Buscar en
Clínica e Investigación en Arteriosclerosis
Toda la web
Inicio Clínica e Investigación en Arteriosclerosis The pleiotropic role of HDL in autoimmune diseases
Información de la revista
Vol. 27. Núm. 2.
Páginas 97-106 (Marzo - Abril 2015)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
3804
Vol. 27. Núm. 2.
Páginas 97-106 (Marzo - Abril 2015)
Revisión
Acceso a texto completo
The pleiotropic role of HDL in autoimmune diseases
El papel pleiotrópico de las partículas HDL en las enfermedades autoinmunes
Visitas
3804
Sandra Parra
Autor para correspondencia
sandra.parra@grupsagessa.com

Corresponding author.
, Antoni Castro, Luis Masana
Internal Medicine, Sant Joan University Hospital, Reus, Spain
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (2)
Abstract

As is widely known, the classic function of HDL is reverse cholesterol transport (RCT), thus removing cholesterol from peripheral tissues. Early epidemiological studies, such as Framingham's, stated that increased HDL levels were associated with a significant decrease in relative risk for cardiovascular disease (CVD) mortality. However, those with heightened expectations in recent years for the development of therapeutic targets to increase HDL levels have been disappointed, because efforts have demonstrated the opposite effect on cardiovascular and global mortality.

However, in contrast, studies have highlighted the complexity and the intriguing role of HDL in different pathological conditions, such as infections, neoplasms, and autoimmune diseases.

In this review an attempt is made to summarize some biological pathways that link HDL function with the immune system, and its possible clinical repercussions in autoimmune diseases.

Keywords:
HDL
Autoimmune diseases
SLE
Acute phase response
Abbreviations:
RCT
CVD
CAD
ApoA1
APR
PON1
ABCA1
ABCG1
PAF-AH
LCAT
GSPx
SAA
HDL
LDL
PTX3
IL-1
TNF
IL-6
APC
CE
TLR
BLR
DC
LPS
MHC
S1P
ApoM
SLE
RA
MS
LPL
β2-GLP1
Apo H
Resumen

La función clásica de las partículas de colesterol HDL en el transporte reverso de colesterol está ampliamente establecida. Estudios epidemiológicos clásicos, tales como Framingham ya establecieron la relación inversa entre el incremento de los niveles de HDL y la mortalidad por riesgo cardiovascular.

Las grandes expectativas para el desarrollo de terapias que incrementen los niveles de colesterol HDL han creado grandes decepciones en estudios relativamente recientes. A pesar de todo, estos estudios han destacado la complejidad de las partículas HDL en diferentes condiciones patológicas como infecciones, neoplasias y enfermedades autoinmunes.

En esta revisión intentamos resumir algunos mecanismos biológicos que unen las HDL con las funciones dentro del sistema inmune y sus posibles repercusiones clínicas en las enfermedades autoinmunes.

Palabras clave:
HDL
Enfermedades autoinmunes
Lupus eritematoso sistémico
Respuesta de fase aguda
Texto completo
Introduction

As is widely known, the classic function of HDL is reverse cholesterol transport (RCT), removing cholesterol from peripheral tissues. Early epidemiological studies, such as Framingham's, stated that increased HDL levels were associated with a significant decrease in relative risk for cardiovascular disease (CVD) mortality.1 However, those with heightened expectations in recent years for the development of therapeutic targets to increase HDL levels have been disappointed because efforts have demonstrated the opposite effect on cardiovascular and global mortality.2

In contrast, however, studies have highlighted the complexity and the intriguing role of HDL in different pathologic conditions, such as infections, neoplasms and autoimmune diseases. These heterogenic functions of HDL have not been well understood, but what appears to be clear is that not only HDL-c levels but also lipoprotein particle composition appear to be important for the function of this complex lipoprotein.3,4

The predominant lipoprotein content in the plasma of several species is HDL. The main apolipoprotein associated with HDL is Apolipoprotein A1 (ApoA1), which is associated with cholesterol transport in cell surfaces via ABCA1 and ABCG1.5 ApoA1's structure is conserved throughout its evolution, and recent studies have associated HDL function not only with the homeostasis of cholesterol metabolism but also with immune system regulation,6 the acute phase response after infections; environmental stresses, such as severe burns; autoimmune diseases; and cancer.7–11

The proper knowledge of the role of HDL particles in pathologic conditions other than atherosclerosis has increased interest in the development of new therapeutic strategies for clinical conditions beyond cardiovascular diseases.4,12 In this review, we attempt to summarize the possible clinical relevance of HDL's functioning in the immune system in relation to its possible clinical implications in autoimmune diseases (Fig. 1).

Figure 1.

Main HDL-bounded proteins linked to immune system and acute phase response.

(0,09MB).
HDL and the acute phase response

The potential protective nature of HDL has been primarily attributed to its role in RCT. However, the mechanisms by which HDL may impact cardiovascular health and disease remain complex and not fully understood.3,4 HDL possesses a number of heterogenic functions that impact cardiovascular health. The heterogenic functions of HDL involve anti-inflammatory, antioxidant, antithrombotic, antiapoptotic and nitric oxide synthesis mechanisms. These different functions are related to the complex and heterogenic structure of HDL particles. This heterogeneity is the result of changes in the content of the apolipoproteins, lipids and proteins that are associated with HDL and are related not only to cholesterol metabolism but also to regulating the complement system and the acute phase response.13–17

The acute phase response (APR) is a systemic reaction to infectious and noninfectious tissue destruction. Multiple physiologic adaptations occur, including changes in the hepatic synthesis of a number of plasma proteins, termed acute-phase reactants.18 Two acute-phase reactants, C-reactive protein (CRP) and serum amyloid A protein (SAA), are known to interact with lipoproteins.19,20 CRP binds to apolipoprotein B, which is contained in atherogenic lipoproteins, whereas SAA circulates primarily with HDL.

Apolipoprotein A1(ApoA1) and enzymes associated with HDL with antioxidant properties such as paraoxonase-1 (PON1), platelet-activating factor acetylhydrolase (PAF-AH), lecithin: cholesterol acyltransferase (LCAT) and glutathione selenoperoxidase (GSPx) are replaced by SAA after the acute phase response.21–28

Under these pro-inflammatory conditions, the composition of HDL particles changes; they evolve into pro-inflammatory, pro-atherogenic particles that have been associated with the presence of coronary artery disease (CAD) and an increased risk for cardiovascular diseases.17,21,29–35

HDL and immune cell functionLipid rafts and immune cell function

Cholesterol membrane homeostasis is linked to the innate and adaptive immune response. Lipid rafts are membrane micro-domains that are enriched in cholesterol, phospholipids and proteins. They play an important role in activating signaling pathways in immune cells.6,36–38

HDL particles and cholesterol efflux transport mediated by the ATP-binding cassette A1 (ABCA1) or ABCG1 alter the structure and lipid composition of the cell membranes.39 Cholesterol depletion of lipid rafts by ApoA1 in antigen-presenting cells (APC) inhibits dendritic cells differentiation and the ability of macrophages and dendritic cells to stimulate T-cell activation by reducing the number of major histocompatibility (MHC) class II molecules.40,41

In another noteworthy study, it was demonstrated that ABCG1 negatively regulates thymocyte and peripheral lymphocyte proliferation, another novel mechanism by which cholesterol can alter the signaling pathways and proliferation of immune cells. These results showed that ABCG1 is a negative regulator of lymphocyte proliferation.42

It can be hypothesized that HDL may inhibit antigen presentation to T-cells by reducing the lipid raft cholesterol.7

