Next Article in Journal
NOX Dependent ROS Generation and Cell Metabolism
Next Article in Special Issue
Pharmacological Utility of PPAR Modulation for Angiogenesis in Cardiovascular Disease
Previous Article in Journal
Kappa Opioid Receptors Reduce Serotonin Uptake and Escitalopram Efficacy in the Mouse Substantia Nigra Pars Reticulata
Previous Article in Special Issue
Modified mRNA Therapeutics for Heart Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Monomeric C-Reactive Protein in Atherosclerotic Cardiovascular Disease: Advances and Perspectives

1
National Medical Research Centre of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia
2
State Research Center of the Russian Federation, Institute of Biomedical Problems of Russian Academy of Sciences, 76A Khoroshevskoye Shosse, 123007 Moscow, Russia
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2079; https://doi.org/10.3390/ijms24032079
Submission received: 30 December 2022 / Revised: 16 January 2023 / Accepted: 16 January 2023 / Published: 20 January 2023
(This article belongs to the Special Issue Molecular Mechanisms of Cardiac Development and Disease)

Abstract

:
This review aimed to trace the inflammatory pathway from the NLRP3 inflammasome to monomeric C-reactive protein (mCRP) in atherosclerotic cardiovascular disease. CRP is the final product of the interleukin (IL)-1β/IL-6/CRP axis. Its monomeric form can be produced at sites of local inflammation through the dissociation of pentameric CRP and, to some extent, local synthesis. mCRP has a distinct proinflammatory profile. In vitro and animal-model studies have suggested a role for mCRP in: platelet activation, adhesion, and aggregation; endothelial activation; leukocyte recruitment and polarization; foam-cell formation; and neovascularization. mCRP has been shown to deposit in atherosclerotic plaques and damaged tissues. In recent years, the first published papers have reported the development and application of mCRP assays. Principally, these studies demonstrated the feasibility of measuring mCRP levels. With recent advances in detection techniques and the introduction of first assays, mCRP-level measurement should become more accessible and widely used. To date, anti-inflammatory therapy in atherosclerosis has targeted the NLRP3 inflammasome and upstream links of the IL-1β/IL-6/CRP axis. Large clinical trials have provided sufficient evidence to support this strategy. However, few compounds target CRP. Studies on these agents are limited to animal models or small clinical trials.

1. Introduction

Atherosclerosis and its complications, primarily coronary artery disease (CAD) and ischemic stroke, remain the leading causes of mortality and disability worldwide. The incidence of atherosclerotic cardiovascular disease and major adverse cardiovascular events (MACE) increases with age. The probability of the clinical manifestation of atherosclerosis and the development of MACE is determined by the total plaque burden [1]. The total plaque burden is characterized by the concentration and duration of exposure to circulating atherogenic apolipoprotein B (apoB)-containing lipoproteins [1]. As plaque burden increases, the probability of atherosclerotic cardiovascular disease onset increases [1,2]. Therefore, reducing low-density lipoprotein cholesterol (LDL-C) level, the main fraction of apoB-containing lipoproteins in the blood, is the mainstay of atherosclerosis prevention. The reduction in LDL-C levels is achieved through diet, lifestyle modification, and pharmacological treatment with statin monotherapy, statin in combination with ezetimibe or proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors [3].
Large randomized clinical trials demonstrated that aggressive lipid-lowering therapy with statins in combination with PCSK9 inhibitors produced a formidable reduction in LDL-C levels, approaching extremely low values in some patients. For example, a subgroup of 504 patients in the FOURIER trial achieved an LDL-C level of 0.18 mmol/L [4], whereas a subgroup of 3357 patients in the ODYSSEY OUTCOMES trial achieved an LDL-C level of less than 0.65 mmol/L [5]. Nevertheless, the MACE rate observed in these patients was substantial [6], which could be ascribed to the large total plaque burden that had already accumulated in these patients prior to treatment [1,2].
The cardiovascular risk that persists despite aggressive lipid-lowering therapy and correction of modifiable risk factors is called residual cardiovascular risk [7]. One of its main types is the residual inflammatory risk resulting from low-grade inflammation in atherosclerotic plaques [8]. It is determined by the level of the main inflammatory biomarker C-reactive protein (CRP), measured using a high-sensitivity assay (hsCRP), with a value of 2.0 mg/L or more [9]. The hsCRP assay measures the level of the pentameric form of CRP (pCRP), which is produced in the liver under the stimulation by interleukin (IL)-6 [10]. There exists another form of CRP, monomeric CRP (mCRP), which is formed at sites of local inflammation through the dissociation of pCRP and, to some extent, local synthesis. mCRP is essentially different from pCRP in its functions [10,11]. mCRP has long remained the subject of basic research; however, in recent years, reports measuring the levels of mCRP in the blood of patients and healthy individuals have been published.
This review aimed to trace the inflammatory pathway from the NLRP3 inflammasome to mCRP in atherosclerotic cardiovascular disease. The role of mCRP in other diseases, such as cancer, venous thromboembolism, and age-related and neurodegenerative diseases, has been discussed in various reports [12,13,14,15]. Hereafter, the abbreviations CRP, pCRP, hsCRP, and mCRP may be used. CRP is used when referring to studies that did not distinguish between forms of CRP. pCRP represents the pentameric liver-produced form of CRP. hsCRP is pCRP measured using a high-sensitivity assay. Finally, mCRP represents the monomeric dissociated form of CRP.

2. NLRP3 Inflammasome and Inflammatory IL-1β/IL-6/CRP Axis in Pathogenesis of Subclinical Vascular Inflammation

Cellular immune responses play a key role in all stages of atherosclerotic lesion development, from fatty streaks to plaque rupture [16]. A detailed discussion of cellular immunity in the pathogenesis of atherosclerosis has been provided elsewhere [17,18]. Four types of cells are involved in the pathogenesis of atherosclerotic lesions: smooth muscle cells (SMCs), T-lymphocytes, macrophages, and neutrophils. The cytoplasm of myeloid immune cells, including macrophages and neutrophils, and SMC contains pattern-recognition receptors (PRRs) [19,20]. These receptors recognize exogenous pathogen-associated molecular patterns (PAMPs) and endogenous damage-associated molecular patterns (DAMPs). DAMPs arise from signals produced by injured cells and tissues in the absence of an exogenous pathogen [20]. One type of PRR is the nucleotide-binding oligomerization domain-like receptor (NLR) [21]. The NLRP3 receptor in this group recognizes non-pathogenic DAMPs. NLRP3 activation leads to the formation of a molecular complex, the inflammasome, in the cytoplasm of myeloid immune cells. The NLRP3 inflammasome is a cytoplasmic protein complex that serves as a molecular platform for the activation of the cysteine protease caspase-1 [21]. Caspase-1 proteolyzes three proteins: pro-interleukin (pro-IL)-1β, pro-IL-18, and gasdermin D. Pro-IL-1β is converted into the active proinflammatory form of IL-1β and pro-IL-18 is converted into active proinflammatory IL-18. The activation of gasdermin D can induce pyroptosis, an inflammatory cell death, with the ensuing release of cytoplasmic content into surrounding tissues. This is particularly observed in the apoptosis of foam cells in atherosclerotic lesions [22,23].
The NLRP3 inflammasome formation has been observed in a number of diseases characterized by sterile inflammation. Uric-acid crystals can activate the NLRP3 inflammasome in gout [24]. Furthermore, the NLRP3 inflammasome plays an important role in the development of abdominal aortic aneurysm [25], myocardial damage in ischemia-reperfusion injury [26], kidney damage in diabetes, gout, and acute renal failure [27]. Active oxygen species and free fatty acids activate the NLRP3 inflammasome in obesity [28] and diabetes [29], contributing to the development of insulin resistance [30].
In 2010, Duewell et al. demonstrated for the first time the activation of the NLRP3 inflammasome by cholesterol crystals [31]. CD36-mediated uptake of oxidized LDL by macrophages results in intracellular cholesterol crystallization. This disrupts phagocytosis and leads to the accumulation of cholesterol crystals in macrophage lysosomes [32]. Subsequently, cholesterol crystals damage lysosome membranes and are released along with the lysosomal protease cathepsin B. They interact with PRR in the cytoplasm and induce the NLRP3 inflammasome formation [33]. Calcium-phosphate crystals accumulated in calcified atherosclerotic plaques can also activate the NLRP3 inflammasome [34]. Moreover, a recent study showed that CRP could activate the NLRP-3 inflammasome via the nuclear factor kappa B (NF-kB) pathway [35].
The NLRP3 inflammasome activation triggers a central cascade of inflammatory signaling represented by IL-1β, IL-6, and CRP [36]. IL-1β and IL-6 are mainly produced by myeloid cells [37,38], whereas CRP is by hepatocytes [10]. IL-1β stimulates the release of chemokines and the expression of cell-adhesion molecules by endotheliocytes, facilitating leukocyte recruitment to the site of inflammation. IL-1β stimulates the proliferation of SMC and the secretion of chemokines and collagenases by macrophages, thus contributing to the destabilization of atherosclerotic plaques [39].
IL-1β induces IL-6 synthesis. IL-6 has a wide range of proinflammatory and anti-inflammatory properties. For example, IL-6 stimulates the chemotaxis of neutrophils and macrophages to the site of inflammation [40]. It also stimulates chemokine release and expression of cell-adhesion molecules by endotheliocytes, facilitating leukocyte recruitment and platelet activation [41]. IL-6 produces a proatherogenic effect by stimulating SMC proliferation and modifying LDL uptake by macrophages and SMC [42]. Simultaneously, it stimulates an increase in LDL receptor expression, acting in an anti-atherogenic way [43]. IL-6 exhibits anti-inflammatory action by inhibiting IL-1 and tumor necrosis factor-α (TNF-α) synthesis and increasing IL-1 receptor antagonists and soluble p55 TNF-α receptor release [44]. Despite both proinflammatory and anti-inflammatory effects, higher levels of IL-1β and IL-6 are unequivocally associated with increased cardiovascular risk [45]. IL-6 stimulates CRP production in hepatocytes.

3. Assessment of Residual Inflammatory Risk

3.1. C-Reactive Protein as a Biomarker of Subclinical Vascular Inflammation

CRP is the final product of the central inflammatory cascade. Owing to its excellent reproducibility, it is considered the main biomarker of inflammation that reflects the activity of the IL-1β/IL-6/CRP axis [36]. The CRP level is consistent over time. It is not affected by hematocrit or other blood-protein levels [46]. Moreover, it is unaffected by the circadian rhythm, time of food intake, blood sampling, anticoagulants, delay in specimen processing up to 6 h, or storage conditions. Nor is it affected by the specimen type: CRP measurements gave similar results in fresh, thawed, and even repeatedly thawed and refrozen plasma and serum. CRP can be measured using standard laboratory methods such as enzyme-linked immunosorbent assay (ELISA), turbidimetry, or nephelometry without significant discrepancies in results [46]. Thus, CRP is a convenient laboratory biomarker widely used in clinical practice and basic research.
CRP levels rise manifold in response to infection or tissue damage: from 5–10 mg/L in mild cases to 320–550 mg/L in the most severe cases [47,48]. However, in atherosclerosis CRP levels are usually below 5 mg/L. A high-sensitivity assay with a threshold of 0.28 mg/L was developed to measure CRP below this level [49]. Large prospective observational studies have demonstrated that in surveyed populations, CRP levels were within tertiles of less than 1.0 mg/L, 1–3 mg/L, and more than 3.0 mg/L. In meta-analyses, the odds ratio for MACE between the lower and upper tertiles was between 1.58 and 2.0 [50,51]. The PROVE IT-TIMI 22 trial demonstrated that, in patients on aggressive statin therapy, the median CRP level was 2.0 mg/L. Patients with a CRP level of 2.0 mg/L or more had a 30% higher relative risk of MACE [52]. Similar CRP-level medians and cardiovascular risk ratios were observed in subsequent large clinical trials of statin therapy [53,54]. Currently, a CRP level 2.0 mg/L or more is suggested by the American College of Cardiology/American Heart Association guidelines on cardiovascular disease prevention as a cardiovascular risk factor [9].
The association between the CRP level and MACE rate has been examined in large observational studies in postmenopausal women [55], healthy volunteers in the Physicians’ Health Study [56], and MRFIT [57]. A meta-analysis of 52 prospective studies that included 246,669 individuals without cardiovascular disease showed that increased CRP levels worsened the 10-year prognosis of cardiovascular risk [58]. In addition, a meta-analysis of the East Asian population showed an association between elevated CRP and higher cardiovascular risk [59]. Furthermore, the USPSTF meta-analysis that explored studies published from 1966 to 2007 demonstrated that relative cardiovascular risk is 1.58-fold higher in individuals with a CRP level more than 3.0 mg/L than in those with a CRP level less than 1.0 mg/L [50].

