eISSN: 1897-4252
ISSN: 1731-5530
Kardiochirurgia i Torakochirurgia Polska/Polish Journal of Thoracic and Cardiovascular Surgery
Current issue Archive Manuscripts accepted About the journal Supplements Editorial board Reviewers Abstracting and indexing Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
1/2006
vol. 3
 
Share:
Share:

Forum ekspertów
The role of C-reactive protein as a cardiovascular risk predictor

George Boguslawski
,
Carlos A. Labarrere

Kardiochir Torakochir Pol 2006; 3, 1: 16-28
Online publish date: 2006/05/19
Article file
Get citation
 
 
Introduction
The importance of acute-phase molecules as risk factors for human cardiovascular disease has been emphasized in a number of reports [1, 2]. The principal acute-phase protein, C-reactive protein (CRP), so named for its ability to precipitate the C-polysaccharide of Streptococcus pneumoniae, has been linked to atherosclerosis and its complications. CRP is highly stable, and its serum concentration, normally less than 1 mg/L, can increase 10000-fold during the acute phase response [3-5]. Over the past decade, multiple clinical studies have associated CRP with an increased risk of developing myocardial infarction, stroke, peripheral arterial disease, cardiovascular and sudden death [1, 6]. In addition, elevated levels of CRP are one of the strongest predictors of progressive vascular disease [7] and future cardiovascular events in apparently healthy men and women [6, 8, 9]. The relationship between CRP levels and the acute phase response to infection, inflammatory disease, surgery, trauma and cancer is well known [3]. However, its specific roles in the acute phase response remain unclear. In contrast to acute inflammation, CRP levels are minimally elevated (<10 mg/L) in patients at risk for atherosclerotic disease and remain elevated for many months to years. The biological effects of minimal elevations of CRP and the subsequent development of cardiovascular disease remains unexplained [1].
Biochemical and immunological characteristics of CRP
The possible involvement of CRP in atherosclerotic cardiovascular disease has attracted an increasing attention in recent years and gave the impetus to intense molecular and physiological investigations of the protein. CRP, a pentamer composed of five identical non-glycosylated 23 kDa subunits, is a member of a highly conserved family of proteins known as pentraxins. Along with more than 40 other molecules, the pentraxins are recognized as acute-phase response proteins, and are involved in innate immunity and protection against a variety of pathogens through opsonization and activation of the complement cascade. CRP is rapidly synthesized (mainly, but not exclusively, by hepatocytes) in response to an acute inflammatory stimulus, and is internalized through specific receptors on a variety of cells. The circulating protein is a pentamer, but may be dissociated into a (denatured) monomeric form that exhibits distinct immunological and biochemical properties. The pentameric CRP binds to phosphocholine on membranes of damaged and apoptotic cells, to C1q component of the complement cascade and to the immunoglobulin Fc gamma receptors I and II (CD64 and CD32, respectively). In contrast, the monomer displays unique antigenic epitopes and binds preferentially to Fc gamma receptor III (CD16). Recent results from our laboratory indicate that the denatured CRP – but not the pentamer – has a promiscuous ability to bind to IgG molecules regardless of their source and antigenic specificity. This property appears restricted to CRP because other pentraxins, (serum amyloid component P and long pentraxin PTX3) do not interact with antibodies other than those specifically directed against them (Fig. 1).
Clinical and epidemiological relevance of CRP blood levels: CRP as a risk marker
During the past decade, numerous primary prevention studies associated elevated CRP blood levels with increased numbers of cardiovascular events. However, traditional assays for CRP lacked the ability to accurately measure CRP levels that were below 8 mg/L, levels associated with vascular disease prediction. The newer high-sensitivity assays are able to determine CRP levels within the 0.3-10 mg/L ranges [10]. CRP levels of <1.0, 1.0 to 3.0, and >3.0 mg/L are associated with low-, moderate-, and high-risk for future cardiovascular events. Parenthetically, we suggest that the term „high sensitivity” be used in reference to the assay methodology, and not to give an impression that one is detecting a novel form of CRP. Nested case-control studies, as well as a significant number of large prospective studies and meta analyses, established the prognostic power of the high-sensitivity CRP measurement for prediction of myocardial infarction, stroke, peripheral arterial disease, and sudden cardiac death, even among individuals without a previous history of cardiovascular disease [11]. The relationship between levels of CRP and future vascular risk has been consistent in studies performed in both the United States and Europe. The predictive power of CRP measurements for cardiovascular events holds for men as well as women, middle age as well as older individuals, non-smokers and smokers, as well as diabetic and non-diabetic, hyperlipidemic and normolipidemic, and hypertensive and normotensive individuals. CRP levels also predict the risk of both recurrent ischemia and death among individuals with known atherosclerotic disease such as stable and unstable angina, patients undergoing percutaneous angioplasty, and those individuals presenting to the emergency room with acute coronary syndromes. CRP levels consistently predict the likelihood of new coronary events in patients with unstable angina and acute myocardial infarction [12-21]. However, in acute coronary syndromes, CRP predicts recurrent myocardial infarction independent of troponin levels (an indicator of myocardial cell damage), suggesting that CRP is not a marker associated with the presence of myocardial damage [19-21]. Elevated CRP levels also seem to predict the prognosis of recurrent events in patients with stroke [22, 23], and may be a marker for risk of restenosis following percutaneous coronary interventions [24, 25]; this issue remains controversial [26]. In many studies, CRP was a stronger predictor of cardiovascular events than LDL and HDL cholesterol. In addition, CRP levels contribute to the cardiovascular risk associated with other risk factors such as hypertension, diabetes mellitus, smoking, and obesity. The additive value of CRP to lipid screening in terms of coronary risk prediction has been demonstrated in several settings [11, 27-30].

