v
Search
Advanced Search

Publications > Journals > Gene Expression > Article Full Text

  • OPEN ACCESS

Proatherogenic Disorders of Blood Lipid and Lipoprotein Metabolism in Patients with Rheumatoid Arthritis

  • Elena V. Gerasimova1,* ,
  • Tatiana V. Popkova1,
  • Maria V. Shalygina1 and
  • Daria A. Gerasimova2
 Author information
Gene Expression   2024

doi: 10.14218/GE.2024.00036

Abstract

Disorders of blood lipids and lipoproteins are a global problem and a high-risk factor for atherosclerosis in patients with rheumatoid arthritis (RA). This article presents data on the influence of inflammation on proatherogenic disorders of lipid and lipoprotein metabolism, with an emphasis on proinflammatory cytokines. It analyzes the blood lipid profile in RA patients and identifies the need to study subfractions of high-density lipoproteins and their function in reverse cholesterol transport in RA patients as a more promising direction for clarifying cardiovascular risk. Depending on their type and metabolites, lipids may either promote disease progression or protect against RA. Supported by the close connection between altered lipid metabolism and chronic autoimmune inflammation, specific lipid profiles are emerging as unique disease biomarkers with diagnostic, predictive, and prognostic potential. Studying the influence of the immunoinflammatory process on lipids and lipoproteins in the blood of patients with RA will not only deepen knowledge about the pathogenesis of chronic inflammation but also expand understanding of the pathogenetic and prognostic significance of lipids, allowing for early diagnosis of dyslipidemia in RA at a qualitatively new level.

Keywords

Lipid, Lipoprotein, Cholesterol, Cholesterol efflux capacity, Proinflammatory cytokines, Rheumatoid arthritis

Introduction

In chronic inflammation, lipid abnormalities are important components of atherogenesis that determine the progression and clinical manifestations of atherosclerosis.1,2

During the acute phase of inflammation associated with trauma or infection, there are marked changes in the composition and concentration of plasma lipids and lipoproteins.3 Early studies have shown that experimental modeling of systemic inflammatory processes leads to changes in lipid metabolism, characterized by hypertriglyceridemia, increased levels of free fatty acids (FA), and decreased levels of low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C).4 Hypertriglyceridemia associated with acute-phase inflammatory proteins develops due to increased formation and decreased clearance of very-low-density lipoproteins.5 Reduction in lipoprotein lipase (LPL) and hepatic lipase activity also contributes to hypertriglyceridemia and suppresses the formation of a specific HDL subclass associated with cholesterol efflux capacity (CEC).6 Furthermore, significant changes occur in the protein and lipid composition of lipoproteins, which may not only redefine their function but also increase their atherogenic and inflammatory properties.7

Dyslipidemia often occurs in autoimmune diseases such as rheumatoid arthritis (RA), type 1 diabetes, psoriasis, inflammatory bowel disease, and other diseases. An imbalance in lipid metabolism accelerates inflammatory reactions and contributes to the development of atherosclerosis. While there have been numerous studies on the relationship between abnormal lipid metabolism and RA, and evidence suggests that lipid abnormalities occur in the preclinical stages of RA, it remains unclear whether dyslipidemia plays a unique role in the onset and progression of RA.

The purpose of this review is to present the latest data on the effect of inflammation on proatherogenic disorders of lipid and lipoprotein metabolism, with an emphasis on proinflammatory cytokines. It aimed to analyze lipid and lipidomic blood profiles based on disease phase, inflammation activity, and research methods. The review identifies the necessity to study high-density lipoprotein subfractions and their role in reverse cholesterol transport in RA patients as a promising direction for clarifying cardiovascular risk.

Effects of proinflammatory cytokines on lipid and lipoprotein metabolism

Numerous studies have shown that inflammation and inflammatory cytokines are closely related to lipid metabolism. Pro-inflammatory cytokines are involved in the regulation of adipocyte proliferation and apoptosis by stimulating lipolysis, inhibiting lipid synthesis, and reducing blood lipid concentration. Under the influence of pro-inflammatory cytokines during the inflammatory process, structural modifications and a decrease in HDL-C concentration occur, which are associated with the reduction of CEC proteins.8,9

Key cytokines affecting lipid metabolism include tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and IL-1β. The most important source of pro-inflammatory cytokines is adipose tissue, consisting mainly of adipocytes, macrophages, and endothelial cells.10 Increased TNF-α levels in adipose tissue are associated with increased lipolysis, disruption of insulin action on glucose transport, decreased LPL expression, inhibition of fat cell recruitment, and induction of apoptosis in fat cells.4

Several studies have demonstrated a negative effect of TNF-α on lipid metabolism.11,12 TNF-α affects LPL reduction and triglyceride (TG) hydrolysis and enhances lipid mobilization from fat depots, which probably causes proatherogenic changes in the blood lipid profile.13 In parallel, TNF-α stimulates the production of LDL in the liver, leading to the development of hypertriglyceridemia, which modulates the movement of cholesterol esters from HDL to LDL in exchange for TG mediated by cholesteryl ester transfer protein (CETP). Researchers found that lipolysis is mediated by TNF-α through the regulation of lipid droplet-associated perilipin protein expression, hormone-sensitive adipose tissue lipase, and triglyceride lipase.14,15 It has been suggested that the main physiological goal of increased TNF-α expression may be to limit adipose tissue overgrowth and subsequently induce insulin resistance and further disorders of glucose and lipid metabolism.16

The central inflammatory mediator IL-6 has a similar effect. IL-6 is known to be produced by visceral adipose tissue cells, including adipocytes and resident immune cells, primarily macrophages.17,18 In obesity, the number of macrophages significantly increases, and adipocyte hypertrophy is accompanied by increased IL-6 production.19

Administration of recombinant human IL-6 to rhesus macaques resulted in changes in lipid metabolism, including a reduction in apolipoprotein (apo) A1, apoA2, and apoB levels, a decrease in HDL-C and LDL-C concentrations, and an increase in TG.20 Under the influence of IL-6, the ratio of proatherogenic and antiatherogenic lipids, lipoproteins, and their protein components (apoB/apoA1) is disturbed, which obviously increases cardiovascular risk.21

It is important to note that overexpression of IL-6 and its receptors is found in areas of the vascular bed that are more exposed to atherosclerotic lesions (coronary, brachiocephalic, and peripheral arteries).22,23 Cytokine involvement and the protective function of IL-6 receptor inhibitors have been demonstrated in ischemia-reperfusion injury of human cardiac myocytes.24

There was a marked increase in plasma concentrations of IL-1, IL-6, IL-8, IL-10, and TNF-α receptor antagonists in men after marathon running.25 The increase in plasma concentrations of free FA, IL-6, and TNF-α during marathon running may be related to the release of angiopoietin-related protein 4. This mechanism is considered compensatory against lipotoxicity and oxidative stress.26 IL-6 and TNF-α can stimulate adipose tissue lipolysis during prolonged exercise independently of catecholamines and other factors.

The effects of pro-inflammatory cytokines on lipid metabolism may be mediated through increased formation of acute-phase proteins.27 Inflammation primarily increases the production of C-reactive protein (CRP), synthesized under the influence of IL-6, IL-1β, and TNF-α in liver and adipose tissue cells.28

The native circulating form of CRP is the pentameric form, which is released into the bloodstream after exposure to inflammatory stimuli.29 Native CRP is able to bind specifically to LDL when the pentameric structure of CRP changes to a monomeric structure or when LDL is modified/oxidized.30,31 Several in vitro studies have convincingly demonstrated CRP-mediated opsonization of LDL with phagocytic cells through interaction with the immunoglobulin receptors, Fc gamma receptors I and II.32–34

In earlier studies, native CRP was shown to induce the release of proinflammatory cytokines from endothelial cells (vascular cellular adhesion molecule-1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and E-selectin) and monocytes (Monocyte chemoattractant protein-1), endothelial dysfunction, and monocyte adhesion to the endothelium.35 The negative effect of CRP on the vascular wall has been confirmed by studies of its interaction with angiotensin II and its effects on the integrity of the endothelial glycocalyx,36,37 as well as on increased expression of the lectin-like oxidized low-density lipoprotein receptor-1 involved in the damaging effect of oxidized low-density lipoproteins (ox-LDL).38 Moreover, LDL-associated CRP deposits have been detected in foci of atherosclerotic lesions and myocardial damage induced by ischemia/reperfusion in experimental animals and humans.39 These observations served as a basis to consider CRP as an active participant in atherosclerosis. Further study of the structural and functional relations of CRP in vitro and in vivo, and clearer diagnostics with separation of pentameric and monomeric forms of CRP, led to an understanding of the atheroprotective role of CRP.33,40

On the surface of activated platelets, apoptotic cells, and circulating cell-derived microparticles, upon ligand binding or under conditions of high oxidative potential of the extracellular environment, pentameric CRP irreversibly dissociates into insoluble monomers that have different pathophysiological functions from pentameric CRP.41,42

Monomeric CRP is thought to be involved in innate immunity processes through activation of the complement cascade,43 angiogenesis, and atherothrombosis.44 Pentameric CRP is not involved in thrombogenesis, whereas monomeric CRP induces platelet activation and thrombus growth. Additionally, pentameric and monomeric CRP have different pathways for stimulating endothelial cells and neutrophils, and different ways of binding to ligands, including LDL and C1q.33,40

The atheroprotective role of monomeric CRP is evidenced by its ability to retard foam cell formation by suppressing the aggressive response of macrophages to ox-LDL and to remove native LDL from the extracellular space, reducing the risk of modifications.45,46 Moreover, monomeric CRP reduces the proatherogenic effects of macrophages in binding to lysophosphatidylcholine in ox-LDL and inhibits the association of ox-LDL with macrophages. These effects may partially slow the progression of atherosclerosis.47

Thus, CRP has conflicting proangiogenic and antiangiogenic effects that establish tissue remodeling in an atherosclerotic plaque (ASP) and in infarct-affected tissues.33

Myeloperoxidase (MPO) is considered a key element in oxidative damage to lipoproteins. MPO is a lysosomal enzyme that catalyzes the oxidation of various substrates, with hydrogen peroxide as a co-substrate. The specific oxidation products produced by MPO are found in ASP in large quantities.48

The products of MPO-catalyzed reactions are strong oxidants that initiate lipid peroxidation and cause protein modification.49 The enzyme increases the expression of P-selectin on the platelet surface and significantly raises the formation of oxygen radicals by platelets.50

MPO is thought to be a potent producer of oxidized LDL in vivo. Oxidized LDL accumulates in macrophages via class A1 scavenger receptors (SR) and SR-B1.51 Another target of MPO is apoA1 HDL; by oxidizing this lipoprotein, MPO reduces its atheroprotective functions.52 Modified HDLs are less effective in stimulating cholesterol efflux and are rapidly degraded by macrophages. Macrophages continue to engulf modified lipoproteins, and large amounts of lipids accumulate intracellularly, resulting in foam cell formation. Foam cells, in turn, have a pro-inflammatory effect by producing cytokines, chemokines, and growth factors, and by stimulating the secretion of adhesive molecules.53

Thus, systemic inflammation plays an important role in the development of disorders related to the blood cholesterol transport system. Data from numerous studies indicate the influence of cytokines (TNF-α, IL-6) on the proatherogenic potential of blood lipids and lipoproteins, supporting the concept of the contribution of chronic inflammation to the development of atherosclerosis.