3.2. Pentameric C-Reactive Protein

pCRP belongs to the pentraxin family of acute-phase proteins. It is the primary acute-phase reactant in humans. pCRP is synthesized in hepatocytes and secreted into the bloodstream upon stimulation with IL-6. Circulating pCRP consists of five monomeric subunits bound with disulfide bonds in a ring-shaped disk [60]. Each subunit of the pentameric disk has a calcium-dependent binding site for lysophosphatidylcholine on one side and the complement component C1q on the other [61,62].
Phosphatidylcholine is a major structural component of cell and extracellular vesicle membranes. It is mainly present on the outer leaflet of the membrane phospholipid bilayer [63]. Secretory phospholipase A2 (PLA2) hydrolyzes it to lysophosphatidylcholine. Normally, phosphatidylcholine does not interact with PLA2. However, during apoptosis and cell injury, phospholipids of the inner leaflet translocate to the outer leaflet of the cell membrane. These phospholipids include phosphatidylserine and phosphatidylethanolamine, which are PLA2 ligands. In the presence of these two phospholipids, PLA2 hydrolyzes phosphatidylcholine to biologically active lysophosphatidylcholine [63]. In oxidized LDL, lipoprotein-associated PLA2 cleaves phosphatidylcholine in the lipid monolayer to lysophosphatidylcholine [64].
Lysophosphatidylcholine stimulates the endothelial synthesis of several chemokines, impairs endothelium-dependent arterial relaxation, increases oxidative stress, suppresses endotheliocyte migration and proliferation, and facilitates macrophage activation and polarization to the inflammatory M1 phenotype [65]. Lysophosphatidylcholine of oxidized LDL contributes to lysosomal damage and the NLRP-3 inflammasome activation in foam cells [66]. Lysophosphatidylcholine can activate the NLRP-3 inflammasome in adipose tissue, contributing to the development of insulin resistance [67].
Lysophosphatidylcholine is a ligand for pCRP [68]. Circulating pCRP acts as an opsonin that binds to lysophosphatidylcholine on the surface of cell membranes and oxidized lipoproteins. The sites on the reverse side of the CRP disc interact with the complement component C1q. This results in activation of the classical complement cascade up to the C4 component. Thus, CRP-induced complement activation facilitates phagocytosis of damaged cells and oxidized lipoproteins but does not initiate the formation of the membrane-attack complex C5b–C9 [68]. pCRP can also interact with factor H and activate the complement cascade up to the C4 component via the alternative pathway [69]. Furthermore, pCRP can opsonize nuclear antigens released by apoptotic and necrotic cells [70]. Therefore, the biological role of circulating pCRP is characterized by the facilitation of the clearance of cell-destruction products formed during trauma, infection, or sterile inflammation.

3.3. Monomeric C-Reactive Protein

Upon binding to lysophosphatidylcholine, the pentameric disk of CRP undergoes dissociation through intermediate forms into the final product, mCRP [71,72]. Dissociation occurs through the disintegration of disulfide bonds between the pCRP subunits [73]. This process involves lysophosphatidylcholine but also requires other cell-membrane components and calcium. Soluble lysophosphatidylcholine does not dissociate pCRP in the absence of cell membranes [72]. Dissociation opens a neoepitope (octapeptide Phe-Thr-Lys-Pro-Gly-Leu-Trp-Pro) on the C-terminal end of monomeric subunits, which is concealed in the pentameric disk [72]. This dramatically changes the antigenic specificity and biological functions of CRP [74].
mCRP has reduced aqueous solubility and remains predominantly bound to the cell membranes [75]. It has been detected in extracellular vesicles circulating in the bloodstream. A pronounced increase in the number of mCRP-positive extracellular vesicles has been observed in patients with acute myocardial infarction [76] and peripheral artery disease [77]. mCRP in monocyte-derived exosomes has also been detected in patients with stable coronary artery disease [78].
mCRP may contribute to thromboinflammation. Thromboinflammation has recently been introduced to describe the complex interplay between blood coagulation and inflammation [79]. Immobilized on a collagen substrate, mCRP substantially increased platelet adhesion and thrombus growth rate at the shear rate of 1500 s−1, characteristic of arteries with mild stenosis [80]. Perfusion of mCRP-preincubated whole blood over a collagen type I-coated flow chamber yielded a similar result [80]. Unlike pCRP, mCRP induced platelet glycoprotein (GP) IIb/IIIa activation in a dose-dependent manner. mCRP facilitated platelet adhesion via activation of GP IIb/IIIa receptors. Moreover, pCRP dissociated into mCRP on activated adhered platelets during the perfusion of whole blood through a flow chamber [81]. Without dissociation, pCRP did not stimulate platelet adhesion or thrombus growth [80,81]. pCRP was attached to platelet membranes and dissociated into mCRP in another experiment with perfusion of whole blood over a surface coated with activated adhered platelets. GP IIb/IIIa inhibition with an antibody (abciximab) prevented pCRP dissociation [82]. mCRP stimulated platelet adhesion to the endothelial cells [83] and induced tissue-factor expression and fibrin formation on endothelial cells [84].
When dissociated on platelets and adhering to the vessel wall, mCRP can induce endothelial activation and leukocyte recruitment. mCRP enhanced endothelial activation and neutrophil attachment to the endothelium [83,85]; monocyte adhesion to the collagen [86], fibrinogen [87], and fibronectin matrix [88]; and T-lymphocyte extravasation [89]. In vitro, mCRP decreased nitric-oxide release and increased production of proinflammatory IL-8 and monocyte chemoattractant protein-1 by endothelial cells via the NF-kB pathway [90]. Moreover, mCRP stimulated leukocyte recruitment to the vessel wall, inducing the expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin, as well as the production of IL-6 and IL-8 by the endothelium [83,90,91]. mCRP induced IL-8 production [75,92] and prevented neutrophil apoptosis [75]. In addition, mCRP stimulated macrophage and T-cell polarization to inflammatory M1 and Th1 phenotypes [93]. mCRP stimulated oxidized LDL uptake by macrophages [94]. The in vivo evidence that mCRP can stimulate monocyte infiltration into damaged tissues was obtained from recent studies on a murine model of myocardial infarction [95] and a rat model of renal ischemia/reperfusion injury [96]. Compared with controls, the infarcted myocardium of mCRP-pretreated mice demonstrated increased accumulation of macrophages of the inflammatory M1 phenotype [95]. Furthermore, the damaged renal tissue of pCRP-pretreated rats demonstrated increased infiltration with monocytes colocalized with mCRP [96]. In addition, mCRP has been shown to stimulate neoangiogenesis and stabilize novel microvessels in vitro (bovine aortic endothelial cells and SMC) and in vivo (chorioallantoic membrane) [97,98]. In summary, the role of mCRP has been suggested in: platelet activation, adhesion, and aggregation; endothelial activation; leukocyte recruitment and polarization; foam-cell formation; and neovascularization (Figure 1).
The problem of mCRP deposition into atherosclerotic plaques has been addressed in several immunohistochemical studies. Herein, we discuss studies of human tissues. mCRP deposits have been detected in atherosclerotic plaques of the aorta [86], carotid [86,87,99], coronary [100,101], and femoral arteries [102], as well as diseased coronary artery venous bypass grafts [103]. Furthermore, mCRP deposits have been found in inflamed human striated muscles and infarcted myocardium [87]. CRP colocalized with leukocytes in thrombotic masses. CRP deposits in atheromatous tissues were larger in patients with increased CRP levels in blood plasma [100]. CRP accumulation in carotid atherosclerotic plaques was lower in patients treated with aspirin, angiotensin-converting enzyme inhibitors, or angiotensin-receptor blockers than in those who did not receive this therapy [104]. mCRP was deposited predominantly in the necrotic core and around clusters of macrophages, T-cells, and SMC, as well as neovessels in atherosclerotic plaques. Notably, CRP deposits were not found in intact arteries or fibrous or calcific plaques [86,87,99,100,102,103,104].
Some of the mentioned studies clearly distinguished between the two forms of CRP and confirmed that mCRP, but not pCRP, was deposited into damaged tissues [86,87,101], whereas other studies did not discriminate between CRP forms [99,100,102,103,104]. Nevertheless, it is unclear how mCRP accumulates in tissues. It can cross the endothelial barrier after dissociation [87] or be synthesized locally. The problem of local mCRP production has been explored in several studies. However, most of these studies did not discriminate between CRP forms. This creates difficulties in interpreting the locally synthesized form. An in vitro study demonstrated CRP production in an adipocyte culture stimulated by proinflammatory cytokines [105]. The expression of membrane-associated mCRP in human monocytes and CRP mRNA expression has been shown in another study [106]. Lipopolysaccharide-stimulated cultured macrophages synthesized mCRP and expressed CRP mRNA [107]. CRP mRNA expression in lipopolysaccharide-stimulated cultured macrophages was also observed in our study [78]. In an ex vivo study, monocytes collected from lipopolysaccharide-treated volunteers expressed CRP mRNA and produced CRP [108]. Moreover, CRP mRNA expression was detected in SMC and endotheliocytes from atheromatous tissues of coronary arteries [100], in atheromatous tissues of the femoral [102] and carotid [104] arteries, and diseased coronary artery venous bypass grafts [103]. In contrast, tissues of intact arteries did not express CRP mRNA [100,102,103,104]. Nonetheless, the contribution of local synthesis to the total concentration of mCRP in the tissues and bloodstream is unknown.

3.4. Level of Monomeric C-Reactive Protein in Health and Disease

To date, nine studies on the measurement of mCRP levels in human serum or plasma have been published (Table 1).
Wang et al. measured mCRP levels in 101 patients with acute myocardial infarction, 38 with unstable angina, 41 with stable angina, and 43 without coronary artery disease [109]. The mCRP level was 20.96 ± 1.64 μg/L in patients with myocardial infarction and 0.0 μg/L in other groups. No difference was found in mCRP levels between patients with ST-segment elevation and non-ST-segment-elevation myocardial infarction. mCRP level was substantially higher in patients deceased within 30 days from the onset of myocardial infarction than in survivors (36.70 ± 10.26 μg/L vs. 19.41 ± 1.43 μg/L) [109].
Zhang et al. measured mCRP levels in patients with autoimmune skin disorders (urticaria, eczema, and psoriasis; 20 patients in each group) and 20 healthy volunteers [110]. The mCRP level in patients with urticaria was 59.8 (44.5; 79.1) μg/L; psoriasis, 35.4 (17.0; 48.6) μg/L; and eczema, 30.0 (4.77; 36.3) μg/L, whereas in controls it was lower: 15.2 (5.05; 27.1) μg/L [110].
Williams et al. measured mCRP levels in 40 patients with markedly elevated CRP levels (more than 100 mg/L). The mean mCRP level in samples was 1.03 mg/L (±0.11 SE) [111].
Wu et al. measured mCRP levels in 37 patients with anti-neutrophil cytoplasmic antibody-associated vasculitis (AAV) and 20 control participants [112]. The mCRP level was 244.1 (226.1; 331.7) mg/L in patients with AAV and 170.0 (135.7; 199.3) mg/L in controls. The highest mCRP level was detected in four patients with AAV who had myocardial infarction: 581.4 (508.7; 647.3) mg/L and 240.8 (219.2; 292.1) mg/L in patients with and without myocardial infarction, respectively [112].
Munuswamy et al. measured mCRP levels in 38 patients with chronic obstructive pulmonary disease (COPD) and 18 non-COPD individuals. mCRP level was 0.66 (0.38; 1.03) mg/L in patients with COPD and 0.0 (0.0; 0.29) mg/L in non-COPD controls [113].
Liang et al. measured the mCRP levels in 206 patients with osteoarthritis and 60 healthy controls [114]. The mCRP level was 12.5 (7.8; 24.8) μg/L in patients with osteoarthritis and 5.0 (3.5; 9.8) μg/L in controls. Higher mCRP levels were associated with a more severe course of osteoarthritis [114].
Melnikov et al. reported a 7-year follow-up study of 80 patients with subclinical carotid atherosclerosis and an initially moderate cardiovascular risk [115]. The median mCRP level of the entire cohort was 5.2 (3.3; 7.1) μg/L. Patients with a median mCRP level or higher were more likely to have an increased number and total height of carotid atherosclerotic plaques at the end of the 7-year follow-up [115].
Karlsson et al. measured mCRP levels in 160 patients with systemic lupus erythematosus and 30 with AAV [116]. mCRP level was 3.7 (1.3; 7.4) μg/L in patients with systemic lupus erythematosus and 11 (5.8; 22) μg/L in those with AAV. No association was found between mCRP levels and any type of irreversible organ damage [116].
Fujita et al. measured the mCRP levels in 70 patients with autoimmune skin diseases, 50 with infectious diseases, and 30 healthy controls [117]. In the autoimmune skin-disease group, the mean mCRP levels were 477 (100; 2570) μg/L in patients with adult-onset Still’s disease, 186 (0.035; 626) μg/L in those with polymyalgia rheumatica, and 77 (0.035; 501) μg/L in those with rheumatoid arthritis. The mCRP level was 228 (0.035; 1086) μg/L in the infection group and 1.2 (0.035; 10) μg/L in the control group [117].
It is noteworthy that no correlation between mCRP and hsCRP level was found in five out of nine mentioned studies [111,113,115,116,117], whereas in the study by Wu et al., it was negative [112]. In contrast, Liang et al. reported a strong positive correlation between mCRP and hsCRP in patients with rheumatoid arthritis [114]. The studies by Zhang et al. and Wang et al. did not specifically examine this correlation [109,110].
None of these studies explored the correlation between mCRP levels and other proteins of the pentraxin family, in particular, pentraxin 3, which is considered a biomarker of cardiovascular disease [118]. It is synthesized locally by a range of stromal and myeloid cells in response to tissue damage and inflammation. It was suggested that pentraxin 3 and hsCRP could be measured complementary to assess local and systemic inflammatory response. Pentraxin 3 and hsCRP levels rarely correlate [118]. The association between pentraxin 3 and mCRP requires further research.
In summary, these studies demonstrated the feasibility of measuring mCRP levels. Most of these studies reported mCRP values in a comparable range. The nonconforming data on the correlation between mCRP and hsCRP levels may be at least partially explained by the different conditions in which they were analyzed. Some studies have reported an association between mCRP levels and disease severity and outcome. Nevertheless, large prospective studies are required to provide sufficient evidence to support this association or even establish mCRP levels as a diagnostic or prognostic tool.