Vascular inflammation: a role for CRP
Accumulating evidence suggests that atherosclerosis is an inflammatory vascular disease and that inflammation plays a central role in the development and progression of atherosclerosis and its complications [31, 32]. Among potential risk factors associated with inflammatory processes, the following are of particular note: CRP, serum amyloid A, selectins and adhesion molecules ICAM-1 and VCAM-1 (intercellular and vascular adhesion molecules, respectively). An intriguing question is whether some of these inflammatory molecules represent merely reporters of systemic inflammation or whether they actively contribute to the formation of atherosclerotic lesions and the associated complications. Atherosclerosis begins with the formation of fibrofatty and fibrous lesions and ends with the generation of more advanced lesions characterized by fibrous plaques (Fig. 2), as a response to insults to the endothelium and smooth muscle cells of the arterial wall. These insults include diet (generation of oxidized or modified LDL-cholesterol, high concentrations of omega-6 polyunsaturated fatty acids), mechanical shear, hypertension, infection (viruses, bacteria), homocysteine, hyperglycemia, obesity, immunologic damage, toxins (tobacco smoke), and other agents. Acute-phase proteins such as CRP can also directly contribute to the generation of a dysfunctional endothelium (Fig. 3) and the recruitment of macrophages and leukocytes into the arterial intima. This recruitment process occurs in a series of steps tightly regulated by cytokines and adhesion molecules [33] (Fig. 3). Significantly, CRP activates endothelium and induces adhesion molecule expression on human aortic endothelial cells to a level comparable to that induced by cytokines [34, 35]. Furthermore, CRP induces the expression of monocyte chemoattractant protein-1 [35, 36], and interleukin-8 [37] which facilitate monocyte chemotaxis and entry of monocytes into the sub-endothelial space and recruitment into the arterial intima. Interestingly, CRP-mediated monocyte chemotaxis is abolished by a specific monoclonal antibody to CRP [38]. Recent studies demonstrate that CRP can increase chemokine CC receptor 2 expression on monocytes [39], which – together with interleukin-8 – may facilitate adherence of the monocytes to the arterial endothelium and subsequent migration into the intima [37, 40]. Macrophage-derived foam cells in the intimal space are active participants in the development of the vascular lesions of atherosclerosis [31, 32, 41]. The development of macrophage foam cells containing large amounts of cholesterol esters is a hallmark of atherosclerosis and emphasizes the importance of low-density lipoprotein (LDL)-cholesterol in the development of these lesions. Macrophage foam cells sustain a proinflammatory milieu within the arterial intima through increased synthesis and secretion of cytokines and CRP (Fig. 3). Recruitment and transmigration of phagocytes, especially neutrophils, across the endothelium into inflammatory sites within advanced, complicated or unstable fibrous plaques could be facilitated by interleukin-8, a member of the CXC family of chemokines. Recent findings have demonstrated that CRP induces interleukin-8 expression at mRNA and protein levels through nuclear factor kB (NFkB) activation in human aortic and coronary artery endothelial cells [37]

CRP and atherosclerosis A. CRP and endothelial activation
Activation of arterial endothelium is associated with the development of the native form as well as the transplant-associated form of atherosclerosis [32, 41, 42]. An aspect of activation is an increase in ICAM-1 production, and CRP is capable of directly inducing expression of ICAM-1 on human aortic endothelial cells (Fig. 4) [34]. Importantly, induction of adhesion molecule expression is achieved with concentrations of CRP found in patients at risk of developing cardiovascular disease. A positive relationship between elevated CRP levels and ICAM-1 expression on endothelium and in blood was recently found in heart transplant patients who subsequently developed coronary artery disease [42]. Additionally, elevated plasma levels of ICAM-1 have been found in apparently healthy men who subsequently developed myocardial infarction, suggesting adhesion molecules play a significant role in the etiology of cardiovascular disease [43]. CRP can facilitate the release of cytokines such as interleukin 1β, IL-6 and tumor necrosis factor-a and increase the release of soluble IL-6 receptor by macrophages and foam cells in the neointima (Fig. 4) [35, 44, 45]. The production of IL-6 in arteries is likely due to the presence of macrophages within the vascular wall, since IL-6 is produced by venous but not arterial endothelium [46]. In addition, CRP can evoke the production of the potent endothelium-derived vasoactive factor, endothelin-1 [35]. Endothelin-1 and IL-6 are two mediators of CRP-induced expression of adhesion molecules, monocyte chemoattractant protein-1 secretion and LDL uptake by macrophages. The release of IL-6 and soluble IL-6 receptor by endothelial cells and inflammatory cells within the areas of atherogenesis can lead to the appearance of complexes of IL-6 and IL-6 receptors in circulation. These complexes bind to arterial endothelium through glycoprotein 130, which is constitutively expressed on endothelial cells (Fig. 4) [47], sustaining a proinflammatory milieu in these arteries. Such a milieu could favor further synthesis and release of CRP by human arterial endothelial cells. This possibility is strengthened by the recent demonstration of intracellular CRP mRNA and protein in, as well as increased CRP secretion by, human aortic endothelial cells following incubation with interleukins-1β and -6 [48]. Increased synthesis of both intracellular and secreted CRP was also achieved following incubation with macrophage conditioned media, and these effects were reversed by neutralizing antibodies directed against interleukins-1β and -6 [48]. These data support the idea that macrophages within the atherosclerotic plaques could facilitate cytokine-mediated CRP synthesis and release by arterial endothelial cells. Moreover, a proinflammatory microenvironment within the arterial intima can favor CRP production, since it has been demonstrated that interleukins-1 and -6 induce CRP mRNA and protein in human coronary artery smooth muscle cells [49]. CRP can activate endothelium through modulation of the endothelial nitric oxide synthase-nitric oxide pathway (Fig. 4). Elevated levels of CRP are associated with a significant decrease in nitric oxide production that is the consequence of CRP acting to lower the endothelial nitric oxide synthase mRNA stability and protein levels [50, 51]. Increased CRP levels also result in an increase in monocyte adhesion to the aortic endothelium. These responses are associated with an increased expression of endothelial ICAM-1 and VCAM-1. The effect of decreased nitric oxide production on endothelial adhesion molecule expression is partially mediated via activation of NFκβ (Fig. 4) [52]. Such activation represents one mechanism for CRP-associated proinflammatory activity within the arterial vasculature. This idea is strengthened by the recent demonstration that circulating monocytes from unstable angina patients with persistently elevated levels of CRP exhibited activation of NFκB [53]. In addition, CRP activated NFkβ in human saphenous vein endothelial cells [54] and in human aortic endothelial cells [37, 55]. CRP increased interleukin-8 mRNA and protein in human aortic endothelial cells, and NFκβ inhibitors diminished this upregulation. Since activation of NFκβ is associated with a concomitant increase of IL-6 levels, it is possible that proinflammatory effects of CRP are mediated through NFκβ [56]. On the basis of its documented effects in vitro, CRP may function as a procoagulant, being able to reduce the endothelial nitric oxide synthase and prostacyclin, and to increase plasminogen activator inhibitor-1 and, indirectly, tissue factor [57], a protein expressed on monocytes/macrophages and endothelial cells following activation [58]. Tissue factor represents a linkage between inflammation and coagulation. The presence of this molecule in atherosclerotic lesions contributes to thrombosis in coronary arteries following plaque rupture and it could explain the prothrombogenic status found in patients that develop the transplant-associated form of the disease [59]. The participation of CRP in atherothrombosis is further supported by the recent demonstration that CRP increases plasminogen activator inhibitor-1 expression and activity in human aortic endothelial cells [46]; moreover, it decreases tissue plasminogen activator antigen expression and activity in human aortic endothelial cells through generation of proinflammatory cytokines interleukin-1β and tumor necrosis factor-a [60]. Additionally, a proatherogenic and prothrombotic role for CRP is suggested by the experiments in which CRP decreased the release of the stable metabolite of prostacyclin, prostaglandin F-1α, in both human aortic and human coronary artery endothelial cells [55, 61]. The effects of CRP upon arterial endothelium seem to be mediated via Fcg receptors II and I, since it was recently demonstrated that the increase in interleukin-8, ICAM-1 and VCAM-1, as well as the decrease in endothelial nitric oxide synthase and prostacyclin induced by CRP, was abrogated with antibodies to CD32 and CD64 [62]. A direct role for CRP in the development of vascular disease is supported by recent studies performed in CRP transgenic mice [63-65]. Compared with normal mice, the CRP transgenic mice have a significantly increased and faster rate of arterial thrombosis following vascular injury [65]. In addition, CRP accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice [66]. CRP transgenic/apolipoprotein E-deficient mice demonstrated increased arterial deposition of CRP and complement C3, and enhanced expression of angiotensin type 1 receptor, VCAM-1, and collagen, an observation that supports a proatherogenic role for CRP in vivo.