Disruption of cholesterol efflux via ApoA-I can be used as a basis for interpreting the effects on immune cell function. One notable study demonstrated for the first time that cholesterol enrichment cells stimulate T-cell activation and expansion, developing an “autoimmune phenotype” that was resolved by subcutaneous injection of lipid-free ApoA1. The study's authors experimented with mice that lacked both LDLr (LDLr−/−) and apoA1 (LDLr−/−, apoA-I−/−) mice. As expected, when the LDLr−/− and apoA-I−/− (DKO) mice were fed with an atherogenic diet, they developed increased atherosclerosis compared with the LDLr−/− (SKO) controls that were fed the same diet. Unexpectedly, the mice also displayed an unusual expansion and activation of the T-cells in their skin, which drained the lymph nodes and led to an autoimmune phenotype. Furthermore, when the T- and B-cells were fluorescence-activated and cell sorted from the lymph nodes of the atherogenic-diet-fed DKO mice, the cells were found to be nearly filled with CE, as measured by mass spectrometry, whereas the cells from the atherogenic-diet-fed SKO mice were not.43,44

Cholesterol enrichers other than macrophages and monocytes induce cell activation and cell dysfunction, developing autoimmune phenotypes.38

In addition to enriching the cholesterol in lipid rafts, it was demonstrated in another study that the decreased cholesterol in these micro-domains from neutrophiles by HDL downregulated the activation, adhesion, spread and migration of the neutrophiles.45,46

All of these data indicate that the RCT and HDL regulation of cholesterol homeostasis was not primarily intended for atheroprotective functionality. It is reasonable to consider HDL lipoproteins as part of the innate immune system that regulates the host response after infections. However, cholesterol's enriching of membrane micro-domains is associated with a stimulation of the immune system and possibly with a risk of autoimmune diseases and atherosclerosis.7,47

Activation of toll-like receptors mediated by HDL particles

Toll-like receptors (TLRs) are molecules that are expressed in the monocytes, macrophages and dendritic cells (DC) involved in the innate immune response against infections. They are responsible for recognizing the conserved molecular patterns present in pathogens. In atherosclerotic lesions, TLRs play a role in recruiting leukocytes and in forming foam cells by activating T-lymphocytes. HDL interacts with TLR4 and inhibits the antiviral response in macrophages induced by lipopolysaccharides (LPS). ApoA1 and cholesterol removal from lipid rafts by ApoA-1 and the apoA1 mimetic peptide 4F also reduce TLR4 expression.48–51

Moreover, although direct evidence for any HDL effects on B-cell functioning has not yet been demonstrated, it seems possible that removing cholesterol from lipid rafts could also affect B-cell activation via the expression of BLR.7,52,53

T regulatory cell (Treg-CD4+CD25+FoxP3+)

Regulatory T-cells (Tregs) are the guardians of peripheral tolerance, acting to prevent autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Defects in Tregs have been reported in these two diseases despite significant differences in their clinical phenotypes and pathogeneses. In both diseases, the potency of the Tregs fails to keep pace with the activation of effector cells, and the Tregs are unable to resist the ensuing inflammation.54,55

The importance of Tregs is underscored by the overwhelming inflammation and autoimmunity that result from their absence. Treg cells have been linked not only to autoimmune diseases but also to the atherosclerotic process by inhibiting pro-inflammatory T-cells in atherosclerotic vessels.56–58

Of note, in an autoimmune animal model in mice, apoA1 treatment increased Tregs and decreased the percentage of effector/effector memory T-cells (Teff), suggesting that apoA1 may control T-cell homeostasis in peripheral lymphoid tissues by regulating the Treg/Teff balance. How apoA1 modulates Treg/Teff balance remains unknown, but the process likely involves cellular cholesterol and lipid raft homeostasis.43,59,60

HDL and humoral immunityComplement system and HDL

A number of complement system proteins, activation products, receptors and regulatory proteins have been detected in atherosclerotic lesions, and C5b-9 deposits have been shown to correlate with the atherosclerotic process.61–63

Furthermore, complement component 3 (C3) plasma levels have been related to coronary artery disease measured by coronary angiography and to clinical ischemic events.64,65

Lipid and glucose metabolism are also related to the complement system. A number of studies have associated C3 and C4 levels with higher levels of triglycerides and postprandial hypertriglyceridemia, low levels of HDL and insulin resistance, which are the metabolic syndrome compounds.66

These data suggest the relevance of the complement system in atherogenesis.67

Furthermore, a fragment of C3 (C3a-des-Arg) is common to a portion of the acylation-stimulating protein (ASP), which is the most potent stimulant of triglyceride synthesis and glucose membrane transport in human adipocytes.68 This could help to explain the association between the complement system, lipid metabolism, insulin resistance and postprandial lipemia.69

More recently, it has been stated that HDL plasma levels interact with complement system activation because an inverse correlation has been identified between HDL level and the circulating terminal complex C5b-C9 levels70.

Recently, new proteomic technologies applied to HDL composition have revealed that HDL plays a previously unsuspected role in regulating the complement system. The complement components C3, C4 and C9, and complement-regulatory proteins, such as vitronectin and clusterin, have been found in HDL. These studies also showed that the HDL in patients with CAD was enriched with C3 and C4.14,15,23

Our group described in a previous work that in a cohort of patients affected with SLE, there was a positive correlation between small, dense HDL particles, C3 and C4 levels and the activation of the complement cascade.71 These small HDL particles were pro-atherogenic, and they were also associated with the presence of subclinical atherosclerosis and inflammatory serum markers such as ESR and CRP. These data highlight that in SLE patients, different HDL subpopulations with different compositions have different roles in autoimmune systemic disease, but there is a close association with the complement system. These small HDL particles were pro-atherogenic and pro-inflammatory, but the positive correlation with the complement system components may be important in the clinical course of lupus because a decrease in the complement proteins is associated with flare-ups and worse clinical courses.

Pentraxins

Pentraxins are a superfamily of acute-phase proteins that are highly conserved during evolution and characterized by the presence of a multimeric structural motif, the pentraxin motif. They can be classified as short pentraxins, such as C reactive protein (CRP), or long pentraxins, such as PTX3. PTX3 was the first recognized member of this family. It is produced in a number of cell types (mononuclear phagocytes, dendritic cells, fibroblasts and endothelial cells) in response to interleukin (IL)-1 and tumor necrotic factor (TNF), in contrast with CRP, which is produced only in the liver and is stimulated by IL-6.72 It would appear that PTX3 plays a role in the innate immune response given that it has been reported that PTX3 deficiency leads to invasive pulmonary aspergillosis owing to the defective recognition of conidia by alveolar macrophages and dendritic cells.73,74

PTX3 has also been associated with some types of cardiovascular disease, including atherosclerosis.75–77

Norata et al. demonstrated in a noteworthy study that HDL induces mRNA expression and the protein release of PTX3.78 Moreover, PTX3-C1q binding promotes complement-mediated clearance of apoptotic cells, limiting inflammatory tissue damage after injury. These data suggest that part of the atheroprotective effects of HDL could result from the modulation of molecules that act as sensors of the immunoinflammatory balance in the vascular wall, but no additional studies have confirmed this atheroprotective association with PTX3.72

Sphingosine 1-phosphate (S1P)

Sphingosine 1-phosphate (S1P) is a membrane-derived lysophospholipid that acts primarily as an extracellular signaling molecule.79 Recent evidence on the physiology of the S1P-S1PR axis in the homeostasis of immune-mediated cells is receiving increased attention because these cells have become attractive therapeutic targets in diseases such as chronic inflammatory pathologies (asthma, arthritis), autoimmune disorders (multiple sclerosis), neoplasms and atherosclerosis.

