-8. Circ Res 2005;97(12):1245-52.

[218] Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 1999;257(1):79-83.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 34

[219] Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y, et al. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 2000;20(6):1595-9.

[220] Oh DK, Ciaraldi T, Henry RR. Adiponectin in health and disease. Diabetes Obes Metab 2007;9(3):282-9.

[221] Kistorp C, Faber J, Galatius S, Gustafsson F, Frystyk J, Flyvbjerg A, et al. Plasma adiponectin, body mass index, and mortality in patients with chronic heart failure. Circulation 2005;112(12):1756-62.

[222] Pischon T, Girman CJ, Hotamisligil GS, Rifai N, Hu FB, Rimm EB. Plasma adiponectin levels and risk of myocardial infarction in men. JAMA 2004;291(14):1730-7.

[223] Frystyk J, Berne C, Berglund L, Jensevik K, Flyvbjerg A, Zethelius B. Serum Adiponectin Is a Predictor of Coronary Heart Disease: A Population-Based 10-Year Follow-Up Study in Elderly Men. J Clin Endocrinol Metab 2007;92(2):571-6.

[224] te Velde AA, Huijbens RJ, Heije K, de Vries JE, Figdor CG. Interleukin-4 (IL-4) inhibits secretion of IL-1 beta, tumor necrosis factor alpha, and IL-6 by human monocytes. Blood 1990;76(7):1392-7.

[225] Lampa J, Westman M, Kadetoff D, Agreus AN, Le Maitre E, Gillis-Haegerstrand C, et al. Peripheral inflammatory disease associated with centrally activated IL-1 system in humans and mice. Proc Natl Acad Sci U S A 2012.

[226] van Meegeren ME, Roosendaal G, Jansen NW, Wenting MJ, van Wesel AC, van Roon JA, et al. IL-4 alone and in combination with IL-10 protects against blood-induced cartilage damage. Osteoarthritis Cartilage 2012;20(7):764-72.

[227] Stanisic M, Aasen AO, Pripp AH, Lindegaard KF, Ramm-Pettersen J, Lyngstadaas SP, et al. Local and systemic pro-inflammatory and anti-inflammatory cytokine patterns in patients with chronic subdural hematoma: a prospective study. Inflamm Res 2012;61(8):845-52.

[228] Payne N, Dantanarayana A, Sun G, Moussa L, Caine S, McDonald C, et al. Early intervention with gene-modified mesenchymal stem cells overexpressing interleukin-4 enhances anti-inflammatory responses and functional recovery in experimental autoimmune demyelination. Cell Adh Migr 2012;6(3).

[229] Lee YW, Kuhn H, Hennig B, Neish AS, Toborek M. IL-4-induced Oxidative Stress Upregulates VCAM-1 Gene Expression in Human Endothelial Cells. Journal of Molecular and Cellular Cardiology 2001;33(1):83-94.

[230] Lee YW, Hennig B, Toborek M. Redox-regulated mechanisms of IL-4-induced MCP-1 expression in human vascular endothelial cells. Am J Physiol Heart Circ Physiol 2003;284(1):H185-92.

[231] King VL, Szilvassy SJ, Daugherty A. Interleukin-4 Deficiency Decreases Atherosclerotic Lesion Formation in a Site-Specific Manner in Female LDL Receptor-/- Mice. Arterioscler Thromb Vasc Biol 2002;22(3):456-61.

[232] George J, Mulkins M, Shaish A, Casey S, Schatzman R, Sigal E, et al. Interleukin (IL)-4 deficiency does not influence fatty streak formation in C57BL/6 mice. Atherosclerosis 2000;153(2):403-11.

[233] Szkodzinski J, Hudzik B, Osuch M, Romanowski W, Szygula-Jurkiewicz B, Polonski L, et al. Serum concentrations of interleukin-4 and interferon-gamma in relation to severe left ventricular dysfunction in patients with acute myocardial infarction undergoing percutaneous coronary intervention. Heart Vessels 2011;26(4):399-407.

[234] Hopps E, Canino B, Caimi G. Effects of exercise on inflammation markers in type 2 diabetic subjects. Acta Diabetol 2011;48(3):183-9.

[235] Balducci S, Zanuso S, Nicolucci A, Fernando F, Cavallo S, Cardelli P, et al. Anti-inflammatory effect of exercise training in subjects with type 2 diabetes and the metabolic syndrome is dependent on exercise modalities and independent of weight loss. Nutr Metab Cardiovasc Dis 2010;20(8):608-17.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 35

[236] Wakkach A, Cottrez F, Groux H. Can interleukin-10 be used as a true immunoregulatory cytokine? Eur Cytokine Netw 2000;11(2):153-60.

[237] Bogdan C, Vodovotz Y, Nathan C. Macrophage deactivation by interleukin 10. J Exp Med 1991;174(6):1549-55.

[238] Tedgui A, Mallat Z. Anti-Inflammatory Mechanisms in the Vascular Wall. Circ Res 2001;88(9):877-87.

[239] Pinderski Oslund LJ, Hedrick CC, Olvera T, Hagenbaugh A, Territo M, Berliner JA, et al. Interleukin-10 Blocks Atherosclerotic Events In Vitro and In Vivo. Arterioscler Thromb Vasc Biol 1999;19(12):2847-53.

[240] Potteaux S, Esposito B, van Oostrom O, Brun V, Ardouin P, Groux H, et al. Leukocyte-Derived Interleukin 10 Is Required for Protection Against Atherosclerosis in Low-Density Lipoprotein Receptor Knockout Mice. Arterioscler Thromb Vasc Biol 2004;24(8):1474-8.

[241] Smith DA, Irving SD, Sheldon J, Cole D, Kaski JC. Serum levels of the antiinflammatory cytokine interleukin-10 are decreased in patients with unstable angina. Circulation 2001;104(7):746-9.

[242] Heeschen C, Dimmeler S, Hamm CW, Fichtlscherer S, Boersma E, Simoons ML, et al. Serum level of the antiinflammatory cytokine interleukin-10 is an important prognostic determinant in patients with acute coronary syndromes. Circulation 2003;107(16):2109-14.

[243] Grainger DJ. Transforming Growth Factor {beta} and Atherosclerosis: So Far, So Good for the Protective Cytokine Hypothesis. Arterioscler Thromb Vasc Biol 2004;24(3):399-404.

[244] Bobik A. Transforming growth factor-betas and vascular disorders. Arterioscler Thromb Vasc Biol 2006;26(8):1712-20.

[245] Cipollone F, Fazia M, Mincione G, Iezzi A, Pini B, Cuccurullo C, et al. Increased expression of transforming growth factor-beta1 as a stabilizing factor in human atherosclerotic plaques. Stroke 2004;35(10):2253-7.

[246] Gojova A, Brun V, Esposito B, Cottrez F, Gourdy P, Ardouin P, et al. Specific abrogation of transforming growth factor-{beta} signaling in T cells alters atherosclerotic lesion size and composition in mice. Blood 2003;102(12):4052-8.

[247] Robertson AK, Rudling M, Zhou X, Gorelik L, Flavell RA, Hansson GK. Disruption of TGF-beta signaling in T cells accelerates atherosclerosis. J Clin Invest 2003;112(9):1342-50.

[248] Mallat Z, Gojova A, Marchiol-Fournigault C, Esposito B, Kamate C, Merval R, et al. Inhibition of transforming growth factor-beta signaling accelerates atherosclerosis and induces an unstable plaque phenotype in mice. Circ Res 2001;89(10):930-4.

[249] Wahl SM, McCartney-Francis N, Mergenhagen SE. Inflammatory and immunomodulatory roles of TGF-beta. Immunol Today 1989;10(8):258-61.