Immunomodulatory effects of HDL

In recent years, there has been increasing evidence for the immunomodulatory properties of HDL. The determination of pro- or anti-inflammatory properties of HDL depends on the lipoprotein composition, the cholesterol content in macrophages, and the leading signaling pathways.54,55

HDLs undergo continuous remodeling processes in circulation, characterized by a wide variety of subtypes differing in size, density, shape, charge, and composition. This allows researchers to distinguish dysfunctional or even “proatherogenic” particles from “antiatherogenic” HDL.56 The inflammatory process helps to reduce HDL levels and eliminate cholesterol from cells, leading to the production of proinflammatory HDL.57,58 Proteins and enzymes responsible for the proinflammatory activity of HDL have been identified. The content of acute-phase proteins (such as serum amyloid A, fibrinogen, haptoglobin, apoJ, and complement factors B, C3, and C9) is elevated in proinflammatory HDL complexes compared to normal HDL.59 In chronic inflammation, elevated concentrations of amyloid A, apoJ, and pancreatic phospholipase A2 are found in serum and can displace the usual HDL components (apoA1, HDL, etc.). Moreover, changes in the acetylhydrolase activity of HDL enzymes were observed: a decrease in paraoxonase 1 (PON1) and an increase in platelet-activating factor acetylhydrolase.56 Together, these changes contribute to the proatherogenic potential of HDL.

On the other hand, HDLs inhibit the production of pro-inflammatory cytokines and chemokines, which may ultimately prevent immune cell activation and reduce inflammation.60 The ability of HDL to inhibit the expression of ICAM-1, VCAM-1, and E-selectin has been demonstrated in endothelial cells.61,62

HDL and apoA1 can suppress inflammation by binding directly to lipopolysaccharide or lipoteichoic acid, thereby neutralizing them.63,64 Several studies have clarified the immunomodulatory activity of HDL occurring without neutralization of lipopolysaccharide. HDL can block the ability of serum amyloid A to induce the production of reactive oxygen species and activate the NOD-like receptor protein 3 inflammasome involved in the formation of the active forms of IL-1β and IL-18.65 Pretreatment of human endothelial cells with HDL reduced TNF-α-induced expression of ICAM-1, VCAM-1, and E-selectin.66,67 Taborda et al.67 studied the ability of HDL to mediate immune-inhibitory effects on innate immunostimulatory compounds via Toll-like receptors and inflammasomes without affecting the expression of pattern recognition receptors. These results suggest that HDL may modulate the immune response induced by various stimuli, thereby influencing the inflammatory response.68

The comparative characterization of normal and proinflammatory HDL is presented in Table 1.

Table 1

Comparative characteristics of normal and pro-inflammatory HDL

EffectsAnti-inflammatory (normal) HDLPro-inflammatory HDL
InflammatoryApo AI reflects the antiatherogenic activity of lipoproteins and suppresses the atherogenic activity of ox-LDL; TNF-α regulation in macrophages; Prevention of EC IL-8, MCP-1 expressionPromote the oxidation of LDL; The synthesis of enzymes decreases under the influence of inflammation; Serum amyloid A, ceruloplasmin replace the usual components of HDL
Anti-oxidant propertiesPON1 prevents the formation of oxidation products; Endothelial protection; Upregulation of EC and NO production; EC express receptors for apoA-I and HDLThe connection between PON1 and altered apoA1 is disrupted; PAPC interacts with the oxidation products of arachidonic acid to form biologically active oxidation products
Reverse cholesterol transportApoA1 and apoJ in HDL transport cholesterol from arterial vascular cells and macrophages to other lipids and liver cells for processing and disposalThe functions of apoA1 and apoJ are impaired by oxidative stress; Lipoprotein synthesis is reduced under the influence of inflammation
Platelet responsesBinding of mildly and native ox-LDL to apoER2′ and SR-BI on the platelet membrane induces signaling pathways that suppress platelet activationPlatelet activation through binding via CD36 to heavily oxidized HDL

In recent years, understanding of the mechanism of cholesterol transfer from biomembranes to HDL has greatly advanced through studies of plasma factors and cellular proteins in mouse models.69 In 2022, researchers identified previously unknown membrane proteins associated with the regulation of lipoprotein metabolism and the cell’s ability to release membrane cholesterol, thereby clarifying mechanisms of reverse cholesterol transport.68,70 HDL directly transports cholesterol to SR-B1 receptors expressed in the liver. Alternatively, cholesterol from HDL transfers to LDL and very-low-density lipoproteins via CETP, then returns to the liver through LDL receptors, where it is metabolized into bile acids and excreted into the intestinal tract. Cholesterol exported from cells is transferred to large spherical HDL by the ATP-binding cholesterol transporter (ABC) G1. A related transporter, ABCA1, delivers cellular cholesterol to apoA1 and small dense HDL.68

The importance of the reverse cholesterol transport system has become evident from large cohort studies in humans, which consistently show an inverse association between the ability of apoB-containing lipoprotein-depleted plasma to uptake macrophage-derived cholesterol and cardiovascular disease (CVD) risk.71,72 In healthy subjects with varying HDL-C and apoA1 levels, the ability of HDL to CEC from macrophages in vitro correlates inversely with measures of carotid intima-media thickness, independent of HDL-C and apoA1 concentrations.73

However, even with efficient acceptors, there may be factors limiting cholesterol efflux from membranes, potentially contributing to various proatherogenic abnormalities.74–76

The ability of serum to induce ABCA1-mediated CEC was reduced in patients with type 2 diabetes mellitus with microalbuminemia and proteinuria. The authors attributed this impaired CEC to low levels of preβ-1 HDL.77 Additionally, depletion of preβ-1 HDL induced by chymase (a mast cell-secreted proteinase) therapy impaired ABCA1-mediated but not SR-B1-mediated, cholesterol efflux from J774 macrophages.78 Specific genomic variants of lipoprotein receptors, apoB-100, Proprotein Convertase Subtilisin/Kexin type 9, and other loci may contribute to lipid metabolism abnormalities complicating vascular disorders in type 2 diabetes mellitus.79,80

Disorders of blood lipid and lipoprotein metabolism in patients with rheumatoid arthritis

RA is an autoimmune disease of unknown etiology characterized by chronic erosive arthritis with a progressive course, leading to serious complications and severe comorbidity. RA is associated with early disability and reduced life expectancy among patients.81 CVDs play a significant role in the development of adverse outcomes in RA.82 Numerous studies have shown that the relative risk of cardiovascular events in RA patients ranges from 1.4 to 3.7.83 In a recent large cohort study, mortality rates remained high among RA patients (n = 16,047) observed from 2005 to 2018: the relative risk for death from all causes was 1.22 (95% confidence interval (CI) 1.15–1.30), from CVD - 1.63 (95% CI 1.51–1.75), from myocardial infarction - 2.00 (95% CI 1.78–2.26), and from stroke - 1.39 (95% CI 1.22–1.58). The risk of CVD in RA patients increases from the early years after diagnosis and does not decrease over time.84

The main mechanism leading to the development of CVD in RA is atherosclerotic vascular damage.85 The multifactorial pathogenesis of atherosclerosis involves vascular, metabolic, and inflammatory components.86 Disorders of blood lipid and lipoprotein metabolism are implicated in all three components and play a direct role in the formation of atherosclerotic vascular lesions in RA.87,88 According to meta-analyses, patients with RA and hypercholesterolemia have a 73% higher incidence of CVD compared to patients without it.87

Blood lipid profile of RA patients

Inflammation in RA occurs with increased catabolism of lipoproteins, often leading to decreased serum levels of HDL and LDL. Despite this so-called “lipid paradox,” patients remain at high risk of CVD.89,90 Many researchers have observed lower concentrations of total cholesterol, LDL-C, and HDL-C in patients with active, untreated RA compared to healthy controls and patients with osteoarthritis.91–96 In a prospective study, VanEvery et al.97 have found an association between low LDL-C concentrations and RA risk. Other authors suggest that early-stage RA is associated with normal or reduced serum TG and total cholesterol levels, which, combined with a more pronounced reduction in HDL-C, may increase atherogenic potential.98 Additionally, smaller, more atherogenic lipoprotein phenotypes have been detected in early RA patients compared to controls.94 The formation of small dense LDL particles has been attributed to the acute phase reaction.99

In recent years, the atherogenic index of plasma (AIP), the logarithm of the ratio of TG/HDL-C, has been well-established as a predictor of coronary heart disease in the general population and in type 2 diabetes mellitus.100–103 Hammam et al.104 found associations between AIP and 10-year CVD risk as determined by the Framingham scale in patients with RA and systemic lupus erythematosus (SLE). The potential role of AIP in predicting and managing CVD risk in autoimmune rheumatic diseases has been investigated in several other studies.105–108

Similar data were obtained when analyzing the blood lipid spectrum in patients with untreated early RA included in the REMARCA (Russian invEstigation of MethotrexAte and biologicals in eaRly aCtive inflammatory Arthritis) trial. Decreased total cholesterol and HDL-C levels, along with increased AIP, were found in RA patients compared to healthy controls (Table 2).

Table 2

Blood lipid spectrum in patients with early RA and in control (REMARCA trial), Me [25th; 75th percentile]

Blood lipidsRA (n = 154)Сontrol (n = 104)p
Total cholesterol, mmol/L5.22 [4.63; 5.98]5.88 [5.45; 6.63]0.036
LDL-C, mmol/L3.41 [2.84; 4.14]3.78 [3.53; 4.26]0.01
HDL-C, mmol/L1.32 [1.03; 1.62]1.73 [1.43; 1.97]0.023
TG, mmol/L1.13 [0.86; 1.63]1.03 [0.85; 1.39]0.12
AIP (TG/HDL-C)1.1 (0.6; 1.9)0.6 (0.4; 1.5)<0.001

In contrast, other studies have found higher levels of total cholesterol, LDL-C, TG, and, to a lesser extent, low HDL-C, resulting in significantly higher AIP in patients with early RA compared to healthy individuals.109,110 It is important to note that total cholesterol and TG levels increase even before the clinical manifestations of RA, and the subsequent development of chronic inflammation can explain the decrease in these indicators.98

Lipidomics has shown that serum lipid profiles in patients with active RA are similar to those of patients with preclinical RA, confirming alterations in lipid metabolism before disease onset.111,112 In a prospective cohort study of anti-citrullinated protein antibody (ACPA) positive individuals, higher levels of monohydroxyeicosatetraenoic acids (5-HETE, an important precursor to leukotrienes) appeared to be associated with the subsequent development of inflammatory arthritis.111 Other predictors of RA development in ACPA-positive individuals were decreased levels of apoA1 and ω-3 FA.113,114 A body of research suggests a role for polyunsaturated FA metabolites in preclinical RA. However, the mechanism of their action is still poorly understood.115,116