Signals initiated by S1P are conducted by five G protein-coupled receptors (GPCR), named S1P15.79 Cellular and temporal expression of the S1P receptors determine their specific roles in various organ systems, but they are particularly critical for regulating the cardiovascular, immune, and nervous systems, with the most well-known contributions of S1PR signaling being modulation of vascular barrier function, vascular tone and regulation of lymphocyte trafficking.80

Approximately 35% of plasma S1P is bound to albumin and 65% to ApoM, which is found on a small percentage (approximately 5%) of high-density lipoprotein (HDL) particles.80 This ApoM+HDL-bound1P has been proposed as a primary contributor to the antiatherogenic properties of HDL.81

There are a number of well-characterized S1PR agonists and antagonists; however, most compounds have been directed toward modulating S1P1 activity. FTY720 (fingolimod; Gilenya, Novartis) is the prototypical S1PR agonist and was approved by the U.S. Food and Drug Administration as a first-line oral therapy for relapsing-remitting multiple sclerosis (MS).82–84 FTY720 acts as an agonist on S1P1 and S1P3–5, and it also acts as a functional S1P1 antagonist by inducing receptor endocytosis and degradation of this receptor.85–88 This promiscuity may be responsible for the adverse effects, such as bradycardia and hypertension, seen in fingolimod-treated patients.89

S1P receptors regulate many aspects of immune cell biology. The best known is S1P1's regulation of lymphocyte migration out of the secondary lymphoid organs into the blood and lymph nodes.

The contribution of S1PRs to regulating the immune response has been studied extensively in the context of experimental autoimmune encephalomyelitis (EAE), the most commonly used animal model of multiple sclerosis (MS).90 The mechanism of action of the S1P1 agonist fingolimod has been presumed to be the trapping of autoreactive T- and B-cells in the lymphoid organs, away from the central nervous system.91 However, T-cell S1P1 may also regulate the activation and differentiation of these immune cells. Deletion of T-cell S1P1 significantly suppresses the ability of these cells to be polarized to Th17 in vitro.92

S1P1 is also expressed on CD4 T-cells that have been isolated from human rheumatoid arthritis (RA) patients.93 S1P enhances the expression by these cells of the receptor activator of nuclear factor kB (RANK) ligand, an effect that is replicated in a synovial cell-like cell line. In collagen-induced RA models, an S1P1-specific antagonist prevented or ameliorated disease by up-regulating lymphocyte CD69 expression, which downregulates S1P1 surface expression, blocking thymic egress.93–95

S1P1 suppresses Treg development via the AKT/mTOR pathway and affects their migration from the thymus and out of the periphery by counteracting CCR7 retention signals, similar to the mechanism that regulates the egress of effector T-cells from lymph nodes.90,96,97 FTY720 significantly increased the number of regulatory T-cells while decreasing central memory T-cells.98

In LDL-receptor-deficient mice, fingolimod inhibited atherosclerosis by modulating lymphocyte and macrophage function; in this study, lipids were unchanged, but the lymphocyte blood count decreased.99

Although S1P1 has been the focus of much research, little is known of the roles of the S1PR with a cell-subtype-specific effect on certain cell lines in the immune system.

Research focusing on knowledge of the diverse biological functions of the S1PR family will provide an opportunity to discover new treatment strategies for these autoimmune and chronic inflammatory disorders. The study of the function of S1P that is transported by apoM-HDL is also of interest in inflammatory response and vascular permeability after infection. In one transversal study, decreased ApoM levels were associated with the presence of sepsis and systemic inflammatory response syndrome (SIRS) after infection.100

With all of these data, the HDL-apoM-S1P axis is of interest as a novel therapeutic approach with diverse biological functions in different pathogenic processes that involve the immune system.

HDL and autoinmune diseases

Plasma HDL-c levels are modified by a number of clinical conditions, such as autoimmune diseases. HDL-c is elevated in multiple sclerosis and reduced in systemic lupus erythematosus (SLE), rheumatoid arthritis (RA),68 Sjögren's syndrome,69 ankylosing spondylitis,70 psoriatic arthritis, and inflammatory bowel disease.71

Patients affected with chronic autoimmune diseases also maintain a chronic inflammatory status that appears to accelerate atherosclerosis and cardiovascular diseases.

Research on nonclassical cardiovascular risk factors and the interaction of these diseases with lipid metabolism is of interest because these diseases are characterized by chronic inflammation and hyperactive immune systems.101,102

HDL and SLE

SLE is the prototype of autoimmune diseases with multisystem involvement. The presence of antibodies with different targets is associated with the diversity of clinical manifestations of this systemic disease.

Patients affected with systemic lupus erythematosus (SLE) show an increase in cardiovascular mortality and morbidity despite improvements in the control of disease activity and complications.103,104 These data are supported by the results of a number of studies that have shown a higher prevalence of subclinical atherosclerosis in this young population.105–107 The accelerated atherosclerosis observed in patients with SLE cannot be entirely explained by the traditional cardiovascular risk factors. The presence of some metabolic disturbances, such as atherogenic dyslipidemia9 and metabolic syndrome (MetS), appears to be more prevalent in SLE patients owing to inflammatory mechanisms.108,109 Other nonclassical cardiovascular risk factors related to inflammation that have been associated with this accelerated atherosclerotic process include C-reactive protein (CRP), cytokines, the complement system110–112 and some adipokines.113,114

HDL metabolism is affected by the presence of lupus. A “lupus pattern” of dyslipidemia has been defined by elevated levels of very-low-density lipoprotein cholesterol (VLDL) and triglycerides (TG) and lower high-density lipoprotein cholesterol (HDL) levels.9,115 A notable finding has been that activity aggravates these alterations – striking increases in VLDL and TG levels and decreased HDL levels were directly correlated with SLEDAI scores.115 Another consideration regarding the lipid profiles of SLE patients is that ordinal biochemical analyses may not reflect changes in lipoprotein subpopulations that could give additional information about SLE's pro-atherogenic lipid profile, which has been described in studies on lipid profiles measured by magnetic nuclear resonance (MNR) and the presence of subclinical atherosclerosis.112

The abnormalities observed in SLE that are associated with low HDL-cholesterol levels could be explained by a number of mechanisms that involve inactivating lipoprotein lipase activity and the presence of antibodies against LPL and ApoA1. OxLDL/β2GPI complexes and IgG anti-β2GPI antibodies were also found in lupus patients with high TG and low HDL levels.116

As was described above, these HDL particles also show qualitative changes in composition and protein cargo, with HDL lipoproteins evolving into pro-inflammatory molecules.35,117,118

These “pro-inflammatory HDLs” were associated with the presence of subclinical atherosclerosis in SLE patients and also in RA patients.21

Changes in HDL composition after the acute phase response and these chronic inflammatory conditions impair the functionality and antioxidant properties of HDL.

One of the most important antioxidant enzymes bound to HDL is PON1. Decreased PON1 serum enzyme activity has been reported in SLE119,120 and also in other rheumatic disorders such as RA121 EM122 and psoriasis.123 The impaired antioxidant ability of HDL has been associated with increased atherosclerosis and also with the degree of disease activity.

Titers of antibodies against HDL and ApoA1 were associated in SLE patients with persistent inflammatory disease (measured with the SLEDAI index),124,125 and they have also been inversely correlated with paraoxonase activity.126

Investigation of HDL's impaired cholesterol transport capacity in autoimmune disorders is of interest owing to HDL's association with modulating the immune system. In SLE and RA patients, impaired HDL function as measured by efflux cholesterol capability has been described.127 Cholesterol efflux capacity was associated with the degree of disease activity in RA127 and with the extent of psoriatic areas and the severity index in another study.128 In this study, it was found that SLE patients showed a more complex pattern of modifications, with marked reduction of ATP-binding cassette G1- and ATP-binding cassette A1-mediated cholesterol efflux capacity unrelated to the disease activity.127

In another study with SLE patients, antibodies against ATP-binding cassette transporter A1 were associated with the presence of atherosclerosis, but it was not the study's objective to investigate ABCA1's association with the disease activity.

Therapeutic strategies using ApoA and ApoA-mimetic peptides as targets have been initiated in animal models of SLE.