4. Treatment to Reduce CRP Level and Ameliorate Residual Inflammatory Risk

4.1. Lifestyle Modifications

Lifestyle interventions can decrease subclinical vascular inflammation. According to the 10-year UCC-SMART study, smoking cessation, body-weight reduction, and regular physical activity decreased CRP levels [119]. Regular aerobic exercise decreased CRP levels independently of body-weight reduction or statin therapy [120]. Moreover, a meta-analysis of 83 studies including 3769 patients demonstrated the effect of regular physical activity on CRP levels [121]. A meta-analysis of 76 studies involving 6742 patients with obesity confirmed the association between body-weight reduction and a decrease in proinflammatory cytokines and CRP levels [122]. Furthermore, the consumption of foods rich in wholegrain cereals, dietary fiber, omega-3 fatty acids, and vitamins E and C reduced CRP levels [120].

4.2. Statins

Randomized clinical trials have demonstrated that statins influence subclinical vascular inflammation. The PROVE IT-TIMI 22 and IMPROVE-IT trials showed that a CRP-level reduction to less than 2.0 mg/L decreased MACE rate by 28–33%, which was virtually identical to patients with a LDL-C-level reduction of less than 1.8 mmol/L [52,123]. In the REVERSAL trial, the slowest progression of atheroma occurred in patients with LDL-C- and CRP-level reductions above the median values [124]. The JUPITER trial demonstrated the efficacy of statin therapy in the primary prevention of atherosclerosis in individuals with an initial CRP level of 2.0 mg/L or more and an LDL-C level less than 3.4 mmol/L, resulting in a 44% reduction in the relative risk of MACE [125]. In the FOURIER trial, the MACE rate was associated with a CRP level independent of LDL-C. In the group that achieved an LDL-C level of less than 0.52 mmol/L on high-dose statin plus evolocumab, MACE occurred in 9.0% of patients with a CRP level less than 1.0 mg/L vs. 13.1% of patients with a CRP level more than 3 mg/L [126]. The rate of MACE events per 100 human-years in the SPIRE-1/SPIRE-2 trials was 1.96 in patients with a CRP level less than 1.0 mg/L vs. 3.59 in patients with a CRP level more than 3.0 mg/L [127]. Two retrospective studies of patients undergoing percutaneous coronary intervention also demonstrated that MACE occurred less frequently in patients with initially low CRP levels or those who responded to statin therapy with a reduction in CRP level below 2.0 mg/L [128,129].
Although statins ameliorate subclinical vascular inflammation, their effect is sufficient only in approximately half of intensively treated patients. Despite aggressive lipid-lowering with statins, CRP levels remained above 2.0 mg/L in 43% of patients in PROVE IT-TIMI 22, 47% in IMPROVE-IT, 47% in SPIRE-1/SPIRE-2 [130], 43.6% in ODYSSEY OUTCOMES [131], and 45.6% and 48% in percutaneous coronary intervention trials [128,129]. The prevalence of residual inflammatory risk is higher in the general population [132].

4.3. Other Lipid-Lowering Agents

The IMPROVE-IT trial showed that the combination of statins with ezetimibe more often led to the achievement of both LDL-C and CRP target levels, which was associated with the lowest MACE rate. However, this effect was ascribed to the achievement of combined target levels, not direct ezetimibe action [123]. Ezetimibe monotherapy had no effect on CRP levels [133,134,135]. The addition of ezetimibe to statin therapy did not result in an additional reduction in CRP levels [136]. PCSK9 blockers also do not affect inflammatory activity or alter CRP levels [137,138].

4.4. Anti-Inflammatory Agents

4.4.1. Upstream Agents

The data from randomized clinical trials of anti-inflammatory agents support the hypothesis that MACE reduction can be achieved by directly targeting the NLRP3 inflammasome and IL-1β/IL-6/CRP axis (Figure 2). In contrast, agents that did not affect this inflammatory cascade failed to alter the MACE rate.
The CIRT trial studied the efficacy of low-dose methotrexate in the secondary prevention of atherosclerosis in patients with type 2 diabetes mellitus or metabolic syndrome [139]. The initial CRP level in the patients was 1.5 mg/L. During the 2.3-year follow-up, methotrexate neither reduced IL-1β, IL-6, or CRP levels nor altered the MACE rate. A possible explanation for the ineffectiveness of methotrexate is that a low baseline CRP level indicated an initially low residual inflammatory risk in patients. Moreover, the mechanism of the anti-inflammatory action of methotrexate does not affect the NLRP3 inflammasome and IL-1β/IL-6/CRP axis [36]. Two other anti-inflammatory agents that do not affect the NLRP3 inflammasome and the IL-1β/IL-6/CRP axis also failed to reduce MACE in patients with acute myocardial infarction [36]. These agents are losmapimod, a p38 MAPK inflammatory pathway inhibitor studied in the LATITUDE-TIMI 60 trial [140] and darapladib, a lipoprotein-associated PLA2 inhibitor studied in the SOLID-TIMI 52 trial [141].
Conversely, colchicine, which inhibits the NLRP3 inflammasome, was effective in reducing MACE. This anti-inflammatory agent is routinely used to suppress crystal-mediated inflammation in gout and Mediterranean fever [142]. The COLCOT trial showed that low-dose colchicine treatment resulted in a 23% reduction in the relative risk of MACE in patients with acute myocardial infarction [143]. Furthermore, the LoDoCo2 trial demonstrated that low-dose colchicine treatment reduced the relative risk of MACE by 31% in patients with stable coronary artery disease [144]. Nevertheless, the COPS trial failed to demonstrate a reduction in MACE in patients with acute myocardial infarction following low-dose colchicine treatment [145]. Colchicine ineffectiveness in the COPS trial could be related to the clinical characteristics of the study group, drug dosage, and time of therapy initiation [145].
The CANTOS trial examined the efficacy of the IL-1β antagonist canakinumab for the secondary prevention of atherosclerosis in patients with CRP levels of 2 mg/L or more [146]. Canakinumab reduced CRP and IL-6 levels by 36–40% without altering the lipid profile. The canakinumab group demonstrated a 14.2% reduction in the relative risk of MACE [146]. The subgroup analysis showed a 25% MACE reduction in patients who achieved a CRP level of less than 2.0 mg/L and no MACE reduction in non-responders to canakinumab with a CRP level of 2.0 mg/L or more compared with that in the placebo group [147]. A decrease in IL-6 level below the median of 1.65 ng/L on canakinumab treatment was associated with a 32% reduction in the relative risk of MACE [148].
The RESCUE trial studied the efficacy and safety of ziltivekimab, a monoclonal antibody against IL-6, in patients with an hsCRP level of 2.0 mg/L or more and moderate to severe chronic kidney disease [149]. Twelve weeks of treatment resulted in a dose-dependent 77–92% reduction in the median CRP level. Ziltivekimab also produced a dose-dependent reduction in the levels of fibrinogen, serum amyloid A, haptoglobin, secretory phospholipase A2, and lipoprotein(a) [149]. In late 2021, the ZEUS trial was initiated to test whether ziltivekimab treatment of patients with cardiovascular disease, chronic kidney disease, and elevated CRP levels would result in a reduction in the MACE rate [150].

4.4.2. Anti-CRP Agents

Attempts to ameliorate inflammation by directly influencing CRP (Table 2) aim at one of three targets: translation of CRP, dissociation of pCRP into mCRP, and mCRP.
One approach is to use antisense oligonucleotides that promote degradation of CRP mRNA and prevent CRP translation. Treatment with an antisense oligonucleotide ISIS 280290 reduced CRP levels in a rabbit model of atherosclerosis but did not result in an antiatherogenic effect [151]. In another study, treatment with an antisense oligonucleotide ISIS 329993 produced a pronounced decrease in CRP levels and ameliorated collagen-induced arthritis symptoms in CRP transgenic mice. In addition, it substantially reduced CRP levels and was well tolerated in healthy volunteers with initial CRP levels between 2 and 10 mg/L [152]. Additionally, ISIS 329993 administration markedly reduced CRP levels in endotoxin-pretreated healthy volunteers [153]. Nevertheless, it did not reduce atrial fibrillation burden in patients with paroxysmal atrial fibrillation in a phase II trial [154]. Furthermore, its effectiveness was unclear in a phase II trial of patients with rheumatoid arthritis [155].
Another approach is to target pCRP-mCRP dissociation. 1,6-bis (phosphocholine)-hexane (1,6-bisPC) inhibits pCRP to mCRP dissociation by cross-linking two pCRP disks in a “decameric” form that conceals phosphatidylcholine-binding sites. In a rat model of ischemia/reperfusion injury, this agent abrogated mCRP deposition and reduced leukocyte infiltration into the damaged myocardium [87]. In another study, 1,6-bisPC administration decreased infarct size and cardiac dysfunction induced by the injection of human CRP into rats with induced acute myocardial infarction [156]. Furthermore, 1,6-bisPC administration decreased the inflammatory response in damaged renal tissue and improved blood-urea nitrogen levels in a rat model of renal ischemia/reperfusion injury [96]. However, the suboptimal pharmacokinetic profile of 1,6-bisPC hinders its further development as an anti-inflammatory agent [87,156]. A recent study introduced a novel phosphatidylcholine-mimetic compound, C10M, that binds to phosphatidylcholine-specific sites on the pCRP disk [157]. This prevents pCRP from interacting with phosphatidylcholine and disrupts dissociation. In vitro, the C10M compound decreased pCRP binding to activated platelets, mCRP-induced endotheliocyte activation, and monocyte adhesion. In a rat model of CRP-induced aggravation of renal ischemia/reperfusion injury, C10M abrogated mCRP deposition in the damaged tissues and improved excretory renal function. Furthermore, this novel compound inhibited CRP-mediated allograft rejection in a rat model of hindlimb transplantation [157]. The current form of this compound may need further development, considering its relatively low bioavailability and short half-life [158].
The third approach is to directly target mCRP. A monoclonal antibody against mCRP attenuated rheumatoid arthritis symptoms, joint inflammation, pannus formation, and bone destruction in a murine arthritis model [159]. The same antibody also ameliorated glomerular damage and the progression of proteinuria in a murine lupus nephritis model [159].
To date, anti-inflammatory therapy has targeted the NLRP3 inflammasome and upstream links of the IL-1β/IL-6/CRP axis. Large clinical trials have provided sufficient evidence to support this strategy. Few known compounds target CRP. Studies on these agents are limited to basic research and animal models. Antisense oligonucleotides that target CRP mRNA were studied in small clinical trials; however, they did not demonstrate a clinical benefit. It is not clear whether targeting CRP might provide an advantage over upstream agents. On the one hand, upstream targeting reduces CRP level as well, and this is explicitly demonstrated in clinical trials. In contrast, mCRP production at sites of local inflammation (either by local synthesis or dissociation of circulating pCRP) may be independent of the overall CRP production in the liver. In this case, targeting mCRP or pCRP-mCRP dissociation might prove effective. However, to date, no trials have explored this strategy.