A recent study by Koh et al.117 demonstrated a marked shift in the lipidome profile in the synovial fluid of RA patients, correlating with disease activity and the degree of synovitis on ultrasound. In particular, lipid subclasses were decreased in lysophosphatidylcholine (LPC) and increased in phosphatidylcholine, its ester, triacylglycerol, and sphingomyelin. Similar changes, although less pronounced, were found in the serum lipidome profile. It is believed that the immunomodulatory effect of LPC depends on its biochemical structure: saturated and monounsaturated LPC species have a pro-inflammatory effect, whereas polyunsaturated species have an anti-inflammatory effect.118

Additionally, depletion of short-chain acylcarnitine was detected in the serum of women predisposed to RA.119 The blood lipid spectrum in patients with active RA and high laboratory activity (CRP and erythrocyte sedimentation rate (ESR) levels) was similar to the lipid profile in patients with preclinical RA and normal CRP/ESR levels. This confirms the occurrence of dyslipidemia at the preclinical stage of RA.119

The relationship of lipid abnormalities with RA activity and acute-phase blood indices is also indicated in several other studies. Boers et al.120 found the lowest levels of LDL-C and HDL-C in high RA activity. An association was found between levels of total cholesterol, CRP, and DAS28 activity index.121 Park et al.,122 in their study of lipid disorders in untreated RA patients, revealed a negative association of HDL-C and apoA1 concentrations with CRP. In the work of White et al.,123 they confirmed the relationship between low HDL-C and high CRP/ESR levels. According to other data, lower serum HDL-C and apoA1 levels were observed in RA patients not only with high CRP levels but also with RF positivity and hand synovitis.124,125

During inflammation, lipoprotein (a) (Lp(a)) synthesis and expression increase, similar to acute-phase proteins.126 Lp(a) is a cholesterol-rich plasma lipoprotein subclass consisting of LDL particles attached to apoA. In RA patients, blood Lp(a) concentration and apoB/apoA1 ratio were significantly higher than in the control group.122,124 Higher levels of Lp(a), proinflammatory, and proatherogenic modified LDL associated with RA are independent risk factors for CVD.127

The comparative characterization of serum blood lipids in RA patients and healthy controls is presented in Table 3.91-93,96,99,109,110,112,122-125,128-139

Table 3

Lipid spectrum of the blood of rheumatoid arthritis (RA) patients, healthy controls (HC)

ResearchersYearNumber of RA patients/HCResults
Lazarevic et al.128199269/65↓ LDL-C, TC, HDL-C, more pronounced in patients with high RA activity
Park et al.122199942/42↓ HDL-C и apoА1, ↑ Lp(а) and AI; correlations between HDL-C and CRP (r = −0.35), apoA1 and CRP (r = −0.44)
Hurt-Camejo et al.99200131/28TC, LDL-C, TG, HDL-C, apoB, apoA1 did not change. ↑ subfractions LDL1, ↓ HDL
Kim et al.91200454/50↓ TC, LDL-C
Choi et al.1252005104/NHANES (>60 лет)↓ HDL-C and apoA1; inverse correlations HDL-C levels with CRP, RF, and synovitis of the hands
White et al.1232006204/75↑ LDL-C; ↓ HDL-C; ↑ AI; negative correlations HDL-C and CRP, HDL-C and ESR and positive – LDL-C and ESR
Georgiadis et al.109200658/63↑ TC, LDL-С, TG; ↓ HDL-С; ↑ AI
Popkova et al.129200784/15↑ TG, apoА1; ↓ LDL-С; TC and HDL-С did not differ
Gonzalez et al.1302008603/603TC, HDL-C, LDL-C, TG, apo did not change
Myasoedova et al.922010577/540↓ TC, LDL-С: 5 years before the development of RA
Boyer et al.13120112,956/3,713↓ HDL-С
Liao et al.9320132,005/NHANES↓ TC and LDL-С
Pozzi et al.132201530/30TC, LDL-C, TG, apoA1 did not differ, ↑ HDL-C, apoЕ, ↓ apoВ
Chavan et al.110201550/50↑ TC, LDL-C; ↓ HDL-C
Govindan et al.124201530/30↓ HDL-C and apoA1, ↑ AI
Kim et al.133201633/13↓ HDL-C, ↑TG, СЕТР, AGEs, apoА1
Ormseth et al.1342016169/92↓ LDL-C; CEC did not differ
Rodríguez-Carrio et al.1352017113/113↑ HDL-C, TG, ↓PON1; TGhighHDLlow profile did not differ and was associated with increased TNF-α, MCP-1, leptin, and INF- γ-inducible protein-10 (p < 0.05 in all cases)
Tejera-Segura et al.962017178/223↓ HDL-C, LDL-C, apoА; ↑ Lp(a). In patients with RA activity, a decrease in CEC was detected, in contrast to patients with remission.
Burggraaf et al.1362018312/57No differences were detected in the lipid profile; ↑ аpoAI, аpoB48. ApoB48 correlated with TG (r = 0.645, p < 0.001)
Dessie et al.137202073/40↑ TC, TG; ↑TC/HDL and LDL/HDL ratios; ↓ HDL-C; LDL-C did not differ. HDL-C negative correlation with CRP, and TC/HDL-C and LDL/HDL-C had а direct association with CRP (p < 0.05).
Alisik et al.1382021130/67No differences were detected in the lipid profile; ↑ MPO/PON1 ratios in RA patients, especially high with CVD history. MPO/PON1 ratios were significant positive correlated with DAS28 scores (r = 0.357, p < 0.001)
Rodríguez-Carrio et al.112202160/28In RA patients, 2 oxylipin clusters (cluster I and cluster II) were identified in contrast to the control group (p = 0.003).
Chang et al.139202380/15↑ particle number, cholesterol ester, free cholesterol, and phospholipids in large/very large-sized HDL; small-sized HDL negative correlation with CRP; ACPA-positivity was associated with HDL- metabolites.

In a study by Memon et al.,140 it was observed that experimentally induced infection increases the level of oxidized lipids in serum, which may induce LDL oxidation in vivo and be a mechanism leading to increased CVD in patients with chronic infections and inflammatory diseases. The results of several subsequent studies confirmed higher blood levels of ox-LDL in RA patients compared to healthy controls.141,142 LPC is known to be a major component of оx-LDL.143 Recently published research in RA patients demonstrated a possible atherogenic effect of IgG anti-ox-LDL antibodies.144,145 Specifically, it was shown that anti-ox-LDL titers (p = 0.020) may predict the presence of coronary ASP (noncalcified, partially calcified, and high-risk plaque) in RA patients with lower LDL-C levels (<1.8 mmol/L).144

The influence of pro-inflammatory cytokines on lipid metabolism and the pro-atherogenic potential of immune cells in RA is presented in Figure 1.

The influence of pro-inflammatory cytokines on lipid metabolism and the pro-atherogenic potential of immune cells in RA.
Fig. 1  The influence of pro-inflammatory cytokines on lipid metabolism and the pro-atherogenic potential of immune cells in RA.

Pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) are produced in large quantities locally in the joints and subsequently enter the bloodstream. TNF-α and IL-6 may promote LDL metabolism by increasing the expression of LDLR and SR-B1 on the surface of liver cells. Moreover, TNF-α and IL-1β can reduce the formation of pro-ApoA1 particles in the liver by suppressing HDL production. Presentation of antigenic peptides from ApoB by antigen-presenting cells (APCs) promotes the formation of effector T cells from naïve (CD4+) T helper cells. APCs oxidize LDL-C particles, process, and present peptides from ApoB on major histocompatibility complex molecules. The specific T cell receptor is responsible for recognizing antigen fragments in the form of peptides bound to major histocompatibility complex molecules. Under the influence of co-stimulatory signals and cytokines secreted by APCs, T cells express transcription factors that promote differentiation into distinct Th types, which produce specific cytokines that can act both atheroprotectively and proatherogenic. anti-apoA1, antibodies against apolipoprotein A1; anti-HDL-Ab, anti-HDL antibodies; APCs, antigen-presenting cells; CRP, C-reactive protein; HDL, high-density lipoproteins; ICAM-1, intercellular adhesion molecule 1; IL, interleukin; LDL-C, low-density lipoprotein cholesterol; LDLR, low density lipoprotein receptor; LPC, lysophosphatidylcholine; MHC, major histocompatibility complex; ox-LDL, oxidized low-density lipoproteins; RA, rheumatoid arthritis; SR, scavenger receptor; TCR, T-cell receptor; Th, T helper; TNF, tumor necrosis factor; VCAM-1, vascular cellular adhesion molecule-1.

The most negatively charged LDL subcomponent associated with atherosclerosis is L5.146 In RA, L5 may contribute to atherosclerosis by enhancing foam cell formation by macrophages and increasing the expression of M1 macrophage-associated markers (TNF-α, IL-6, and IL-8).147

Heterogeneity and functions of HDL

The first study on the determination of HDL subfractions in RA patients was carried out by Hurt-Camejo et al. in 2001.99 The authors found a decrease in HDL2 in RA patients compared to healthy individuals, while the levels of total cholesterol, LDL-C, HDL-C, TG, apoB, and apoA1 did not differ.99 In another study, lower levels of HDL2 and HDL3 were found in 45 patients with RA (especially women) compared to healthy controls.148 The study demonstrated a moderate effect of RA activity on HDL2 levels. HDL2 concentration was reduced by 0.06 mmol/L with increasing DAS28 adjusted for sex, age, RA duration, and glucocorticoid use.

The inflammatory process in RA may attenuate the potential anti-atherogenic effect of HDL, causing a decrease in HDL-C levels and a reduction in antioxidant capacity. This reduction in antioxidant capacity may reduce the ability to eliminate cholesterol from cells, potentially leading to proinflammatory properties of lipoproteins.57,58 Proinflammatory HDL has been detected in the blood of patients with CVD,149 RA, and SLE.150 Charles-Schoeman et al.151 showed that proinflammatory HDLs were more frequently found in RA patients than in healthy individuals (20% and 4%, respectively, p < 0.006). Age, disease activity, erosive process, smoking, and non-Caucasoid race were correlated with proinflammatory HDL.

Further examination of the composition of proinflammatory HDL in RA patients revealed elevated levels of acute-phase proteins and complement factors compared to normal HDL.59 Moreover, changes in the acetylhydrolase activity of HDL enzymes were observed: a decrease in PON1 and an increase in platelet-activating factor acetylhydrolase.

MPO may contribute to the atheroprotective effect of HDL in RA. MPO is an enzyme found in macrophages and neutrophils, acting as a mediator of inflammation and oxidative stress.152,153 Several studies have found increased MPO concentration and activity in RA patients compared to healthy controls and patients with other rheumatic diseases (osteoarthritis, ankylosing spondylitis, SLE, and Sjögren’s syndrome).154–157 The high inflammatory status of RA and markers of acute-phase response (CRP, ESR) were associated with increased MPO activity.154,156,158 MPO content in synovial fluid was higher in untreated RA patients compared to treated patients; moreover, in untreated patients, MPO concentration and activity correlated with IL-8 and IL-18 content.157

In a recent study by Alisik et al.,138 MPO levels were higher in RA patients with concomitant CVD compared to RA patients without CVD. According to receiver operating characteristic analysis, the ratio of MPO to PON1, which characterizes HDL dysfunction, appeared to be more associated with CVD in RA.