B. CRP and LDL uptake, foam cell and plaque formation
Native LDL is not taken up by macrophages rapidly enough to generate foam cells, leading to the proposition that LDL needs to be modified in the vessel wall before being transported inside the macrophage (Fig. 4) [67, 68]. Foam cells in atherosclerotic lesions are generated through the activity of scavenger receptors, which allow macrophages to take up such oxidized or modified LDL particles. Because CRP influences the production of reactive oxygen species by macrophages and smooth muscle cells [69, 70] it is likely that CRP can also facilitate LDL oxidation in atherosclerotic lesions. CRP deposited within the arterial intima can bind oxidized LDL through recognition of phosphorylcholine residues on oxidized phospholipids [67, 71]. Chang et al. [72] demonstrated recently the binding of CRP to oxidized LDL via phosphorylcholine and the recognition of these complexes by scavenger receptors. CRP displays calcium-dependent binding to native LDL and to very low-density LDL [73], facilitating LDL uptake into macrophages and contributing to the formation of foam cells [74] (Fig. 4). The binding of LDL to extracellular matrix proteins in the arterial wall can expose phosphoryl choline residues on native LDL allowing the binding of CRP [72]. The fact that foam cells in early atherosclerotic lesions show the presence of CRP receptors as well as of CRP [38] is consistent with the hypothesis that CRP participates in foam cell formation through opsonization of lipid particles. The colocalization of CRP with enzymatically modified LDL was recently demonstrated in early atherosclerotic lesions [67]. In addition to the evidence that foam cell formation can occur through the uptake of modified LDL by scavenger receptors, the uptake could be further facilitated by the binding of CRP to such modified LDL and subsequent incorporation via macrophage-associated CRP receptors (Fig. 4). The major CRP receptor in human macrophages seems to be Fcg receptor II (CD 32). This receptor could play a significant role in the generation of foam cells [75]. Other CRP-specific receptors are thought to exist in monocytes/macrophages [76], but the relevance of these receptors for subsequent foam cell generation remains to be established. Complement can facilitate the opsonization of CRP-bound LDL by macrophages, and it has been demonstrated that the CD32 CRP receptor can cluster with other receptors such as complement receptors [75]. This is particularly relevant when one considers that CRP can directly activate complement [77]. The interactions between complement and CRP can be facilitated by the synthesis and release of CRP and complement components by macrophages and smooth muscle cells within the plaques (Fig. 4) [78, 79]. However, CRP also appears to upregulate complement-inhibitory proteins and protects endothelial cells from complement-mediated cell injury [80], suggesting that a balance of proatherogenic and antiatherogenic effects of CRP on the vessel wall may be important in the development of atherosclerosis. Elevated circulating levels of CRP can increase levels of CRP within vascular lesions by way of diffusion or receptor-mediated uptake by endothelial cells [46]. CRP within the plaques can be also increased by additional synthesis and release by intimal macrophages and smooth muscle cells [38, 75, 77, 78] (Fig. 4). CRP can directly contribute to plaque generation and progression through smooth muscle cell migration and proliferation via angiotensin 1 receptor upregulation [70]. CRP also increases smooth muscle cell basal reactive oxygen species production and potentiates the effects of angiotensin II on reactive oxygen species formation. CRP that accumulates in atherosclerotic plaques might enhance vascular oxidative stress via p22phox-based NADH/NADPH oxidase [81] CRP could also inhibit adrenomedullin in arterial endothelial and smooth muscle cells, thereby facilitating oxidative stress [82]. It has been recently demonstrated that CRP stimulates matrix metalloproteinase-1 expression by human macrophages through FcgRII and extracellular signal-regulated kinase pathway [83]. This activity of CRP may promote matrix degradation and thus contribute to plaque vulnerability. It has been proposed that there are distinct forms of CRP that are formed during inflammation. Conformationally rearranged CRP molecules (monomeric species) express epitopes not seen in native CRP, and these molecules are referred to as modified CRP (mCRP). Patients with more monomeric CRP might exhibit a greater proinflammatory phenotype and hence be at a greater risk for adverse cardiovascular events [84]. Indeed, the loss of pentameric symmetry in CRP, resulting in formation of mCRP, promotes a proinflammatory phenotype in human aortic endothelial cells characterized by the expression of ICAM-1, E-selectin and VCAM-1 protein and mRNA (Fig. 4). Incubation of human aortic endothelial cells with mCRP also results in concentration-dependent increases in interleukin-8 and monocyte chemoattractant protein-1 secretion and mRNA levels. These effects are mediated by a p38 MAP kinase-dependent mechanism (Fig. 4) [84]. The presence of cytokines such as interleukin-8 in the arterial intima could facilitate the recruitment of polymorphonuclear leukocytes, and these cells may play an important role in the pathogenesis of thrombosis and atherosclerosis by exerting concentration-dependent regulatory effects on the tissue factor production by mononuclear cells via the release of reactive oxygen species [85]. In contrast to native CRP that predominantly binds the low-affinity IgG FcγRIIa (CD32) receptor [86], mCRP utilizes the low-affinity immune-complex FcγRIII (CD16) on leukocytes [87]. This seems to apply also to arterial endothelial cells since an anti-FcγRIII (CD16) but not an anti- FcγRIIa (CD32) antibody significantly reduced the mCRP effects [84]. The role of mCRP in atherosclerosis is still controversial since calcium is always present in the lesions and mCRP has not yet been demonstrated in atherosclerotic plaques [88]. The role of CRP promoting a proinflammatory status leading to the development and progression of atherosclerosis is depicted in Fig. 5. Briefly, several inflammatory states generate cytokines that in turn induce synthesis and release of CRP by the liver. Elevated CRP can act as an injurious agent upon arterial endothelium, promoting generation and progression of atherosclerosis. Atherosclerotic lesions can be the source of further cytokine and CRP synthesis and release. In this „self-perpetuating” manner atherosclerosis-mediated ischemia and inflammation can contribute to the creation and maintenance of a proinflammatory and proatherosclerotic environment.
C. CRP and atherosclerosis: therapeutic approaches
Numerous clinical studies indicate that CRP levels are associated with the development and progression of atherosclerotic disease. These studies also indicate that CRP is a direct mediator of biochemical processes associated with atherosclerosis such as inflammation and activation of the coagulation cascade (Tab. I). In support of a role for CRP in inflammation and coagulation activation, infusion of recombinant human CRP into healthy volunteers significantly increased serum levels of interleukins-6 and -8, serum amyloid A, serum phospholipase A2, prothrombin 1 and 2, D-dimer and plasminogen activator inhibitor 1 [89]. Furthermore, although to-date there is no proof that reduction of CRP levels alone moderates the progression of atherosclerotic disease, treatments that reduce CRP levels have been associated with a diminished cardiovascular risk. Thus, CRP may be a target for the prevention and modulation of atherosclerotic lesions. Definitive proof of independent benefits of reducing CRP levels in humans awaits the development of specific inhibitors of CRP synthesis or action.
Dietary supplements
Diet-induced weight loss is a primary means of reducing CRP levels and the cardiovascular risk in patients who are overweight. It is probably not accidental that obesity is an independent risk factor for the development of cardiovascular disease and that adipocytes (fat cells) synthesize IL-6, the principal CRP activator. There is a positive association of weight and body mass index with CRP and IL-6 levels. Insulin resistance, which is common in obese patients, is also associated with elevated CRP levels. Diet-induced (ie. low calorie, low fat) or surgery-related (gastric surgery) weight loss decreases CRP and IL-6 levels [90-92]. Reduction of insulin resistance through weight loss or with insulin-sensitizing agents also decreases CRP levels. Some dietary supplements have known anti-inflammatory properties. Omega-3 long chain polyunsaturated fatty acids exhibit anti-inflammatory activities and have been associated with a decreased incidence of atherosclerotic cardiovascular disease and mortality following myocardial infarction. The effect of omega-3 fatty acids on CRP levels remains unclear [93] with some studies suggesting that they reduce CRP [94, 95] while other studies reporting no effects [96, 97]. Moderate alcohol intake is associated with decreased CRP levels in both cross-sectional population-based studies [98] and in a prospective randomized trial [99]. Multivitamins have been shown to reduce CRP levels [100]. Similarly, diets high in plant sterols, alpha-tocopherol and arginine have been reported to lower CRP levels [101]. Also effective are diets rich in soy protein, viscous fiber and almonds, and foods with low glycemic indexes.