5. Conclusions

pCRP is the main biomarker of inflammation that reflects the activity of the IL-1β/IL-6/CRP axis. mCRP is a form of CRP with a distinct proinflammatory profile. mCRP has long remained the subject of basic research; however, recent studies have introduced mCRP assays and demonstrated the feasibility of mCRP-level measurement. Notably, most studies have reported mCRP values in a comparable range. Some studies have reported an association between mCRP levels and disease severity and outcome. Nevertheless, large prospective studies are required to provide sufficient evidence to support this association or even establish mCRP levels as a diagnostic or prognostic tool. Regarding anti-inflammatory therapy, there are few known compounds that target CRP. Studies on these agents are limited to animal models or small clinical trials.

Author Contributions

Conceptualization, I.M., K.G. and Z.G.; methodology, I.M., S.K., O.S. and Z.G.; resources, S.K. and Z.G.; data curation, I.M., Y.A. and K.G.; writing—original draft preparation, I.M., O.S. and Y.A.; writing—review and editing, K.G., S.K. and Z.G.; visualization, O.S. and Y.A.; supervision, S.K. and Z.G.; project administration, I.M. and S.K.; funding acquisition, S.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Russian Science Foundation project #22-25-00054.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ference, B.A.; Graham, I.; Tokgozoglu, L.; Catapano, A.L. Impact of Lipids on Cardiovascular Health. J. Am. Coll. Cardiol. 2018, 72, 1141–1156. [Google Scholar] [CrossRef] [PubMed]
  2. Robinson, J.G.; Williams, K.J.; Gidding, S.; Borén, J.; Tabas, I.; Fisher, E.A.; Packard, C.; Pencina, M.; Fayad, Z.A.; Mani, V.; et al. Eradicating the Burden of Atherosclerotic Cardiovascular Disease by Lowering Apolipoprotein B Lipoproteins Earlier in Life. JAHA 2018, 7, e009778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Giugliano, R.P.; Pedersen, T.R.; Park, J.-G.; De Ferrari, G.M.; Gaciong, Z.A.; Ceska, R.; Toth, K.; Gouni-Berthold, I.; Lopez-Miranda, J.; Schiele, F.; et al. Clinical efficacy and safety of achieving very low LDL-cholesterol concentrations with the PCSK9 inhibitor evolocumab: A prespecified secondary analysis of the FOURIER trial. Lancet 2017, 390, 1962–1971. [Google Scholar] [CrossRef]
  5. Schwartz, G.G.; Gabriel Steg, P.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Goodman, S.G.; Jukema, J.W.; Kim, Y.-U.; Li, Q.H.; Manvelian, G.; et al. Clinical Efficacy and Safety of Alirocumab After Acute Coronary Syndrome According to Achieved Level of Low-Density Lipoprotein Cholesterol: A Propensity Score–Matched Analysis of the ODYSSEY OUTCOMES Trial. Circulation 2021, 143, 1109–1122. [Google Scholar] [CrossRef]
  6. Dimmitt, S.B.; Stampfer, H.G.; Martin, J.H.; Warren, J.B. Clinical benefits of evolocumab appear less than hoped. Lancet 2018, 391, 933–934. [Google Scholar] [CrossRef] [Green Version]
  7. Lawler, P.R.; Bhatt, D.L.; Godoy, L.C.; Lüscher, T.F.; Bonow, R.O.; Verma, S.; Ridker, P.M. Targeting cardiovascular inflammation: Next steps in clinical translation. Eur. Heart J. 2020, 42, ehaa099. [Google Scholar] [CrossRef]
  8. Ridker, P.M. Residual inflammatory risk: Addressing the obverse side of the atherosclerosis prevention coin. Eur. Heart J. 2016, 37, 1720–1722. [Google Scholar] [CrossRef]
  9. Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 140, e596–e646. [Google Scholar] [CrossRef]
  10. McFadyen, J.D.; Kiefer, J.; Braig, D.; Loseff-Silver, J.; Potempa, L.A.; Eisenhardt, S.U.; Peter, K. Dissociation of C-Reactive Protein Localizes and Amplifies Inflammation: Evidence for a Direct Biological Role of C-Reactive Protein and Its Conformational Changes. Front. Immunol. 2018, 9, 1351. [Google Scholar] [CrossRef]
  11. Rajab, I.M.; Hart, P.C.; Potempa, L.A. How C-Reactive Protein Structural Isoforms With Distinctive Bioactivities Affect Disease Progression. Front. Immunol. 2020, 11, 2126. [Google Scholar] [CrossRef] [PubMed]
  12. Dix, C.; Zeller, J.; Stevens, H.; Eisenhardt, S.U.; Shing, K.S.C.T.; Nero, T.L.; Morton, C.J.; Parker, M.W.; Peter, K.; McFadyen, J.D. C-reactive protein, immunothrombosis and venous thromboembolism. Front. Immunol. 2022, 13, 1002652. [Google Scholar] [CrossRef]
  13. Potempa, L.A.; Rajab, I.M.; Olson, M.E.; Hart, P.C. C-Reactive Protein and Cancer: Interpreting the Differential Bioactivities of Its Pentameric and Monomeric, Modified Isoforms. Front. Immunol. 2021, 12, 744129. [Google Scholar] [CrossRef] [PubMed]
  14. McFadyen, J.D.; Zeller, J.; Potempa, L.A.; Pietersz, G.A.; Eisenhardt, S.U.; Peter, K. C-Reactive Protein and Its Structural Isoforms: An Evolutionary Conserved Marker and Central Player in Inflammatory Diseases and Beyond. In Vertebrate and Invertebrate Respiratory Proteins, Lipoproteins and other Body Fluid Proteins; Hoeger, U., Harris, J.R., Eds.; Subcellular Biochemistry; Springer International Publishing: Cham, Switzerland, 2020; Volume 94, pp. 499–520. ISBN 978-3-030-41768-0. [Google Scholar]
  15. Luan, Y.; Yao, Y. The Clinical Significance and Potential Role of C-Reactive Protein in Chronic Inflammatory and Neurodegenerative Diseases. Front. Immunol. 2018, 9, 1302. [Google Scholar] [CrossRef] [Green Version]
  16. Ross, R. Atherosclerosis—An Inflammatory Disease. N. Engl. J. Med. 1999, 340, 115–126. [Google Scholar] [CrossRef] [PubMed]
  17. Soehnlein, O.; Libby, P. Targeting inflammation in atherosclerosis—From experimental insights to the clinic. Nat. Rev. Drug Discov. 2021, 20, 589–610. [Google Scholar] [CrossRef]
  18. Wolf, D.; Ley, K. Immunity and Inflammation in Atherosclerosis. Circ. Res. 2019, 124, 315–327. [Google Scholar] [CrossRef]
  19. Burger, F.; Baptista, D.; Roth, A.; da Silva, R.F.; Montecucco, F.; Mach, F.; Brandt, K.J.; Miteva, K. NLRP3 Inflammasome Activation Controls Vascular Smooth Muscle Cells Phenotypic Switch in Atherosclerosis. Int. J. Mol. Sci. 2021, 23, 340. [Google Scholar] [CrossRef]
  20. Li, D.; Wu, M. Pattern recognition receptors in health and diseases. Sig. Transduct. Target. Ther. 2021, 6, 291. [Google Scholar] [CrossRef]
  21. Karasawa, T.; Takahashi, M. Role of NLRP3 Inflammasomes in Atherosclerosis. J. Atheroscler. Thromb. 2017, 24, 443–451. [Google Scholar] [CrossRef]
  22. Silvis, M.J.M.; Demkes, E.J.; Fiolet, A.T.L.; Dekker, M.; Bosch, L.; van Hout, G.P.J.; Timmers, L.; de Kleijn, D.P.V. Immunomodulation of the NLRP3 Inflammasome in Atherosclerosis, Coronary Artery Disease, and Acute Myocardial Infarction. J. Cardiovasc. Trans. Res. 2021, 14, 23–34. [Google Scholar] [CrossRef] [PubMed]
  23. Gui, Y.; Zheng, H.; Cao, R.Y. Foam Cells in Atherosclerosis: Novel Insights Into Its Origins, Consequences, and Molecular Mechanisms. Front. Cardiovasc. Med. 2022, 9, 845942. [Google Scholar] [CrossRef]
  24. Martinon, F.; Pétrilli, V.; Mayor, A.; Tardivel, A.; Tschopp, J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006, 440, 237–241. [Google Scholar] [CrossRef] [Green Version]
  25. Usui, F.; Shirasuna, K.; Kimura, H.; Tatsumi, K.; Kawashima, A.; Karasawa, T.; Yoshimura, K.; Aoki, H.; Tsutsui, H.; Noda, T.; et al. Inflammasome Activation by Mitochondrial Oxidative Stress in Macrophages Leads to the Development of Angiotensin II–Induced Aortic Aneurysm. ATVB 2015, 35, 127–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kawaguchi, M.; Takahashi, M.; Hata, T.; Kashima, Y.; Usui, F.; Morimoto, H.; Izawa, A.; Takahashi, Y.; Masumoto, J.; Koyama, J.; et al. Inflammasome Activation of Cardiac Fibroblasts Is Essential for Myocardial Ischemia/Reperfusion Injury. Circulation 2011, 123, 594–604. [Google Scholar] [CrossRef] [Green Version]
  27. Hutton, H.L.; Ooi, J.D.; Holdsworth, S.R.; Kitching, A.R. The NLRP3 inflammasome in kidney disease and autoimmunity: Inflammasomes in kidney disease. Nephrology 2016, 21, 736–744. [Google Scholar] [CrossRef]
  28. Esser, N.; Legrand-Poels, S.; Piette, J.; Scheen, A.J.; Paquot, N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res. Clin. Pract. 2014, 105, 141–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Ding, S.; Xu, S.; Ma, Y.; Liu, G.; Jang, H.; Fang, J. Modulatory Mechanisms of the NLRP3 Inflammasomes in Diabetes. Biomolecules 2019, 9, 850. [Google Scholar] [CrossRef] [Green Version]
  30. Wen, H.; Gris, D.; Lei, Y.; Jha, S.; Zhang, L.; Huang, M.T.-H.; Brickey, W.J.; Ting, J.P.-Y. Fatty acid–induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 2011, 12, 408–415. [Google Scholar] [CrossRef] [Green Version]
  31. Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010, 464, 1357–1361. [Google Scholar] [CrossRef]
  32. Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; Becker, C.E.; Ediriweera, H.N.; Mullick, A.E.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013, 14, 812–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tall, A.R.; Westerterp, M. Inflammasomes, neutrophil extracellular traps, and cholesterol. J. Lipid Res. 2019, 60, 721–727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Usui, F.; Shirasuna, K.; Kimura, H.; Tatsumi, K.; Kawashima, A.; Karasawa, T.; Hida, S.; Sagara, J.; Taniguchi, S.; Takahashi, M. Critical role of caspase-1 in vascular inflammation and development of atherosclerosis in Western diet-fed apolipoprotein E-deficient mice. Biochem. Biophys. Res. Commun. 2012, 425, 162–168. [Google Scholar] [CrossRef] [PubMed]
  35. Bian, F.; Yang, X.-Y.; Xu, G.; Zheng, T.; Jin, S. CRP-Induced NLRP3 Inflammasome Activation Increases LDL Transcytosis Across Endothelial Cells. Front. Pharmacol. 2019, 10, 40. [Google Scholar] [CrossRef]
  36. Ridker, P.M. Anticytokine Agents: Targeting Interleukin Signaling Pathways for the Treatment of Atherothrombosis. Circ Res 2019, 124, 437–450. [Google Scholar] [CrossRef]