[250] Gamble JR, Khew-Goodall Y, Vadas MA. Transforming growth factor-beta inhibits E-selectin expression on human endothelial cells. J Immunol 1993;150(10):4494-503.

[251] Ruscetti FW, Dubois CM, Jacobsen SE, Keller JR. Transforming growth factor beta and interleukin-1: a paradigm for opposing regulation of haemopoiesis. Baillieres Clin Haematol 1992;5(3):703-21.

[252] Stefoni S, Cianciolo G, Donati G, Dormi A, Silvestri MG, Colì L, et al. Low TGF-beta1 serum levels are a risk factor for atherosclerosis disease in ESRD patients. Kidney international 2002;61(1):324-35.

[253] Koch W, Hoppmann P, Mueller JC, Schomig A, Kastrati A. Association of transforming growth factor-beta1 gene polymorphisms with myocardial infarction in patients with angiographically proven coronary heart disease. Arterioscler Thromb Vasc Biol 2006;26(5):1114-9.

[254] Holweg CT, Baan CC, Niesters HG, Vantrimpont PJ, Mulder PG, Maat AP, et al. TGF-beta1 gene polymorphisms in patients with end-stage heart failure. J Heart Lung Transplant 2001;20(9):979-84.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 36

[255] Tran-Fadulu V, Pannu H, Kim DH, Vick GW, 3rd, Lonsford CM, Lafont AL, et al. Analysis of multigenerational families with thoracic aortic aneurysms and dissections due to TGFBR1 or TGFBR2 mutations. J Med Genet 2009;46(9):607-13.

[256] Inamoto S, Kwartler CS, Lafont AL, Liang YY, Fadulu VT, Duraisamy S, et al. TGFBR2 mutations alter smooth muscle cell phenotype and predispose to thoracic aortic aneurysms and dissections. Cardiovasc Res 2010;88(3):520-9.

[257] Li G, Borger MA, Williams WG, Weisel RD, Mickle DA, Wigle ED, et al. Regional overexpression of insulin-like growth factor-I and transforming growth factor-beta1 in the myocardium of patients with hypertrophic obstructive cardiomyopathy. J Thorac Cardiovasc Surg 2002;123(1):89-95.

[258] Morrell NW, Yang X, Upton PD, Jourdan KB, Morgan N, Sheares KK, et al. Altered growth responses of pulmonary artery smooth muscle cells from patients with primary pulmonary hypertension to transforming growth factor-beta(1) and bone morphogenetic proteins. Circulation 2001;104(7):790-5.

[259] Cambien F, Ricard S, Troesch A, Mallet C, Generenaz L, Evans A, et al. Polymorphisms of the transforming growth factor-beta 1 gene in relation to myocardial infarction and blood pressure. The Etude Cas-Temoin de l'Infarctus du Myocarde (ECTIM) Study. Hypertension 1996;28(5):881-7.

[260] Chou HT, Chen CH, Tsai CH, Tsai FJ. Association between transforming growth factor-beta1 gene C-509T and T869C polymorphisms and rheumatic heart disease. Am Heart J 2004;148(1):181-6.

[261] Sie MP, Uitterlinden AG, Bos MJ, Arp PP, Breteler MM, Koudstaal PJ, et al. TGF-beta 1 polymorphisms and risk of myocardial infarction and stroke: the Rotterdam Study. Stroke 2006;37(11):2667-71.

[262] Kim Y, Lee C. The gene encoding transforming growth factor beta 1 confers risk of ischemic stroke and vascular dementia. Stroke 2006;37(11):2843-5.

[263] Devaux Y, Bousquenaud M, Rodius S, Marie PY, Maskali F, Zhang L, et al. Transforming growth factor beta receptor 1 is a new candidate prognostic biomarker after acute myocardial infarction. BMC Med Genomics 2010;5(4):83.

[264] Najar RA, Ghaderian SM, Panah AS. Association of transforming growth factor-beta1 gene polymorphisms with genetic susceptibility to acute myocardial infarction. Am J Med Sci 2011;342(5):365-70.

[265] Sovari AA, Morita N, Weiss JN, Karagueuzian HS. Serum transforming growth factor-beta1 as a risk stratifier of sudden cardiac death. Med Hypotheses 2008;71(2):262-5.

[266] Zernecke A, Shagdarsuren E, Weber C. Chemokines in atherosclerosis: an update. Arterioscler Thromb Vasc Biol 2008;28(11):1897-908.

[267] Zernecke A, Weber C. Chemokines in the vascular inflammatory response of atherosclerosis. Cardiovasc Res 2010;86(2):192-201.

[268] von Hundelshausen P, Koenen RR, Sack M, Mause SF, Adriaens W, Proudfoot AE, et al. Heterophilic interactions of platelet factor 4 and RANTES promote monocyte arrest on endothelium. Blood 2005;105(3):924-30.

[269] Weber C, Belge KU, von Hundelshausen P, Draude G, Steppich B, Mack M, et al. Differential chemokine receptor expression and function in human monocyte subpopulations. J Leukoc Biol 2000;67(5):699-704.

[270] Apostolakis S, Vogiatzi K, Amanatidou V, Spandidos DA. Interleukin 8 and cardiovascular disease. Cardiovasc Res 2009;84(3):353-60.

[271] Remick DG. Interleukin-8. Crit Care Med 2005;33(12 Suppl):S466-7. [272] Bochkov VN, Philippova M, Oskolkova O, Kadl A, Furnkranz A, Karabeg E, et al. Oxidized

phospholipids stimulate angiogenesis via autocrine mechanisms, implicating a novel role for lipid oxidation in the evolution of atherosclerotic lesions. Circ Res 2006;99(8):900-8.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 37

[273] Yoshida S, Ono M, Shono T, Izumi H, Ishibashi T, Suzuki H, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol 1997;17(7):4015-23.

[274] Tuschil A, Lam C, Haslberger A, Lindley I. Interleukin-8 stimulates calcium transients and promotes epidermal cell proliferation. J Invest Dermatol 1992;99(3):294-8.

[275] Hechtman DH, Cybulsky MI, Fuchs HJ, Baker JB, Gimbrone MA, Jr. Intravascular IL-8. Inhibitor of polymorphonuclear leukocyte accumulation at sites of acute inflammation. J Immunol 1991;147(3):883-92.

[276] Boisvert WA, Santiago R, Curtiss LK, Terkeltaub RA. A leukocyte homologue of the IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J Clin Invest 1998;101(2):353-63.

[277] Zhou RH, Shi Q, Gao HQ, Shen BJ. Changes in serum interleukin-8 and interleukin-12 levels in patients with ischemic heart disease in a Chinese population. J Atheroscler Thromb 2001;8(1):30-2.

[278] Hashmi S, Zeng QT. Role of interleukin-17 and interleukin-17-induced cytokines interleukin-6 and interleukin-8 in unstable coronary artery disease. Coron Artery Dis 2006;17(8):699-706.

[279] Dominguez-Rodriguez A, Abreu-Gonzalez P, Garcia-Gonzalez M, Ferrer J. Prognostic value of interleukin-8 as a predictor of heart failure in patients with myocardial infarction and percutaneous intervention. Int J Cardiol 2006;111(1):158-60.

[280] Inoue T, Komoda H, Nonaka M, Kameda M, Uchida T, Node K. Interleukin-8 as an independent predictor of long-term clinical outcome in patients with coronary artery disease. Int J Cardiol 2008;124(3):319-25.

[281] Herder C, Baumert J, Thorand B, Martin S, Lowel H, Kolb H, et al. Chemokines and incident coronary heart disease: results from the MONICA/KORA Augsburg case-cohort study, 1984-2002. Arterioscler Thromb Vasc Biol 2006;26(9):2147-52.