Aspirin and statins
Aspirin use modifies the cardiovascular risk associated with elevated CRP levels in both primary prevention trials and unstable angina [6]. However, the direct effects of aspirin on CRP levels are still controversial. Some studies report a decrease in CRP levels with aspirin [102] while other studies report no effects [103]. No such uncertainty exists in the case of statins (3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors). These compounds reduce cardiovascular events and CRP levels, independent of their reduction in LDL levels [104-106]. For example, Albert et al. [105] studied 1702 individuals with no prior history of cardiovascular disease (primary prevention trial) and 1182 patients with cardiovascular disease (secondary prevention trial). Patients were randomized with respect to pravastatin or placebo for 24 weeks. In the primary prevention trial, median CRP levels were reduced by 17% at 24 weeks in the pravastatin group (no change in the placebo group). In the secondary prevention trial, median CRP levels were reduced 13% in the pravastatin group. The benefits of statins therapy are the greatest among the patients with high levels of CRP, regardless of the level of cholesterol or LDL. In a 5 year study of the use of statins for primary prevention of coronary events, lovastatin reduced CRP levels by 15% [28]. The drug was effective in preventing coronary events in patients with elevated CRP levels regardless of the ratio of cholesterol to high-density lipoprotein. In another study, the effects of simvastatin, pravastatin, and atorvastatin upon CRP levels were evaluated in a randomized double-blind crossover trial in patients with combined hyperlipidemia [106]. After 6 weeks of treatment, all three statins decreased CRP levels to a similar extent. Statins may also be useful for reducing the development of transplant-associated coronary artery disease and stroke, both diseases associated with elevated CRP levels. These beneficial actions of statins may be related in part to the cholesterol-independent effect of statins on inflammatory processes [107], perhaps mediated through a reduction in CRP levels. Similarly, for coronary artery disease patients undergoing intensive (as contrasted with moderate) statin therapy, the diminished rate of progression of atherosclerosis was significantly associated with reductions in the levels of both atherogenic lipoproteins and CRP [108].
Hypertension, hyperlipidemia and CRP
Studies on a large population of cardiovascular disease patients treated with either angiotensin-converting enzyme inhibitors or angiotensin type 1 receptor antagonists demonstrated some additive effects of these drugs on the cardiovascular endpoints [109-111]. These effects were unrelated to the degree of blood pressure lowering. The additivity of actions could be related partly to the blockade of the renin-angiotensin system. Interestingly, recent investigations have shown that the blockade of the renin-angiotensin system is associated with reduction of CRP levels in patients with coronary artery disease, acute myocardial infarction and stroke [112-114], suggesting a potential use for angiotensin-converting enzyme inhibitors or angiotensin type 1 receptor antagonists in atherosclerosis and its complications. This possibility is supported by the observation that the combination of renin-angiotensin system blockers and aspirin reduces nuclear factor kB activation and CRP expression within atherosclerotic plaques [115]. Both peroxisome proliferator-activated receptor (PPAR)-a and PPAR-g agonists (fibrates and thiazolidinediones, respectively) currently used for the management of hyperlipidemic disorders or diabetes, have been shown to possess anti-inflammatory and antiatherosclerotic properties [36, 116-118]. Treatment of non-diabetic hypertensive patients with rosiglitazone (PPAR-g agonist) was characterized by a significant decline in CRP levels, together with blood pressure reduction and improvement in insulin sensitivity [119]. Fibric acid derivatives reduced CRP levels in patients with combined hyperlipidemia [120]. Bezafibrate was also effective in reducing CRP levels in patients with severe hypertriglyceridemia and insulin resistance [121]. Furthermore, fenofibrate significantly lowered plasma concentrations of CRP in patients with coronary artery disease [122]. CRP levels can be reduced using heparin-induced extra-corporeal low-density lipoprotein precipitation (HELP)-apheresis [123]. HELP-apheresis has been effective in the treatment of the native as well as transplant-associated forms of atherosclerosis. The use of a combination of HELP-apheresis and statins significantly prevented the development of transplant associated coronary artery disease when compared to patients receiving only statin therapy [124]. Preliminary studies indicate that HELP-apheresis results in a rapid and long-term reduction of CRP levels in patients with familial hypercholesterolemia and advanced coronary artery disease [123].
D. Clinical usefulness of measuring CRP levels
Numerous reports to date support the clinical use of high-sensitivity CRP measurements as independent predictors of an increased cardiovascular risk such as ischemic coronary artery disease (angina, myocardial infarction, sudden death) and stroke. CRP levels are independently associated with risk in patients with known atherosclerotic disease, healthy individuals, and in patients with other risk factors. The levels of high-sensitivity CRP do not correlate with the degree of angiographically defined atherosclerotic disease [18] and therefore are of little value in the selection of patients for coronary artery or peripheral artery procedures. The predictive value of CRP for other cardiovascular events such as renal failure and occlusive peripheral vascular disease requires an additional study. By evaluating genetic variations in the CRP gene, at-risk genotypes may be identified, providing additional information for overall risk assessment [125]. Baseline levels of CRP show a clear heritability of approximately 40% in family studies [126]. To-date, three polymorphisms that are associated with changes in CRP levels have been identified in the CRP gene [127-129]. Genotype-specific risk categories may identify individuals who have relatively low serum CRP levels yet display an enhanced proinflammatory phenotype. Such identification could lead to more targeted therapies directed to reduce the proinflammatory load in the individuals at risk. There is no direct evidence to support independent treatment of CRP levels. However, measurement of high-sensitivity CRP levels is reasonable to consider in patients who develop cardiovascular complications in the absence of known cardiovascular risk factors. Although not proven, such patients may benefit from treatments that lower CRP levels. However, proof for such a therapeutic strategy requires a clinical study. An interesting but not yet tested use for high-sensitivity CRP measurement is as a means for motivating individuals with moderate to high cardiovascular risk to improve their lifestyles (i.e., smoking cessation, dietary modification, exercise, weight loss) or to comply with drug therapies. However, it is unclear whether CRP would add anything over that of monitoring other risk factors. Furthermore, the use of CRP as a predictor for cardiovascular disease and its complications still needs to be proved in prospective studies since a recent study showed CRP concentration was a relatively moderate predictor of risk of coronary artery disease and added only marginally to the predictive value of already established risk factors for coronary heart disease [130]. If utilized to predict the cardiovascular risk, patients should be free of known inflammatory processes and CRP levels should be measured at least twice over a period of several weeks [131, 132]. Repeated measurement provides an assurance that a subtle undiagnosed inflammatory state was not present. Levels can be measured in the fasting or non-fasting state, since they are minimally affected by food intake and demonstrate no circadian variation. Relative risk categories (low, moderate, high) based upon population studies correspond to the following CRP values: <1.0 mg/L, 1.0 to 3.0 mg/L, and >3.0 mg/L. The finding of a high relative risk level (CRP >3.0 mg/L) should lead to a complete reassessment of therapeutic strategies and drug therapies to maximally lower all cardiovascular risk factors [131-134]. Once the risk is established, there is little evidence upon which to base repeated assessment of CRP levels. Future trials are needed to determine the benefits of repeated testing of CRP levels upon cardiovascular events and other outcomes.
Acknowledgements
The authors are indebted to Christopher M Brown, medical illustrator at the Office of Visual Media, Indiana University School of Medicine, for the artwork. The help of Marcelo J Sosa in compiling the list of references is also gratefully acknowledged.