  37. Kaneko, N.; Kurata, M.; Yamamoto, T.; Morikawa, S.; Masumoto, J. The role of interleukin-1 in general pathology. Inflamm. Regener. 2019, 39, 12. [Google Scholar] [CrossRef] [Green Version]
  38. Rose-John, S. Interleukin-6 signalling in health and disease. F1000Research 2020, 9, 1013. [Google Scholar] [CrossRef]
  39. Libby, P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy. J. Am. Coll. Cardiol. 2017, 70, 2278–2289. [Google Scholar] [CrossRef]
  40. Kaplanski, G. IL-6: A regulator of the transition from neutrophil to monocyte recruitment during inflammation. Trends Immunol. 2003, 24, 25–29. [Google Scholar] [CrossRef]
  41. Roldan, V. Interleukin-6, endothelial activation and thrombogenesis in chronic atrial fibrillation. Eur. Heart J. 2003, 24, 1373–1380. [Google Scholar] [CrossRef]
  42. Morimoto, S.; Nabata, T.; Koh, E.; Shiraishi, T.; Fukuo, K.; Imanaka, S.; Kitano, S.; Miyashita, Y.; Ogihara, T. Interleukin-6 Stimulates Proliferation of Cultured Vascular Smooth Muscle Cells Independently of Interleukin-1β. J. Cardiovasc. Pharmacol. 1991, 17, S117–S118. [Google Scholar] [CrossRef] [PubMed]
  43. Gierens, H.; Nauck, M.; Roth, M.; Schinker, R.; Schürmann, C.; Scharnagl, H.; Neuhaus, G.; Wieland, H.; März, W. Interleukin-6 Stimulates LDL Receptor Gene Expression via Activation of Sterol-Responsive and Sp1 Binding Elements. ATVB 2000, 20, 1777–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Scheller, J.; Chalaris, A.; Schmidt-Arras, D.; Rose-John, S. The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim. Et Biophys. Acta (BBA)–Mol. Cell Res. 2011, 1813, 878–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Tousoulis, D.; Oikonomou, E.; Economou, E.K.; Crea, F.; Kaski, J.C. Inflammatory cytokines in atherosclerosis: Current therapeutic approaches. Eur. Heart J. 2016, 37, 1723–1732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Aziz, N.; Fahey, J.L.; Detels, R.; Butch, A.W. Analytical Performance of a Highly Sensitive C-Reactive Protein-Based Immunoassay and the Effects of Laboratory Variables on Levels of Protein in Blood. Clin. Vaccine Immunol. 2003, 10, 652–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Clyne, B.; Olshaker, J.S. The C-reactive protein. J. Emerg. Med. 1999, 17, 1019–1025. [Google Scholar] [CrossRef] [PubMed]
  48. Póvoa, P.; Almeida, E.; Moreira, P.; Fernandes, A.; Mealha, R.; Aragão, A.; Sabino, H. C-reactive protein as an indicator of sepsis. Intensive Care Med. 1998, 24, 1052–1056. [Google Scholar] [CrossRef]
  49. Eda, S.; Kaufmann, J.; Molwitz, M.; Vorberg, E. A new method of measuring C-reactive protein, with a low limit of detection, suitable for risk assessment of coronary heart disease. Scand. J. Clin. Lab Investig. Suppl. 1999, 230, 32–35. [Google Scholar] [CrossRef]
  50. Buckley, D.I.; Fu, R.; Freeman, M.; Rogers, K.; Helfand, M. C-Reactive Protein as a Risk Factor for Coronary Heart Disease: A Systematic Review and Meta-analyses for the U.S. Preventive Services Task Force. Ann. Intern. Med. 2009, 151, 483. [Google Scholar] [CrossRef] [Green Version]
  51. Pearson, T.A.; Mensah, G.A.; Alexander, R.W.; Anderson, J.L.; Cannon, R.O.; Criqui, M.; Fadl, Y.Y.; Fortmann, S.P.; Hong, Y.; Myers, G.L.; et al. 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. [Google Scholar] [CrossRef]
  52. Ridker, P.M.; Cannon, C.P.; Morrow, D.; Rifai, N.; Rose, L.M.; McCabe, C.H.; Pfeffer, M.A.; Braunwald, E. C-Reactive Protein Levels and Outcomes after Statin Therapy. N. Engl. J. Med. 2005, 352, 20–28. [Google Scholar] [CrossRef] [PubMed]
  53. Nissen, S.E.; Tuzcu, E.M.; Schoenhagen, P.; Crowe, T.; Sasiela, W.J.; Tsai, J.; Orazem, J.; Magorien, R.D.; O’Shaughnessy, C.; Ganz, P. Statin Therapy, LDL Cholesterol, C-Reactive Protein, and Coronary Artery Disease. N. Engl. J. Med. 2005, 352, 29–38. [Google Scholar] [CrossRef] [PubMed]
  54. Morrow, D.A.; de Lemos, J.A.; Sabatine, M.S.; Wiviott, S.D.; Blazing, M.A.; Shui, A.; Rifai, N.; Califf, R.M.; Braunwald, E. Clinical Relevance of C-Reactive Protein During Follow-Up of Patients With Acute Coronary Syndromes in the Aggrastat-to-Zocor Trial. Circulation 2006, 114, 281–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ridker, P.M.; Hennekens, C.H.; Buring, J.E.; Rifai, N. C-Reactive Protein and Other Markers of Inflammation in the Prediction of Cardiovascular Disease in Women. N. Engl. J. Med. 2000, 342, 836–843. [Google Scholar] [CrossRef] [PubMed]
  56. Ridker, P.M.; Cushman, M.; Stampfer, M.J.; Tracy, R.P.; Hennekens, C.H. Inflammation, Aspirin, and the Risk of Cardiovascular Disease in Apparently Healthy Men. N. Engl. J. Med. 1997, 336, 973–979. [Google Scholar] [CrossRef]
  57. MRFIT Research Group; Kuller, L.H.; Tracy, R.P.; Shaten, J.; Meilahn, E.N. Relation of C-Reactive Protein and Coronary Heart Disease in the MRFIT Nested Case-Control Study. Am. J. Epidemiol. 1996, 144, 537–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. The Emerging Risk Factors Collaboration C-Reactive Protein, Fibrinogen, and Cardiovascular Disease Prediction. N. Engl. J. Med. 2012, 367, 1310–1320. [CrossRef] [Green Version]
  59. Saito, I.; Maruyama, K.; Eguchi, E. C-Reactive Protein and Cardiovascular Disease in East Asians: A Systematic Review. Clin. Med. Insights Cardiol. 2014, 8, CMC-S17066. [Google Scholar] [CrossRef] [Green Version]
  60. Lv, J.-M.; Lü, S.-Q.; Liu, Z.-P.; Zhang, J.; Gao, B.-X.; Yao, Z.-Y.; Wu, Y.-X.; Potempa, L.A.; Ji, S.-R.; Long, M.; et al. Conformational folding and disulfide bonding drive distinct stages of protein structure formation. Sci. Rep. 2018, 8, 1494. [Google Scholar] [CrossRef] [Green Version]
  61. Volanakis, J.E.; Wirtz, K.W.A. Interaction of C-reactive protein with artificial phosphatidylcholine bilayers. Nature 1979, 281, 155–157. [Google Scholar] [CrossRef]
  62. Thompson, D.; Pepys, M.B.; Wood, S.P. The physiological structure of human C-reactive protein and its complex with phosphocholine. Structure 1999, 7, 169–177. [Google Scholar] [CrossRef] [PubMed]
  63. Hack, C.E.; Wolbink, G.-J.; Schalkwijk, C.; Speijer, H.; Hermens, W.T.; van den Bosch, H. A role for secretory phospholipase A2 and C-reactive protein in the removal of injured cells. Immunol. Today 1997, 18, 111–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wu, R.; Huang, Y.H.; Elinder, L.S.; Frostegård, J. Lysophosphatidylcholine Is Involved in the Antigenicity of Oxidized LDL. ATVB 1998, 18, 626–630. [Google Scholar] [CrossRef] [Green Version]
  65. Law, S.-H.; Chan, M.-L.; Marathe, G.K.; Parveen, F.; Chen, C.-H.; Ke, L.-Y. An Updated Review of Lysophosphatidylcholine Metabolism in Human Diseases. Int. J. Mol. Sci. 2019, 20, 1149. [Google Scholar] [CrossRef] [Green Version]
  66. Corrêa, R.; Silva, L.F.F.; Ribeiro, D.J.S.; das Neves Almeida, R.; de Oliveira Santos, I.; Corrêa, L.H.; de Sant’Ana, L.P.; Assunção, L.S.; Bozza, P.T.; Magalhães, K.G. Lysophosphatidylcholine Induces NLRP3 Inflammasome-Mediated Foam Cell Formation and Pyroptosis in Human Monocytes and Endothelial Cells. Front. Immunol. 2020, 10, 2927. [Google Scholar] [CrossRef] [Green Version]
  67. Zhang, S.-Y.; Dong, Y.-Q.; Wang, P.; Zhang, X.; Yan, Y.; Sun, L.; Liu, B.; Zhang, D.; Zhang, H.; Liu, H.; et al. Adipocyte-derived Lysophosphatidylcholine Activates Adipocyte and Adipose Tissue Macrophage Nod-Like Receptor Protein 3 Inflammasomes Mediating Homocysteine-Induced Insulin Resistance. EBioMedicine 2018, 31, 202–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Du Clos, T.W.; Mold, C. Pentraxins (CRP, SAP) in the process of complement activation and clearance of apoptotic bodies through Fcγ receptors. Curr. Opin. Organ Transplant. 2011, 16, 15–20. [Google Scholar] [CrossRef] [Green Version]
  69. Mold, C.; Gewurz, H.; Du Clos, T.W. Regulation of complement activation by C-reactive protein. Immunopharmacology 1999, 42, 23–30. [Google Scholar] [CrossRef]
  70. Du Clos, T.W. The interaction of C-reactive protein and serum amyloid P component with nuclear antigens. Mol. Biol. Rep. 1996, 23, 253–260. [Google Scholar] [CrossRef]
  71. Braig, D.; Nero, T.L.; Koch, H.-G.; Kaiser, B.; Wang, X.; Thiele, J.R.; Morton, C.J.; Zeller, J.; Kiefer, J.; Potempa, L.A.; et al. Transitional changes in the CRP structure lead to the exposure of proinflammatory binding sites. Nat. Commun. 2017, 8, 14188. [Google Scholar] [CrossRef]
  72. Ji, S.; Wu, Y.; Zhu, L.; Potempa, L.A.; Sheng, F.; Lu, W.; Zhao, J. Cell membranes and liposomes dissociate C-reactive protein (CRP) to form a new, biologically active structural intermediate: mCRPm. FASEB J. 2007, 21, 284–294. [Google Scholar] [CrossRef]
  73. Zhang, C.-M.; Tan, Y.-B.; Zhou, H.-H.; Ge, Z.-B.; Feng, J.-R.; Lv, G.-B.; Sun, Z.-Y.; Fu, Y.; Wang, M.-Y. Intra-subunit Disulfide Determines the Conversion and Structural Stability of CRP Isoforms. Inflammation 2020, 43, 466–477. [Google Scholar] [CrossRef]
  74. Li, H.-Y.; Wang, J.; Meng, F.; Jia, Z.-K.; Su, Y.; Bai, Q.-F.; Lv, L.-L.; Ma, F.-R.; Potempa, L.A.; Yan, Y.-B.; et al. An Intrinsically Disordered Motif Mediates Diverse Actions of Monomeric C-reactive Protein. J. Biol. Chem. 2016, 291, 8795–8804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Khreiss, T.; József, L.; Hossain, S.; Chan, J.S.D.; Potempa, L.A.; Filep, J.G. Loss of Pentameric Symmetry of C-reactive Protein Is Associated with Delayed Apoptosis of Human Neutrophils. J. Biol. Chem. 2002, 277, 40775–40781. [Google Scholar] [CrossRef] [Green Version]
  76. Habersberger, J.; Strang, F.; Scheichl, A.; Htun, N.; Bassler, N.; Merivirta, R.-M.; Diehl, P.; Krippner, G.; Meikle, P.; Eisenhardt, S.U.; et al. Circulating microparticles generate and transport monomeric C-reactive protein in patients with myocardial infarction. Cardiovasc. Res. 2012, 96, 64–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Crawford, J.R.; Trial, J.; Nambi, V.; Hoogeveen, R.C.; Taffet, G.E.; Entman, M.L. Plasma Levels of Endothelial Microparticles Bearing Monomeric C-reactive Protein are Increased in Peripheral Artery Disease. J. Cardiovasc. Trans. Res. 2016, 9, 184–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Melnikov, I.; Kozlov, S.; Saburova, O.; Zubkova, E.; Guseva, O.; Domogatsky, S.; Arefieva, T.; Radyukhina, N.; Zvereva, M.; Avtaeva, Y.; et al. CRP Is Transported by Monocytes and Monocyte-Derived Exosomes in the Blood of Patients with Coronary Artery Disease. Biomedicines 2020, 8, 435. [Google Scholar] [CrossRef]