[282] Rothenbacher D, Muller-Scholze S, Herder C, Koenig W, Kolb H. Differential expression of chemokines, risk of stable coronary heart disease, and correlation with established cardiovascular risk markers. Arterioscler Thromb Vasc Biol 2006;26(1):194-9.

[283] Troseid M, Lappegard KT, Claudi T, Damas JK, Morkrid L, Brendberg R, et al. Exercise reduces plasma levels of the chemokines MCP-1 and IL-8 in subjects with the metabolic syndrome. Eur Heart J 2004;25(4):349-55.

[284] Deshmane SL, Kremlev S, Amini S, Sawaya BE. Monocyte chemoattractant protein-1 (MCP-1): an overview. J Interferon Cytokine Res 2009;29(6):313-26.

[285] Gosling J, Slaymaker S, Gu L, Tseng S, Zlot CH, Young SG, et al. MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress human apolipoprotein B. J Clin Invest 1999;103(6):773-8.

[286] Gu L, Okada Y, Clinton SK, Gerard C, Sukhova GK, Libby P, et al. Absence of monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol Cell 1998;2(2):275-81.

[287] Deo R, Khera A, McGuire DK, Murphy SA, Meo Neto Jde P, Morrow DA, et al. Association among plasma levels of monocyte chemoattractant protein-1, traditional cardiovascular risk factors, and subclinical atherosclerosis. J Am Coll Cardiol 2004;44(9):1812-8.

[288] Martinovic I, Abegunewardene N, Seul M, Vosseler M, Horstick G, Buerke M, et al. Elevated monocyte chemoattractant protein-1 serum levels in patients at risk for coronary artery disease. Circ J 2005;69(12):1484-9.

[289] de Lemos JA, Morrow DA, Sabatine MS, Murphy SA, Gibson CM, Antman EM, et al. Association Between Plasma Levels of Monocyte Chemoattractant Protein-1 and Long-Term Clinical Outcomes in Patients With Acute Coronary Syndromes. Circulation 2003;107(5):690-5.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 38

[290] de Lemos JA, Morrow DA, Sabatine MS, Murphy SA, Gibson CM, Antman EM, et al. Association between plasma levels of monocyte chemoattractant protein-1 and long-term clinical outcomes in patients with acute coronary syndromes. Circulation 2003;107(5):690-5.

[291] Kervinen H, Manttari M, Kaartinen M, Makynen H, Palosuo T, Pulkki K, et al. Prognostic usefulness of plasma monocyte/macrophage and T-lymphocyte activation markers in patients with acute coronary syndromes. Am J Cardiol 2004;94(8):993-6.

[292] Braunersreuther V, Mach F, Steffens S. The specific role of chemokines in atherosclerosis. Thromb Haemost 2007;97(5):714-21.

[293] Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 2007;13(5):587-96.

[294] Morand EF, Leech M, Bernhagen J. MIF: a new cytokine link between rheumatoid arthritis and atherosclerosis. Nat Rev Drug Discov 2006;5(5):399-410.

[295] Calandra T, Echtenacher B, Roy DL, Pugin J, Metz CN, Hultner L, et al. Protection from septic shock by neutralization of macrophage migration inhibitory factor. Nat Med 2000;6(2):164-70.

[296] Donnelly SC, Haslett C, Reid PT, Grant IS, Wallace WA, Metz CN, et al. Regulatory role for macrophage migration inhibitory factor in acute respiratory distress syndrome. Nat Med 1997;3(3):320-3.

[297] Kleemann R, Bucala R. Macrophage migration inhibitory factor: critical role in obesity, insulin resistance, and associated comorbidities. Mediators Inflamm 2010;2010(610479.

[298] Santos LL, Morand EF. Macrophage migration inhibitory factor: a key cytokine in RA, SLE and atherosclerosis. Clin Chim Acta 2009;399(1-2):1-7.

[299] Burger-Kentischer A, Goebel H, Seiler R, Fraedrich G, Schaefer HE, Dimmeler S, et al. Expression of Macrophage Migration Inhibitory Factor in Different Stages of Human Atherosclerosis. Circulation 2002;105(13):1561-6.

[300] Lin SG, Yu XY, Chen YX, Huang XR, Metz C, Bucala R, et al. De novo expression of macrophage migration inhibitory factor in atherogenesis in rabbits. Circ Res 2000;87(12):1202-8.

[301] Schober A, Bernhagen J, Thiele M, Zeiffer U, Knarren S, Roller M, et al. Stabilization of atherosclerotic plaques by blockade of macrophage migration inhibitory factor after vascular injury in apolipoprotein E-deficient mice. Circulation 2004;109(3):380-5.

[302] Boekholdt SM, Peters RJ, Day NE, Luben R, Bingham SA, Wareham NJ, et al. Macrophage migration inhibitory factor and the risk of myocardial infarction or death due to coronary artery disease in adults without prior myocardial infarction or stroke: the EPIC-Norfolk Prospective Population study. Am J Med 2004;117(6):390-7.

[303] Herder C, Illig T, Baumert J, Muller M, Klopp N, Khuseyinova N, et al. Macrophage migration inhibitory factor (MIF) and risk for coronary heart disease: results from the MONICA/KORA Augsburg case-cohort study, 1984-2002. Atherosclerosis 2008;200(2):380-8.

[304] Makino A, Nakamura T, Hirano M, Kitta Y, Sano K, Kobayashi T, et al. High plasma levels of macrophage migration inhibitory factor are associated with adverse long-term outcome in patients with stable coronary artery disease and impaired glucose tolerance or type 2 diabetes mellitus. Atherosclerosis 2010;213(2):573-8.

[305] Sies H, Cadenas E. Oxidative stress: damage to intact cells and organs. Philos Trans R Soc Lond B Biol Sci 1985;311(1152):617-31.

[306] Refsgaard HH, Tsai L, Stadtman ER. Modifications of proteins by polyunsaturated fatty acid peroxidation products. Proc Natl Acad Sci U S A 2000;97(2):611-6.

[307] Morrow JD. Quantification of Isoprostanes as Indices of Oxidant Stress and the Risk of Atherosclerosis in Humans. Arterioscler Thromb Vasc Biol 2005;25(2):279-86.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 39

[308] Pratico D, Rokach J, Lawson J, FitzGerald GA. F2-isoprostanes as indices of lipid peroxidation in inflammatory diseases. Chem Phys Lipids 2004;128(1-2):165-71.

[309] Mallat Z, Nakamura T, Ohan J, Leseche G, Tedgui A, Maclouf J, et al. The relationship of hydroxyeicosatetraenoic acids and F2-isoprostanes to plaque instability in human carotid atherosclerosis. J Clin Invest 1999;103(3):421-7.

[310] Zhang ZJ. Systematic review on the association between F2-isoprostanes and cardiovascular disease. Annals of Clinical Biochemistry 2012.

[311] Milne GL, Gao B, Terry ES, Zackert WE, Sanchez SC. Measurement of F2- isoprostanes and isofurans using gas chromatography–mass spectrometry. Free Radical Biology and Medicine In Press0).

[312] Six DA, Dennis EA. The expanding superfamily of phospholipase A(2) enzymes: classification and characterization. Biochim Biophys Acta 2000;1488(1-2):1-19.

[313] Stafforini DM, Prescott SM, McIntyre TM. Human plasma platelet-activating factor acetylhydrolase. Purification and properties. J Biol Chem 1987;262(9):4223-30.

[314] Zalewski A, Macphee C. Role of Lipoprotein-Associated Phospholipase A2 in Atherosclerosis: Biology, Epidemiology, and Possible Therapeutic Target. Arterioscler Thromb Vasc Biol 2005;25(5):923-31.

[315] Caslake MJ, Packard CJ. Lipoprotein-associated phospholipase A2 (platelet-activating factor acetylhydrolase) and cardiovascular disease. Curr Opin Lipidol 2003;14(4):347-52.