The best-studied effects of MPO on CVD include the formation of dysfunctional atherogenic lipoproteins, decreased nitric oxide availability, and endothelial dysfunction leading to vasoreactivity and ASP instability.159 These effects strongly suggest that MPO is directly involved in the pathophysiology of CVD in RA.160,161

Thus, inflammation and high RA activity may reduce antioxidant properties and contribute to the proatherogenic potential of HDL. However, there is no clear consensus on whether this mechanism affects CVD risk in RA.161

HDL becomes proatherogenic against the backdrop of inflammation due to decreased CEC from macrophages, which is an independent risk factor for CVD.96,162

The efficacy of CEC is known to correlate directly with the properties of plasma lipid transport proteins. Decreases in CETP mass and activity were found in RA patients compared to healthy subjects,96 with the lowest rates observed in RA patients on glucocorticoids.163

Studies have shown no difference or a decrease in CEC in RA patients compared with healthy donors.96,155,161,164,165 A meta-analysis confirmed no differences in HDL-C efflux capacity and concentration between patients with RA and controls.166

In a recent study including 195 SLE patients and 265 RA patients, more severe CEC impairment was noted in SLE patients compared to RA patients.167 Although both diseases share common features of inflammatory dyslipidemia, RA patients had lower HDL-C levels and higher apoB levels than SLE patients.

Importantly, patients with high RA activity (DAS28 > 5.1) had lower CEC than patients in remission.96,164 The association of high RA activity with suppression of CEC may be indicated by inverse correlations of cholesterol efflux with DAS28 (r = −0.39, р = 0.01) and ESR (r = −0.41, р < 0.001).164 Multivariate analysis revealed that smoking, diabetes, ESR, and glucocorticoid use were correlated with CEC. However, according to Ormseth et al.,161 cholesterol efflux was not associated with RA activity, the systemic inflammation index CRP, oxidative stress (urinary F2-isoprostanes), or insulin resistance measured by the HOMA index. The authors concluded that net cholesterol efflux capacity by HDL-enriched serum is not altered in patients with well-controlled RA.

Liao et al.165 studied the blood lipid spectrum and CEC in RA patients with CRP concentration ≥10 mg/L. After one year of therapy and a 23.5 mg/L decrease in CRP, patients showed a 7.2% increase in HDL-C (р = 0.02) and a 5.7% increase in CEC (р = 0.002). The decrease in CRP concentration was accompanied by an increase in apoA1 (r = 0.27, p = 0.01) and CEC (r = 0.24, р = 0.002).

When CEC mediated by SR-B1, ABCG1, and ABCA1 were studied using apoB-depleted serum, Ronda et al.168 found that only ABCG1-mediated CEC was lower in active RA patients compared with controls. The same authors later conducted a comparative study of CEC before and after 6 months of methotrexate as monotherapy and in combination with the TNF-α inhibitor adalimumab.169 Methotrexate monotherapy moderately increased SR-B1- and ABCG1-mediated CEC by 6% and 7%, respectively. Multidirectional associations of ABCG1-mediated CEC with serum HDL-C concentration (r = 0.18, p = 0.047) and DAS28 (r = −0.247, p = 0.018) were observed in RA patients after 6 months of combination therapy with methotrexate and a TNF-α inhibitor. The study found no correlation between CEC and inflammatory markers.

As noted by Ormseth & Stein in their review, there is no uniform data on the significance of impaired CEC in RA patients and the influence of disease activity or inflammation on it.161 A meta-analysis of HDL cholesterol efflux capacity and concentration in RA showed improvement in CEC with early administration of antirheumatic therapy and control of inflammation.166 However, the authors noted that the results should be interpreted with caution due to the heterogeneity of groups and methodologies in those studies.

CEC is considered a sensitive predictor of cardiovascular risk in the general population.170,171 Nevertheless, the association of CEC with CVD risk in RA appears to be ambiguous.

In a study by Vivekanandan-Giri et al.,155 no differences were found between RA patients with and without CVD. According to Ormseth et al.,134 an increase in net CEC by HDL-enriched serum was not significantly associated with a decrease in coronary calcium score (odds ratio (OR) = 0.78 [95% CI 0.51–1.19], p = 0.24), adjusting for age, sex, race, the Framingham cardiovascular risk scale, and the presence of diabetes. Another study demonstrated the association of high CEC with a low risk of carotid ASP development in RA patients (OR = 0.94 [95% CI 0.89–0.98], p = 0.015).96

Convincing data on the impact of rheumatoid inflammation on CEC and its contribution to the development of atherosclerosis and associated CVD were reported in 2023. Karpouzas et al.172 confirmed the inverse association of systemic inflammation measured by CRP and disease activity measured by DAS28-CRP with ABCA1-mediated CEC. ABCA1-mediated CEC was associated with less progression and fewer coronary ASPs in patients with low baseline and cumulative CRP levels, in patients not receiving glucocorticoids, and in patients on disease-modifying antirheumatic drug therapy. In contrast, ABCA1-mediated CEC was associated with accelerated development of coronary atherosclerosis in patients with baseline high CRP levels and in those receiving glucocorticoids but not methotrexate or biologic disease-modifying antirheumatic drugs.173

In another study by these authors,172 ABCG1-mediated CEC was inversely correlated with a large number (≥5) of carotid ASPs (adjusted OR 0.50 [95% CI 0.28–0.88]), numbers of partially calcified (rate ratio 0.71 [0.53–0.94]), and low-attenuation plaques (rate ratio 0.63 [0.43–0.91] per standard deviation increment). Low-attenuation plaques on coronary computed tomography angiography are known to be associated with ASP progression and instability.

The general form of atherothrombosis in RA is shown schematically in Figure 2.

LDL passes through the endothelium.
Fig. 2  LDL passes through the endothelium.

Under the influence of MPO, LDL is oxidized. Under the influence of macrophages, ox-LDL is taken up and turns into foam cells. Proinflammatory HDL shows marked changes in proteins (↑ SAA and MPO and ↓ apoA1 and PON1). This circumstance leads to the inability of HDL to block the expression of ICAM/VCAM on endothelial cells (1), remove cholesterol from foam cells (2), and exhibit its antioxidant ability (3). Ox-LDL triggers the proliferation and migration of smooth muscle cells. A conglomerate of foam cells and smooth muscle cells forms an atherosclerotic plaque. Apo, apolipoprotein; HDL, high-density lipoproteins; ICAM-1, intercellular adhesion molecule 1; IFN-α, interferon-alpha; IL, interleukin; LDL, low-density lipoproteins; MPO, myeloperoxidase; ox-LDL, oxidized low-density lipoproteins; PON1, paraoxonase-1; SAA, serum amyloid A; TNF-α, tumor necrosis factor alpha; VCAM-1, vascular cellular adhesion molecule-1.

Thus, in well-treated RA with controlled disease activity, ABCA1-CEC may exhibit atheroprotective actions. Conversely, in uncontrolled RA, inflammation promotes the development of proatherogenic properties of ABCA1-CEC.166,173

Conclusions

Interest in the study of lipids and lipoproteins in RA remains high. New data on the development of lipid abnormalities have been obtained in the preclinical phase of RA research. Studies have demonstrated differences in lipid and lipidome profiles depending on disease phase, inflammation activity, and study methods. Lipids may contribute to progression or exert a protective effect in RA depending on their type and metabolites. Inflammation or disease activity appears to have a deleterious effect on HDL-mediated reverse cholesterol transport activity. Effective treatment of inflammation by adequate control of RA activity can offset proatherogenic abnormalities in lipid and lipoprotein metabolism.

Declarations

Acknowledgement

None.

Funding

This work was supported by the Russian Science Foundation (Grant # 22-15-00199).

Conflict of interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Authors’ contributions

Study concept and design (EG, TP), acquisition of data (EG, MS), analysis and interpretation of data (EG, TP, DG), drafting of the manuscript (EG, MS, DG), critical revision of the manuscript for important intellectual content (TP, DG), administrative, technical, or material support (TP), and study supervision (TP). All authors have made significant contributions to this study and have approved the final manuscript.