As we described earlier, an animal model of apoA-I−/−LDLr−/− mice displayed an autoimmune-like phenotype similar to SLE, including increased plasma antibodies against double-stranded DNA, β2-glycoprotein I, and oxidized LDL. Treatment of these mice with lipid-free apoA-I reversed the autoimmune phenotype and lowered the number of lymphatic nodules.44,129

Another study, with a murine lupus model that was treated with apolipoprotein A-1 mimetic peptide (L-4F) both alone and with pravastatine, showed decreased levels of IgG anti-dsDNA, IgG anti-oxPLs, proteinuria glomerulonephritis and osteopenia.130

Apolipoprotein H and antiphospholipid syndrome

Apolipoprotein H (Apo H), also known as beta2-glycoprotein, is a glycoprotein involved in clotting mechanisms and lipid pathways. ApoH, is the main autoantigen responsible for negatively charged antiphospholipid antibodies production in the antiphospholipid syndrome (SAF) a prothrombotic clinical condition in patients with autoimmune diseases as lupus but also in the general population.131

Polz and Kostner described for the first time the presence of a beta2-glycoprotein in human apolipoproteins, and it was designated with the name of ApoH. After that, it was described that only a small percentage of ApoH (4–13%) is associated with plasma lipoproteins, for this reason the plasma levels of apoH have also been known as beta2-glycoprotein (beta2-GPI).132,133

Patients affected by SAF and SLE are under an oxidative stress environment. This oxidative stress is one of the mechanisms that promote important structural changes in beta2-GPI protein. One of these changes is the formation of beta2-GPI-oxLDL complexes that are associated with arterial thrombosis. Antibodies against these complexes have been reported both in SLE patients and in diabetic patients and were associated with an increase of the cardiovascular risk.134

To conclude, the parallelism between autoimmune diseases and the atherosclerotic process focuses on the role of HDL in modulating cholesterol homeostasis and the pathogenesis of both diseases. This promising approach could highlight new therapeutic strategies to modulate the immune system and disease activity and to avoid the metabolic disturbances that are associated with increased cardiovascular risk factors and the atherosclerotic process in patients with increased morbidity and mortality (Fig. 2).

Figure 2.

Interactions between HDL particles and the immune system. SAA: serum amyloid A; PON: paraoxonase-1; CRP: C-Reactive protein; PTX3: pentraxin 3; S1P: sphingosine 1 phosphate, apoM: apolipoprotein-M; Tregs: T regulatory cell; APC: antigen presentation cell; MHC-II: major histocompatibility complex II; SLE: Systemic lupus erythematosus; Rheumatoid Artritis, SAF: antiphospholipid syndrome; ApoA1: apolipoprotein-1.

(0,15MB).
Ethical disclosuresProtection of human and animal subjects

The authors declare that no experiments were performed on humans or animals for this investigation.

Confidentiality of Data

The authors declare that no patient data appears in this article.

Right to privacy and informed consent

The authors declare that no patient data appears in this article.

Conflict of interest

The authors confirm that this article content has no conflict of interest.