  79. d’Alessandro, E.; Becker, C.; Bergmeier, W.; Bode, C.; Bourne, J.H.; Brown, H.; Buller, H.R.; ten Cate-Hoek, A.J.; ten Cate, V.; van Cauteren, Y.J.M.; et al. Thrombo-Inflammation in Cardiovascular Disease: An Expert Consensus Document from the Third Maastricht Consensus Conference on Thrombosis. Thromb. Haemost. 2020, 120, 538–564. [Google Scholar] [CrossRef] [Green Version]
  80. Molins, B.; Peña, E.; Vilahur, G.; Mendieta, C.; Slevin, M.; Badimon, L. C-Reactive Protein Isoforms Differ in Their Effects on Thrombus Growth. ATVB 2008, 28, 2239–2246. [Google Scholar] [CrossRef] [Green Version]
  81. Molins, B.; Peña, E.; de la Torre, R.; Badimon, L. Monomeric C-reactive protein is prothrombotic and dissociates from circulating pentameric C-reactive protein on adhered activated platelets under flow. Cardiovasc. Res. 2011, 92, 328–337. [Google Scholar] [CrossRef]
  82. de la Torre, R.; Peña, E.; Vilahur, G.; Slevin, M.; Badimon, L. Monomerization of C-reactive protein requires glycoprotein IIb-IIIa activation: Pentraxins and platelet deposition. J. Thromb. Haemost. 2013, 11, 2048–2058. [Google Scholar] [CrossRef] [PubMed]
  83. Khreiss, T.; József, L.; Potempa, L.A.; Filep, J.G. Conformational Rearrangement in C-Reactive Protein Is Required for Proinflammatory Actions on Human Endothelial Cells. Circulation 2004, 109, 2016–2022. [Google Scholar] [CrossRef] [Green Version]
  84. Li, R.; Ren, M.; Luo, M.; Chen, N.; Zhang, Z.; Luo, B.; Wu, J. Monomeric C-reactive protein alters fibrin clot properties on endothelial cells. Thromb. Res. 2012, 129, e251–e256. [Google Scholar] [CrossRef]
  85. Zouki, C.; Haas, B.; Chan, J.S.D.; Potempa, L.A.; Filep, J.G. Loss of Pentameric Symmetry of C-Reactive Protein Is Associated with Promotion of Neutrophil-Endothelial Cell Adhesion. J. Immunol. 2001, 167, 5355–5361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Eisenhardt, S.U.; Habersberger, J.; Murphy, A.; Chen, Y.-C.; Woollard, K.J.; Bassler, N.; Qian, H.; von zur Muhlen, C.; Hagemeyer, C.E.; Ahrens, I.; et al. Dissociation of Pentameric to Monomeric C-Reactive Protein on Activated Platelets Localizes Inflammation to Atherosclerotic Plaques. Circ. Res. 2009, 105, 128–137. [Google Scholar] [CrossRef] [Green Version]
  87. Thiele, J.R.; Habersberger, J.; Braig, D.; Schmidt, Y.; Goerendt, K.; Maurer, V.; Bannasch, H.; Scheichl, A.; Woollard, K.J.; von Dobschütz, E.; et al. Dissociation of Pentameric to Monomeric C-Reactive Protein Localizes and Aggravates Inflammation: In Vivo Proof of a Powerful Proinflammatory Mechanism and a New Anti-Inflammatory Strategy. Circulation 2014, 130, 35–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Ullah, N.; Ma, F.-R.; Han, J.; Liu, X.-L.; Fu, Y.; Liu, Y.-T.; Liang, Y.-L.; Ouyang, H.; Li, H.-Y. Monomeric C-reactive protein regulates fibronectin mediated monocyte adhesion. Mol. Immunol. 2020, 117, 122–130. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, Z.; Na, H.; Gan, Q.; Tao, Q.; Alekseyev, Y.; Hu, J.; Yan, Z.; Yang, J.B.; Tian, H.; Zhu, S.; et al. Monomeric C-reactive protein via endothelial CD31 for neurovascular inflammation in an ApoE genotype-dependent pattern: A risk factor for Alzheimer’s disease? Aging Cell 2021, 20, e13501. [Google Scholar] [CrossRef]
  90. Li, H.-Y.; Wang, J.; Wu, Y.-X.; Zhang, L.; Liu, Z.-P.; Filep, J.G.; Potempa, L.A.; Wu, Y.; Ji, S.-R. Topological Localization of Monomeric C-reactive Protein Determines Proinflammatory Endothelial Cell Responses. J. Biol. Chem. 2014, 289, 14283–14290. [Google Scholar] [CrossRef] [Green Version]
  91. Ji, S.-R.; Ma, L.; Bai, C.-J.; Shi, J.-M.; Li, H.-Y.; Potempa, L.A.; Filep, J.G.; Zhao, J.; Wu, Y. Monomeric C-reactive protein activates endothelial cells via interaction with lipid raft microdomains. FASEB J. 2009, 23, 1806–1816. [Google Scholar] [CrossRef]
  92. Khreiss, T.; József, L.; Potempa, L.A.; Filep, J.G. Loss of Pentameric Symmetry in C-Reactive Protein Induces Interleukin-8 Secretion Through Peroxynitrite Signaling in Human Neutrophils. Circ. Res. 2005, 97, 690–697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Trial, J.; Potempa, L.A.; Entman, M.L. The role of C-reactive protein in innate and acquired inflammation: New perspectives. Inflamm. Cell Signal. 2016, 3, e1409. [Google Scholar] [PubMed]
  94. Ji, S.; Wu, Y.; Potempa, L.; Qiu, Q.; Zhao, J. Interactions of C-reactive protein with low-density lipoproteins: Implications for an active role of modified C-reactive protein in atherosclerosis. Int. J. Biochem. Cell Biol. 2006, 38, 648–661. [Google Scholar] [CrossRef] [PubMed]
  95. Zha, Z.; Cheng, Y.; Cao, L.; Qian, Y.; Liu, X.; Guo, Y.; Wang, J. Monomeric CRP Aggravates Myocardial Injury After Myocardial Infarction by Polarizing the Macrophage to Pro-Inflammatory Phenotype Through JNK Signaling Pathway. JIR 2021, 14, 7053–7064. [Google Scholar] [CrossRef]
  96. Thiele, J.R.; Zeller, J.; Kiefer, J.; Braig, D.; Kreuzaler, S.; Lenz, Y.; Potempa, L.A.; Grahammer, F.; Huber, T.B.; Huber-Lang, M.; et al. A Conformational Change in C-Reactive Protein Enhances Leukocyte Recruitment and Reactive Oxygen Species Generation in Ischemia/Reperfusion Injury. Front. Immunol. 2018, 9, 675. [Google Scholar] [CrossRef] [Green Version]
  97. Boras, E.; Slevin, M.; Alexander, M.Y.; Aljohi, A.; Gilmore, W.; Ashworth, J.; Krupinski, J.; Potempa, L.A.; Al Abdulkareem, I.; Elobeid, A.; et al. Monomeric C-reactive protein and Notch-3 co-operatively increase angiogenesis through PI3K signalling pathway. Cytokine 2014, 69, 165–179. [Google Scholar] [CrossRef] [PubMed]
  98. Turu, M.M.; Slevin, M.; Matou, S.; West, D.; Rodríguez, C.; Luque, A.; Grau-Olivares, M.; Badimon, L.; Martinez-Gonzalez, J.; Krupinski, J. C-reactive protein exerts angiogenic effects on vascular endothelial cells and modulates associated signalling pathways and gene expression. BMC Cell Biol. 2008, 9, 47. [Google Scholar] [CrossRef] [Green Version]
  99. Krupinski, J.; Turu, M.M.; Martinez-Gonzalez, J.; Carvajal, A.; Juan-Babot, J.O.; Iborra, E.; Slevin, M.; Rubio, F.; Badimon, L. Endogenous Expression of C-Reactive Protein Is Increased in Active (Ulcerated Noncomplicated) Human Carotid Artery Plaques. Stroke 2006, 37, 1200–1204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Kobayashi, S.; Inoue, N.; Ohashi, Y.; Terashima, M.; Matsui, K.; Mori, T.; Fujita, H.; Awano, K.; Kobayashi, K.; Azumi, H.; et al. Interaction of Oxidative Stress and Inflammatory Response in Coronary Plaque Instability: Important Role of C-Reactive Protein. ATVB 2003, 23, 1398–1404. [Google Scholar] [CrossRef] [Green Version]
  101. Melnikov, I.S.; Kozlov, S.G.; Chumachenko, P.V.; Saburova, O.S.; Guseva, O.A.; Prokofyeva, L.V.; Gabbasov, Z.A. Monomeric C-reactive protein and local inflammatory reaction in the wall of the coronary arteries in patients with stable coronary artery disease. Russ. J. Cardiol. 2019, 24, 56–61. [Google Scholar] [CrossRef]
  102. Vainas, T.; Stassen, F.R.M.; de Graaf, R.; Twiss, E.L.L.; Herngreen, S.B.; Welten, R.J.T.J.; van den Akker, L.H.J.M.; van Dieijen-Visser, M.P.; Bruggeman, C.A.; Kitslaar, P.J.E.H.M. C-reactive protein in peripheral arterial disease: Relation to severity of the disease and to future cardiovascular events. J. Vasc. Surg. 2005, 42, 243–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Jabs, W.J.; Theissing, E.; Nitschke, M.; Bechtel, J.F.M.; Duchrow, M.; Mohamed, S.; Jahrbeck, B.; Sievers, H.-H.; Steinhoff, J.; Bartels, C. Local Generation of C-Reactive Protein in Diseased Coronary Artery Venous Bypass Grafts and Normal Vascular Tissue. Circulation 2003, 108, 1428–1431. [Google Scholar] [CrossRef] [PubMed]
  104. Sattler, K.J.E.; Woodrum, J.E.; Galili, O.; Olson, M.; Samee, S.; Meyer, F.B.; Zhu, X.-Y.; Lerman, L.O.; Lerman, A. Concurrent Treatment With Renin-Angiotensin System Blockers and Acetylsalicylic Acid Reduces Nuclear Factor κB Activation and C-Reactive Protein Expression in Human Carotid Artery Plaques. Stroke 2005, 36, 14–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Calabro, P.; Chang, D.W.; Willerson, J.T.; Yeh, E.T.H. Release of C-Reactive Protein in Response to Inflammatory Cytokines by Human Adipocytes: Linking Obesity to Vascular Inflammation. J. Am. Coll. Cardiol. 2005, 46, 1112–1113. [Google Scholar] [CrossRef] [Green Version]
  106. Kolb-Bachofen, V.; Puchta-Teudt, N.; Egenhofer, C. Expression of membrane-associated C-reactive protein by human monocytes: Indications for a selectin-like activity participating in adhesion. Glycoconj. J. 1995, 12, 122–127. [Google Scholar] [CrossRef]
  107. Ciubotaru, I.; Potempa, L.A.; Wander, R.C. Production of Modified C-Reactive Protein in U937-Derived Macrophages. Exp. Biol. Med. 2005, 230, 762–770. [Google Scholar] [CrossRef] [Green Version]
  108. Haider, D.G.; Leuchten, N.; Schaller, G.; Gouya, G.; Kolodjaschna, J.; Schmetterer, L.; Kapiotis, S.; Wolzt, M. C-reactive protein is expressed and secreted by peripheral blood mononuclear cells. Clin. Exp. Immunol. 2006, 146, 533–539. [Google Scholar] [CrossRef]
  109. Wang, J.; Tang, B.; Liu, X.; Wu, X.; Wang, H.; Xu, D.; Guo, Y. Increased monomeric CRP levels in acute myocardial infarction: A possible new and specific biomarker for diagnosis and severity assessment of disease. Atherosclerosis 2015, 239, 343–349. [Google Scholar] [CrossRef]