References

  1. Wolf D, Ley K. Immunity and Inflammation in Atherosclerosis. Circ Res 2019;124(2):315-327 View Article PubMed/NCBI
  2. Getz GS, Reardon CA. Atherosclerosis: cell biology and lipoproteins. Curr Opin Lipidol 2020;31(5):286-290 View Article PubMed/NCBI
  3. Carpentier YA, Scruel O. Changes in the concentration and composition of plasma lipoproteins during the acute phase response. Curr Opin Clin Nutr Metab Care 2002;5(2):153-158 View Article PubMed/NCBI
  4. Read TE, Grunfeld C, Kumwenda ZL, Calhoun MC, Kane JP, Feingold KR, et al. Triglyceride-rich lipoproteins prevent septic death in rats. J Exp Med 1995;182(1):267-272 View Article PubMed/NCBI
  5. Hardardóttir I, Grünfeld C, Feingold KR. Effects of endotoxin and cytokines on lipid metabolism. Curr Opin Lipidol 1994;5(3):207-215 View Article PubMed/NCBI
  6. Li Y, Xu Y, Jadhav K, Zhu Y, Yin L, Zhang Y. Hepatic Forkhead Box Protein A3 Regulates ApoA-I (Apolipoprotein A-I) Expression, Cholesterol Efflux, and Atherogenesis. Arterioscler Thromb Vasc Biol 2019;39(8):1574-1587 View Article PubMed/NCBI
  7. Niisuke K, Kuklenyik Z, Horvath KV, Gardner MS, Toth CA, Asztalos BF. Composition-function analysis of HDL subpopulations: influence of lipid composition on particle functionality. J Lipid Res 2020;61(3):306-315 View Article PubMed/NCBI
  8. Tahir A, Martinez PJ, Ahmad F, Fisher-Hoch SP, McCormick J, Gay JL, et al. An evaluation of lipid profile and pro-inflammatory cytokines as determinants of cardiovascular disease in those with diabetes: a study on a Mexican American cohort. Sci Rep 2021;11(1):2435 View Article PubMed/NCBI
  9. McGillicuddy FC, de la Llera Moya M, Hinkle CC, Joshi MR, Chiquoine EH, Billheimer JT, et al. Inflammation impairs reverse cholesterol transport in vivo. Circulation 2009;119(8):1135-1145 View Article PubMed/NCBI
  10. Ren Y, Zhao H, Yin C, Lan X, Wu L, Du X, et al. Adipokines, Hepatokines and Myokines: Focus on Their Role and Molecular Mechanisms in Adipose Tissue Inflammation. Front Endocrinol (Lausanne) 2022;13:873699 View Article PubMed/NCBI
  11. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 1993;259(5091):87-91 View Article PubMed/NCBI
  12. Kapłon-Cieślicka A, Postuła M, Rosiak M, Peller M, Kondracka A, Serafin A, et al. Association of adipokines and inflammatory markers with lipid control in type 2 diabetes. Pol Arch Med Wewn 2015;125(6):414-423 View Article PubMed/NCBI
  13. Park S, Zhang T, Yue Y, Wu X. Effects of Bile Acid Modulation by Dietary Fat, Cholecystectomy, and Bile Acid Sequestrant on Energy, Glucose, and Lipid Metabolism and Gut Microbiota in Mice. Int J Mol Sci 2022;23(11):5935 View Article PubMed/NCBI
  14. Souza SC, de Vargas LM, Yamamoto MT, Lien P, Franciosa MD, Moss LG, et al. Overexpression of perilipin A and B blocks the ability of tumor necrosis factor alpha to increase lipolysis in 3T3-L1 adipocytes. J Biol Chem 1998;273(38):24665-24669 View Article PubMed/NCBI
  15. Kralisch S, Klein J, Lossner U, Bluher M, Paschke R, Stumvoll M, et al. Isoproterenol, TNFalpha, and insulin downregulate adipose triglyceride lipase in 3T3-L1 adipocytes. Mol Cell Endocrinol 2005;240(1-2):43-49 View Article PubMed/NCBI
  16. Hauner H. Secretory factors from human adipose tissue and their functional role. Proc Nutr Soc 2005;64(2):163-169 View Article PubMed/NCBI
  17. Päth G, Bornstein SR, Gurniak M, Chrousos GP, Scherbaum WA, Hauner H. Human breast adipocytes express interleukin-6 (IL-6) and its receptor system: increased IL-6 production by beta-adrenergic activation and effects of IL-6 on adipocyte function. J Clin Endocrinol Metab 2001;86(5):2281-2288 View Article PubMed/NCBI
  18. Tanaka T, Narazaki M, Kishimoto T. Interleukin (IL-6) Immunotherapy. Cold Spring Harb Perspect Biol 2018;10(8):a028456 View Article PubMed/NCBI
  19. Hoene M, Weigert C. The role of interleukin-6 in insulin resistance, body fat distribution and energy balance. Obes Rev 2008;9(1):20-29 View Article PubMed/NCBI
  20. Ettinger WH, Sun WH, Binkley N, Kouba E, Ershler W. Interleukin-6 causes hypocholesterolemia in middle-aged and old rhesus monkeys. J Gerontol A Biol Sci Med Sci 1995;50(3):M137-M140 View Article PubMed/NCBI
  21. Khovidhunkit W, Kim MS, Memon RA, Shigenaga JK, Moser AH, Feingold KR, et al. Effects of infection and inflammation on lipid and lipoprotein metabolism: mechanisms and consequences to the host. J Lipid Res 2004;45(7):1169-1196 View Article PubMed/NCBI
  22. Gotsman I, Stabholz A, Planer D, Pugatsch T, Lapidus L, Novikov Y, et al. Serum cytokine tumor necrosis factor-alpha and interleukin-6 associated with the severity of coronary artery disease: indicators of an active inflammatory burden?. Isr Med Assoc J 2008;10(7):494-498 PubMed/NCBI
  23. Ridker PM, Lüscher TF. Anti-inflammatory therapies for cardiovascular disease. Eur Heart J 2014;35(27):1782-1791 View Article PubMed/NCBI
  24. Cheng HF, Feng Y, Jiang DM, Tao KY, Kong MJ. Protective function of tocilizumab in human cardiac myocytes ischemia reperfusion injury. Asian Pac J Trop Med 2015;8(1):48-52 View Article PubMed/NCBI
  25. Bernecker C, Scherr J, Schinner S, Braun S, Scherbaum WA, Halle M. Evidence for an exercise induced increase of TNF-α and IL-6 in marathon runners. Scand J Med Sci Sports 2013;23(2):207-214 View Article PubMed/NCBI
  26. Górecka M, Krzemiński K, Mikulski T, Ziemba AW. ANGPTL4, IL-6 and TNF-α as regulators of lipid metabolism during a marathon run. Sci Rep 2022;12(1):19940 View Article PubMed/NCBI
  27. Zimmermann O, Li K, Zaczkiewicz M, Graf M, Liu Z, Torzewski J. C-reactive protein in human atherogenesis: facts and fiction. Mediators Inflamm 2014;2014:561428 View Article PubMed/NCBI
  28. Calabro P, Chang DW, Willerson JT, Yeh ET. Release of C-reactive protein in response to inflammatory cytokines by human adipocytes: linking obesity to vascular inflammation. J Am Coll Cardiol 2005;46(6):1112-1113 View Article PubMed/NCBI
  29. Pepys MB, Hirschfield GM. C-reactive protein: a critical update. J Clin Invest 2003;111(12):1805-1812 View Article PubMed/NCBI
  30. Wang MS, Black JC, Knowles MK, Reed SM. C-reactive protein (CRP) aptamer binds to monomeric but not pentameric form of CRP. Anal Bioanal Chem 2011;401(4):1309-1318 View Article PubMed/NCBI
  31. Fu T, Borensztajn J. Macrophage uptake of low-density lipoprotein bound to aggregated C-reactive protein: possible mechanism of foam-cell formation in atherosclerotic lesions. Biochem J 2002;366(Pt 1):195-201 View Article PubMed/NCBI
  32. Lu J, Marnell LL, Marjon KD, Mold C, Du Clos TW, Sun PD. Structural recognition and functional activation of FcgammaR by innate pentraxins. Nature 2008;456(7224):989-992 View Article PubMed/NCBI
  33. Boncler M, Wu Y, Watala C. The Multiple Faces of C-Reactive Protein-Physiological and Pathophysiological Implications in Cardiovascular Disease. Molecules 2019;24(11):2062 View Article PubMed/NCBI
  34. Lu J, Mold C, Du Clos TW, Sun PD. Pentraxins and Fc Receptor-Mediated Immune Responses. Front Immunol 2018;9:2607 View Article PubMed/NCBI
  35. Pathak A, Agrawal A. Evolution of C-Reactive Protein. Front Immunol 2019;10:943 View Article PubMed/NCBI
  36. Wang CH, Li SH, Weisel RD, Fedak PW, Dumont AS, Szmitko P, et al. C-reactive protein upregulates angiotensin type 1 receptors in vascular smooth muscle. Circulation 2003;107(13):1783-1790 View Article PubMed/NCBI
  37. Devaraj S, Yun JM, Adamson G, Galvez J, Jialal I. C-reactive protein impairs the endothelial glycocalyx resulting in endothelial dysfunction. Cardiovasc Res 2009;84(3):479-484 View Article PubMed/NCBI
  38. Li L, Roumeliotis N, Sawamura T, Renier G. C-reactive protein enhances LOX-1 expression in human aortic endothelial cells: relevance of LOX-1 to C-reactive protein-induced endothelial dysfunction. Circ Res 2004;95(9):877-883 View Article PubMed/NCBI
  39. Pegues MA, McCrory MA, Zarjou A, Szalai AJ. C-reactive protein exacerbates renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2013;304(11):F1358-F1365 View Article PubMed/NCBI
  40. Singh SK, Agrawal A. Functionality of C-Reactive Protein for Atheroprotection. Front Immunol 2019;10:1655 View Article PubMed/NCBI
  41. Thiele JR, Habersberger J, Braig D, Schmidt Y, Goerendt K, Maurer V, 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(1):35-50 View Article PubMed/NCBI
  42. Strang F, Schunkert H. C-reactive protein and coronary heart disease: all said—is not it?. Mediators Inflamm 2014;2014:757123 View Article PubMed/NCBI
  43. Hakobyan S, Harris CL, van den Berg CW, Fernandez-Alonso MC, de Jorge EG, de Cordoba SR, et al. Complement factor H binds to denatured rather than to native pentameric C-reactive protein. J Biol Chem 2008;283(45):30451-30460 View Article PubMed/NCBI
  44. Badimon L, Peña E, Arderiu G, Padró T, Slevin M, Vilahur G, et al. C-Reactive Protein in Atherothrombosis and Angiogenesis. Front Immunol 2018;9:430 View Article PubMed/NCBI
  45. Ji SR, Wu Y, Potempa LA, 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(4):648-661 View Article PubMed/NCBI
  46. Krayem I, Bazzi S, Karam M. The combination of CRP isoforms with oxLDL decreases TNF-α and IL-6 release by U937-derived macrophages. Biomed Rep 2017;7(3):272-276 View Article PubMed/NCBI
  47. 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(3):2079 View Article PubMed/NCBI
  48. Podrez EA, Abu-Soud HM, Hazen SL. Myeloperoxidase-generated oxidants and atherosclerosis. Free Radic Biol Med 2000;28(12):1717-1725 View Article PubMed/NCBI
  49. Kamal FZ, Lefter R, Jaber H, Balmus IM, Ciobica A, Iordache AC. The Role of Potential Oxidative Biomarkers in the Prognosis of Acute Ischemic Stroke and the Exploration of Antioxidants as Possible Preventive and Treatment Options. Int J Mol Sci 2023;24(7):6389 View Article PubMed/NCBI
  50. O’Flynn J, Dixon KO, Faber Krol MC, Daha MR, van Kooten C. Myeloperoxidase directs properdin-mediated complement activation. J Innate Immun 2014;6(4):417-425 View Article PubMed/NCBI