References
[1]
T. Gordon, W.B. Kannel, W.P. Castelli, T.R. Dawber.
Lipoproteins, cardiovascular disease, and death the Framingham study.
Arch Intern Med, 141 (1981), pp. 1128-1131
[2]
P.J. Barter, M. Caulfield, M. Eriksson, S.M. Grundy, J.J.P. Kastelein, M. Komajda, et al.
Effects of torcetrapib in patients at high risk for coronary events.
N Engl J Med, 357 (2007), pp. 2109-2122
[3]
C. Mineo, P.W. Shaul.
Novel biological functions of high-density lipoprotein cholesterol.
Circ Res, 111 (2012), pp. 1079-1090
[4]
R.S. Rosenson, H.B. Brewer, B. Ansell, P. Barter, M.J. Chapman, J.W. Heinecke, et al.
Translation of high-density lipoprotein function into clinical practice: current prospects and future challenges.
Circulation, 128 (2013), pp. 1256-1267
[5]
X. Collet, Y.L. Marcel, N. Tremblay, C. Lazure, R.W. Milne, B. Perret, et al.
Evolution of mammalian apolipoprotein A-I and conservation of antigenicity: correlation with primary and secondary structure.
J Lipid Res, 38 (1997), pp. 634-644
[6]
M.G. Sorci-Thomas, M.J. Thomas.
High density lipoprotein biogenesis, cholesterol efflux, and immune cell function.
Arterioscler Thromb Vasc Biol, 32 (2012), pp. 2561-2565
[7]
G.D. Norata, A. Pirillo, E. Ammirati, A.L. Catapano.
Emerging role of high density lipoproteins as a player in the immune system.
Atherosclerosis, 220 (2012), pp. 11-21
[8]
F. Rassoul, V. Richter, C. Kistner, D. Wisser, B. Reichert.
Soluble cell adhesion molecules and parameters of lipoprotein metabolism in patients with severe burns.
West Indian Med J, 58 (2009), pp. 417-421
[9]
E.F. Borba, J.F. Carvalho, E. Bonfá.
Mechanisms of dyslipoproteinemuas ins systemic lupus erythematosus.
Dev Immunol, 13 (2006), pp. 203-208
[10]
O.F. Sharifov, X. Xu, A. Gaggar, W.E. Grizzle, V.K. Mishra, J. Honavar, et al.
Anti-inflammatory mechanisms of apolipoprotein A-I mimetic peptide in acute respiratory distress syndrome secondary to sepsis.
[11]
D. McGrowder, C. Riley, E.Y. Morrison, L. Gordon.
The role of high-density lipoproteins in reducing the risk of vascular diseases, neurogenerative disorders, and cancer.
Cholesterol, 2011 (2011), pp. 496925
[12]
X. Zhu, J.S. Parks.
New roles of HDL in inflammation and hematopoiesis.
Annu Rev Nutr, 32 (2012), pp. 161-182
[13]
T. Vaisar.
Proteomics investigations of HDL. Challenges and promise.
Curr Vasc Pharmacol, 10 (2012), pp. 410-421
[14]
A.S. Shah, L. Tan, J.L. Long, W.S. Davidson.
Proteomic diversity of high density lipoproteins: our emerging understanding of its importance in lipid transport and beyond.
J Lipid Res, 54 (2013), pp. 2575-2585
[15]
S.M. Gordon, J. Deng, L.J. Lu, W.S. Davidson.
Proteomic characterization of human plasma high density lipoprotein fractionated by gel filtration chromatography.
J Proteome Res, 9 (2010), pp. 5239-5249
[16]
M.C. Cheung, T. Vaisar, X. Han, J.W. Heinecke, J.J. Albers.
Phospholipid transfer protein in human plasma associates with proteins linked to immunity and inflammation.
Biochemistry, 49 (2010), pp. 7314-7322
[17]
A.M. Fogelman.
The complexity of high-density lipoproteins.
Circulation, 122 (2010), pp. 1900-1901
[18]
M.B. Pepys, M.L. Baltz.
Acute phase proteins with special reference to C-reactive protein and related proteins (pentaxins) and serum amyloid A protein.
Adv Immunol, 34 (1983), pp. 141-212
[19]
G. Marhaug, K. Sletten, G. Husby.
Characterization of amyloid related protein SAA complexed with serum lipoproteins (apoSAA).
Clin Exp Immunol, 50 (1982), pp. 382-389
[20]
I.F. Rowe, K. Soutar a, I.M. Trayner, G.R. Thompson, M.B. Pepys.
Circulating human C-reactive protein binds very low density lipoproteins.
Clin Exp Immunol, 58 (1984), pp. 237-244
[21]
B.J. Van Lenten, S.Y. Hama, F.C. de Beer, D.M. Stafforini, T.M. McIntyre, S.M. Prescott, 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, 96 (1995), pp. 2758-2767
[22]
A. Jahangiri.
HDL the acute phase response.
Curr Opin Endocrinol Diab Obes, 17 (2010), pp. 156-160
[23]
T. Vaisar, S. Pennathur, P.S. Green, S.A. Gharib, A.N. Hoofnagle, M.C. Cheung, et al.
Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of HDL.
(2007), pp. 117
[24]
M. Riwanto, L. Rohrer, B. Roschitzki, C. Besler, P. Mocharla, M. Mueller, et al.
Altered activation of endothelial anti- and proapoptotic pathways by high-density lipoprotein from patients with coronary artery disease: role of high-density lipoprotein-proteome remodeling.
Circulation, 127 (2013), pp. 891-904
[25]
M. Tölle, T. Huang, M. Schuchardt, V. Jankowski, N. Prüfer, J. Jankowski, et al.
High-density lipoprotein loses its anti-inflammatory capacity by accumulation of pro-inflammatory-serum amyloid A.
Cardiovasc Res, 94 (2012), pp. 154-162
[26]
J. Watanabe, C. Charles-Schoeman, Y. Miao, D. Elashoff, Y.Y. Lee, G. Katselis, 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, 64 (2012), pp. 1828-1837
[27]
K. Alwaili, D. Bailey, Z. Awan, S.D. Bailey, I. Ruel, A. Hafiane, et al.
The HDL proteome in acute coronary syndromes shifts to an inflammatory profile.
Biochim Biophys Acta, 1821 (2012), pp. 405-415
[28]
C.Y. Han, T. Chiba, J.S. Campbell, N. Fausto, M. Chaisson, G. Orasanu, et al.
Reciprocal and coordinate regulation of serum amyloid A versus apolipoprotein A-I and paraoxonase-1 by inflammation in murine hepatocytes.
Arterioscler Thromb Vasc Biol, 26 (2006), pp. 1806-1813
[29]
H.B. G, V.S. Rao, V.V. Kakkar.
Friend turns foe: transformation of anti-inflammatory HDL to proinflammatory HDL during acute–phase response.
Cholesterol, 2011 (2011), pp. 274629
[30]
M. Navab, G.M. Ananthramaiah, S.T. Reddy, B.J. Van Lenten, B.J. Ansell, S. Hama, et al.
The double jeopardy of HDL.
Ann Med, 37 (2005), pp. 173-178
[31]
E.a. Fisher, J.E. Feig, B. Hewing, S.L. Hazen, J.D. Smith.
High-density lipoprotein function, dysfunction, and reverse cholesterol transport.
Arterioscler Thromb Vasc Biol, 32 (2012), pp. 2813-2820
[32]
M. Navab, S.T. Reddy, B.J. Van Lenten, G.M. Anantharamaiah, A.M. Fogelman.
The role of dysfunctional HDL in atherosclerosis.
J Lipid Res, 50 (2009), pp. S145-S149
[33]
G.H. Raterman, H. Levels, A.E. Voskuyl, W.F. Lems, B.A. Dijkmans, M.T. Nurmohamed.
HDL protein composition alters from proatherogenic into less atherogenic and proinflammatory in rheumatoid arthritis patients responding to rituximab.
Ann Rheum Dis, 72 (2013), pp. 560-565
[34]
M. Navab, G.M. Anantharamaiah, S.T. Reddy, B.J. Van Lenten, B.J. Ansell, A.M. Fogelman.
Mechanisms of disease: proatherogenic HDL—an evolving field.
Nat Clin Pract Endocrinol Metab, 2 (2006), pp. 504-511
[35]
M. Mcmahon, J. Grossman, B. Skaggs, D. Ph, L. Sahakian, N. Ragavendra, et al.
Dysfunctional pro-inflammatory high density lipoproteins confer increased risk foratherosclerosis in women with systemic lupus erythematosus.
Arthritis Rheum, 60 (2009), pp. 2428-2437
[36]
P.S. Kabouridis, J. Janzen, A.L. Magee, S.C. Ley.
Cholesterol depletion disrupts lipid rafts and modulates the activity of multiple signaling pathways in T lymphocytes.
[37]
H.A. Anderson, E.M. Hiltbold, P.A. Roche.