[316] Stafforini DM, McIntyre TM, Zimmerman GA, Prescott SM. Platelet-activating Factor Acetylhydrolases. J Biol Chem 1997;272(29):17895-8.

[317] Watson AD, Navab M, Hama SY, Sevanian A, Prescott SM, Stafforini DM, et al. Effect of platelet activating factor-acetylhydrolase on the formation and action of minimally oxidized low density lipoprotein. J Clin Invest 1995;95(2):774-82.

[318] Tselepis AD, John Chapman M. Inflammation, bioactive lipids and atherosclerosis: potential roles of a lipoprotein-associated phospholipase A2, platelet activating factor-acetylhydrolase. Atheroscler Suppl 2002;3(4):57-68.

[319] Dada N, Kim NW, Wolfert RL. Lp-PLA2: an emerging biomarker of coronary heart disease. Expert Review of Molecular Diagnostics 2002;2(1):17-22.

[320] Yamada Y, Yoshida H, Ichihara S, Imaizumi T, Satoh K, Yokota M. Correlations between plasma platelet-activating factor acetylhydrolase (PAF-AH) activity and PAF-AH genotype, age, and atherosclerosis in a Japanese population. Atherosclerosis 2000;150(1):209-16.

[321] Yamada Y, Ichihara S, Fujimura T, Yokota M. Identification of the G994--> T missense in exon 9 of the plasma platelet-activating factor acetylhydrolase gene as an independent risk factor for coronary artery disease in Japanese men. Metabolism 1998;47(2):177-81.

[322] Ballantyne CM, Hoogeveen RC, Bang H, Coresh J, Folsom AR, Heiss G, et al. Lipoprotein-Associated Phospholipase A2, High-Sensitivity C-Reactive Protein, and Risk for Incident Coronary Heart Disease in Middle-Aged Men and Women in the Atherosclerosis Risk in Communities (ARIC) Study. Circulation 2004;109(7):837-42.

[323] Brilakis ES, McConnell JP, Lennon RJ, Elesber AA, Meyer JG, Berger PB. Association of lipoprotein-associated phospholipase A2 levels with coronary artery disease risk factors, angiographic coronary artery disease, and major adverse events at follow-up. Eur Heart J 2005;26(2):137-44.

[324] Mohiuddin I, Chai H, Lin PH, Lumsden AB, Yao Q, Chen C. Nitrotyrosine and chlorotyrosine: clinical significance and biological functions in the vascular system. J Surg Res 2006;133(2):143-9.

[325] Peluffo G, Radi R. Biochemistry of protein tyrosine nitration in cardiovascular pathology. Cardiovascular research 2007;75(2):291-302.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 40

[326] Gaut JP, Byun J, Tran HD, Lauber WM, Carroll JA, Hotchkiss RS, et al. Myeloperoxidase produces nitrating oxidants in vivo. J Clin Invest 2002;109(10):1311-9.

[327] Fernandez AP, Serrano J, Castro S, Salazar FJ, Lopez JC, Rodrigo J, et al. Distribution of nitric oxide synthases and nitrotyrosine in the kidney of spontaneously hypertensive rats. J Hypertens 2003;21(12):2375-88.

[328] Bro S, Bentzon JF, Falk E, Andersen CB, Olgaard K, Nielsen LB. Chronic Renal Failure Accelerates Atherogenesis in Apolipoprotein E-Deficient Mice. J Am Soc Nephrol 2003;14(10):2466-74.

[329] Zou M-H, Cohen RA, Ullrich V. Peroxynitrite and Vascular Endothelial Dysfunction in Diabetes Mellitus. Endothelium 2004;11(2):89 - 97.

[330] Pennathur S, Bergt C, Shao B, Byun J, Kassim SY, Singh P, et al. Human Atherosclerotic Intima and Blood of Patients with Established Coronary Artery Disease Contain High Density Lipoprotein Damaged by Reactive Nitrogen Species. J Biol Chem 2004;279(41):42977-83.

[331] Da Ros R, Quagliaro L, Gasparini D, Barillari G, Ceriello A. Nitrotyrosine in peripheral vascular disease. Journal of Thrombosis and Haemostasis 2003;1(2):382-3.

[332] Shishehbor Mh ARJBM, et al. Association of nitrotyrosine levels with cardiovascular disease and modulation by statin therapy. JAMA: The Journal of the American Medical Association 2003;289(13):1675-80.

[333] Thomson L, Tenopoulou M, Lightfoot R, Tsika E, Parastatidis I, Martinez M, et al. Immunoglobulins Against Tyrosine-Nitrated Epitopes in Coronary Artery Disease: Clinical Perspective. Circulation 2012;126(20):2392-401.

[334] Shishehbor MH, Brennan ML, Aviles RJ, Fu X, Penn MS, Sprecher DL, et al. Statins promote potent systemic antioxidant effects through specific inflammatory pathways. Circulation 2003;108(4):426-31.

[335] Feng Q, Lu X, Jones DL, Shen J, Arnold JMO. Increased inducible nitric oxide synthase expression contributes to myocardial dysfunction and higher mortality after myocardial infarction in mice. Circulation 2001;104(6):700-4.

[336] Steinberg D. Low Density Lipoprotein Oxidation and Its Pathobiological Significance. J Biol Chem 1997;272(34):20963-6.

[337] Witztum JL, Steinberg D. The Oxidative Modification Hypothesis of Atherosclerosis: Does It Hold for Humans? Trends in Cardiovascular Medicine 2001;11(3-4):93-102.

[338] Boulanger Cm Fau - Tanner FC, Tanner Fc Fau - Bea ML, Bea Ml Fau - Hahn AW, Hahn Aw Fau - Werner A, Werner A Fau - Luscher TF, Luscher TF. Oxidized low density lipoproteins induce mRNA expression and release of endothelin from human and porcine endothelium. SO - Circ Res 1992 Jun;70(6):1191-7.

[339] Ruef J, Moser M, Kubler W, Bode C. Induction of endothelin-1 expression by oxidative stress in vascular smooth muscle cells. Cardiovasc Pathol 2001;10(6):311-5.

[340] Tsimikas S, Willeit P, Willeit J, Santer P, Mayr M, Xu Q, et al. Oxidation-Specific Biomarkers, Prospective 15-Year Cardiovascular and Stroke Outcomes, and Net Reclassification of Cardiovascular Events. Journal of the American College of Cardiology 2012;60(21):2218-29.

[341] Laczik R, Szodoray P, Veres K, Szomjak E, Csipo I, Sipka S, Jr., et al. Assessment of IgG antibodies to oxidized LDL in patients with acute coronary syndrome. Lupus 2011;20(7):730-5.

[342] Matsuo Y, Kubo T, Okumoto Y, Ishibashi K, Komukai K, Tanimoto T, et al. Circulating malondialdehyde-modified low-density lipoprotein levels are associated with the presence of thin-cap fibroatheromas determined by optical coherence tomography in coronary artery disease. Eur Heart J Cardiovasc Imaging 2012.

[343] Imazu M, Ono K, Tadehara F, Kajiwara K, Yamamoto H, Sumii K, et al. Plasma levels of oxidized low density lipoprotein are associated with stable angina pectoris and modalities of acute coronary syndrome. Int Heart J 2008;49(5):515-24.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 41

[344] Stellos K, Sauter R, Fahrleitner M, Grimm J, Stakos D, Emschermann F, et al. Binding of oxidized low-density lipoprotein on circulating platelets is increased in patients with acute coronary syndromes and induces platelet adhesion to vascular wall in vivo--brief report. Arterioscler Thromb Vasc Biol 2012;32(8):2017-20.