  51. Delporte C, Van Antwerpen P, Vanhamme L, Roumeguère T, Zouaoui Boudjeltia K. Low-density lipoprotein modified by myeloperoxidase in inflammatory pathways and clinical studies. Mediators Inflamm 2013;2013:971579 View Article PubMed/NCBI
  52. Getz GS, Reardon CA. Myeloperoxidase-mediated dysfunctional high-density lipoprotein. Arterioscler Thromb Vasc Biol 2014;34(4):695-696 View Article PubMed/NCBI
  53. Ross EA, Devitt A, Johnson JR. Macrophages: The Good, the Bad, and the Gluttony. Front Immunol 2021;12:708186 View Article PubMed/NCBI
  54. van der Vorst EPC, Theodorou K, Wu Y, Hoeksema MA, Goossens P, Bursill CA, et al. High-Density Lipoproteins Exert Pro-inflammatory Effects on Macrophages via Passive Cholesterol Depletion and PKC-NF-κB/STAT1-IRF1 Signaling. Cell Metab 2017;25(1):197-207 View Article PubMed/NCBI
  55. Fotakis P, Kothari V, Thomas DG, Westerterp M, Molusky MM, Altin E, et al. Anti-Inflammatory Effects of HDL (High-Density Lipoprotein) in Macrophages Predominate Over Proinflammatory Effects in Atherosclerotic Plaques. Arterioscler Thromb Vasc Biol 2019;39(12):e253-e272 View Article PubMed/NCBI
  56. Pirillo A, Catapano AL, Norata GD. Biological Consequences of Dysfunctional HDL. Curr Med Chem 2019;26(9):1644-1664 View Article PubMed/NCBI
  57. Van Lenten BJ, Hama SY, de Beer FC, Stafforini DM, McIntyre TM, Prescott SM, et al. Anti-inflammatory HDL becomes pro-inflammatory during the acute phase response. Loss of protective effect of HDL against LDL oxidation in aortic wall cell cocultures. J Clin Invest 1995;96(6):2758-2767 View Article PubMed/NCBI
  58. Van Lenten BJ, Navab M, Shih D, Fogelman AM, Lusis AJ. The role of high-density lipoproteins in oxidation and inflammation. Trends Cardiovasc Med 2001;11(3-4):155-161 View Article PubMed/NCBI
  59. Watanabe J, Charles-Schoeman C, Miao Y, Elashoff D, Lee YY, Katselis G, et al. Proteomic profiling following immunoaffinity capture of high-density lipoprotein: association of acute-phase proteins and complement factors with proinflammatory high-density lipoprotein in rheumatoid arthritis. Arthritis Rheum 2012;64(6):1828-1837 View Article PubMed/NCBI
  60. Spirig R, Schaub A, Kropf A, Miescher S, Spycher MO, Rieben R. Reconstituted high-density lipoprotein modulates activation of human leukocytes. PLoS One 2013;8(8):e71235 View Article PubMed/NCBI
  61. Wadham C, Albanese N, Roberts J, Wang L, Bagley CJ, Gamble JR, et al. High-density lipoproteins neutralize C-reactive protein proinflammatory activity. Circulation 2004;109(17):2116-2122 View Article PubMed/NCBI
  62. Holzwirth E, Fischer-Schaepmann T, Obradovic D, von Lucadou M, Schwedhelm E, Daum G, et al. Anti-inflammatory HDL effects are impaired in atrial fibrillation. Heart Vessels 2022;37(1):161-171 View Article PubMed/NCBI
  63. Morin EE, Guo L, Schwendeman A, Li XA. HDL in sepsis - risk factor and therapeutic approach. Front Pharmacol 2015;6:244 View Article PubMed/NCBI
  64. De Geest B, Mishra M. Impact of High-Density Lipoproteins on Sepsis. Int J Mol Sci 2022;23(21):12965 View Article PubMed/NCBI
  65. Shridas P, De Beer MC, Webb NR. High-density lipoprotein inhibits serum amyloid A-mediated reactive oxygen species generation and NLRP3 inflammasome activation. J Biol Chem 2018;293(34):13257-13269 View Article PubMed/NCBI
  66. Cockerill GW, Rye KA, Gamble JR, Vadas MA, Barter PJ. High-density lipoproteins inhibit cytokine-induced expression of endothelial cell adhesion molecules. Arterioscler Thromb Vasc Biol 1995;15(11):1987-1994 View Article PubMed/NCBI
  67. Taborda NA, Blanquiceth Y, Urcuqui-Inchima S, Latz E, Hernandez JC. High-Density Lipoproteins Decrease Proinflammatory Activity and Modulate the Innate Immune Response. J Interferon Cytokine Res 2019;39(12):760-770 View Article PubMed/NCBI
  68. Ogura M. HDL, cholesterol efflux, and ABCA1: Free from good and evil dualism. J Pharmacol Sci 2022;150(2):81-89 View Article PubMed/NCBI
  69. Rothblat GH, Phillips MC. High-density lipoprotein heterogeneity and function in reverse cholesterol transport. Curr Opin Lipidol 2010;21(3):229-238 View Article PubMed/NCBI
  70. von Eckardstein A, Nordestgaard BG, Remaley AT, Catapano AL. High-density lipoprotein revisited: biological functions and clinical relevance. Eur Heart J 2023;44(16):1394-1407 View Article PubMed/NCBI
  71. Anastasius M, Luquain-Costaz C, Kockx M, Jessup W, Kritharides L. A critical appraisal of the measurement of serum ‘cholesterol efflux capacity’ and its use as surrogate marker of risk of cardiovascular disease. Biochim Biophys Acta Mol Cell Biol Lipids 2018;1863(10):1257-1273 View Article PubMed/NCBI
  72. Nordestgaard LT, Christoffersen M, Lauridsen BK, Afzal S, Nordestgaard BG, Frikke-Schmidt R, et al. Long-term Benefits and Harms Associated With Genetic Cholesteryl Ester Transfer Protein Deficiency in the General Population. JAMA Cardiol 2022;7(1):55-64 View Article PubMed/NCBI
  73. Cuchel M, Rader DJ. Macrophage reverse cholesterol transport: key to the regression of atherosclerosis?. Circulation 2006;113(21):2548-2555 View Article PubMed/NCBI
  74. Attie AD, Kastelein JP, Hayden MR. Pivotal role of ABCA1 in reverse cholesterol transport influencing HDL levels and susceptibility to atherosclerosis. J Lipid Res 2001;42(11):1717-1726 View Article PubMed/NCBI
  75. Ogasawara F, Kodan A, Ueda K. ABC proteins in evolution. FEBS Lett 2020;594(23):3876-3881 View Article PubMed/NCBI
  76. Liu SL, Sheng R, Jung JH, Wang L, Stec E, O’Connor MJ, et al. Orthogonal lipid sensors identify transbilayer asymmetry of plasma membrane cholesterol. Nat Chem Biol 2017;13(3):268-274 View Article PubMed/NCBI
  77. Zhou H, Tan KC, Shiu SW, Wong Y. Cellular cholesterol efflux to serum is impaired in diabetic nephropathy. Diabetes Metab Res Rev 2008;24(8):617-623 View Article PubMed/NCBI
  78. Favari E, Lee M, Calabresi L, Franceschini G, Zimetti F, Bernini F, et al. Depletion of pre-beta-high density lipoprotein by human chymase impairs ATP-binding cassette transporter A1- but not scavenger receptor class B type I-mediated lipid efflux to high density lipoprotein. J Biol Chem 2004;279(11):9930-9936 View Article PubMed/NCBI
  79. Kane JP, Pullinger CR, Goldfine ID, Malloy MJ. Dyslipidemia and diabetes mellitus: Role of lipoprotein species and interrelated pathways of lipid metabolism in diabetes mellitus. Curr Opin Pharmacol 2021;61:21-27 View Article PubMed/NCBI
  80. Brunetti A, Chiefari E, Foti D. Recent advances in the molecular genetics of type 2 diabetes mellitus. World J Diabetes 2014;5(2):128-140 View Article PubMed/NCBI
  81. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011;365(23):2205-2219 View Article PubMed/NCBI
  82. Figus FA, Piga M, Azzolin I, McConnell R, Iagnocco A. Rheumatoid arthritis: Extra-articular manifestations and comorbidities. Autoimmun Rev 2021;20(4):102776 View Article PubMed/NCBI
  83. Avina-Zubieta JA, Thomas J, Sadatsafavi M, Lehman AJ, Lacaille D. Risk of incident cardiovascular events in patients with rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis 2012;71(9):1524-1529 View Article PubMed/NCBI
  84. Baviera M, Cioffi G, Colacioppo P, Tettamanti M, Fortino I, Roncaglioni MC. Temporal trends from 2005 to 2018 in deaths and cardiovascular events in subjects with newly diagnosed rheumatoid arthritis. Intern Emerg Med 2021;16(6):1467-1475 View Article PubMed/NCBI
  85. Dijkshoorn B, Raadsen R, Nurmohamed MT. Cardiovascular Disease Risk in Rheumatoid Arthritis Anno 2022. J Clin Med 2022;11(10):2704 View Article PubMed/NCBI
  86. Gusev E, Sarapultsev A. Atherosclerosis and Inflammation: Insights from the Theory of General Pathological Processes. Int J Mol Sci 2023;24(9):7910 View Article PubMed/NCBI
  87. Baghdadi LR, Woodman RJ, Shanahan EM, Mangoni AA. The impact of traditional cardiovascular risk factors on cardiovascular outcomes in patients with rheumatoid arthritis: a systematic review and meta-analysis. PLoS One 2015;10(2):e0117952 View Article PubMed/NCBI
  88. Yan J, Yang S, Han L, Ba X, Shen P, Lin W, et al. Dyslipidemia in rheumatoid arthritis: the possible mechanisms. Front Immunol 2023;14:1254753 View Article PubMed/NCBI
  89. Myasoedova E, Crowson CS, Kremers HM, Roger VL, Fitz-Gibbon PD, Therneau TM, et al. Lipid paradox in rheumatoid arthritis: the impact of serum lipid measures and systemic inflammation on the risk of cardiovascular disease. Ann Rheum Dis 2011;70(3):482-487 View Article PubMed/NCBI
  90. Venetsanopoulou AI, Pelechas E, Voulgari PV, Drosos AA. The lipid paradox in rheumatoid arthritis: the dark horse of the augmented cardiovascular risk. Rheumatol Int 2020;40(8):1181-1191 View Article PubMed/NCBI
  91. Kim SH, Lee CK, Lee EY, Park SY, Cho YS, Yoo B, et al. Serum oxidized low-density lipoproteins in rheumatoid arthritis. Rheumatol Int 2004;24(4):230-233 View Article PubMed/NCBI
  92. Myasoedova E, Crowson CS, Kremers HM, Fitz-Gibbon PD, Therneau TM, Gabriel SE. Total cholesterol and LDL levels decrease before rheumatoid arthritis. Ann Rheum Dis 2010;69(7):1310-1314 View Article PubMed/NCBI
  93. Liao KP, Cai T, Gainer VS, Cagan A, Murphy SN, Liu C, et al. Lipid and lipoprotein levels and trend in rheumatoid arthritis compared to the general population. Arthritis Care Res (Hoboken) 2013;65(12):2046-2050 View Article PubMed/NCBI
  94. Rizzo M, Spinas GA, Cesur M, Ozbalkan Z, Rini GB, Berneis K. Atherogenic lipoprotein phenotype and LDL size and subclasses in drug-naïve patients with early rheumatoid arthritis. Atherosclerosis 2009;207(2):502-506 View Article PubMed/NCBI
  95. Mathieu S, Lambert C, Fayet F, Couderc M, Beauger M, Malochet S, et al. Comparison of the cardiovascular risk profile of rheumatoid arthritis versus hand osteoarthritis patients. Rheumatol Int 2023;43(11):2065-2072 View Article PubMed/NCBI