Concentration of MHC class II molecules in lipid rafts facilitates antigen presentation.
Nat Immunol, 1 (2000), pp. 156-162
[38]
G.D. Norata, A.L. Catapano.
HDL and adaptive immunity: a tale of lipid rafts.
Atherosclerosis, 225 (2012), pp. 34-35
[39]
M.G. Sorci-Thomas, J.S. Owen, B. Fulp, S. Bhat, X. Zhu, J.S. Parks, et al.
Nascent high density lipoproteins formed by ABCA1 resemble lipid rafts and are structurally organized by three apo A-I monomers.
J Lipid Res, 53 (2012), pp. 1890-1909
[40]
K.D. Kim, H.Y. Lim, H.G. Lee, D.Y. Yoon, Y.K. Choe, I. Choi, et al.
Apolipoprotein A-I induces IL-10 and PGE2 production in human monocytes and inhibits dendritic cell differentiation and maturation.
Biochem Biophys Res Commun, 338 (2005), pp. 1126-1136
[41]
S-H. Wang, S-G. Yuan, D.Q. Peng, S.P. Zhao.
HDL ApoA-I inhibit antigen presentation-mediated T cell activation by disrupting lipid rafts in antigen presenting cells.
Atherosclerosis, 225 (2012), pp. 105-114
[42]
A.J. Armstrong, A.K. Gebre, J.S. Parks, C.C. Hedrick.
ATP-binding cassette transporter G1 negatively regulates thymocyte and peripheral lymphocyte proliferation.
J Immunol, 184 (2010), pp. 173-183
[43]
A.J. Wilhelm, M. Zabalawi, J.S. Owen, D. Shah, J.M. Grayson, A.S. Major, et al.
Apolipoprotein A-I modulates regulatory T cells in autoimmune LDLr−/−, ApoA-I−/− mice.
J Biol Chem, 285 (2010), pp. 36158-36169
[44]
A.J. Wilhelm, M. Zabalawi, J.M. Grayson, A.E. Weant, A.S. Major, J. Owen, et al.
Apolipoprotein A-I and its role in lymphocyte cholesterol homeostasis and autoimmunity.
Arterioscler Thromb Vasc Biol, 29 (2009), pp. 843-849
[45]
A.J. Murphy, K.J. Woollard, A. Suhartoyo, R.a. Stirzaker, J. Shaw, D. Sviridov, et al.
Neutrophil activation is attenuated by high-density lipoprotein and apolipoprotein A-I in in vitro and in vivo models of inflammation.
Arterioscler Thromb Vasc Biol, 31 (2011), pp. 1333-1341
[46]
A.J. Murphy, M. Westerterp, L. Yvan-Charvet, A.R. Tall.
Anti-atherogenic mechanisms of high density lipoprotein: effects on myeloid cells.
Biochim Biophys Acta, 1821 (2012), pp. 513-521
[47]
H. Kaji.
High-density lipoproteins and the immune system.
J Lipids, 2013 (2013), pp. 684903
[48]
L. Perrin-Cocon, O. Diaz, M. Carreras, S. Dollet, A. Guironnet-Paquet, P. André, et al.
High-density lipoprotein phospholipids interfere with dendritic cell Th1 functional maturation.
Immunobiology, 217 (2012), pp. 91-99
[49]
K. Edfeldt, J.H. Swedenborg, G.K. Hansson, Z.Q. Yan.
Expression of toll-like receptors in human atherosclerotic lesions.
Circulation, 105 (2002), pp. 1158-1161
[50]
A.J. Sadler, B.R. Wiliams.
Interferon-inducible antiviral effectors.
Nat Rev Immunol, 8 (2008), pp. 559-568
[51]
D.K. Suzuki Masashi Pritchard, A.N. Becker Lev Hoofnagle, N. Tanimura, T.K. Bammler, R.P. Beyer, K. Miyake, et al.
HDL suppresses the type I interferon response, a family of potent antiviral immunoregullators, in macrophages challenged with lipopolysaccharide.
Circulation, 122 (2010), pp. 1919-1927
[52]
L.E. Smythies, C.R. White, A. Maheshwari, M.N. Palgunachari, G.M. Anantharamaiah, M. Chaddha, et al.
Apolipoprotein A-I mimetic 4F alters the function of human monocyte-derived macrophages.
Am J Physiol Cell Physiol, 298 (2010), pp. C1538-C1548
[53]
A.M. Scanu, C. Edelstein.
HDL: bridging past and present with a look at the future.
FASEB J, 22 (2008), pp. 4044-4054
[54]
K.M. Chavele, M.R. Ehrenstein.
Regulatory T-cells in systemic lupus erythematosus and rheumatoid arthritis.
FEBS Lett, 585 (2011), pp. 3603-3610
[55]
C. Lahoute, O. Herbin, Z. Mallat, A. Tedgui.
Adaptive immunity in atherosclerosis: mechanisms and future therapeutic targets.
Nat Rev Cardiol, 8 (2011), pp. 348-358
[56]
A.H. Lichtman.
T cell costimulatory and coinhibitory pathways in vascular inflammatory diseases.
Front Physiol, 3 (2012), pp. 18
[57]
C. Foks a, V. Frodermann, M. ter Borg, K.L.L. Habets, I. Bot, Y. Zhao, et al.
Differential effects of regulatory T cells on the initiation and regression of atherosclerosis.
Atherosclerosis, 218 (2011), pp. 53-60
[58]
T. Van Es, G.H.M. van Puijvelde, A.C. Foks, K.L.L. Habets, I. Bot, E. Gilboa, et al.
Vaccination against Foxp3(+) regulatory T cells aggravates atherosclerosis.
Atherosclerosis, 209 (2010), pp. 74-80
[59]
N. Hyka, J. Dayer, C. Modoux, T. Kohno, C.K. Edwards, P. Roux-lombard, et al.
Apolipoprotein A-I inhibits the production of interleukin-1 and tumor necrosis factor by blocking contact-mediated activation of monocytes by T lymphocytes.
Blood, 97 (2001), pp. 2381-2389
[60]
E. Maganto-García, M.L. Tarrio, N. Grabie, D. Bu, A.H. Lichtman.
Dynamic changes in regulatory T cells are linked to levels of diet-induced hypercholesterolemia.
Circulation, 124 (2011), pp. 185-195
[61]
P.S. Seifert, G.K. Hansson.
Complement receptors and regulatory proteins in human atherosclerotic lesions.
Arterioscler Thromb Vasc Biol, 9 (1989), pp. 802-811
[62]
R. Vlaicu, F. Niculescu, H.G. Rus.
Immunohistochemical localization of the terminal C5b-9 complement complex in human aortic fibrous plaque.
Atherosclerosis, 57 (1985), pp. 163-177
[63]
R. Oksjoki, H. Jarva, P.T. Kovanen, P. Laine, S. Meri, M.O. Pentikäinen.
Association between complement factor H and proteoglycans in early human coronary atherosclerotic lesions: implications for local regulation of complement activation.
Arterioscler Thromb Vasc Biol, 23 (2003), pp. 630-636
[64]
R. Ajjan, P.J. Grant, T.S. Futers, J.M. Brown, C.M. Cymbalista, M. Boothby, et al.
Complement C3 and C-reactive protein levels in patients with stable coronary artery disease.
Thromb Haemost, 9 (2005), pp. 1048-1053
[65]
a. Muscari, G. Massarelli, L. Bastagli, G. Poggiopollini, V. Tomassetti, G. Drago, et al.
Relationship of serum C3 to fasting insulin, risk factors and previous ischaemic events in middle-aged men.
Eur Heart J, 21 (2000), pp. 1081-1090
[66]
A. Baldo, A.D. Sniderman, S. St-luce, R.K. Avramoglu, M. Maslowska, B. Hoang, et al.
The adipsin-acylation stimulating protein system and regulation of intracellular triglyceride synthesis.
J Clin Invest, 92 (1993), pp. 1543-1547
[67]
D.O. Haskard, J.J. Boyle, J. Mason.
The role of complement in atherosclerosis.
Curr Opin Lipidol, 19 (2008), pp. 478-482
[68]
Y.L. Mamane, C. Chan, G. Lavallee, N. Morin, L.J. Xu, J. Huang, et al.
The C3a anaphylatoxin receptor is a key mediator of insulin resistance and functions by modulating adipose tissue macrophage infiltration and activation.
Diabetes, 58 (2009), pp. 2006-2017
[69]
A.J. Van Oostrom, A. Alipour, T.W. Plokker, A.D. Sniderman, M.C. Cabezas.
The metabolic syndrome in relation to complement component 3 and postprandial lipemia in patients from an outpatient lipid clinic and healthy volunteers.
Atherosclerosis, 190 (2007), pp. 167-173
[70]
A.L. Pasqui, L. Puccetti, G. Bova, M. Di Renzo, F. Bruni, M. Pastorelli, et al.
Relationship between serum complement and different lipid disorders.
Clin Exp Med, 2 (2002), pp. 33-38
[71]
S. Parra, G. Vives, R. Ferré, M. González, M. Guardiola, J. Ribalta, et al.