[345] Tsimikas S, Bergmark C, Beyer RW, Patel R, Pattison J, Miller E, et al. Temporal increases in plasma markers of oxidized low-density lipoprotein strongly reflect the presence of acute coronary syndromes. J Am Coll Cardiol 2003;41(3):360-70.

[346] Ehara S, Naruko T, Shirai N, Itoh A, Hai E, Sugama Y, et al. Small coronary calcium deposits and elevated plasma levels of oxidized low density lipoprotein are characteristic of acute myocardial infarction. J Atheroscler Thromb 2008;15(2):75-81.

[347] Ehara S, Ueda M, Naruko T, Haze K, Itoh A, Otsuka M, et al. Elevated levels of oxidized low density lipoprotein show a positive relationship with the severity of acute coronary syndromes. Circulation 2001;103(15):1955-60.

[348] Dominguez-Rodriguez A, Abreu-Gonzalez P, Garcia-Gonzalez M, Ferrer-Hita J, Vargas M, Reiter RJ. Elevated levels of oxidized low-density lipoprotein and impaired nocturnal synthesis of melatonin in patients with myocardial infarction. Atherosclerosis 2005;180(1):101-5.

[349] Yamashita H, Ehara S, Yoshiyama M, Naruko T, Haze K, Shirai N, et al. Elevated plasma levels of oxidized low-density lipoprotein relate to the presence of angiographically detected complex and thrombotic coronary artery lesion morphology in patients with unstable angina. Circ J 2007;71(5):681-7.

[350] Andican G, Seven A, Uncu M, Cantasdemir M, Numan F, Burcak G. Oxidized LDL and anti-oxLDL antibody levels in peripheral atherosclerotic disease. Scand J Clin Lab Invest 2008;68(6):473-8.

[351] Yamaji M, Tsutamoto T, Kawahara C, Nishiyama K, Yamamoto T, Fujii M, et al. Serum cortisol as a useful predictor of cardiac events in patients with chronic heart failure: the impact of oxidative stress. Circ Heart Fail 2009;2(6):608-15.

[352] Jorde UP, Colombo PC, Ahuja K, Hudaihed A, Onat D, Diaz T, et al. Exercise-induced increases in oxidized low-density lipoprotein are associated with adverse outcomes in chronic heart failure. J Card Fail 2007;13(9):759-64.

[353] Uno M, Kitazato KT, Nishi K, Itabe H, Nagahiro S. Raised plasma oxidised LDL in acute cerebral infarction. J Neurol Neurosurg Psychiatry 2003;74(3):312-6.

[354] Kassner SS, Kollmar R, Bonaterra GA, Hildebrandt W, Schwab S, Kinscherf R. The early immunological response to acute ischemic stroke: differential gene expression in subpopulations of mononuclear cells. Neuroscience 2009;160(2):394-401.

[355] Ghio M, Fabbi P, Contini P, Fedele M, Brunelli C, Indiveri F, et al. OxLDL- and HSP-60 antigen-specific CD8(+) T lymphocytes are detectable in the peripheral blood of patients suffering from coronary artery disease. Clin Exp Med 2012.

[356] Koenig W, Karakas M, Zierer A, Herder C, Baumert J, Meisinger C, et al. Oxidized LDL and the risk of coronary heart disease: results from the MONICA/KORA Augsburg Study. Clin Chem 2011;57(8):1196-200.

[357] Faviou E, Vourli G, Nounopoulos C, Zachari A, Dionyssiou-Asteriou A. Circulating oxidized low density lipoprotein, autoantibodies against them and homocysteine serum levels in diagnosis and estimation of severity of coronary artery disease. Free Radic Res 2005;39(4):419-29.

[358] Huang B, Chen Y, Xie Q, Lin G, Wu Y, Feng Y, et al. CagA-positive Helicobacter pylori strains enhanced coronary atherosclerosis by increasing serum OxLDL and HsCRP in patients with coronary heart disease. Dig Dis Sci 2011;56(1):109-14.

[359] Meuwissen M, van der Wal AC, Niessen HW, Koch KT, de Winter RJ, van der Loos CM, et al. Colocalisation of intraplaque C reactive protein, complement, oxidised low density lipoprotein,

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 42

and macrophages in stable and unstable angina and acute myocardial infarction. J Clin Pathol 2006;59(2):196-201.

[360] Naruko T, Ueda M, Ehara S, Itoh A, Haze K, Shirai N, et al. Persistent high levels of plasma oxidized low-density lipoprotein after acute myocardial infarction predict stent restenosis. Arterioscler Thromb Vasc Biol 2006;26(4):877-83.

[361] Itabe H. Oxidized low-density lipoprotein as a biomarker of in vivo oxidative stress: from atherosclerosis to periodontitis. J Clin Biochem Nutr 2012;51(1):1-8.

[362] Itabe H, Ueda M. Measurement of plasma oxidized low-density lipoprotein and its clinical implications. J Atheroscler Thromb 2007;14(1):1-11.

[363] Kalen A, Norling B, Appelkvist EL, Dallner G. Ubiquinone biosynthesis by the microsomal fraction from rat liver. Biochim Biophys Acta 1987;926(1):70-8.

[364] Overvad K, Diamant B, Holm L, Holmer G, Mortensen SA, Stender S. Coenzyme Q10 in health and disease. Eur J Clin Nutr 1999;53(10):764-70.

[365] Singh U, Devaraj S, Jialal I. Coenzyme Q10 supplementation and heart failure. Nutr Rev 2007;65(6 Pt 1):286-93.

[366] Mellors A, Tappel A. The inhibition of mitochondrial peroxidation by ubiquinone and ubiquinol. Journal of Biological chemistry 1966;241(19):4353-6.

[367] Stocker R, Bowry VW, Frei B. Ubiquinol-10 protects human low density lipoprotein more efficiently against lipid peroxidation than does alpha-tocopherol. Proceedings of the National Academy of Sciences 1991;88(5):1646-50.

[368] Kettawan A, Takahashi T, Kongkachuichai R, Charoenkiatkul S, Kishi T, Okamoto T. Protective effects of coenzyme Q10 on decreased oxidative stress resistance induced by simvastatin. Journal of clinical biochemistry and nutrition 2007;40(3):194.

[369] Shi H, Noguchi N, Niki E. Comparative study on dynamics of antioxidative action of alpha-tocopheryl hydroquinone, ubiquinol, and alpha-tocopherol against lipid peroxidation. Free Radic Biol Med 1999;27(3-4):334-46.

[370] Chan AC. Partners in defense, vitamin E and vitamin C. Can J Physiol Pharmacol 1993;71(9):725-31.

[371] Constantinescu A, Maguire JJ, Packer L. Interactions between ubiquinones and vitamins in membranes and cells. Mol Aspects Med 1994;15 Suppl(s57-65.

[372] Villalba JM, Navarro F, Cordoba F, Serrano A, Arroyo A, Crane FL, et al. Coenzyme Q Reductase from Liver Plasma Membrane: Purification and Role in Trans-Plasma-Membrane Electron Transport. Proceedings of the National Academy of Sciences 1995;92(11):4887-91.

[373] Alleva R, Tomasetti M, Battino M, Curatola G, Littarru GP, Folkers K. The Roles of Coenzyme Q10 and Vitamin E on the Peroxidation of Human Low Density Lipoprotein Subfractions. Proceedings of the National Academy of Sciences 1995;92(20):9388-91.

[374] Folkers K, Littarru GP, Nakamura R, Scholler J. Survey and new clinical studies on conenzyme Q in human muscular dystrophy. Int J Vitam Nutr Res 1972;42(1):139-63.

[375] Folkers K, Wolaniuk J, Simonsen R, Morishita M, Vadhanavikit S. Biochemical Rationale and the Cardiac Response of Patients with Muscle Disease to Therapy with Coenzyme Q10. Proceedings of the National Academy of Sciences 1985;82(13):4513-6.