  96. Tejera-Segura B, Macía-Díaz M, Machado JD, de Vera-González A, García-Dopico JA, Olmos JM, et al. HDL cholesterol efflux capacity in rheumatoid arthritis patients: contributing factors and relationship with subclinical atherosclerosis. Arthritis Res Ther 2017;19(1):113 View Article PubMed/NCBI
  97. VanEvery H, Yang W, Su J, Olsen N, Bao L, Lu B, et al. Low-Density Lipoprotein Cholesterol and the Risk of Rheumatoid Arthritis: A Prospective Study in a Chinese Cohort. Nutrients 2022;14(6):1240 View Article PubMed/NCBI
  98. Choy E, Sattar N. Interpreting lipid levels in the context of high-grade inflammatory states with a focus on rheumatoid arthritis: a challenge to conventional cardiovascular risk actions. Ann Rheum Dis 2009;68(4):460-469 View Article PubMed/NCBI
  99. Hurt-Camejo E, Paredes S, Masana L, Camejo G, Sartipy P, Rosengren B, et al. Elevated levels of small, low-density lipoprotein with high affinity for arterial matrix components in patients with rheumatoid arthritis: possible contribution of phospholipase A2 to this atherogenic profile. Arthritis Rheum 2001;44(12):2761-2767 View Article PubMed/NCBI
  100. Wu J, Zhou Q, Wei Z, Wei J, Cui M. Atherogenic Index of Plasma and Coronary Artery Disease in the Adult Population: A Meta-Analysis. Front Cardiovasc Med 2021;8:817441 View Article PubMed/NCBI
  101. Zhou K, Qin Z, Tian J, Cui K, Yan Y, Lyu S. The Atherogenic Index of Plasma: A Powerful and Reliable Predictor for Coronary Artery Disease in Patients With Type 2 Diabetes. Angiology 2021;72(10):934-941 View Article PubMed/NCBI
  102. Si Y, Fan W, Han C, Liu J, Sun L. Atherogenic Index of Plasma, Triglyceride-Glucose Index and Monocyte-to-Lymphocyte Ratio for Predicting Subclinical Coronary Artery Disease. Am J Med Sci 2021;362(3):285-290 View Article PubMed/NCBI
  103. Kosmas CE, Rodriguez Polanco S, Bousvarou MD, Papakonstantinou EJ, Peña Genao E, Guzman E, et al. The Triglyceride/High-Density Lipoprotein Cholesterol (TG/HDL-C) Ratio as a Risk Marker for Metabolic Syndrome and Cardiovascular Disease. Diagnostics (Basel) 2023;13(5):929 View Article PubMed/NCBI
  104. Hammam N, Abdel-Wahab N, Gheita TA. Atherogenic Index of Plasma in Women with Rheumatoid Arthritis and Systemic Lupus Erythematosus: A 10-Year Potential Predictor of Cardiovascular Disease. Curr Rheumatol Rev 2021;17(1):122-130 View Article PubMed/NCBI
  105. Uslu AU, Kucuk A, Icli A, Cure E, Sakiz D, Arslan S, et al. Plasma Atherogenic Index is an Independent Indicator of Subclinical Atherosclerosis in Systemic Lupus Erythematosus. Eurasian J Med 2017;49(3):193-197 View Article PubMed/NCBI
  106. Min HK, Kim HR, Lee SH, Shin K, Kim HA, Park SH, et al. Four-year follow-up of atherogenicity in rheumatoid arthritis patients: from the nationwide Korean College of Rheumatology Biologics Registry. Clin Rheumatol 2021;40(8):3105-3113 View Article PubMed/NCBI
  107. Dessie G. Association of atherogenic indices with C-reactive protein and risk factors to assess cardiovascular risk in rheumatoid arthritis patient at Tikur Anbessa Specialized Hospital, Addis Ababa. PLoS One 2022;17(6):e0269431 View Article PubMed/NCBI
  108. Ye L, Zhang X, Wu H, Chen Y, Zhou H, Wang Q, et al. Insulin resistance and adverse lipid profile in untreated very early rheumatoid arthritis patients: A single-center, cross-sectional study in China. Arch Rheumatol 2022;37(4):593-602 View Article PubMed/NCBI
  109. Georgiadis AN, Papavasiliou EC, Lourida ES, Alamanos Y, Kostara C, Tselepis AD, et al. Atherogenic lipid profile is a feature characteristic of patients with early rheumatoid arthritis: effect of early treatment—a prospective, controlled study. Arthritis Res Ther 2006;8(3):R82 View Article PubMed/NCBI
  110. Chavan VU, Ramavataram D, Patel PA, Rupani MP. Evaluation of serum magnesium, lipid profile and various biochemical parameters as risk factors of cardiovascular diseases in patients with rheumatoid arthritis. J Clin Diagn Res 2015;9(4):BC01-BC05 View Article PubMed/NCBI
  111. Polinski KJ, Bemis EA, Yang F, Crume T, Demoruelle MK, Feser M, et al. Association of Lipid Mediators With Development of Future Incident Inflammatory Arthritis in an Anti-Citrullinated Protein Antibody-Positive Population. Arthritis Rheumatol 2021;73(6):955-962 View Article PubMed/NCBI
  112. Rodríguez-Carrio J, Coras R, Alperi-López M, López P, Ulloa C, Ballina-García FJ, et al. Profiling of Serum Oxylipins During the Earliest Stages of Rheumatoid Arthritis. Arthritis Rheumatol 2021;73(3):401-413 View Article PubMed/NCBI
  113. van de Stadt LA, van Sijl AM, van Schaardenburg D, Nurmohamed MT. Dyslipidaemia in patients with seropositive arthralgia predicts the development of arthritis. Ann Rheum Dis 2012;71(11):1915-1916 View Article PubMed/NCBI
  114. Gan RW, Young KA, Zerbe GO, Demoruelle MK, Weisman MH, Buckner JH, et al. Lower omega-3 fatty acids are associated with the presence of anti-cyclic citrullinated peptide autoantibodies in a population at risk for future rheumatoid arthritis: a nested case-control study. Rheumatology (Oxford) 2016;55(2):367-376 View Article PubMed/NCBI
  115. de Pablo P, Romaguera D, Fisk HL, Calder PC, Quirke AM, Cartwright AJ, et al. High erythrocyte levels of the n-6 polyunsaturated fatty acid linoleic acid are associated with lower risk of subsequent rheumatoid arthritis in a southern European nested case-control study. Ann Rheum Dis 2018;77(7):981-987 View Article PubMed/NCBI
  116. Gan RW, Bemis EA, Demoruelle MK, Striebich CC, Brake S, Feser ML, et al. The association between omega-3 fatty acid biomarkers and inflammatory arthritis in an anti-citrullinated protein antibody positive population. Rheumatology (Oxford) 2017;56(12):2229-2236 View Article PubMed/NCBI
  117. Koh JH, Yoon SJ, Kim M, Cho S, Lim J, Park Y, et al. Lipidome profile predictive of disease evolution and activity in rheumatoid arthritis. Exp Mol Med 2022;54(2):143-155 View Article PubMed/NCBI
  118. Law SH, Chan ML, Marathe GK, Parveen F, Chen CH, Ke LY. An Updated Review of Lysophosphatidylcholine Metabolism in Human Diseases. Int J Mol Sci 2019;20(5):1149 View Article PubMed/NCBI
  119. Chu SH, Cui J, Sparks JA, Lu B, Tedeschi SK, Speyer CB, et al. Circulating plasma metabolites and risk of rheumatoid arthritis in the Nurses’ Health Study. Rheumatology (Oxford) 2020;59(11):3369-3379 View Article PubMed/NCBI
  120. Boers M, Nurmohamed MT, Doelman CJ, Lard LR, Verhoeven AC, Voskuyl AE, et al. Influence of glucocorticoids and disease activity on total and high density lipoprotein cholesterol in patients with rheumatoid arthritis. Ann Rheum Dis 2003;62(9):842-845 View Article PubMed/NCBI
  121. Attar SM. Hyperlipidemia in rheumatoid arthritis patients in Saudi Arabia. Correlation with C-reactive protein levels and disease activity. Saudi Med J 2015;36(6):685-691 View Article PubMed/NCBI
  122. Park YB, Lee SK, Lee WK, Suh CH, Lee CW, Lee CH, et al. Lipid profiles in untreated patients with rheumatoid arthritis. J Rheumatol 1999;26(8):1701-1704 PubMed/NCBI
  123. White D, Fayez S, Doube A. Atherogenic lipid profiles in rheumatoid arthritis. N Z Med J 2006;119(1240):U2125 PubMed/NCBI
  124. Govindan KP, Basha S, Ramesh V, Kumar CN, Swathi S. A comparative study on serum lipoprotein (a) and lipid profile between rheumatoid arthritis patients and normal subjects. J Pharm Bioallied Sci 2015;7(Suppl 1):S22-S25 View Article PubMed/NCBI
  125. Choi HK, Seeger JD. Lipid profiles among US elderly with untreated rheumatoid arthritis—the Third National Health and Nutrition Examination Survey. J Rheumatol 2005;32(12):2311-2316 PubMed/NCBI
  126. Ramharack R, Barkalow D, Spahr MA. Dominant negative effect of TGF-beta1 and TNF-alpha on basal and IL-6-induced lipoprotein(a) and apolipoprotein(a) mRNA expression in primary monkey hepatocyte cultures. Arterioscler Thromb Vasc Biol 1998;18(6):984-990 View Article PubMed/NCBI
  127. Orsó E, Schmitz G. Lipoprotein(a) and its role in inflammation, atherosclerosis and malignancies. Clin Res Cardiol Suppl 2017;12(Suppl 1):31-37 View Article PubMed/NCBI
  128. Lazarevic MB, Vitic J, Mladenovic V, Myones BL, Skosey JL, Swedler WI. Dyslipoproteinemia in the course of active rheumatoid arthritis. Semin Arthritis Rheum 1992;22(3):172-178 View Article PubMed/NCBI
  129. Popkova TV, Novikova DS, Novikov AA, Alexandrova EN, Mach ES, Gawa TN, et al. Role of blood cholesterol transport system disturbances in atherosclerosis development in rheumatoid arthritis. Rheumatology Science and Practice 2007;45(5):4-10 View Article
  130. Gonzalez A, Maradit Kremers H, Crowson CS, Ballman KV, Roger VL, Jacobsen SJ, et al. Do cardiovascular risk factors confer the same risk for cardiovascular outcomes in rheumatoid arthritis patients as in non-rheumatoid arthritis patients?. Ann Rheum Dis 2008;67(1):64-69 View Article PubMed/NCBI
  131. Boyer JF, Gourraud PA, Cantagrel A, Davignon JL, Constantin A. Traditional cardiovascular risk factors in rheumatoid arthritis: a meta-analysis. Joint Bone Spine 2011;78(2):179-183 View Article PubMed/NCBI
  132. Pozzi FS, Maranhão RC, Guedes LK, Borba EF, Laurindo IM, Bonfa E, et al. Plasma kinetics of an LDL-like non-protein nanoemulsion and transfer of lipids to high-density lipoprotein (HDL) in patients with rheumatoid arthritis. J Clin Lipidol 2015;9(1):72-80 View Article PubMed/NCBI
  133. Kim JY, Lee EY, Park JK, Song YW, Kim JR, Cho KH. Patients with Rheumatoid Arthritis Show Altered Lipoprotein Profiles with Dysfunctional High-Density Lipoproteins that Can Exacerbate Inflammatory and Atherogenic Process. PLoS One 2016;11(10):e0164564 View Article PubMed/NCBI
  134. Ormseth MJ, Yancey PG, Yamamoto S, Oeser AM, Gebretsadik T, Shintani A, et al. Net cholesterol efflux capacity of HDL enriched serum and coronary atherosclerosis in rheumatoid arthritis. IJC Metab Endocr 2016;13:6-11 View Article PubMed/NCBI
  135. Rodríguez-Carrio J, Alperi-López M, López P, López-Mejías R, Alonso-Castro S, Abal F, et al. High triglycerides and low high-density lipoprotein cholesterol lipid profile in rheumatoid arthritis: A potential link among inflammation, oxidative status, and dysfunctional high-density lipoprotein. J Clin Lipidol 2017;11(4):1043-1054.e2 View Article PubMed/NCBI