Complement system and small HDL particles are associated with subclinical atherosclerosis in SLE patients.
Atherosclerosis, 225 (2012), pp. 224-230
[72]
Z. Mallat, A. Tedgui.
HDL,PTX3, and vascular protection.
Arterioscler Thromb Vasc Biol, 28 (2008), pp. 809-811
[73]
C. Garlanda, E. Hirsch, S. Bozza, A. Salustri, M. De Acetis, R. Nota, et al.
Non-redundant role of the long pentraxin PTX3 in anti-fungal innate immune response.
Nature, 420 (2002), pp. 182-186
[74]
G.D. Norata, P. Marchesi, A. Pirillo, P. Uboldi, G. Chiesa, V. Maina, et al.
Long pentraxin 3, a key component of innate immunity, is modulated by high-density lipoproteins in endothelial cells.
Arterioscler Thromb Vasc Biol, 28 (2008), pp. 925-931
[75]
E. Barbati, C. Specchia, M. Villella, M.L. Rossi, S. Barlera, B. Bottazzi, et al.
Influence of pentraxin 3 (PTX3) genetic variants on myocardial infarction risk and PTX3 plasma levels.
[76]
G.D. Norata, P. Marchesi, V.K. Pulakazhi Venu, F. Pasqualini, A. Anselmo, F. Moalli, et al.
Deficiency of the long pentraxin PTX3 promotes vascular inflammation and atherosclerosis.
Circulation, 120 (2009), pp. 699-708
[77]
M. Zanetti, A. Bosutti, C. Ferreira, P. Vinci, G. Biolo, M. Fonda, et al.
Circulating pentraxin 3 levels are higher in metabolic syndrome with subclinical atherosclerosis: evidence for association with atherogenic lipid profile.
Clin Exp Med, 9 (2009), pp. 243-248
[78]
G.D. Norata, C. Garlanda, A.L. Catapano.
The long pentraxin PTX3: a modulator of the immunoinflammatory response in atherosclerosis and cardiovascular diseases.
Trends Cardiovasc Med, 20 (2010), pp. 35-40
[79]
V.A. Blaho, T. Hla.
An update on the biology of sphingosine 1-phosphate receptors.
J Lipid Res, (2014),
[80]
C. Christoffersen, H. Obinata, S.B. Kumaraswamy, S. Galvani, J. Ahnström, M. Sevvana, et al.
Endothelium-protective sphingosine-1-phosphate provided by HDL-associated apolipoprotein M.
Proc Natl Acad Sci U S A, 108 (2011), pp. 9613-9618
[81]
K.M. Argraves, W.S. Argraves.
HDL serves as a S1P signaling platform mediating a multitude of cardiovascular effects.
J Lipid Res, 48 (2007), pp. 2325-2333
[82]
J. Quancard, B. Bollbuck, P. Janser, D. Angst, F. Berst, P. Buehlmayer, et al.
A potent and selective S1P(1) antagonist with efficacy in experimental autoimmune encephalomyelitis.
Chem Biol, 19 (2012), pp. 1142-1151
[83]
J. Chun, T. Hla, K.R. Lynch, S. Spiegel, W.H. Moolenaar.
International Union of Basic and Clinical Pharmacology. LXXVIII. Lysophospholipid receptor nomenclature.
Pharmacol Rev, 62 (2010), pp. 579-587
[84]
J. Chun, V. Brinkmann.
A mechanistically novel, first oral therapy for multiple sclerosis: the development of fingolimod (FTY720, Gilenya).
Discov Med, 12 (2011), pp. 213-228
[85]
S. Mandala, R. Hajdu, J. Bergstrom, E. Quackenbush, J. Xie, J. Milligan, et al.
Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists.
Science, 12 (2002), pp. 346-349
[86]
A. Billich, T. Baumruker, P. Heining, R. Schmouder, G. Francis, A.S. Fingolimod, et al.
(FTY720): discovery and development of an oral drug to treat multiple sclerosis. Brinkmann V1.
Nat Rev Drug Discov, 9 (2010), pp. 883-897
[87]
V. Brinkmann, M.D. Davis, C.E. Heise, R. Albert, S. Cottens, R. Hof, et al.
The immune modulator FTY720 targets sphingosine 1-phosphate receptors.
J Biol Chem, 277 (2002), pp. 21453-21457
[88]
M.L. Oo, S. Thangada, M.T. Wu, C.H. Liu, T.L. Macdonald, K.R. Lynch, et al.
Immunosuppressive and anti-angiogenic sphingosine 1-phosphate receptor-1 agonists induce ubiquitinylation and proteasomal degradation of the receptor.
J Biol Chem, 282 (2007), pp. 9082-9089
[89]
J.a. Cohen, F. Barkhof, G. Comi, H.P. Hartung, B.O. Khatri, X. Montalban, et al.
Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis.
N Engl J Med, 362 (2010), pp. 402-415
[90]
D.P. McCarthy, M.H. Richards, S.D. Miller.
Mouse models of multiple sclerosis: experimental autoimmune encephalomyelitis and Theiler's virus-induced demyelinating disease.
Methods Mol Biol, 900 (2012), pp. 381-401
[91]
J.a. Cohen, J. Chun.
Mechanisms of fingolimod's efficacy and adverse effects in multiple sclerosis.
Ann Neurol, 69 (2011), pp. 759-777
[92]
C.S1 Garris, L. Wu, S. Acharya, A. Arac, V.A. Blaho, Y. Huang, et al.
Defective sphingosine 1-phosphate receptor 1 (S1P1) phosphorylation exacerbates TH17-mediated autoimmune neuroinflammation.
Nat Immunol, 14 (2013), pp. 1166-1172
[93]
H. Takeshita, M. Kitano, T. Iwasaki, S. Kitano, S. Tsunemi, C. Sato, et al.
Sphingosine 1-phosphate (S1P)/S1P receptor 1 signaling regulates receptor activator of NF-(B ligand (RANKL) expression in rheumatoid arthritis.
Biochem Biophys Res Commun, 419 (2012), pp. 154-159
[94]
L.R. Shiow, D.B. Rosen, N. Brdicková, Y. Xu, J. An, L.L. Lanier, et al.
CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs.
Nature, 440 (2006), pp. 540-544
[95]
A.J. Bankovich, L.R. Shiow, J.G. Cyster.
CD69 suppresses sphingosine 1-phosophate receptor-1 (S1P1) function through interaction with membrane helix 4.
J Biol Chem, 285 (2010), pp. 22328-22337
[96]
L. Guangwei, Y. Kai, B. Samir, S. Sharad, C. Hongbo.
S1P1-mTOR axis directs the reciprocal differentiation of TH1 and regulatory T cells.
Nat Immunol, 11 (2011), pp. 1047-1056
[97]
N. Ishimaru, A. Yamada, T. Nitta, R. Arakaki, M. Lipp, Y.H.Y. Takahama.
CCR7 with S1P1 signaling through AP-1 for migration of Foxp3+ regulatory T-cells controls autoimmune exocrinopathy.
Am J Pathol, 180 (2012), pp. 199-208
[98]
L.D. Serpero, G. Filaci, A. Parodi, F. Battaglia, F. Kalli, D. Brogi, et al.
Fingolimod modulates peripheral effector and regulatory T cells in MS patients.
J Neuroimmune Pharmacol, 8 (2013), pp. 1106-1113
[99]
J-R. Nofer, M. Bot, M. Brodde, P.J. Taylor, P. Salm, V. Brinkmann, et al.
FTY720, a synthetic sphingosine 1 phosphate analogue, inhibits development of atherosclerosis in low-density lipoprotein receptor-deficient mice.
Circulation, 115 (2007), pp. 501-508
[100]
S.B. Kumaraswamy, A. Linder, P. Åkesson, B. Dahlbäck.
Decreased plasma concentrations of apolipoprotein M in sepsis and systemic inflammatory response syndromes.
Crit Care, 16 (2012), pp. R60
[101]
Y. Shoenfeld, R. Gerli, A. Doria, E. Matsuura, M.M. Cerinic, N. Ronda, et al.
Accelerated atherosclerosis in autoimmune rheumatic diseases.
Circulation, 112 (2005), pp. 3337-3347
[102]
J.E. Salmon, M.J. Roman.
Subclinical atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus.
[103]
J. Nossent, N. Cikes, E. Kiss, a. Marchesoni, V. Nassonova, M. Mosca, et al.
Current causes of death in systemic lupus erythematosus in Europe, 2000–2004: relation to disease activity and damage accrual.
Lupus, 16 (2007), pp. 309-317
[104]
M.M. Ward.
Premature morbidity from cardiovascular and cerebrovascular diseases in women with systemic lupus erythematosus.
[105]
M.J. Roman, B-A. Shanker, A. Davis, M.D. Lockshin, L. Sammaritano, R. Simantov, et al.
Prevalence and correlates of accelerated atherosclerosis in systemic lupus erythematosus.
N Engl J Med, 349 (2003), pp. 2399-2406
[106]
M.J. Roman, M.K. Crow, M.D. Lockshin, R.B. Devereux, S.a. Paget, L. Sammaritano, et al.