Supported by the Showalter Foundation, Indianapolis, IN
References
1. Kuller LH, Tracy RP, Shaten J, Meilahn EN. Relation of C-reactive protein and coronary heart disease in the MRFIT nested case-control study. Multiple Risk Factor Intervention Trial. Am J Epidemiol 1996; 144: 537-47. 2. Tracy RP, Lemaitre RN, Psaty BM, Ives DG, Evans RW, Cushman M, Meilahn EN, Kuller LH.. Relationship of C-reactive protein to risk of cardiovascular disease in the elderly: Results from the Cardiovascular Health Study and the Rural Health Promotion Project. Arterioscler Thomb Vasc Biol 1997; 17: 1121-7. 3. Pepys MB, Hirschfield GM. C-reactive protein: a critical update. [Erratum in: J Clin Invest. 2003; 112: 299]. J Clin Invest 2003; 111: 1805-12. 4. Labarrere CA, Zaloga GP. C-Reactive Protein: From innocent bystander to pivotal mediator of atherosclerosis. Am J Med 2004; 117: 499-507. 5. Black S, Kushner I, Samols D. C-reactive Protein. J Biol Chem 2004; 279: 48487-90. 6. Ridker PM, Cushman M, Stampfer MJ, Tracy RP, Hennekens CH. Inflammation, aspirin, and the risk of cardiovascular disease in apparently healthy men. N Engl J Med 1997; 336: 973-9. 7. Van Der Meer IM, De Maat MP, Hak AE, Kiliaan AJ, Del Sol AI, Van Der Kuip DA, Nijhuis RL, Hofman A, Witteman JC. C-reactive protein predicts progression of atherosclerosis measured at various sites in the arterial tree: The Rotterdam Study. Stroke 2002; 33: 2750-5. 8. Ridker PM, Buring JE, Shih J, Matias M, Hennekens CH. Prospective study of C-reactive protein and the risk of future cardiovascular events among apparently healthy women. Circulation 1998; 98: 731-3. 9. Koenig W, Sund M, Frohlich M, Fischer HG, Lowel H, Doring A, Hutchinson WL, Pepys MB. C-Reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: results from the MONICA (Monitoring Trends and Determinants in Cardiovascular Disease) Augsburg Cohort Study, 1984 to 1992. Circulation 1999; 99: 237-42. 10. Roberts WL, Moulton L, Law TC, Farrow G, Cooper-Anderson M, Savory J, Rifai N. Evaluation of nine automated high-sensitivity C-reactive protein methods: implications for clinical and epidemiological applications. Part 2.[erratum in Clin Chem 2001; 47:980]. Clin Chem 2001; 47: 418-25. 11. Ridker PM. High-sensitivity C-reactive protein: potential adjunct for global risk assessment in the primary prevention of cardiovascular disease. Circulation 2001; 103: 1813-8. 12. Haverkate F, Thompson SG, Pyke SDM, Gallimore JR, Pepys MB. Production of C-reactive protein and risk of coronary events in stable and unstable angina. Lancet 1997; 349: 462-6. 13. Lindahl B, Toss H, Siegbahn A, Venge P, Wallentin L. Markers of myocardial damage and inflammation in relation to long-term mortality in unstable coronary artery disease. N Engl J Med 2000; 343: 1139-47. 14. Ridker PM, Rifai N, Pfeffer MA, Sacks FM, Moye LA, Goldman S, Flaker GC, Braunwald E. Inflammation, pravastatin, and the risk of coronary events after myocardial infarction in patients with average cholesterol levels. Circulation 1998; 98: 839-44. 15. Milazzo D, Biasucci LM, Luciani N, Martinelli L, Canosa C, Schiavello R, Maseri A, Possati G. Elevated levels of C-reactive protein before coronary artery bypass grafting predict recurrence of ischemic events. Am J Cardiol 1999; 84: 459-61. 16. Ferreiros ER, Boissonnet CP, Pizarro R, Merletti PF, Corrado G, Cagide A, Bazzino OO. Independent prognostic value of elevated C-reactive protein in unstable angina. Circulation 1999; 100: 1958-63. 17. Bazzino O, Ferreiros ER, Pizarro R, Corrado G. C-reactive protein and the stress tests for the risk stratification of patients recovering from unstable angina pectoris. Am J Cardiol 2001; 87: 1235-39. 18. Zebrack JS, Muhlestein JB, Horne BD, Anderson JL. For the Intermountain Heart Collaboration Study, Group. C-reactive protein and angiographic coronary artery disease: independent and additive predictors of risk in subjects with angina. J Am Coll Cardiol 2002; 39: 632-7. 19. Morrow DA, Rifai N, Antman EM, Weiner DL, McCabe CH, Cannon CP, Braunwald E. C-reactive protein is a potent predictor of mortality independently of and in combination with troponin T in acute coronary syndromes: a TIMI 11A substudy. J Am Coll Cardiol 1998; 31: 1460-5. 20. Benamer H, Steg PG, Benessiano J, Vicaut E, Gaultier CJ, Boccara A, Aubry P, Nicaise P, Brochet E, Juliard JM, Himbert D, Assayag P. Comparison of the prognostic value of C-reactive protein and troponin I in patients with unstable angina pectoris. Am J Cardiol 1998; 82: 845-50. 21. Rebuzzi AG, Quaranta G, Liuzzo G, Caligiuri G, Lanza GA, Gallimore JR, Grillo RL, Cianflone D, Biasucci LM, Maseri A. Incremental prognostic value of serum levels of troponin T and C- reactive protein on admission in patients with unstable angina pectoris. Am J Cardiol 1998; 82: 715-9. 22. Di Napoli M, Papa F, Bocola V. C-reactive protein in ischemic stroke: an independent prognostic factor. Stroke 2001; 32: 917-24. 23. Di Napoli M, Papa F, Bocola V. Prognostic influence of increased C-reactive protein and fibrinogen levels in ischemic stroke. Stroke 2001; 32: 133-8. 24. Chew DP, Bhatt DL, Robbins MA, Penn MS, Schneider JP, Lauer MS, Topol EJ, Ellis SG. Incremental prognostic value of elevated baseline C-reactive protein among established markers of risk in percutaneous coronary intervention. Circulation 2001; 104: 992-7. 25. Walter DH, Fichtlscherer S, Britten MB, et al. Statin therapy, inflammation and recurrent coronary events in patients following coronary stent implantation. J Am Coll Cardiol 2001; 38: 2006-2012. 26. Zhou YF, Csako G, Grayston JT, Wang SP, Yu ZX, Shou M, Leon M, Epstein SE. Lack of association of restenosis following coronary angioplasty with elevated C-reactive protein levels or seropositivity to Chlamydia pneumoniae. Am J Cardiol 1999; 84: 595-8. 27. Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med 2000; 342: 836-43. 28. Ridker PM, Rifai N, Clearfield M, Downs JR, Weis SE, Miles JS, Gotto AM Jr; Air Force/Texas Coronary Atherosclerosis Prevention Study Investigators. Measurement of C-reactive protein for the targeting of statin therapy in the primary prevention of acute coronary events. N Engl J Med 2001; 344: 1959-65. 29. 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: 1557-65. 30. Ridker PM, Glynn RJ, Hennekens CH. C-reactive protein adds to the predictive value of total and HDL cholesterol in determining risk of first myocardial infarction. Circulation 1998; 97: 2007-11. 31. Ross R. Atherosclerosis – an inflammatory disease. N Engl J Med 1999; 340: 115-26. 32. Lusis AJ. Atherosclerosis. Nature 2000; 407: 233-41. 33. Pober JS, Cotran RS. Cytokines and endothelial cell biology. Physiol Rev 1990; 70: 427-51. 34. Pasceri V, Willerson JT, Yeh ETH. Direct proinflammatory effect of C-reactive protein on human endothelial cells. Circulation 2000; 102: 2165-8. 35. Verma S, Li SH, Badiwala MV, Weisel RD, Fedak PW, Li RK, Dhillon B, Mickle DA. Endothelin antagonism and interleukin-6 inhibition attenuate the proatherogenic effects of C-reactive protein. Circulation 2002; 105: 1890-6. 36. Pasceri V, Chang J, Willerson JT, Yeh ETH. Modulation of C-reactive protein – mediated monocyte chemattractant protein-1 induction in human endothelial cells by anti-athersclerosis drugs. Circulation 2001; 103: 2531-34. 37. Devaraj S, Kumaresan PR, Jialal I. Effect of C-reactive protein on chemokine expression in human aortic endothelial cells. J Mol Cell Cardiol 2004; 36: 405-410. 38. Torzewski M, Rist C, Mortensen RF, Zwaka TP, Bienek M, Waltenberger J, Koenig W, Schmitz G, Hombach V, Torzewski J. C-reactive protein in the arterial intima: Role of C-reactive protein receptor-dependent monocyte recruitment in atherogenesis. Arterioscler Thromb Vasc Biol 2000; 20: 2094-9. 