[376] Hanaki Y, Sugiyama S, Ozawa T, Ohno M. Ratio of low-density lipoprotein cholesterol to ubiquinone as a coronary risk factor. N Engl J Med 1991;325(11):814-5.

[377] Littarru GP, Jones D, Scholler J, Folkers K. Deficiency of coenzyme Q 10 in a succinate-CoQ 10 -Enzyme in the dystrophic rabbit on an antioxidant deficient diet. Int J Vitam Nutr Res 1972;42(1):127-38.

[378] Kontush A, Reich A, Baum K, Spranger T, Finckh B, Kohlschutter A, et al. Plasma ubiquinol-10 is decreased in patients with hyperlipidaemia. Atherosclerosis 1997;129(1):119-26.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 43

[379] Hasegawa G, Yamamoto Y, Zhi JG, Tanino Y, Yamasaki M, Yano M, et al. Daily profile of plasma %CoQ10 level, a biomarker of oxidative stress, in patients with diabetes manifesting postprandial hyperglycaemia. Acta Diabetol 2005;42(4):179-81.

[380] Dröge W, Breitkreutz R. Glutathione and immune function. Proceedings of the Nutrition Society 2000;59(04):595-600.

[381] Gmünder H, Roth S, Eck HP, Gallas H, Mihm S, Dröge W. Interleukin-2 mRNA expression, lymphokine production and DNA synthesis in glutathione-depleted T cells. Cellular immunology 1990;130(2):520-8.

[382] Kinscherf R, Fischbach T, Mihm S, Roth S, Hohenhaus-Sievert E, Weiss C, et al. Effect of glutathione depletion and oral N-acetyl-cysteine treatment on CD4+ and CD8+ cells. The FASEB journal 1994;8(6):448-51.

[383] De Chiara B, Mafrici A, Campolo J, Famoso G, Sedda V, Parolini M, et al. Low plasma glutathione levels after reperfused acute myocardial infarction are associated with late cardiac events. Coron Artery Dis 2007;18(2):77-82.

[384] Campolo J, De Maria R, Caruso R, Accinni R, Turazza F, Parolini M, et al. Blood glutathione as independent marker of lipid peroxidation in heart failure. Int J Cardiol 2007;117(1):45-50.

[385] Radovanovic S, Savic-Radojevic A, Pljesa-Ercegovac M, Djukic T, Suvakov S, Krotin M, et al. Markers of oxidative damage and antioxidant enzyme activities as predictors of morbidity and mortality in patients with chronic heart failure. J Card Fail 2012;18(6):493-501.

[386] Kumar A, Nagtilak S, Sivakanesan R, Gunasekera S. Cardiovascular risk factors in elderly normolipidemic acute myocardial infarct patients--a case controlled study from India. Southeast Asian J Trop Med Public Health 2009;40(3):581-92.

[387] Maksimenko AV, Vavaev AV. Antioxidant enzymes as potential targets in cardioprotection and treatment of cardiovascular diseases. Enzyme antioxidants: the next stage of pharmacological counterwork to the oxidative stress. Heart Int 2012;7(1):e3.

[388] Chan PH. Reactive Oxygen Radicals in Signaling and Damage in the Ischemic Brain. J Cereb Blood Flow Metab 2001;21(1):2-14.

[389] Jung O, Marklund SL, Xia N, Busse R, Brandes RP. Inactivation of extracellular superoxide dismutase contributes to the development of high-volume hypertension. Arterioscler Thromb Vasc Biol 2007;27(3):470-7.

[390] Mugge A, Elwell JH, Peterson TE, Harrison DG. Release of intact endothelium-derived relaxing factor depends on endothelial superoxide dismutase activity. 1991;260(2):C219-25.

[391] Omar HA, Cherry PD, Mortelliti MP, Burke-Wolin T, Wolin MS. Inhibition of coronary artery superoxide dismutase attenuates endothelium-dependent and -independent nitrovasodilator relaxation. Circ Res 1991;69(3):601-8.

[392] Gao Y. The multiple actions of NO. Pflügers Arch 2010;459(6):829-39. [393] Michel T, Vanhoutte PM. Cellular signaling and NO production. Pflügers Arch 2010;459(6):807-

16. [394] Moncada S, Higgs EA. Nitric oxide and the vascular endothelium. Handb Exp Pharmacol 2006176

Pt 1):213-54. [395] Napoli C, de Nigris F, Williams-Ignarro S, Pignalosa O, Sica V, Ignarro LJ. Nitric oxide and

atherosclerosis: An update. Nitric Oxide 2006;15(4):265-79. [396] Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and

ugly. 1996;271(5):C1424-37. [397] Darley-Usmar V, Wiseman H, Halliwell B. Nitric oxide and oxygen radicals: a question of balance.

FEBS Lett 1995;369(2-3):131-5. [398] Gotoh N, Niki E. Rates of interactions of superoxide with vitamin E, vitamin C and related

compounds as measured by chemiluminescence. Biochim Biophys Acta 1992;1115(3):201-7.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 44

[399] Marczin N, El-Habashi N, Hoare GS, Bundy RE, Yacoub M. Antioxidants in myocardial ischemia-reperfusion injury: therapeutic potential and basic mechanisms. Arch Biochem Biophys 2003;420(2):222-36.

[400] Ulker S, McMaster D, McKeown PP, Bayraktutan U. Impaired activities of antioxidant enzymes elicit endothelial dysfunction in spontaneous hypertensive rats despite enhanced vascular nitric oxide generation. Cardiovasc Res 2003;59(2):488-500.

[401] Uddin M, Yang H, Shi M, Polley-Mandal M, Guo Z. Elevation of oxidative stress in the aorta of genetically hypertensive mice. Mech Ageing Dev 2003;124(7):811-7.

[402] t Hoen PAC, Van der Lans CAC, Van Eck M, Bijsterbosch MK, Van Berkel TJC, Twisk J. Aorta of ApoE-Deficient Mice Responds to Atherogenic Stimuli by a Prelesional Increase and Subsequent Decrease in the Expression of Antioxidant Enzymes. 2003;93(3):262-9.

[403] Huang TT, Yasunami M, Carlson EJ, Gillespie AM, Reaume AG, Hoffman EK, et al. Superoxide-mediated cytotoxicity in superoxide dismutase-deficient fetal fibroblasts. Arch Biochem Biophys 1997;344(2):424-32.

[404] Macmillan-Crow LA, Cruthirds DL. Invited review: manganese superoxide dismutase in disease. Free Radic Res 2001;34(4):325-36.

[405] Faraci FM, Didion SP. Vascular Protection: Superoxide Dismutase Isoforms in the Vessel Wall. 2004;24(8):1367-73.

[406] Johnson F, Giulivi C. Superoxide dismutases and their impact upon human health. Mol Aspects Med 2005;26(4-5):340-52.

[407] Jung O, Marklund SL, Geiger H, Pedrazzini T, Busse R, Brandes RP. Extracellular Superoxide Dismutase Is a Major Determinant of Nitric Oxide Bioavailability: In Vivo and Ex Vivo Evidence From ecSOD-Deficient Mice. 2003;93(7):622-9.

[408] Gupta S, Sodhi S, Mahajan V. Correlation of antioxidants with lipid peroxidation and lipid profile in patients suffering from coronary artery disease. Expert Opin Ther Targets 2009;13(8):889-94.

[409] Landmesser U, Merten R, Spiekermann S, Buttner K, Drexler H, Hornig B. Vascular extracellular superoxide dismutase activity in patients with coronary artery disease: relation to endothelium-dependent vasodilation. Circulation 2000;101(19):2264-70.