Rate and determinants of progression of atherosclerosis in systemic lupus erythematosus.
Arthritis Rheum, 56 (2007), pp. 3412-3419
[107]
J.M. Esdaile, M. Abrahamowicz, T. Grodzicky, Y. Li, C. Panaritis, R. du Berger, et al.
Traditional Framingham risk factors fail to fully account for accelerated atherosclerosis in systemic lupus erythematosus.
Arthritis Rheum, 44 (2001), pp. 2331-2337
[108]
J.M. Sabio, J. Vargas-Hitos, M. Zamora-Pasadas, J.D. Mediavilla, N. Navarrete, A. Ramirez, et al.
Metabolic syndrome is associated with increased arterial stiffness and biomarkers of subclinical atherosclerosis in patients with systemic lupus erythematosus.
J Rheumatol, 36 (2009), pp. 2204-2211
[109]
C.P. Chung, I. Avalos, A. Oeser, T. Gebretsadik, A. Shintani, P. Raggi, et al.
High prevalence of the metabolic syndrome in patients with systemic lupus erythematosus: association with disease characteristics and cardiovascular risk factors.
Ann Rheum Dis, 66 (2007), pp. 208-214
[110]
C. López-Pedrera, M.Á. Aguirre, N. Barbarroja, M.J. Cuadrado.
Accelerated atherosclerosis in systemic lupus erythematosus: role of proinflammatory cytokines and therapeutic approaches.
J Biomed Biotechnol, (2010), pp. 2010
[111]
R.J. Goldberg, M.B. Urowitz, D. Ibañez, M. Nikpour, D.D. Gladman.
Risk factors for development of coronary artery disease in women with systemic lupus erythematosus.
J Rheumatol, 36 (2009), pp. 2454-2461
[112]
M. Gonzàlez, J. Ribalta, G. Vives, S. Iftimie, R. Ferré, N. Plana, et al.
Nuclear magnetic resonance lipoprotein subclasses and the APOE genotype influence carotid atherosclerosis in patients with systemic lupus erythematosus.
J Rheumatol, 37 (2010), pp. 2259-2267
[113]
M. Vadacca, D. Margiotta, A. Rigon, F. Cacciapaglia, G. Coppolino, A. Amoroso, et al.
Adipokines and systemic lupus erythematosus: relationship with metabolic syndrome and cardiovascular disease risk factors.
J Rheumatol, 36 (2009), pp. 295-297
[114]
S. Parra, a. Cabré, F. Marimon, R. Ferré, J. Ribalta, M. Gonzàlez, et al.
Circulating FABP4 is a marker of metabolic and cardiovascular risk in SLE patients.
Lupus, 23 (2014), pp. 245-254
[115]
J.F. De Carvalho, E. Bonfá, E.F. Borba.
Systemic lupus erythematosus and lupus dyslipoproteinemia.
Autoimmun Rev, 7 (2008), pp. 246-250
[116]
L.R. Lopez, M. Salazar-Paramo, C. Palafox-Sanchez, B.L. Hurley, E. Matsuura, I. Garcia-De La Torre.
Oxidized low-density lipoprotein and beta2-glycoprotein I in patients with systemic lupus erythematosus and increased carotid intima-media thickness: implications in autoimmune-mediated atherosclerosis.
Lupus, 15 (2006), pp. 80-86
[117]
M. McMahon, B.H. Hahn.
Atherosclerosis and systemic lupus erythematosus: mechanistic basis of the association.
Curr Opin Immunol, 19 (2007), pp. 633-639
[118]
M. McMahon, B.J. Skaggs, J.M. Grossman, L. Sahakian, J. Fitzgerald, W.K. Wong, et al.
A panel of biomarkers is associated with increased risk of the presence and progression of atherosclerosis in women with systemic lupus erythematosus.
Arthritis Rheum (Hoboken, NJ), 66 (2014), pp. 130-139
[119]
L.M. Tripi, S. Manzi, Q. Chen, M. Kenney, P. Shaw, A. Kao, et al.
Relationship of serum paraoxonase 1 activity and paraoxonase 1 genotype to risk of systemic lupus erythematosus.
Arthritis Rheum, 54 (2006), pp. 1928-1939
[120]
E. Kiss, I. Seres, T. Tarr, Z. Kocsis, G. Szegedi, G. Paragh.
Reduced paraoxonase1 activity is a risk for atherosclerosis in patients with systemic lupus erythematosus.
Ann N Y Acad Sci, 1108 (2007), pp. 83-91
[121]
C. Charles-Schoeman, Y.Y. Lee, A. Shahbazian, A.H. Gorn, J. Fitzgerald, V.K. Ranganath, et al.
Association of paraoxonase 1 gene polymorphism and enzyme activity with carotid plaque in rheumatoid arthritis.
Arthritis Rheum, 65 (2013), pp. 2765-2772
[122]
A. Kirbas, S. Kirbas, O. Anlar, H. Efe, A. Ylimaz.
Serum paraoxonase and arylesterase activity and oxidative status in patients with multiple sclerosis.
J Clin Neurosci, 20 (2013), pp. 1106-1109
[123]
G1 Ferretti, T. Bacchetti, A. Campanati, O. Simonetti, G.O.A.B.J.D. Liberati.
Correlation between lipoprotein(a) and lipid peroxidation in psoriasis: role of the enzyme paraoxonase-1.
Br J Dermatology, 166 (2012), pp. 204-207
[124]
S.G. O’Neill, I. Giles, A. Lambrianides, J. Manson, D. D’Cruz, L. Schrieber, et al.
Antibodies to apolipoprotein A-I, high-density lipoprotein, and C-reactive protein are associated with disease activity in patients with systemic lupus erythematosus.
Arthritis Rheum, 62 (2010), pp. 845-854
[125]
J.R. Batuca, P.R. Ames, M. Amaral, C. Favas, D.A. Isenberg, J. Delgado Alves.
Anti-atherogenic and anti-inflammatory properties of high-density lipoprotein are affected by specific antibodies in systemic lupus erythematosus.
Rheumatology (Oxford), 48 (2009), pp. 26-31
[126]
J.R. Batuca, P.R. Ames, D.A. Isenberg, J.D. Alves.
Antibodies toward high-density lipoprotein components inhibit paraoxonase activity in patients with systemic lupus erythematosus.
Ann N Y Acad Sci, 1108 (2007), pp. 137-146
[127]
N. Ronda, E. Favari, M.O. Borghi, F. Ingegnoli, M. Gerosa, C. Chighizola, et al.
Impaired serum cholesterol efflux capacity in rheumatoid arthritis and systemic lupus erythematosus.
Ann Rheum Dis, 73 (2014), pp. 609-615
[128]
M. Holzer, P. Wolf, S. Curcic, R. Birner-Gruenberger, W. Weger, M. Inzinger, et al.
Psoriasis alters HDL composition and cholesterol efflux capacity.
J Lipid Res, 53 (2012), pp. 1618-1624
[129]
M. Zabalawi, S. Bhat, T. Loughlin, M.J. Thomas, E. Alexander, M. Cline, et al.
Induction of fatal inflammation in LDL receptor and ApoA-I double-knockout mice fed dietary fat and cholesterol.
Am J Pathol, 163 (2003), pp. 1201-1213
[130]
J.M. Woo, Z. Lin, M. Navab, C. Van Dyck, Y. Trejo-Lopez, K.M. Woo, et al.
Treatment with apolipoprotein A-1 mimetic peptide reduces lupus-like manifestations in a murine lupus model of accelerated atherosclerosis.
Arthritis Res Ther, 12 (2010), pp. R93
[131]
M.I. Kamboh, D.K. Sanghera, H. Mehdi, C.S. Nestlerode, Q. Chen, O. Khalifa, et al.
Single nucleotide polymorphisms in the coding region of the apolipoprotein H (beta2-glycoprotein I) gene and their correlation with the protein polymorphism anti-beta2glycoprotein I antibodies and cardiolipin binding: description of novel haplotypes and their evolution.
Ann Hum Genet, 68 (2004), pp. 285-299
[132]
N.S. Lee, H.B. Brewer, J.C. Osborne.
Beta-2 glycoprotein I. Molecular properties of an unusual apolipoprotein, apolipoprotein H.
J Biol Chem, 258 (1983), pp. 4765
[133]
E. Polz, G. Kostner.
Binding of beta-2-glycoprotein-I to human-serum lipoproteins–distribution among density fractions.
FEBS Lett, 102 (1979), pp. 183-186
[134]
B. Giannakopoulos, S.A. Krilis.
The pathogenesis of the antiphospholipid syndrome.
N Engl J Med, 368 (2013), pp. 1033-1044
Copyright © 2014. Sociedad Española de Arteriosclerosis
Opciones de artículo
Herramientas
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

Quizás le interese:
10.1016/j.arteri.2019.03.004
No mostrar más