39. Han KH, Hong KH, Park JH, Ko J, Kang DH, Choi KJ, Hong MK, Park SW, Park SJ. C-reactive protein promotes monocyte chemoattractant protein-1-mediated chemotaxis through upregulating CC chemokine receptor 2 expression in human monocytes. Circulation 2004; 109: 2566-71. 40. Gerszten RE, Garcia-Zepeda EA, Lim YC, Yoshida M, Ding HA, Gimbrone MA Jr, Luster AD, Luscinskas FW, Rosenzweig A. MCP-1 and IL-8 trigger firm adhesion of monocytes to vascular endothelium under flow conditions. Nature 1999; 398: 718-23. 41. Libby P. Inflammation in atherosclerosis. Nature 2002; 420: 868-74. 42. Labarrere CA, Lee JB, Nelson DR, Al-Hassani M, Miller SJ, Pitts DE. C-reactive protein, arterial endothelial activation and development of transplant coronary artery disease: A prospective study. Lancet 2002; 360: 1462-7. 43. 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: 88-92. 44. Ballou SP, Lozanski G. Induction of inflammatory cytokine release from cultured human monocytes by C-reactive protein. Cytokine 1992; 4: 361-8. 45. Jones SA, Novick D, Horiuchi S, Yamamoto N, Szalai AJ, Fuller GM. C-reactive protein: a physiological activator of interleukin 6 receptor shedding. J Exp Med 1999; 189: 599-604. 46. Devaraj S, Xu DY, Jialal I. C-reactive protein increases plasminogen activator inhibitor-1 expression and acivity in human aortic endothelial cells: Implications for the metabolic syndrome and atherothrombosis. Circulation 2003; 107: 398-404. 47. Mantovani A, Sozzani S, Introna M. Endothelial activation by cytokines. Ann N Y Acad Sci 1997; 832: 93-116. 48. Venugopal SK, Devaraj S, Jialal I. Macrophage conditioned medium induces the expression of C-reactive protein in human aortic endothelial cells. Potential for paracrine/autocrine effects. Am J Pathol 2005; 166: 1265-71. 49. Calabro P, Willerson JT, Yeh ET. Inflammatory cytokines stimulated C-reactive protein production by human coronary artery smooth muscle cells. Circulation 2003; 108: 1930-2. 50. Venugopal SK, Devaraj S, Yuhanna I, Shaul P, Jialal I. Demonstration that C-reactive protein decreases eNOS expression and bioactivity in human aortic endothelial cells. Circulation 2002; 106: 1439-41. 51. Verma S, Wang CH, Li SH, Dumont AS, Fedak PW, Badiwala MV, Dhillon B, Weisel RD, Li RK, Mickle DA, Stewart DJ. A self-fulfilling prophecy: C-reactive protein attenuates nitric oxide production and inhibits angiogenesis. Circulation 2002; 106: 913-9. 52. Iwata A, Sai S, Nitta Y, Chen M, de Fries-Hallstrand R, Dalesandro J, Thomas R, Allen MD. Liposome-mediated gene transfection of endothelial nitric oxide synthase reduces endothelial activation and leukocyte infiltration in transplanted hearts. Circulation 2001; 103: 2753-2759.
53. Liuzzo G, Santamaria M, Ginnetti F, et al. Persistent activation of the NF-kB signaling pathway in patients with unstable angina and elevated levels of C-reactive protein. J Am Coll Cardiol 2002; 39: 321A-2A. 54. Verma S, Badiwala MV, Weisel RD, Li SH, Wang CH, Fedak PW, Li RK, Mickle DA. C-reactive protein activates the nuclear factor-kB signal transduction pathway in saphenous vein endothelial cells: implications for atherosclerosis and restenosis. J Thorac Cardiovasc Surg 2003; 126: 1886-91. 55. Jialal I, Devaraj S, Venugopal SK. C-reactive protein: risk marker or mediator in atherothrombosis? Hypertension 2004; 44: 6-11. 56. Liuzzo G, Colussi C, Ginnetti F, et al. Direct activation of the transcription factor NF-kB by C-reactive protein in human monocytes: A Clue to the inflammatory component of acute coronary syndromes? Circulation 2001; 104: II-200. 57. Paffen E, Vos HL, Bertina RM. C-reactive protein does not directly induce tissue factor in human monocytes. Arterioscler Thromb Vasc Biol 2004; 24: 975-81. 58. Cermak J, Key NS, Bach RR, Balla J, Jacob HS, Vercellotti GM. C-reactive protein induces human peripheral blood monocytes to synthesize tissue factor. Blood 1993; 82: 513-20. 59. Labarrere CA, Nelson DR, Cox CJ, Pitts D, Kirlin P, Halbrook H. Cardiac-specific troponin I levels and risk of coronary artery disease and graft failure following heart transplantation. JAMA 2000; 284: 457-64. 60. Singh U, Devaraj S, Jialal I. C-reactive protein decreases tissue plasminogen activator activity in human aortic endothelial cells. Evidence that C-reactive protein is a procoagulant. Arterioscler Thromb Vasc Biol 2005; 25: 2216-21. 61. Venugopal SK, Devaraj S, Jialal I. C-reactive protein decreases prostacyclin release from human aortic endothelial cells. Circulation 2003; 108: 1676-8. 62. Devaraj S, Du Clos TW, Jialal I. Binding and internalization of C-reactive protein by Fcgamma receptors on human aortic endothelial cells mediates biological effects. Arterioscler Thromb Vasc Biol 2005; 26: 1-5. 63. Szalai AJ. The biological functions of C-reactive protein. Vasc Pharmacol 2002; 39: 105-7. 64. Szalai AJ, McCrory MA. Varied biologic functions of C-reactive protein: lessons learned from transgenic mice. Immunol Res 2002; 26: 279-87. 65. Danenberg HD, Szalai AJ, Swaminathan RV, Peng L, Chen Z, Seifert P, Fay WP, Simon DI, Edelman ER. Increased thrombosis after arterial injury in human C-reactive protein-transgenic mice. Circulation 2003; 108: 512-5. 66. Paul A, Ko KW, Li L, Yechoor V, McCrory MA, Szalai AJ, Chan L. C-reactive protein accelerates the progression of atherosclerosis in apolipoprotein E-deficient mice. Circulation. 2004; 109: 647-55. 67. Bhakdi S, Torzewski M, Klouche M, Hemmes M. Complement and Atherogenesis. Binding of CRP to degraded, nonoxidized LDL enhances complement activation. Arterioscler Thromb Vasc Biol 1999; 19: 2348-54. 68. Bhakdi S, Dorweiler B, Kirchmann R, Torzewski J, Weise E, Tranum-Jensen J, Walev I, Wieland E. On the pathogenesis of atherosclerosis: Enzymatic transformation of human low density lipoprotein to an atherogenic moiety. J Exp Med 1995; 182: 1959-71. 69. Tebo JM, Mortensen RF. Internalization and degradation of receptor bound C-reactive protein by U-937 cells: induction of H2O2 production and tumoricidal activity. Biochim Biophys Acta 1991; 1095: 210-6. 70. Wang CH, Li SH, Weisel RD, Fedak PW, Dumont AS, Szmitko P, Li RK, Mickle DA, Verma S. C-reactive protein upregulates angiotensin type 1 receptors in vascular smooth muscle. Circulation 2003; 107: 1783-90. 71. Pepys MB, Rowe IF, Baltz ML. C-reactive protein: binding to lipids and lipoproteins. Int Rev Exp Pathol 1985; 27: 83-111. 72. Chang MK, Binder CJ, Torzewski M, Witztum JL. C-reactive protein binds to both oxidized LDL and apoptotic cells through recognition of a common ligand: Phosphorylcholine of oxidized phospholipids. Proc Natl Acad Sci USA 2002; 99: 13043-8. 73. Rowe IF, Soutar AK, Trayner IM, Baltz ML, de Beer FC, Walker L, Bowyer D, Herbert J, Feinstein A, Pepys MB. Rabbit and rat C-reactive proteins bind apolipoprotein B-containing lipoproteins. J Exp Med 1984; 159: 604-16. 74. Fu T, Borensztajn J. Macrophage uptake of low-density lipoprotein bound to aggregated C-reactive protein: possible mechanism of foam-cell formation in atherosclerotic lesions. Biochem J 2002; 366: 195-201. 75. Zwaka TP, Hombach V, Torzewski J. C-reactive protein-mediated low density lipoprotein uptake by macrophages. Implications for Atherosclerosis. Circulation 2001; 103: 1194-7. 76. Tebo JM, Mortensen RF. Characterization and isolation of a C-reactive protein receptor from the human monocytic cell line U-937. J Immunol 1990; 144: 231-8. 77. Torzewski J, Torzewski M, Bowyer DE, Frohlich M, Koenig W, Waltenberger J, Fitzsimmons C, Hombach V. C-reactive protein frequently colocalizes with the terminal complement complex in the intima of early atherosclerotic lesions of human coronary arteries. Arterioscler Thromb Vasc Biol 1998; 18: 1386-92.