  110. Zhang, L.; Li, H.-Y.; Li, W.; Shen, Z.-Y.; Wang, Y.-D.; Ji, S.-R.; Wu, Y. An ELISA Assay for Quantifying Monomeric C-Reactive Protein in Plasma. Front. Immunol. 2018, 9, 511. [Google Scholar] [CrossRef] [Green Version]
  111. Williams, R.D.; Moran, J.A.; Fryer, A.A.; Littlejohn, J.R.; Williams, H.M.; Greenhough, T.J.; Shrive, A.K. Monomeric C-Reactive Protein in Serum With Markedly Elevated CRP Levels Shares Common Calcium-Dependent Ligand Binding Properties With an in vitro Dissociated Form of C-Reactive Protein. Front. Immunol. 2020, 11, 115. [Google Scholar] [CrossRef]
  112. Wu, K.-L.; Liang, Q.-H.; Huang, B.-T.; Ding, N.; Li, B.-W.; Hao, J. The plasma level of mCRP is linked to cardiovascular disease in antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Res. Ther. 2020, 22, 228. [Google Scholar] [CrossRef]
  113. Munuswamy, R.; De Brandt, J.; Burtin, C.; Derave, W.; Aumann, J.; Spruit, M.A.; Michiels, L. Monomeric CRP is Elevated in Patients with COPD Compared to Non-COPD Control Persons. JIR 2021, 14, 4503–4507. [Google Scholar] [CrossRef] [PubMed]
  114. Liang, Y.; Xu, K.; Liu, W.; Liu, X.; Yuan, P.; Xu, P.; Li, H. Monomeric C-reactive protein level is associated with osteoarthritis. Exp. Ther. Med. 2022, 23, 277. [Google Scholar] [CrossRef] [PubMed]
  115. Melnikov, I.; Kozlov, S.; Pogorelova, O.; Tripoten, M.; Khamchieva, L.; Saburova, O.; Avtaeva, Y.; Zvereva, M.; Matroze, E.; Kuznetsova, T.; et al. The monomeric C-reactive protein level is associated with the increase in carotid plaque number in patients with subclinical carotid atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 968267. [Google Scholar] [CrossRef] [PubMed]
  116. Karlsson, J.; Wetterö, J.; Weiner, M.; Rönnelid, J.; Fernandez-Botran, R.; Sjöwall, C. Associations of C-reactive protein isoforms with systemic lupus erythematosus phenotypes and disease activity. Arthritis Res. Ther. 2022, 24, 139. [Google Scholar] [CrossRef] [PubMed]
  117. Fujita, C.; Sakurai, Y.; Yasuda, Y.; Homma, R.; Huang, C.-L.; Fujita, M. mCRP as a Biomarker of Adult-Onset Still’s Disease: Quantification of mCRP by ELISA. Front. Immunol. 2022, 13, 938173. [Google Scholar] [CrossRef] [PubMed]
  118. Ristagno, G.; Fumagalli, F.; Bottazzi, B.; Mantovani, A.; Olivari, D.; Novelli, D.; Latini, R. Pentraxin 3 in Cardiovascular Disease. Front. Immunol. 2019, 10, 823. [Google Scholar] [CrossRef] [Green Version]
  119. van ’t Klooster, C.C.; van der Graaf, Y.; Ridker, P.M.; Westerink, J.; Hjortnaes, J.; Sluijs, I.; Asselbergs, F.W.; Bots, M.L.; Kappelle, L.J.; Visseren, F.L.J. The relation between healthy lifestyle changes and decrease in systemic inflammation in patients with stable cardiovascular disease. Atherosclerosis 2020, 301, 37–43. [Google Scholar] [CrossRef]
  120. Gaesser, G.A.; Angadi, S.S.; Ryan, D.M.; Johnston, C.S. Lifestyle Measures to Reduce Inflammation. Am. J. Lifestyle Med. 2012, 6, 4–13. [Google Scholar] [CrossRef]
  121. Fedewa, M.V.; Hathaway, E.D.; Ward-Ritacco, C.L. Effect of exercise training on C reactive protein: A systematic review and meta-analysis of randomised and non-randomised controlled trials. Br. J. Sports Med. 2017, 51, 670–676. [Google Scholar] [CrossRef]
  122. Bianchi, V.E. Weight loss is a critical factor to reduce inflammation. Clin. Nutr. ESPEN 2018, 28, 21–35. [Google Scholar] [CrossRef] [PubMed]
  123. Bohula, E.A.; Giugliano, R.P.; Cannon, C.P.; Zhou, J.; Murphy, S.A.; White, J.A.; Tershakovec, A.M.; Blazing, M.A.; Braunwald, E. Achievement of Dual Low-Density Lipoprotein Cholesterol and High-Sensitivity C-Reactive Protein Targets More Frequent With the Addition of Ezetimibe to Simvastatin and Associated With Better Outcomes in IMPROVE-IT. Circulation 2015, 132, 1224–1233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Nissen, S.E. Effect of Intensive Lipid Lowering on Progression of Coronary Atherosclerosis: Evidence for an Early Benefit from the Reversal of Atherosclerosis with Aggressive Lipid Lowering (REVERSAL) Trial. Am. J. Cardiol. 2005, 96, 61–68. [Google Scholar] [CrossRef] [PubMed]
  125. Ridker, P.M.; Danielson, E.; Fonseca, F.A.H.; Genest, J.; Gotto, A.M.; Kastelein, J.J.P.; Koenig, W.; Libby, P.; Lorenzatti, A.J.; MacFadyen, J.G.; et al. Rosuvastatin to Prevent Vascular Events in Men and Women with Elevated C-Reactive Protein. N. Engl. J. Med. 2008, 359, 2195–2207. [Google Scholar] [CrossRef] [Green Version]
  126. Bohula, E.A.; Giugliano, R.P.; Leiter, L.A.; Verma, S.; Park, J.-G.; Sever, P.S.; Lira Pineda, A.; Honarpour, N.; Wang, H.; Murphy, S.A.; et al. Inflammatory and Cholesterol Risk in the FOURIER Trial. Circulation 2018, 138, 131–140. [Google Scholar] [CrossRef]
  127. Pradhan, A.D.; Aday, A.W.; Rose, L.M.; Ridker, P.M. Residual Inflammatory Risk on Treatment With PCSK9 Inhibition and Statin Therapy. Circulation 2018, 138, 141–149. [Google Scholar] [CrossRef]
  128. Kalkman, D.N.; Aquino, M.; Claessen, B.E.; Baber, U.; Guedeney, P.; Sorrentino, S.; Vogel, B.; de Winter, R.J.; Sweeny, J.; Kovacic, J.C.; et al. Residual inflammatory risk and the impact on clinical outcomes in patients after percutaneous coronary interventions. Eur. Heart J. 2018, 39, 4101–4108. [Google Scholar] [CrossRef]
  129. Guedeney, P.; Claessen, B.E.; Kalkman, D.N.; Aquino, M.; Sorrentino, S.; Giustino, G.; Farhan, S.; Vogel, B.; Sartori, S.; Montalescot, G.; et al. Residual Inflammatory Risk in Patients With Low LDL Cholesterol Levels Undergoing Percutaneous Coronary Intervention. J. Am. Coll. Cardiol. 2019, 73, 2401–2409. [Google Scholar] [CrossRef]
  130. Ridker, P.M. Clinician’s Guide to Reducing Inflammation to Reduce Atherothrombotic Risk. J. Am. Coll. Cardiol. 2018, 72, 3320–3331. [Google Scholar] [CrossRef]
  131. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington, R.A.; et al. Alirocumab and Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Med. 2018, 379, 2097–2107. [Google Scholar] [CrossRef]
  132. Pagidipati, N.J.; Hellkamp, A.S.; Sharma, P.P.; Wang, T.Y.; Fonarow, G.C.; Pencina, M. High-sensitivity C-reactive protein elevation in patients with prior myocardial infarction in the United States. Am. Heart J. 2018, 204, 151–155. [Google Scholar] [CrossRef] [PubMed]
  133. Pearson, T.A.; Ballantyne, C.M.; Veltri, E.; Shah, A.; Bird, S.; Lin, J.; Rosenberg, E.; Tershakovec, A.M. Pooled Analyses of Effects on C-Reactive Protein and Low Density Lipoprotein Cholesterol in Placebo-Controlled Trials of Ezetimibe Monotherapy or Ezetimibe Added to Baseline Statin Therapy. Am. J. Cardiol. 2009, 103, 369–374. [Google Scholar] [CrossRef] [PubMed]
  134. Barbosa, S.P.; Lins, L.C.; Fonseca, F.A.; Matos, L.N.; Aguirre, A.C.; Bianco, H.T.; Amaral, J.B.; França, C.N.; Santana, J.M.; Izar, M.C. Effects of ezetimibe on markers of synthesis and absorption of cholesterol in high-risk patients with elevated C-reactive protein. Life Sci. 2013, 92, 845–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kater, A.-L.A.; Batista, M.C.; Ferreira, S.R. Synergistic effect of simvastatin and ezetimibe on lipid and pro-inflammatory profiles in pre-diabetic subjects. Diabetol. Metab. Syndr. 2010, 2, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Oh, M.S.; Min, Y.J.; Kwon, J.E.; Cho, E.J.; Kim, J.E.; Lee, W.-S.; Lee, K.J.; Kim, S.-W.; Kim, T.H.; Kim, C.J.; et al. Effects of Ezetimibe Added to Ongoing Statin Therapy on C-Reactive Protein Levels in Hypercholesterolemic Patients. Korean Circ. J. 2011, 41, 253. [Google Scholar] [CrossRef] [Green Version]
  137. Ruscica, M.; Tokgözoğlu, L.; Corsini, A.; Sirtori, C.R. PCSK9 inhibition and inflammation: A narrative review. Atherosclerosis 2019, 288, 146–155. [Google Scholar] [CrossRef] [Green Version]
  138. Sahebkar, A.; Di Giosia, P.; Stamerra, C.A.; Grassi, D.; Pedone, C.; Ferretti, G.; Bacchetti, T.; Ferri, C.; Giorgini, P. Effect of monoclonal antibodies to PCSK9 on high-sensitivity C-reactive protein levels: A meta-analysis of 16 randomized controlled treatment arms: PCSK9 inhibitors and hs-CRP levels. Br. J. Clin. Pharmacol. 2016, 81, 1175–1190. [Google Scholar] [CrossRef] [Green Version]
  139. Ridker, P.M.; Everett, B.M.; Pradhan, A.; MacFadyen, J.G.; Solomon, D.H.; Zaharris, E.; Mam, V.; Hasan, A.; Rosenberg, Y.; Iturriaga, E.; et al. Low-Dose Methotrexate for the Prevention of Atherosclerotic Events. N. Engl. J. Med. 2019, 380, 752–762. [Google Scholar] [CrossRef]
  140. O’Donoghue, M.L.; Glaser, R.; Cavender, M.A.; Aylward, P.E.; Bonaca, M.P.; Budaj, A.; Davies, R.Y.; Dellborg, M.; Fox, K.A.A.; Gutierrez, J.A.T.; et al. Effect of Losmapimod on Cardiovascular Outcomes in Patients Hospitalized With Acute Myocardial Infarction: A Randomized Clinical Trial. JAMA 2016, 315, 1591. [Google Scholar] [CrossRef] [Green Version]
  141. O’Donoghue, M.L.; Braunwald, E.; White, H.D.; Steen, D.P.; Lukas, M.A.; Tarka, E.; Steg, P.G.; Hochman, J.S.; Bode, C.; Maggioni, A.P.; et al. Effect of Darapladib on Major Coronary Events After an Acute Coronary Syndrome: The SOLID-TIMI 52 Randomized Clinical Trial. JAMA 2014, 312, 1006. [Google Scholar] [CrossRef]
  142. Martínez, G.J.; Celermajer, D.S.; Patel, S. The NLRP3 inflammasome and the emerging role of colchicine to inhibit atherosclerosis-associated inflammation. Atherosclerosis 2018, 269, 262–271. [Google Scholar] [CrossRef] [PubMed]
  143. Tardif, J.-C.; Kouz, S.; Waters, D.D.; Bertrand, O.F.; Diaz, R.; Maggioni, A.P.; Pinto, F.J.; Ibrahim, R.; Gamra, H.; Kiwan, G.S.; et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N. Engl. J. Med. 2019, 381, 2497–2505. [Google Scholar] [CrossRef] [PubMed]
  144. Nidorf, S.M.; Fiolet, A.T.L.; Mosterd, A.; Eikelboom, J.W.; Schut, A.; Opstal, T.S.J.; The, S.H.K.; Xu, X.-F.; Ireland, M.A.; Lenderink, T.; et al. Colchicine in Patients with Chronic Coronary Disease. N. Engl. J. Med. 2020, 383, 1838–1847. [Google Scholar] [CrossRef]