[410] Tasaki H, Yamashita K, Tsutsui M, Kamezaki F, Kubara T, Tanaka S, et al. Heparin-released extracellular superoxide dismutase is reduced in patients with coronary artery atherosclerosis. Atherosclerosis 2006;187(1):131-8.

[411] Yamashita K, Takahiro K, Kamezaki F, Adachi T, Tasaki H. Decreased plasma extracellular superoxide dismutase level in patients with vasospastic angina. Atherosclerosis 2007;191(1):147-52.

[412] Kariz S, Nikolajevic Starcevic J, Petrovic D. Association of manganese superoxide dismutase and glutathione S-transferases genotypes with myocardial infarction in patients with type 2 diabetes mellitus. Diabetes Res Clin Pract 2012;98(1):144-50.

[413] Neves AL, Mohammedi K, Emery N, Roussel R, Fumeron F, Marre M, et al. Allelic variations in superoxide dismutase-1 (SOD1) gene and renal and cardiovascular morbidity and mortality in type 2 diabetic subjects. Mol Genet Metab 2012;106(3):359-65.

[414] Mulder DJ, van Haelst PL, Gross S, de Leeuw K, Bijzet J, Graaff R, et al. Skin autofluorescence is elevated in patients with stable coronary artery disease and is associated with serum levels of neopterin and the soluble receptor for advanced glycation end products. Atherosclerosis 2007.

[415] Haught WH, Mansour M, Rothlein R, Kishimoto TK, Mainolfi EA, Hendricks JB, et al. Alterations in circulating intercellular adhesion molecule-1 and L-selectin: further evidence for chronic inflammation in ischemic heart disease. Am Heart J 1996;132(1 Pt 1):1-8.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 45

[416] Cheng CH, Lin PT, Liaw YP, Ho CC, Tsai TP, Chou MC, et al. Plasma pyridoxal 5'-phosphate and high-sensitivity C-reactive protein are independently associated with an increased risk of coronary artery disease. Nutrition 2008;24(3):239-44.

[417] Tanner H, Mohacsi P, Fuller-Bicer GA, Rieben R, Meier B, Hess O, et al. Cytokine activation and disease progression in patients with stable moderate chronic heart failure. J Heart Lung Transplant 2007;26(6):622-9.

[418] Yin WH, Chen JW, Jen HL, Chiang MC, Huang WP, Feng AN, et al. Independent prognostic value of elevated high-sensitivity C-reactive protein in chronic heart failure. Am Heart J 2004;147(5):931-8.

[419] Witte KK, Ford SJ, Preston T, Parker JD, Clark AL. Fibrinogen synthesis is increased in cachectic patients with chronic heart failure. Int J Cardiol 2008;129(3):363-7.

[420] Kozdag G, Ertas G, Kilic T, Acar E, Agir A, Sahin T, et al. Elevated level of high-sensitivity C-reactive protein is important in determining prognosis in chronic heart failure. Med Sci Monit 2010;16(3):CR156-61.

[421] Toth MJ, Ades PA, Tischler MD, Tracy RP, LeWinter MM. Immune activation is associated with reduced skeletal muscle mass and physical function in chronic heart failure. Int J Cardiol 2006;109(2):179-87.

[422] Maeda H, Michiue T, Zhu BL, Ishikawa T, Quan L, Bessho Y, et al. Potential risk factors for sudden cardiac death: an analysis of medicolegal autopsy cases. Leg Med (Tokyo) 2009;11 Suppl 1(S263-5.

[423] Iso H, Noda H, Ikeda A, Yamagishi K, Inoue M, Iwasaki M, et al. The impact of C-reactive protein on risk of stroke, stroke subtypes, and ischemic heart disease in middle-aged Japanese: the Japan public health center-based study. J Atheroscler Thromb 2012;19(8):756-66.

[424] Djordjevic VB, Cosic V, Stojanovic I, Kundalic S, Zvezdanovic L, Deljanin-Ilic M, et al. Lipoprotein(a) Is the Best Single Marker in Assessing Unstable Angina Pectoris. Cardiol Res Pract 2011;2011(175363.

[425] Urbonaviciene G, Frystyk J, Flyvbjerg A, Urbonavicius S, Henneberg EW, Lindholt JS. Markers of inflammation in relation to long-term cardiovascular mortality in patients with lower-extremity peripheral arterial disease. Int J Cardiol 2012;160(2):89-94.

[426] Ridker PM, Stampfer MJ, Rifai N. Novel risk factors for systemic atherosclerosis: a comparison of C-reactive protein, fibrinogen, homocysteine, lipoprotein(a), and standard cholesterol screening as predictors of peripheral arterial disease. Jama 2001;285(19):2481-5.

[427] Vainas T, Stassen FR, de Graaf R, Twiss EL, Herngreen SB, Welten RJ, et al. C-reactive protein in peripheral arterial disease: relation to severity of the disease and to future cardiovascular events. J Vasc Surg 2005;42(2):243-51.

[428] Vu JD, Vu JB, Pio JR, Malik S, Franklin SS, Chen RS, et al. Impact of C-reactive protein on the likelihood of peripheral arterial disease in United States adults with the metabolic syndrome, diabetes mellitus, and preexisting cardiovascular disease. Am J Cardiol 2005;96(5):655-8.

[429] Shankar A, Li J, Nieto FJ, Klein BE, Klein R. Association between C-reactive protein level and peripheral arterial disease among US adults without cardiovascular disease, diabetes, or hypertension. Am Heart J 2007;154(3):495-501.

[430] Pande RL, Perlstein TS, Beckman JA, Creager MA. Association of insulin resistance and inflammation with peripheral arterial disease: the National Health and Nutrition Examination Survey, 1999 to 2004. Circulation 2008;118(1):33-41.

[431] De Haro J, Acin F, Medina FJ, Lopez-Quintana A, March JR. Relationship between the plasma concentration of C-reactive protein and severity of peripheral arterial disease. Clin Med Cardiol 2009;3(1-7.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 46

[432] de Haro Miralles J, Gonzalez AF, Varela Casariego C, Garcia FA. Onset of peripheral arterial disease: role of endothelin in endothelial dysfunction. Interact Cardiovasc Thorac Surg 2010;10(5):760-5.

[433] Criqui MH, Ho LA, Denenberg JO, Ridker PM, Wassel CL, McDermott MM. Biomarkers in peripheral arterial disease patients and near- and longer-term mortality. J Vasc Surg 2010;52(1):85-90.

[434] Luo Y, Wang Z, Li J, Xu Y. Serum CRP concentrations and severity of ischemic stroke subtypes. Can J Neurol Sci 2012;39(1):69-73.

[435] Huang Y, Jing J, Zhao XQ, Wang CX, Wang YL, Liu GF, et al. High-sensitivity C-reactive protein is a strong risk factor for death after acute ischemic stroke among Chinese. CNS Neurosci Ther 2012;18(3):261-6.

[436] Dewan KR, Rana PV. C-reactive protein and early mortality in acute ischemic stroke. Kathmandu Univ Med J (KUMJ) 2011;9(36):252-5.

[437] Chan CP, Jiang HL, Leung LY, Wan WM, Cheng NM, Ip WS, et al. Multiple atherosclerosis-related biomarkers associated with short- and long-term mortality after stroke. Clin Biochem 2012;45(16-17):1308-15.

[438] Shen C, Sun X, Wang H, Wang B, Xue Y, Li Y, et al. Association Study of CRP Gene and Ischemic Stroke in a Chinese Han Population. J Mol Neurosci 2012.

[439] Sadeghi M, Pourmoghaddas M, Tavasoli A, Roohafza H. Is there any Relationship Between C-Reactive Protein Level and Complex Coronary Plaques in Patients with Unstable Angina? ARYA Atheroscler 2010;6(1):31-4.