78. Yasojima K, Schwab C, McGeer EG, McGeer PL. Generation of C-reactive protein and complement components in atherosclerotic plaques. Am J Pathol 2001; 158: 1039-51. 79. Jabs WJ, Theissing E, Nitschke M, Bechtel JF, Duchrow M, Mohamed S, Jahrbeck B, Sievers HH, Steinhoff J, Bartels C. Local generation of C-reactive protein in diseased coronary artery venous bypass grafts and normal vascular tissue. Circulation 2003; 108: 1428-31. 80. Li SH, Szmitko PE, Weisel RD, Wang CH, Fedak PW, Li RK, Mickle DA, Verma S. C-reactive protein upregulates complement-inhibitory factors in endothelial cells. Circulation 2004; 109: 833-6. 81. Kobayashi S, Inoue N, Ohashi Y, Terashima M, Matsui K, Mori T, Fujita H, Awano K, Kobayashi K, Azumi H, Ejiri J, Hirata K, Kawashima S, Hayashi Y, Yokozaki H, Itoh H, Yokoyama M. Interaction of oxidative stress and inflammatory response in coronary plaque instability: Important role of C-reactive protein. Arterioscler Thromb Vasc Biol 2003; 23: 1398-404. 82. Nagoshi Y, Kuwasako K, Cao YN, Kitamura K, Eto T. Effects of C-reactive protein on atherogenic mediators and adrenomedullin in human coronary artery endothelial and smooth muscle cells. Biochem Biophys Res Commun 2004; 314: 1057-63. 83. Williams TN, Zhang CX, Game BA, He L, Huang Y. C-reactive protein stimulates MMP-1 expression in U937 histiocytes through FcgRII and extracellular signal-regulated kinase pathway: An implication of CRP involvement in plaque destabilization. Arterioscler Thromb Vasc Biol 2004; 24: 61-6. 84. Khreiss T, Jozsef L, Potempa LA, Filep JG. Conformational rearrangement in C-reactive protein is required for proinflammatory actions on human endothelial cells. Circulation 2004; 109: 2016-22. 85. Cadroy Y, Dupouy D, Boneu B, Plaisancie H. Polymorphonuclear leukocytes modulate tissue factor production by mononuclear cells: Role of reactive oxygen species. J Immunol 2000; 164: 3822-8. 86. Manolov DE, Rocker C, Hombach V, Nienhaus GU, Torzewski J. Ultrasensitive confocal fluorescence microscopy of C-reactive protein interacting with FcgRIIa. Arterioscler Thromb Vasc Biol 2004; 24: 2372-7. 87. Heuertz RM, Schneider GP, Potempa LA, Webster RO. Native and modified C-reactive protein bind different receptors on human neutrophils. Int J Biochem Cell Biol 2005; 37: 320-35. 88. Venugopal SK, Devaraj S, Jialal I. Effect of C-reactive protein on vascular cells: Evidence for a proinflammatory, proatherogenic role. Curr Opin Nephrol Hypertens 2005; 14: 33-7. 89. Bisoendial RJ, Kastelein JJ, Levels JH, Zwaginga JJ, van den Bogaard B, Reitsma PH, Meijers JC, Hartman D, Levi M, Stroes ES. Activation of inflammation and coagulation after infusion of C-reactive protein in humans. Circ Res 2005; 96: 714-716. 90. Tchernof A, Nolan A, Sites CK, Ades PA, Poehlman ET. Weight loss reduces C-reactive protein levels in obese postmenopausal women. Circulation 2002; 105: 564-9. 91. Esposito K, Pontillo A, Di Palo C, Giugliano G, Masella M, Marfella R, Giugliano D. Effect of weight loss and lifestyle changes on vascular inflammatory markers in obese women: A randomized trial. JAMA 2003; 289: 1799-804. 92. Dietrich M, Jialal I. The effect of weight loss on a stable biomarker of inflammation, C-reactive protein. Nutr Rev 2005; 63: 22-8. 93. Mori TA, Beilin LJ. Omega-3 fatty acids and inflammation. Curr Atheroscler Rep 2004; 6: 461-7. 94. Pischon T, Hankinson SE, Hotamisligil GS, Rifai N, Willett WC, Rimm EB. Habitual dietary intake of n-3 and n-6 fatty acids in relation to inflammatory markers among US men and women. Circulation 2003; 108: 155-60. 95. Rallidis LS, Paschos G, Liakos GK, Velissaridou AH, Anastasiadis G, Zampelas A. Dietary alpha-linolenic acid decreases C-reactive protein, serum amyloid A and interleukin-6 in dyslipidaemic patients. Atherosclerosis 2003; 167: 237-42. 96. Chan DC, Watts GF, Barrett PH, Beilin LJ, Mori TA. Effect of atorvastatin and fish oil on plasma high-sensitivity C-reactive protein concentrations in individuals with visceral obesity. Clin Chem 2002; 48: 877-83. 97. Madsen T, Christensen JH, Blom M, Schmidt EB. The effect of dietary n-3 fatty acids on serum concentrations of C-reactive protein: A dose-response study. Br J Nutr 2003; 89: 517-22. 98. Albert MA, Glynn RJ, Ridker PM. Alcohol consumption and plasma concentration of C-reactive protein. Circulation 2003; 107: 443-7. 99. Sierksma A, van der Gaag MS, Kluft C, Hendriks HF. Moderate alcohol consumption reduces plasma C-reactive protein and fibrinogen levels; a randomized, diet-controlled intervention study. Eur J Clin Nutr 2002; 56: 1130-6. 100. Church TS, Earnest CP, Wood KA, Kampert JB. Reduction of C-reactive protein levels through use of a multivitamin. Am J Med 2003; 115: 702-7.