  136. Burggraaf B, van Breukelen-van der Stoep DF, van Zeben J, van der Meulen N, van de Geijn GM, Liem A, et al. Evidence for increased chylomicron remnants in rheumatoid arthritis. Eur J Clin Invest 2018;48(2):e12873 View Article PubMed/NCBI
  137. Dessie G, Tadesse Y, Demelash B, Genet S. Assessment of Serum Lipid Profiles and High-sensitivity C-reactive Protein Among Patients Suffering from Rheumatoid Arthritis at Tikur Anbessa Specialized Hospital, Addis Ababa, Ethiopia: A Cross-Sectional Study. Open Access Rheumatol 2020;12:223-232 View Article PubMed/NCBI
  138. Alisik T, Alisik M, Nacir B, Ayhan FF, Genc H, Erel O. Evaluation of dysfunctional high-density lipoprotein levels with myeloperoxidase/paraoxonase-1 ratio in rheumatoid arthritis. Int J Clin Pract 2021;75(7):e14172 View Article
  139. Chang CK, Chiang EI, Chang KH, Tang KT, Chen PK, Yip HT, et al. The Sizes and Composition of HDL-Cholesterol Are Significantly Associated with Inflammation in Rheumatoid Arthritis Patients. Int J Mol Sci 2023;24(13):10645 View Article PubMed/NCBI
  140. Memon RA, Staprans I, Noor M, Holleran WM, Uchida Y, Moser AH, et al. Infection and inflammation induce LDL oxidation in vivo. Arterioscler Thromb Vasc Biol 2000;20(6):1536-1542 View Article PubMed/NCBI
  141. Cvetkovic JT, Wållberg-Jonsson S, Ahmed E, Rantapää-Dahlqvist S, Lefvert AK. Increased levels of autoantibodies against copper-oxidized low density lipoprotein, malondialdehyde-modified low density lipoprotein and cardiolipin in patients with rheumatoid arthritis. Rheumatology (Oxford) 2002;41(9):988-995 View Article PubMed/NCBI
  142. Paimela L, Helve T, Leirisalo-Repo M, Vaarala O, Alfthan G, Palosuo T, et al. Clinical significance of antibodies against oxidised low density lipoprotein in early RA. Ann Rheum Dis 1996;55(8):558-559 View Article PubMed/NCBI
  143. Huang LS, Hung ND, Sok DE, Kim MR. Lysophosphatidylcholine containing docosahexaenoic acid at the sn-1 position is anti-inflammatory. Lipids 2010;45(3):225-236 View Article PubMed/NCBI
  144. Karpouzas GA, Ormseth SR, Ronda N, Hernandez E, Budoff MJ. Lipoprotein oxidation may underlie the paradoxical association of low cholesterol with coronary atherosclerotic risk in rheumatoid arthritis. J Autoimmun 2022;129:102815 View Article PubMed/NCBI
  145. Papamichail GV, Georgiadis AN, Tellis CC, Rapti I, Markatseli TE, Xydis VG, et al. Antibodies against oxidized LDL and atherosclerosis in rheumatoid arthritis patients treated with biological agents: a prospective controlled study. Clin Rheumatol 2024;43(1):481-488 View Article PubMed/NCBI
  146. Chu CS, Chan HC, Tsai MH, Stancel N, Lee HC, Cheng KH, et al. Range of L5 LDL levels in healthy adults and L5’s predictive power in patients with hyperlipidemia or coronary artery disease. Sci Rep 2018;8(1):11866 View Article PubMed/NCBI
  147. García-Gómez C, Bianchi M, de la Fuente D, Badimon L, Padró T, Corbella E, et al. Inflammation, lipid metabolism and cardiovascular risk in rheumatoid arthritis: A qualitative relationship?. World J Orthop 2014;5(3):304-311 View Article PubMed/NCBI
  148. Arts E, Fransen J, Lemmers H, Stalenhoef A, Joosten L, van Riel P, et al. High-density lipoprotein cholesterol subfractions HDL2 and HDL3 are reduced in women with rheumatoid arthritis and may augment the cardiovascular risk of women with RA: a cross-sectional study. Arthritis Res Ther 2012;14(3):R116 View Article PubMed/NCBI
  149. Ansell BJ, Fonarow GC, Fogelman AM. The paradox of dysfunctional high-density lipoprotein. Curr Opin Lipidol 2007;18(4):427-434 View Article PubMed/NCBI
  150. McMahon M, Grossman J, FitzGerald J, Dahlin-Lee E, Wallace DJ, Thong BY, et al. Proinflammatory high-density lipoprotein as a biomarker for atherosclerosis in patients with systemic lupus erythematosus and rheumatoid arthritis. Arthritis Rheum 2006;54(8):2541-2549 View Article PubMed/NCBI
  151. Charles-Schoeman C, Watanabe J, Lee YY, Furst DE, Amjadi S, Elashoff D, et al. Abnormal function of high-density lipoprotein is associated with poor disease control and an altered protein cargo in rheumatoid arthritis. Arthritis Rheum 2009;60(10):2870-2879 View Article PubMed/NCBI
  152. Undurti A, Huang Y, Lupica JA, Smith JD, DiDonato JA, Hazen SL. Modification of high density lipoprotein by myeloperoxidase generates a pro-inflammatory particle. J Biol Chem 2009;284(45):30825-30835 View Article PubMed/NCBI
  153. Ndrepepa G. Myeloperoxidase - A bridge linking inflammation and oxidative stress with cardiovascular disease. Clin Chim Acta 2019;493:36-51 View Article PubMed/NCBI
  154. Feijóo M, Túnez I, Ruiz A, Tasset I, Muñoz E, Collantes E. [Oxidative stress biomarkers as indicator of chronic inflammatory joint diseases stage]. Reumatol Clin 2010;6(2):91-94 View Article PubMed/NCBI
  155. Vivekanandan-Giri A, Slocum JL, Byun J, Tang C, Sands RL, Gillespie BW, et al. High density lipoprotein is targeted for oxidation by myeloperoxidase in rheumatoid arthritis. Ann Rheum Dis 2013;72(10):1725-1731 View Article PubMed/NCBI
  156. Wang W, Jian Z, Guo J, Ning X. Increased levels of serum myeloperoxidase in patients with active rheumatoid arthritis. Life Sci 2014;117(1):19-23 View Article PubMed/NCBI
  157. Nzeusseu Toukap A, Delporte C, Noyon C, Franck T, Rousseau A, Serteyn D, et al. Myeloperoxidase and its products in synovial fluid of patients with treated or untreated rheumatoid arthritis. Free Radic Res 2014;48(4):461-465 View Article PubMed/NCBI
  158. Feijóo M, Túnez I, Tasset I, Montilla P, Pérez-Guijo V, Muñoz-Gomariz E, et al. Infliximab reduces myeloperoxidase concentration in chronic inflammatory joint diseases. Pharmacology 2009;83(4):211-216 View Article PubMed/NCBI
  159. Maiocchi SL, Ku J, Thai T, Chan E, Rees MD, Thomas SR. Myeloperoxidase: A versatile mediator of endothelial dysfunction and therapeutic target during cardiovascular disease. Pharmacol Ther 2021;221:107711 View Article PubMed/NCBI
  160. Montes EG, Mansani FP, de Oliveira Toledo Júnior A, Schafranski MD, Zardo BQ, Dos Santos FA, et al. Myeloperoxidase as an important predictor of cardiovascular risk in individuals with rheumatoid arthritis. Inflammopharmacology 2021;29(6):1819-1827 View Article PubMed/NCBI
  161. Ormseth MJ, Stein CM. High-density lipoprotein function in rheumatoid arthritis. Curr Opin Lipidol 2016;27(1):67-75 View Article PubMed/NCBI
  162. Ronsein GE, Vaisar T. Inflammation, remodeling, and other factors affecting HDL cholesterol efflux. Curr Opin Lipidol 2017;28(1):52-59 View Article PubMed/NCBI
  163. Ferraz-Amaro I, González-Gay MA, García-Dopico JA, Díaz-González F. Cholesteryl ester transfer protein in patients with rheumatoid arthritis. J Rheumatol 2013;40(7):1040-1047 View Article PubMed/NCBI
  164. Charles-Schoeman C, Lee YY, Grijalva V, Amjadi S, FitzGerald J, Ranganath VK, et al. Cholesterol efflux by high density lipoproteins is impaired in patients with active rheumatoid arthritis. Ann Rheum Dis 2012;71(7):1157-1162 View Article PubMed/NCBI
  165. Liao KP, Playford MP, Frits M, Coblyn JS, Iannaccone C, Weinblatt ME, et al. The association between reduction in inflammation and changes in lipoprotein levels and HDL cholesterol efflux capacity in rheumatoid arthritis. J Am Heart Assoc 2015;4(2):e001588 View Article PubMed/NCBI
  166. Xie B, He J, Liu Y, Liu T, Liu C. A meta-analysis of HDL cholesterol efflux capacity and concentration in patients with rheumatoid arthritis. Lipids Health Dis 2021;20(1):18 View Article PubMed/NCBI
  167. Quevedo-Abeledo JC, Sánchez-Pérez H, Tejera-Segura B, de Armas-Rillo L, Armas-González E, Machado JD, et al. Differences in Capacity of High-Density Lipoprotein Cholesterol Efflux Between Patients With Systemic Lupus Erythematosus and Rheumatoid Arthritis. Arthritis Care Res (Hoboken) 2021;73(11):1590-1596 View Article PubMed/NCBI
  168. Ronda N, Favari E, Borghi MO, Ingegnoli F, Gerosa M, Chighizola C, et al. Impaired serum cholesterol efflux capacity in rheumatoid arthritis and systemic lupus erythematosus. Ann Rheum Dis 2014;73(3):609-615 View Article PubMed/NCBI
  169. Ronda N, Greco D, Adorni MP, Zimetti F, Favari E, Hjeltnes G, et al. Newly identified antiatherosclerotic activity of methotrexate and adalimumab: complementary effects on lipoprotein function and macrophage cholesterol metabolism. Arthritis Rheumatol 2015;67(5):1155-1164 View Article PubMed/NCBI
  170. Anastasius M, Kockx M, Jessup W, Sullivan D, Rye KA, Kritharides L. Cholesterol efflux capacity: An introduction for clinicians. Am Heart J 2016;180:54-63 View Article PubMed/NCBI
  171. Mortensen MB, Afzal S, Nordestgaard BG, Falk E. The high-density lipoprotein-adjusted SCORE model worsens SCORE-based risk classification in a contemporary population of 30,824 Europeans: the Copenhagen General Population Study. Eur Heart J 2015;36(36):2446-2453 View Article PubMed/NCBI
  172. Karpouzas GA, Papotti B, Ormseth SR, Palumbo M, Hernandez E, Adorni MP, et al. ATP-binding cassette G1 membrane transporter-mediated cholesterol efflux capacity influences coronary atherosclerosis and cardiovascular risk in Rheumatoid Arthritis. J Autoimmun 2023;136:103029 View Article PubMed/NCBI
  173. Karpouzas GA, Papotti B, Ormseth SR, Palumbo M, Hernandez E, Adorni MP, et al. Inflammation and immunomodulatory therapies influence the relationship between ATP-binding cassette A1 membrane transporter-mediated cholesterol efflux capacity and coronary atherosclerosis in rheumatoid arthritis. J Transl Autoimmun 2023;7:100209 View Article PubMed/NCBI
  • Gene Expression
  • pISSN 1052-2166
  • eISSN 1555-3884
Back to Top

Proatherogenic Disorders of Blood Lipid and Lipoprotein Metabolism in Patients with Rheumatoid Arthritis

Elena V. Gerasimova, Tatiana V. Popkova, Maria V. Shalygina, Daria A. Gerasimova
  • Reset Zoom
  • Download TIFF