[440] Soysal D, Karakus V, Yavas HH, Biceroglu S, Koseoglu M, Yesil M. C-reactive protein in unstable angina pectoris and its relation to coronary angiographic severity and diffusion scores of coronary lesions. Anadolu Kardiyol Derg 2010;10(5):421-8.

[441] Niccoli G, Biasucci LM, Biscione C, Fusco B, Porto I, Leone AM, et al. Independent prognostic value of C-reactive protein and coronary artery disease extent in patients affected by unstable angina. Atherosclerosis 2008;196(2):779-85.

[442] Mallat Z, Steg PG, Benessiano J, Tanguy ML, Fox KA, Collet JP, et al. Circulating secretory phospholipase A2 activity predicts recurrent events in patients with severe acute coronary syndromes. J Am Coll Cardiol 2005;46(7):1249-57.

[443] Turko IV, Murad F. Protein nitration in cardiovascular diseases. Pharmacological reviews 2002;54(4):619-34.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 47

TABLES

TABLE 1. Inflammatory biomarkers

Biomarker Role Associated with End

point

AGEs Macrophage activation CAD[414], CVD[13-15]

ANG II Induce production of

ROS, cytokines, &

adhesion molecules

CHF[32], HT[31],

UA[33]

E-selectin Leukocyte recruitment ACS[43], CAD[44, 45],

MI[46], UA[47],

SCD[45]

HSP Amplify cytokine

production

CAD[65], MI[66],

SCD[66], Stroke [66]

ICAM-1 Adhesion; leukocyte

recruitment

CAD[118, 120, 121, 124,

415], PAD[123, 130],

[77, 122-124], UA[46]

MMPs Plaque rupture ACS[76, 78-80], MI [75],

UA[76], SCD[77]

MPO LDL-uptake; MMP

activation; plaque

rupture; endothelial

dysfunction

ACS[96], CAD[97, 98],

MI[94, 95]

PECAM-1 Leukocyte

migration/motility

CAD[106-108], MI[109]

VCAM-1 Adhesion; leukocyte

recruitment

ACS, CAD[51, 120],

PAD[130], SCD[45, 77],

stroke[48], UA[46, 47]

ACS, acute coronary syndromes; AGEs, advanced glycation end products; ANG, angiotensin; CAD, coronary artery

disease; CHF, chronic heart failure; CVD, cardiovascular disease; HSP, heat shock protein; HT, hypertension,

ICAM, intracellular cell-adhesion molecule; MI, myocardial infarction; MMPs, matrix metalloproteinases; MPO,

myeloperoxidase; PAD, peripheral arterial disease; PECAM, platelet endothelial cell adhesion molecule; SCD,

sudden cardiac death; UA, unstable angina; VCAM, vascular cell adhesion molecule.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 48

TABLE 2. Pro-inflammatory cytokines

Biomarker Role Associated with End

point

CD40 Thrombosis; MMP

expression; endothelial

dysfunction

ACS[146], MI[144],

SCD[146], stroke[144]

CRP Thrombosis; LDL-

uptake; endothelial

dysfunction

ACS[150], CAD[120,

151, 322, 332, 416],

CHF[417-421], MI[45,

76, 151, 386, 422-424],

PAD[425-433],

SCD[151], stroke[151,

423, 434-438], UA[76,

439-441]

IFN-γ Antiviral;

immunoregulatory;

macrophage activation

CAD[120, 170],

CHF[171], MI[166, 168],

PAD[169]

IL-1 EC & SMC activation CAD [121], UA[175],

MI[176, 206]

IL-6 Induction of acute phase

proteins; plaque rupture;

SMC proliferation

ACS[195], CAD[120,

187], CHF[191], MI [76,

189, 190], UA[76, 188]

IL-8 Chemokine; deactivate

circulating leukocytes

MI[168, 189], UA [277,

278], CHF [279], CAD

[280-282], PAD[169]

MCP-1 Chemokine; leukocyte

recruitment

ACS[288-290],

CAD[154, 281, 282,

288], CVD[287], MI[206,

290], PAD[154, 169],

SCD[290], UA[291]

MIF Chemokine;

immunoregulatory;

macrophage activation

MI[302, 304], SCD[302,

304], UA[304].

TNF-α Leukocyte adhesion, new

vessel formation,

atheroma

CAD[121, 204, 205],

CHF[191], MI[168, 206],

PAD[169]

ACS, acute coronary syndromes; CAD, coronary artery disease; CHF, chronic heart failure; CRP, C-reactive

protein; CVD, cardiovascular disease; EC, endothelial cell; IFN, interferon; IL, interleukin; MCP; monocyte

chemoattractant protein; MI, myocardial infarction; MIF, migration inhibitory factor; PAD, peripheral arterial

disease; SCD, sudden cardiac death; SMC, smooth muscle cell; TNF, tumour necrosis factor-α; UA, unstable

angina.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 49

TABLE 3. Anti-inflammatory cytokines

Biomarker Role Associated with End

point

Adiponectin Anti-inflammatory; anti-

apoptotic; anti-

hypertrophic

CAD[218, 219],

CHF[221], MI[222]

IL-4 Inhibits Th1; proliferation

& differentiation of B

cells

MI[233], CAD [120]

IL-10 Inhibits Th1; proliferation

& differentiation of T

cells

ACS[242], MI[166,

168],UA [241],

TGF-β Immunosuppressive; pro-

fibrotic

SCD[77, 265], CAD

[253], CHF[191, 254],

MI [263, 264],

stroke[261, 262]

ACS, acute coronary syndromes; CAD, coronary artery disease; CHF, chronic heart failure; HT, hypertension; IFN,

interferon; IL, interleukin; MI, myocardial infarction; SCD, sudden cardiac death.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 50

TABLE 4. Oxidative stress biomarkers

Biomarker Role Associated with End

point

CoQ10 Antioxidant; oxidative

phosphorylation; protects

membrane phospholipids

CVD[375, 376],

CHF[375, 376]

GSH Antioxidant; immune

function

MI[383, 386], CHF[384]

Isoprostanes Oxidant CVD [310], MI [307,

308]

Lp-PLA2 LDL oxidation;

inflammation

ACS[442], CAD[152,

153, 322, 323]

Nitrotyrosines Oxidant; mediates MPO;

inflammation

PAD[331], CAD[332-

334], MI[335, 443]

CVD[325]

oxLDL Inflammation; monocytes

& lymphocyte

recruitment; endothelial

dysfunction

ACS [341-345], CAD

[343, 355-359], CHF

[351, 352], CVD [340], ,

MI[346-348], PAD

[350], stroke[340, 353,

354], UA [347, 349]

SOD Major antioxidant

defence against O2- NO

preservation

CAD[409, 410],

MI[412] UA[411],

SCD[413]

ACS, acute coronary syndromes; CAD, coronary artery disease; CHF, chronic heart failure; CoQ10, coenzyme Q10;

CVD, cardiovascular disease; GSH glutathione; Lp-PLA2, lipoprotein-associated phospholipase A2; MI, myocardial

infarction; O2- superoxide anions; oxLDL, oxidized LDL; NO, nitric oxide; PAD, peripheral arterial disease; SCD,

sudden cardiac death; SOD, superoxide dismutase; UA, unstable angina.

ACC

EPTE

D M

ANU

SCR

IPT

ACCEPTED MANUSCRIPTStoner et al Page 51

Highlights

1. Inflammatory/oxidative biomarkers can elucidate cardiovascular events/risk

2. A single biomarker cannot estimate absolute risk of future cardiovascular events

3. This review provides an overview of existing and emerging biomarkers

4. The functions of each biomarker are discussed, along with prognostic data

5. The complexity surrounding the interactions of potential biomarkers is presented


Recommended