101. Wells BJ, Mainous III AG, Everett CJ. Association between dietary arginine and C-reactive protein. Nutrition 2005; 21: 125-30. 102. Ikonomidis I, Andreotti F, Economou E, Stefanadis C, Toutouzas P, Nihoyannopoulos P. Increased proinflammatory cytokines in patients with chronic stable angina and their reduction by aspirin. Circulation 1999; 100: 793-8. 103. Feldman M, Jialal I, Devaraj S, Cryer B. Effects of low-dose aspirin on serum C-reactive protein and thromboxane B2 concentrations: A placebo-controlled study using a highly sensitive C-reactive protein assay. J Am Coll Cardiol 2001; 37: 2036-41. 104. Ridker PM, Rifai N, Pfeffer MA, Sacks F, Braunwald E, for The Cholesterol and Recurrent Events (CARE) Investigators. Long-term effects of pravastatin on plasma concentration of C-reactive protein. Circulation 1999; 100: 230-5. 105. Albert MA, Danielson E, Rifai N, Ridker PM, for The PRINCE Investigators. Effect of statin therapy on C-reactive protein levels. The Pravastatin Inflamation/CRP Evaluation (PRINCE): A randomized trial and cohort study. JAMA 2001; 286: 64-70. 106. Jialal I, Stein D, Balis D, Grundy SM, Adams-Huet B, Devaraj S. Effect of hydroxymethyl glutaryl coenzyme A reductase inhibitor therapy on high sensitive C-reactive protein levels. Circulation 2001; 103: 1933-5. 107. Ridker PM, Cannon CP, Morrow D, Rifai N, Rose LM, McCabe CH, Pfeffer MA, Braunwald E; Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 (PROVE IT-TIMI 22) Investigators. C-reactive protein levels and outcomes after statin therapy. N Engl J Med 2005; 352: 20-8. 108. Nissen SE, Tuzcu EM, Schoenhagen P, Crowe T, Sasiela WJ, Tsai J, Orazem J, Magorien RD, O’Shaughnessy C, Ganz P; Reversal of Atherosclerosis with Aggressive Lipid Lowering (REVERSAL) Investigators. Statin therapy, LDL cholesterol, C-reactive protein, and coronary artery disease. N Engl J Med 2005; 352: 29-38. 109. Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G; for The Heart Outcomes Prevention Evaluation Study Investigators. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. N Engl J Med 2000; 342: 145-53. 110. Dahlof B, Devereux RB, Kjeldsen SE, Julius S, Beevers G, de Faire U, Fyhrquist F, Ibsen H, Kristiansson K, Lederballe-Pedersen O, Lindholm LH, Nieminen MS, Omvik P, Oparil S, Wedel H; LIFE Study Group. Cardiovascular morbidity and mortality in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomised trial against atenolol. Lancet 2002; 359: 995-1003. 111. Fox KM; EURopean trial On reduction of cardiac events with Perindopril in stable coronary Artery disease Investigators. Efficacy of perindopril in reduction of cardiovascular events among patients with stable coronary artery disease: randomised, double-blind, placebo-controlled, multicentre trial (the EUROPA study). Lancet 2003; 362: 782-8. 112. Schieffer B, Bunte C, Witte J, Hoeper K, Boger RH, Schwedhelm E, Drexler H. Comparative effects of AT1-antagonism and angiotensin-converting enzyme inhibition on markers of inflammation and platelet aggregation in patients with coronary artery disease. J Am Coll Cardiol 2004; 44: 362-8. 113. Di Napoli M, Papa F. Angiotensin-converting enzyme inhibitor use is associated with reduced plasma concentration of C-reactive protein in patients with first-ever ischemic stroke. Stroke 2003; 34: 2922-9. 114. Tsikouris JP, Suarez JA, Simoni JS, Ziska M, Meyerrose GE. Exploring the effects of ACE inhibitor tissue penetration on vascular inflammation following acute myocardial infarction. Coron Artery Dis 2004; 15: 211-7. 115. Sattler KJ, Woodrum JE, Galili O, Olson M, Samee S, Meyer FB, Zhu XY, Lerman LO, Lerman A. Concurrent treatment with renin-angiotensin system blockers and acetylsalicylic acid reduces nuclear factor kB activation and C-reactive protein expression in human carotid artery plaques. Stroke 2005; 36: 14-20. 116. Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass CK. Peroxisome proliferator-activated receptor g ligands inhibit development of atherosclerosis in LDL receptor-deficient mice. J Clin Invest 2000; 106: 523-31. 117. Mohanty P, Aljada A, Ghanim H, Hofmeyer D, Tripathy D, Syed T, Al-Haddad W, Dhindsa S, Dandona P Evidence for a potent antiinflammatory effect of rosiglitazone. J Clin Endocrinol Metab 2004; 89: 2728-35. 118. Nesto R. C-reactive protein, its role in inflammation, type 2 diabetes and cardiovascular disease, and the effects of insulin-sensitizing treatment with thiazolidinediones. Diabet Med 2004; 21: 810-7. 119. Raji A, Seely EW, Bekins SA, Williams GH, Simonson DC. Rosiglitazone improves insulin sensitivity and lowers blood pressure in hypertensive patients. Diabetes Care 2003; 26: 172-8. 120. Gomez-Gerique JA, Ros E, Olivan J, Mostaza JM, Vilardell M, Pinto X, Civeira F, Hernandez A, da Silva PM, Rodriguez-Botaro A, Zambon D, Lima J, Diaz C, Aristegui R, Sol JM, Chaves J, Hernandez G; ATOMIX Investigators. Effect of atorvastatin and bezafibrate on plasma levels of C-reactive protein in combined (mixed) hyperlipidemia. Atherosclerosis 2002; 162: 245-51. 121. Jonkers IJ, Mohrschladt MF, Westendorp RG, van der Laarse A, Smelt AH. Severe hypertriglyceridemia with insulin resistance is associated with systemic inflammation: Reversal with bezafibrate therapy in a randomized controlled trial. Am J Med 2002; 112: 275-80. 122. Staels B, Koenig W, Habib A, Merval R, Lebret M, Torra IP, Delerive P, Fadel A, Chinetti G, Fruchart JC, Najib J, Maclouf J, Tedgui A. Activation of human aortic smooth-muscle cells is inhibited by PPARa but not by PPARg activators. Nature 1998; 393: 790-3. 123. Moriarty PM, Gibson CA, Shih J, Matias MS. C-reactive protein and other markers of inflammation among patients undergoing HELP LDL apheresis. Atherosclerosis 2001; 158: 495-8. 124. Jaeger BR, Meiser B, Nagel D, Uberfuhr P, Thiery J, Brandl U, Bruckner W, von Scheidt W, Kreuzer E, Steinbeck G, Reichart B, Seidel D. Aggressive lowering of fibrinogen and cholesterol in the prevention of graft vessel disease after heart transplantation. Circulation 1997; 96: II-15458. 125. Verma S, Szmitko PE, Yeh ET. C-reactive protein: structure affects function. Circulation 2004; 109: 1914-7. 126. Kluft C, de Maat MP. Genetics of C-reactive protein: New possibilities and complications. Arterioscler Thromb Vasc Biol 2003; 23: 1956-9. 127. Brull DJ, Serrano N, Zito F, Jones L, Montgomery HE, Rumley A, Sharma P, Lowe GD, World MJ, Humphries SE, Hingorani AD. Human CRP gene polymorphism influences CRP levels: Implications for the prediction and pathogenesis of coronary heart disease. Arterioscler Thromb Vasc Biol 2003; 23: 2063-9.
128. Suk HJ, Ridker PM, Cook NR, Zee RY. Relation of polymorphism within the C-reactive protein gene and plasma CRP levels. Atherosclerosis 2005; 178: 139-45. 129. Szalai AJ, Wu J, Lange EM, McCrory MA, Langefeld CD, Williams A, Zakharkin SO, George V, Allison DB, Cooper GS, Xie F, Fan Z, Edberg JC, Kimberly RP. Single-nucleotide polymorphisms in the C-reactive protein (CRP) gene promoter that affect transcription factor binding, alter transcriptional activity, and associate with differences in baseline serum CRP level. J Mol Med 2005; 83: 440-7. 130. Danesh J, Wheeler JG, Hirschfield GM, Eda S, Eiriksdottir G, Rumley A, Lowe GD, Pepys MB, Gudnason V. C-reactive protein and other circulating markers of inflammation in the prediction of coronary heart disease. N Engl J Med 2004; 350: 1387-97. 131. Pearson TA, Mensah GA, Alexander RW, Anderson JL, Cannon RO 3rd, Criqui M, Fadl YY, Fortmann SP, Hong Y, Myers GL, Rifai N, Smith SC Jr, Taubert K, Tracy RP, Vinicor F; Centers for Disease Control and Prevention; American Heart Association. Markers of inflammation and cardiovascular disease: Application to clinical and public health practice. A statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation 2003; 107: 499-511. 132. Ridker PM. Clinical application of C-reactive protein for cardiovascular disease detection and prevention. Circulation 2003; 107: 363-9. 133. Ridker PM. on behalf of the JUPITER Study Group. Rosuvastatin in the primary prevention of cardiovascular disease among patients with low levels of low-density lipoprotein cholesterol and elevated high-sensitivity C-reactive protein: Rationale and design of the JUPITER trial. Circulation 2003; 108: 2292-7. 134. Libby P, Ridker PM. Inflammation and atherosclerosis: Role of C-reactive protein in risk assessment. Am J Med 2004; 116:(6A): 9S-16S.
Copyright: © 2006 Polish Society of Cardiothoracic Surgeons (Polskie Towarzystwo KardioTorakochirurgów) and the editors of the Polish Journal of Cardio-Thoracic Surgery (Kardiochirurgia i Torakochirurgia Polska). This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.