  145. Tong, D.C.; Quinn, S.; Nasis, A.; Hiew, C.; Roberts-Thomson, P.; Adams, H.; Sriamareswaran, R.; Htun, N.M.; Wilson, W.; Stub, D.; et al. Colchicine in Patients With Acute Coronary Syndrome: The Australian COPS Randomized Clinical Trial. Circulation 2020, 142, 1890–1900. [Google Scholar] [CrossRef] [PubMed]
  146. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  147. Ridker, P.M.; MacFadyen, J.G.; Everett, B.M.; Libby, P.; Thuren, T.; Glynn, R.J.; Ridker, P.M.; MacFadyen, J.G.; Everett, B.M.; Libby, P.; et al. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: A secondary analysis from the CANTOS randomised controlled trial. Lancet 2018, 391, 319–328. [Google Scholar] [CrossRef]
  148. Ridker, P.M.; Libby, P.; MacFadyen, J.G.; Thuren, T.; Ballantyne, C.; Fonseca, F.; Koenig, W.; Shimokawa, H.; Everett, B.M.; Glynn, R.J. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: Analyses from the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS). Eur. Heart J. 2018, 39, 3499–3507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Ridker, P.M.; Devalaraja, M.; Baeres, F.M.M.; Engelmann, M.D.M.; Hovingh, G.K.; Ivkovic, M.; Lo, L.; Kling, D.; Pergola, P.; Raj, D.; et al. IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2021, 397, 2060–2069. [Google Scholar] [CrossRef]
  150. Ridker, P.M. From RESCUE to ZEUS: Will interleukin-6 inhibition with ziltivekimab prove effective for cardiovascular event reduction? Cardiovasc. Res. 2021, 117, e138–e140. [Google Scholar] [CrossRef]
  151. Yu, Q.; Liu, Z.; Waqar, A.B.; Ning, B.; Yang, X.; Shiomi, M.; Graham, M.J.; Crooke, R.M.; Liu, E.; Dong, S.; et al. Effects of Antisense Oligonucleotides against C-Reactive Protein on the Development of Atherosclerosis in WHHL Rabbits. Mediat. Inflamm. 2014, 2014, 979132. [Google Scholar] [CrossRef]
  152. Jones, N.R.; Pegues, M.A.; McCrory, M.A.; Singleton, W.; Bethune, C.; Baker, B.F.; Norris, D.A.; Crooke, R.M.; Graham, M.J.; Szalai, A.J. A Selective Inhibitor of Human C-reactive Protein Translation Is Efficacious In Vitro and in C-reactive Protein Transgenic Mice and Humans. Mol. Ther. —Nucleic Acids 2012, 1, e52. [Google Scholar] [CrossRef] [PubMed]
  153. Noveck, R.; Stroes, E.S.G.; Flaim, J.D.; Baker, B.F.; Hughes, S.; Graham, M.J.; Crooke, R.M.; Ridker, P.M. Effects of an Antisense Oligonucleotide Inhibitor of C-Reactive Protein Synthesis on the Endotoxin Challenge Response in Healthy Human Male Volunteers. JAHA 2014, 3, e001084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Sugihara, C.; Freemantle, N.; Hughes, S.G.; Furniss, S.; Sulke, N. The effect of C-reactive protein reduction with a highly specific antisense oligonucleotide on atrial fibrillation assessed using beat-to-beat pacemaker Holter follow-up. J. Intervig. Card. Electrophysiol. 2015, 43, 91–98. [Google Scholar] [CrossRef] [PubMed]
  155. Warren, M.S.; Hughes, S.G.; Singleton, W.; Yamashita, M.; Genovese, M.C. Results of a proof of concept, double-blind, randomized trial of a second generation antisense oligonucleotide targeting high-sensitivity C-reactive protein (hs-CRP) in rheumatoid arthritis. Arthritis Res. Ther. 2015, 17, 80. [Google Scholar] [CrossRef] [Green Version]
  156. Pepys, M.B.; Hirschfield, G.M.; Tennent, G.A.; Ruth Gallimore, J.; Kahan, M.C.; Bellotti, V.; Hawkins, P.N.; Myers, R.M.; Smith, M.D.; Polara, A.; et al. Targeting C-reactive protein for the treatment of cardiovascular disease. Nature 2006, 440, 1217–1221. [Google Scholar] [CrossRef] [Green Version]
  157. Zeller, J.; Cheung Tung Shing, K.S.; Nero, T.L.; McFadyen, J.D.; Krippner, G.; Bogner, B.; Kreuzaler, S.; Kiefer, J.; Horner, V.K.; Braig, D.; et al. A novel phosphocholine-mimetic inhibits a pro-inflammatory conformational change in C-reactive protein. EMBO Mol. Med. 2022, 15, e16236. [Google Scholar] [CrossRef]
  158. Kather, M.G.; Zeller, J.; Plattner, D.; Breit, B.; Kreuzaler, S.; Krippner, G.; Peter, K.; Eisenhardt, S.U.; Kammerer, B. Pharmacokinetic study of the novel phosphocholine derivative 3-dibutylaminopropylphosphonic acid by LC-MS coupling. J. Chromatogr. B 2021, 1186, 122998. [Google Scholar] [CrossRef]
  159. Fujita, C.; Sakurai, Y.; Yasuda, Y.; Takada, Y.; Huang, C.-L.; Fujita, M. Anti-Monomeric C-Reactive Protein Antibody Ameliorates Arthritis and Nephritis in Mice. J. Immunol. 2021, 207, 1755–1762. [Google Scholar] [CrossRef]
Figure 1. Proposed roles for mCRP in atherosclerosis. mCRP, monomeric C-reactive protein; pCRP, pentameric C-reactive protein; TF, tissue factor; CAM, cell-adhesion molecules; oxLDL, oxidized low-density lipoproteins.
Figure 1. Proposed roles for mCRP in atherosclerosis. mCRP, monomeric C-reactive protein; pCRP, pentameric C-reactive protein; TF, tissue factor; CAM, cell-adhesion molecules; oxLDL, oxidized low-density lipoproteins.
Ijms 24 02079 g001
Figure 2. Anti-inflammatory agents targeting the NLRP3 inflammasome and IL-1β/IL-6/CRP axis. Upstream agents targeting the NLRP3 inflammasome, interleukin (IL)-1β, and IL-6 reduce the rate of major adverse cardiovascular events, according to large RCTs. Downstream agents target translation of pCRP in the liver, CRP dissociation, and mCRP. Studies on these agents are limited to animal models and small RCTs. CRP, C-reactive protein; mCRP, monomeric C-reactive protein; pCRP, pentameric C-reactive protein; RCTs, randomized clinical trials.
Figure 2. Anti-inflammatory agents targeting the NLRP3 inflammasome and IL-1β/IL-6/CRP axis. Upstream agents targeting the NLRP3 inflammasome, interleukin (IL)-1β, and IL-6 reduce the rate of major adverse cardiovascular events, according to large RCTs. Downstream agents target translation of pCRP in the liver, CRP dissociation, and mCRP. Studies on these agents are limited to animal models and small RCTs. CRP, C-reactive protein; mCRP, monomeric C-reactive protein; pCRP, pentameric C-reactive protein; RCTs, randomized clinical trials.
Ijms 24 02079 g002
Table 1. Monomeric C-reactive protein assays.
Table 1. Monomeric C-reactive protein assays.
Assay TypeAnti-mCRP AntibodyBlood-Specimen
Type
Reported mCRP LevelsCorrelation (mCRP/
hsCRP)
Reference
ELISAUnspecified mAbHeparinized plasma20.96 ± 1.64 μg/L in acute myocardial infarction
0.0 μg/L in angina pectoris, unstable angina, and non-CAD patients
n/aWang et al., 2015 [109]
ELISACRP-8Plasma
(unspecified anticoagulant)
59.8 (44.5; 79.1) μg/L in urticaria
35.4 (17.0; 48.6) μg/L in psoriasis
30.0 (4.77; 36.3) in eczema
15.2 (5.05; 27.1) μg/L in healthy participants
n/aZhang et al., 2018 [110]
ELISACRP-8Serum
purified by chromatography
1.03 ± 0.11 mg/L in patients with hsCRP > 100 mg/LNoneWilliams
et al., 2020 [111]
ELISAUnspecified mAbEDTA
plasma
244.1 (226.1; 331.7) mg/L in AAV
170.0 (135.7; 199.3) mg/L in control participants
NegativeWu et al., 2020 [112]
ELISAAptamerSerum0.66 (0.38; 1.03) mg/L in COPD
0.0 (0.0; 0.29) mg/L in non-COPD patients
NoneMunuswamy et al., 2021 [113]
ELISACRP-8Plasma
(unspecified anticoagulant)
12.5 (7.8; 24.8) μg/L in osteoarthritis
5.0 (3.5; 9.8) μg/L in healthy participants
PositiveLiang et al., 2022 [114]
CBACRP-8Citrated
plasma
5.2 (3.3; 7.1) μg/L in patients with
subclinical carotid atherosclerosis
NoneMelnikov
et al., 2022 [115]
ELISA8C10Serum3.7 (1.3; 7.4) μg/L in systemic lupus erythematosus
11 (5.8; 22) μg/L in AAV patients
NoneKarlsson et al., 2022 [116]
ELISA12CEDTA
plasma
477 (100; 2570) μg/L in adult-onset Still’s disease
186 (0.035; 626) μg/L in polymyalgia rheumatica
77 (0.035; 501) μg/L in rheumatoid arthritis
228 (0.035; 1086) μg/L in acute infections
1.2(0.035; 10) μg/L in healthy participants
NoneFujita et al., 2022 [117]
ELISA, enzyme-linked immunosorbent assay; CBA, cytometric bead array; mAb, monoclonal antibody; mCRP, monomeric C-reactive protein; hsCRP, pentameric C-reactive protein measured using high-sensitivity assay; EDTA, ethylenediaminetetraacetic acid; CAD, coronary artery disease; AAV, anti-neutrophil cytoplasmic antibody-associated vasculitis; COPD, chronic obstructive pulmonary disease; n/a, not assessed.
Table 2. CRP-targeting agents.
Table 2. CRP-targeting agents.
TargetAgentStage of DevelopmentMain Findings
CRP
translation
ISIS 280290Animal modelsNo antiatherogenic effect was shown in a rabbit model of atherosclerosis.
ISIS 329993Phase II
clinical trials
No benefit was shown in atrial fibrillation or rheumatoid arthritis treatment.
CRP
dissociation
1,6-bisPCAnimal modelsMyocardial damage and renal damage were reduced in rat models of myocardial and renal IRI, respectively.
C10MAnimal modelsRenal damage was reduced and allograft rejection prevented in rat models of renal IRI and hindlimb transplantation, respectively.
mCRPmAb C3Animal modelsJoint damage and renal damage were reduced in murine models of rheumatoid arthritis and lupus nephritis, respectively.
CRP, C-reactive protein; mCRP, monomeric C-reactive protein; 1,6-bisPC, 1,6-bis(phosphocholine)-hexane; mAb, monoclonal antibody; IRI, ischemia/reperfusion injury.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Melnikov, I.; Kozlov, S.; Saburova, O.; Avtaeva, Y.; Guria, K.; Gabbasov, Z. Monomeric C-Reactive Protein in Atherosclerotic Cardiovascular Disease: Advances and Perspectives. Int. J. Mol. Sci. 2023, 24, 2079. https://doi.org/10.3390/ijms24032079

AMA Style

Melnikov I, Kozlov S, Saburova O, Avtaeva Y, Guria K, Gabbasov Z. Monomeric C-Reactive Protein in Atherosclerotic Cardiovascular Disease: Advances and Perspectives. International Journal of Molecular Sciences. 2023; 24(3):2079. https://doi.org/10.3390/ijms24032079

Chicago/Turabian Style

Melnikov, Ivan, Sergey Kozlov, Olga Saburova, Yuliya Avtaeva, Konstantin Guria, and Zufar Gabbasov. 2023. "Monomeric C-Reactive Protein in Atherosclerotic Cardiovascular Disease: Advances and Perspectives" International Journal of Molecular Sciences 24, no. 3: 2079. https://doi.org/10.3390/ijms24032079

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop