Role of the complement system in rheumatoid arthritis and psoriatic arthritis: Relationship with...

7
Review Role of the complement system in rheumatoid arthritis and psoriatic arthritis: Relationship with anti-TNF inhibitors Eleonora Ballanti a , Carlo Perricone b , Gioia di Muzio a , Barbara Kroegler a , Maria Sole Chimenti a , Dario Graceffa a , Roberto Perricone a, a Rheumatology, Allergology and Clinical Immunology, Department of Internal Medicine, University of Rome Tor Vergata, Rome, Italy b Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy abstract article info Available online 27 April 2011 Keywords: Complement system Rheumatoid arthritis Psoriatic arthritis Anti-TNF agents Biomarkers The complement system is an essential component of innate immunity and also plays an important role in modulating adaptive immunity. It comprises more than 30 plasma and membrane-bound proteins and can be activated through three pathways: the classical, the alternative and the lectin pathways. Its activation contributes to the pathogenesis of several autoimmune and inammatory conditions. The evidence of complement activation in synovial uid of Rheumatoid Arthritis (RA) patients is abundant, while few data exist in Psoriatic Arthritis (PsA) patients. Levels of complement proteins are generally depressed in the synovial uid of patients with RA, reecting consumption of complement. On the other hand, elevated levels of several complement cleavage products have been observed in synovial uid. Involvement of complement in the pathogenesis of RA was also conrmed in animal models of arthritis: mice decient for complement proteins are protected against the development of collagen-induced arthritis and administration of the anti- C5 monoclonal antibody prevents the onset of this arthritis. In the last decade anti-tumor necrosis factor agents have shown to be effective for the treatment of both RA and PsA and some studies suggest that the interaction between TNFα and complement system may contribute to the pathogenesis of these diseases. Reduction of the complement activation could be one of the mechanism by which TNFα-inhibitors exert their effectiveness in inammatory arthritides. Because of these ndings, complement could be an attractive therapeutic target both in RA and in PsA. © 2011 Elsevier B.V. All rights reserved. Contents 1. Introduction to the complement system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618 1.1. Pathways of complement activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618 1.2. Complement and inammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618 1.3. Complement and autoimmunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 619 2. Complement and rheumatoid arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 619 2.1. Where complement components are synthesized in RA? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 619 2.2. Activation of the complement system in RA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 620 2.3. Complement and collagen-induced arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 620 3. Complement and psoriatic arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 620 4. Complement and anti-TNFα agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621 5. Potential therapies targeting complement cascade in inammatory arthritides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621 6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622 Take-home messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622 Autoimmunity Reviews 10 (2011) 617623 Corresponding author at: Department of Internal Medicine, Unit of Rheumatology, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy. Tel.: + 39 06 72596287; fax: +39 06 20900358. E-mail address: [email protected] (R. Perricone). 1568-9972/$ see front matter © 2011 Elsevier B.V. All rights reserved. doi:10.1016/j.autrev.2011.04.012 Contents lists available at ScienceDirect Autoimmunity Reviews journal homepage: www.elsevier.com/locate/autrev

Transcript of Role of the complement system in rheumatoid arthritis and psoriatic arthritis: Relationship with...

Autoimmunity Reviews 10 (2011) 617–623

Contents lists available at ScienceDirect

Autoimmunity Reviews

j ourna l homepage: www.e lsev ie r.com/ locate /aut rev

Review

Role of the complement system in rheumatoid arthritis and psoriatic arthritis:Relationship with anti-TNF inhibitors

Eleonora Ballanti a, Carlo Perricone b, Gioia di Muzio a, Barbara Kroegler a, Maria Sole Chimenti a,Dario Graceffa a, Roberto Perricone a,⁎a Rheumatology, Allergology and Clinical Immunology, Department of Internal Medicine, University of Rome Tor Vergata, Rome, Italyb Reumatologia, Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy

⁎ Corresponding author at: Department of Internal Mefax: +39 06 20900358.

E-mail address: [email protected] (R. P

1568-9972/$ – see front matter © 2011 Elsevier B.V. Aldoi:10.1016/j.autrev.2011.04.012

a b s t r a c t

a r t i c l e i n f o

Available online 27 April 2011

Keywords:Complement systemRheumatoid arthritisPsoriatic arthritisAnti-TNF agentsBiomarkers

The complement system is an essential component of innate immunity and also plays an important role inmodulating adaptive immunity. It comprises more than 30 plasma and membrane-bound proteins and can beactivated through three pathways: the classical, the alternative and the lectin pathways. Its activationcontributes to the pathogenesis of several autoimmune and inflammatory conditions. The evidence ofcomplement activation in synovial fluid of Rheumatoid Arthritis (RA) patients is abundant, while few dataexist in Psoriatic Arthritis (PsA) patients. Levels of complement proteins are generally depressed in thesynovial fluid of patients with RA, reflecting consumption of complement. On the other hand, elevated levelsof several complement cleavage products have been observed in synovial fluid. Involvement of complementin the pathogenesis of RA was also confirmed in animal models of arthritis: mice deficient for complementproteins are protected against the development of collagen-induced arthritis and administration of the anti-C5 monoclonal antibody prevents the onset of this arthritis. In the last decade anti-tumor necrosis factoragents have shown to be effective for the treatment of both RA and PsA and some studies suggest that theinteraction between TNFα and complement system may contribute to the pathogenesis of these diseases.Reduction of the complement activation could be one of the mechanism by which TNFα-inhibitors exert theireffectiveness in inflammatory arthritides. Because of these findings, complement could be an attractivetherapeutic target both in RA and in PsA.

dicine, Unit of Rheumatology, University of Rome Tor Ver

erricone).

l rights reserved.

© 2011 Elsevier B.V. All rights reserved.

Contents

1. Introduction to the complement system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6181.1. Pathways of complement activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6181.2. Complement and inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6181.3. Complement and autoimmunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 619

2. Complement and rheumatoid arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6192.1. Where complement components are synthesized in RA? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6192.2. Activation of the complement system in RA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6202.3. Complement and collagen-induced arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 620

3. Complement and psoriatic arthritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6204. Complement and anti-TNFα agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6215. Potential therapies targeting complement cascade in inflammatory arthritides . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6216. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622Take-home messages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622

gata, Via Montpellier 1, 00133 Rome, Italy. Tel.: +39 06 72596287;

618 E. Ballanti et al. / Autoimmunity Reviews 10 (2011) 617–623

1. Introduction to the complement system

Complement is part of the innate immune system. The term “innateimmunity” was introduced to define the protective mechanismsagainst infection that operate in the absence of specific “adaptive”immunity [1]. Phylogenetically, themajor function of the complementsystem has been thought to be recognition and elimination ofpathogens through direct killing and/or stimulation of phagocytosis[2,3], but, in recent years, also immunoregulatory functions of thecomplement systemhave become clear and it has been discovered thatthe complement proteins play an important role in modulatingadaptive immunity [4]. Given these multiple functions exerted innormal and pathologic conditions, complement provides an uniqueconnection between innate and adaptive immune responses, with itsinvolvement in the pathogenetic mechanisms of various diseasesranging from protective functions to tissue damage [1,4].

1.1. Pathways of complement activation

The complement system comprises more than 30 plasma andmembrane-bound proteins [5,6]. There are three pathways ofcomplement activation, the classical, the alternative and the lectinpathways. All three pathways are activated according to a cascadesystem, with activation of one factor leading to the activation of thenext (Fig. 1). Activation of the classical pathway is dependent onimmunoglobulins (Ig), IgM or IgG, present in immune complexes,leading to binding of the C1 complex via the C1q subunit [7]. Bindingof C1q to antibody induces conformational changes in the C1 complexand leads to activation of C4 and C2 resulting in the formation of theC4bC2a complex, the classical pathway C3 convertase [8]. In the caseof the alternative pathway, activation of a serine protease, factor D,

Fig. 1. Activation pathways of the complement system. There are three pathways of complemeactivated according to a cascade system, with activation of one factor leading to activation of thactivation of C3. The classical and lectin pathways generate the same C3 convertase (C4b2a), wclassical and lectinpathways generate the sameC5convertase (C4b2a3b),whereas the alternatithe activation of C5 to formC5a (apotent chemoattractant) and themembrane attack complex (complement on self, there aremultiple complement regulators in plasma and on host cells (reppentraxin-3; C1INH, C1-inhibitor;MCP,membrane cofactor protein; DAF, decay-accelerating facMASPs, MBL-associated serine proteases.

cleaves factor B into Ba and Bb when factor B is complexed withspontaneously hydrolyzed iC3b. Bb is a serine protease that generatesthe C3 convertase of the alternative pathway, C3bBb. Properdinincreases the stability of this enzyme [1].

Activation of the lectin pathway occurs in response to recognitionof mannose-binding lectin and ficolins of various carbohydrateligands on the surface of microorganisms [9]. This results in activationof the mannose-binding lectin-associated serine proteases (MASP-1,MASP-2 and MASP-3). MASP-2, which is the key enzyme of the lectinpathway, cleaves C4 and subsequently C2, leading to the formation ofthe same C3 convertase as in the classical pathway, that is C4bC2a [8].Activation of the complement system through any of the threepathways leads to activation of C3. The classical and lectin pathwaysgenerate the same C3 convertase, whereas the alternative pathwaygenerates a different C3 convertase. Likewise, the classical and lectinpathways generate the same C5 convertase (C3bC4bC2a), whereasthe alternative pathway generates a different C5 convertase(C3bBbC3b) [9]. The three pathways converge at the activation ofC5 to form C5a (a potent chemoattractant) and the membrane attackcomplex (MAC) C5b-9, which can lead to cell lysis [9].

To minimize damage induced by over-activation of complementon self, there are multiple complement regulators in plasma and onhost cells that control the complement pathway to maintain theintegrity of the host [10].

1.2. Complement and inflammation

The regulatory mechanisms of complement are finely balanced sothat, on the one hand, the activation of complement is focused on thesurface of invading microorganisms and, on the other hand, thedeposition of complement on normal cells and tissues is limited [5].

nt activation: the classical, the lectin and the alternative pathway. All three pathways aree next. Activation of the complement system through any of the three pathways leads tohereas the alternative pathway generates a different C3 convertase (C3bBb). Likewise, thevepathwaygenerates a different C5convertase (C3bBb3b). The threepathways convergeatMAC)C5b-9,which can lead to cell lysis. Tominimizedamage inducedbyover-activation ofresented in the circles). Ag–Ab, antigen–antibody complex; CRP, C-Reactive Protein; PTX3,tor; CR1, complement receptor 1; C4BP, C4-binding protein;MBL,mannose binding lectin;

619E. Ballanti et al. / Autoimmunity Reviews 10 (2011) 617–623

When themechanisms that regulate this delicate balance go awry, thecomplement system may cause injury, by induction and augmenta-tion of inflammation [5]. Several studies demonstrated the involve-ment of complement in inflammatory tissue destruction. The twomeans by which complement is activated in tissues are throughimmune complexes, which activate the classical complement path-way, and through tissue ischemia and reperfusion, which exposephospholipids and mitochondrial proteins [6]. These activate com-plement directly by binding C1q or mannose-binding lectin orindirectly by binding natural antibodies or C-reactive protein (CRP),which can activate the classical pathway by binding C1q. Necrotic cellsand tissues lack the regulatory molecules that in normal tissuesprevent the binding of complement [6]. Undesired complementactivation results in the formation of several proinflammatorymediators, including the anaphylatoxins C3a and C5a. Furthermore,the MAC can contribute to inflammation and tissue injury [11]. C5a isa potent chemoattractant for neutrophils, monocytes and eosinophilsand both C3a and C5a can stimulate the synthesis of pro-inflammatorymediators [11]. Formation of the terminal complement complex C5b-9 can lead to cell activation, proliferation and cell death by necrosis orapoptosis [11]. There is accumulating evidence that complementactivation is an important contributor to the tissue necrosis thatfollows ischemia. For instance, myocardial infarction and stroke areeach associated with the activation of complement in the area oftissue infarction [6,12].

1.3. Complement and autoimmunity

The complement system participates in the pathogenesis andcontributes to the clinical manifestations of many systemic autoim-mune diseases, such as systemic lupus erythematosus (SLE), vascu-litis, anti-glomerular basement membrane disease, anti-phospholipidantibody syndrome, systemic sclerosis, Sjögren's syndrome, derma-tomyositis and rheumatoid arthritis (RA) [9]. The meticulous study ofpatients with abnormalities of the complement system has illumi-nated our understanding of the immunobiology of complement [5,13].Hereditary homozygous deficiencies of C1q, C1r and C1s, and C4 areeach strongly associated with susceptibility to SLE, with respectivefrequencies of 93%, 57% (since deficiencies of C1r and C1s are usuallyinherited together), and 75% [6]. By contrast, the prevalence of SLEamong subjects with C2 deficiency, which is the most frequenthereditary deficiency in complement classical pathway components,is associated with SLE in 10% of the cases [14]. In the case of C3deficiency, the clinical picture is rather different [6]. It is characterizedby recurrent pyogenic infections, membranoproliferative glomerulo-nephritis and rashes. There are several theories that have beenproposed to explain the association between complement componentdeficiencies and autoimmune disease. Most of this focus on theinability to normally clear immune complexes in the absence ofnormal levels of complements components [15]. Excess immunecomplexes may deposit in tissues, resulting in inflammatory damageand release of autoantigens and initiating an autoimmune response.Alternatively, the presence of apoptotic cells in high concentrationsdue to poor clearance may be sufficient to elicit an autoimmuneresponse [14]. A third hypothesis suggests that the complementsystem plays an important role in the development of toleranceagainst self [16]. Complement is needed for the elimination of self-reactive lymphocytes during maturation of the immune system. Thus,complement deficiency will result in lack of normal B-cell toleranceand production of autoantibodies [17,18].

2. Complement and rheumatoid arthritis

RA is a systemic disease characterized by chronic inflammation ofthe synovium and subsequent destruction of cartilage and bone.

To date, the pathogenesis of RA is not fully understood but there isincreasing evidence for an important role of components of thecomplement cascade in the pathophysiology of RA. In general, thecomplement cascade is involved in the induction and progression ofinflammation reactions and is a major defense system against variouspathogenic agents, including bacteria, viruses, and other antigens[19–21]. Inappropriate activation, however, can lead to tissue damageand manifestation of disease. Although increased complementactivation is potentially related to the occurrence and/or augmenta-tion of inflammation in RA, complement deficiency may induce RA.C1q deficiency and suppression are related to the development of RA[10] as well as C2 deficiency [22]. Association with autoimmunediseases, including RA, has been shown for deficiencies of othercomplement components, including C1r and C1s, C4, C7, C9 and factorI [10]. In the lectin pathway, MBL deficiency while not directlyassociated with the occurrence of RA, is associated with diseaseseverity [23].

2.1. Where complement components are synthesized in RA?

The plasma levels of most complement components are main-tained by hepatic synthesis. Studies on the acute phase response haveshown that complement proteins may act as acute phase proteins,with increased hepatocyte synthesis occurring in response tocytokines such as interleukin 1 (IL-1), interleukin 6 (IL-6), tumornecrosis factor-α (TNF-α) and interferon-γ [24–27]. Complementfactors and receptors have been called to play an important role in RAsince the first report from Brodeur in 1991 [28], and it is evident thatsynthesis and activation of complement take place at distinct siteswithin rheumatoid synovium. Recently, we evaluated plasma levels ofcomplement components in 114 patients with active RA (patientswith an established disease, and suffering from a moderate to severedisease).We demonstrated that the RA patients showmean C3 and C4levels significantly higher than controls and similar CH50. Splitproducts of C3 and factor B were never detected in the patients'plasma (unpublished results). We speculated that the presence ofelevated complement levels may reflect the presence of an under-lining inflammatory process, as previously reported, and complementcomponents can be considered acute phase proteins. Complementactivation probably occurred mainly in the joint space, and these highplasma levels were due to spill over from the joints.

Nevertheless, synthesis and activation of complement proteinsalso occurs in extrahepatic sites. To date it has been shown that allnormal tissues are able to synthesize at least one complementcomponent [29]. In addition, synthesis of complement componentsoccurs in chronically inflamed tissues such as the rheumatoid joint[30]. A large number of different cell types have been shown tosynthesize complement components. These include mononuclearphagocytes, fibroblasts, endothelial cells, epithelial cells, alveolar type2 epithelial cells and adipocytes [31–33]. In particular, the cell ofsynovial membrane which are responsible for the synthesis ofcomplement components are the lining cells (type A-mononuclearphagocyte, type B-fibroblast like), fibroblasts, mononuclear phago-cytes and endothelial cells [34]. In 1974 Ruddy and Coltendemonstrated synthesis of complement components C2, C3, C4 andC5 by rheumatoid synovial tissue by three technics: in vivo metabolicstudies with Radio-labeled C3; in vitro biosynthesis of C2, C3, C4 andC5; staining of cell in rheumatoid synovium with fluorescentantiserums to C3 and C4 [29]. More recently, in a very interestingstudy, Neumann et al. demonstrated that complement componentsand receptors are readily produced in situ in the inflamed RAsynovium [35]. Using in situ hybridization, immunohistochemistry,and Western blot techniques, they localized the sites of complementmessenger RNA (mRNA) expression in rheumatoid synovium.Specifically, they found increased expression of mRNA for C3 andFB, as previously reported [36] and for the first time they reported

620 E. Ballanti et al. / Autoimmunity Reviews 10 (2011) 617–623

increased expression of mRNA for C3a and C5a receptors in synovialtissue [35]. These findings support the idea that they are producedlocally in RA synovium and are not plasma derived.

2.2. Activation of the complement system in RA

Various studies identify complement activation as a main event inthe inflammatory cascade in RA [37–39]. Complement is traditionallyconsidered to be mainly activated by bacteria or immune complexes.RA patients have increased levels of circulating immune complexes[40,41]. Part of these complexes contains rheumatoid factors (RFs),which are autoantibodies against human IgG. RF-containing immunecomplexes are capable of activating complement via the classicalpathway [42–45], with IgM-RF being considerably more effective incomplement activation than IgG-RF [46].

Trouw et al., from in vitro observations, suggested that anti-cycliccitrullinated peptide/protein antibodies (anti-CCP) from RA patientsmay activate complement via both the classical and the alternativepathways [47]. This interesting observation needs to be furtherconfirmed by in vivo analysis.

It is not clear to what extent circulating immune complexescontribute to complement activation in RA. Since there is noconclusive evidence that immune complexes are the main triggerfor complement activation in RA, other potential triggers forcomplement activation in RA should be considered. One of thesetriggers could be C-reactive protein (CRP), since this acute-phaseprotein can activate complement both in vitro and in vivo [48–51]. Inan interesting study Moleenar et al. demonstrated that the plasmalevels of activated complement and CRP–complement complexes areincreased in the majority of patients with RA and that these levels arecorrelated with parameters of disease activity [52]. The observation ofincreased levels of CRP–complement complexes in this study points tothe involvement of CRP in complement activation and to a possiblerole of CRP-mediated complement activation in the pathogenesis ofRA [52].

Furthermore, it appears that molecules present in or released fromcartilage, as fibromodulin (FM), may have a role in activatingcomplement [53]. FM, especially after cartilage degradation producedby metalloproteinases, interacts with the globular head of C1q,activating the classical pathway of complement [53]. Interestingly,other molecules of cartilage, such as decorin and biglycan, are able tobind C1q, but they inhibit the ability of C1 to activate the classicalpathway [54]. Probably the final outcome depends on a balancebetween several local factors.

Both classic and alternative pathways appear to contribute tocomplement activation in RA [55]. If the classical pathway is the maincomplement pathway triggered [55], presumably via binding ofimmune complexes containing rheumatoid factor, the alternativepathway is also activated in RA synovium as demonstrated bydecreased synovial fluid concentration of factor B and properdinand increased levels of Ba [56]. Increased complement activation viathe lectin pathway could also play a role in RA. Changes inglycosylation of IgG in RA cause an increase in binding of mannose-binding lectin resulting in increased complement activation [57].

Evidence of complement activation in synovial fluid is abundant.For example, levels of complement proteins are generally depressedin the synovial fluid of patients with RA, reflecting consumption ofcomplement. Aggarwal et al. recently demonstrated the activation ofthe alternative pathway with formation of the membrane terminalattack complex in patients with juvenile RA [58]. This provided moreconclusive evidence for the pathogenetic significance of complementpathway activation in RA. Moreover, elevated levels of severalcomplement cleavage products, such as C3a, C3c, C5a, sC5b-9, Bb,C1–C1INH complexes have been observed in synovial fluid [59–62].Some studies demonstrated low levels of MAC (membrane attackcomplex) inhibitors in synovial fluid or synovial tissue of patients

with RA, and this might allow lytic or sublytic attacks on local cells[61,63].

One problem with the detection of complement activationproducts is that complement system is susceptible to in vitroactivation, resulting in artificially high levels of activation productsin plasma samples. Wouters et al. recently described novel activationproducts of complement which consists of covalent complexesbetween C1q and activated C4 [64]. This new marker is not onlyspecific for activation of the classical pathway of complement, but alsois remarkably stable in plasma, permitting to avoid artifacts [64].

Several studies failed to demonstrate a correlation between localand systemic activation of complement system in RA and reportedhigher levels of complement cleavage products in synovial fluid thanin plasma [59,65,66], and these findings are consistent with aprevalent local activation of the complement cascade. Associationbetween complement activation and disease activity is another aspectof great interest. Again in the study from Wouters and coworkers, acorrelation between plasma levels of C1q-C4 complexes and diseaseactivity expressed as DAS28 was found [64], likewise Doherty et al.reported raised synovial C3d levels in active compared with inactiveRA joints [59].

2.3. Complement and collagen-induced arthritis

Involvement of complement in the pathogenesis of RA was alsoconfirmed in experimental studies with animal models. In 1977Trentham et al., reported that immunization of rats with type IIcollagen (CII) induce an autoimmune arthritis that in many waysresembled RA [67]. Following immunization with CII, these ratsdeveloped an erosive, polyarticular arthritis mediated by an autoim-mune response to the rat CII. Similarly, this same collagen-inducedarthritis (CIA) model has been reproduced in the mouse [68] and inprimates [69,70]. The significance of this model is that CII is the majorconstituent protein of cartilage in diarthrodial joints, the predominantsite of inflammation in RA. In addition, the pathogenesis of CIA is inmany ways similar to that of RA [71].

In 2002Hietala et al. demonstrated that themice deficient for C3 orFB and immunized with bovine CII displayed reduction or almostcomplete absence of clinically or histologically verifiable arthritis [72].These results showed that the classical pathway and possibly also thealternative pathway are involved in the pathogenesis of CIA [72].

According to these findings, Wang et al. had previously shown thatsystemic administration of the anti-C5 monoclonal antibody (mAb)effectively inhibits terminal complement activation in vivo and thatanti-C5 mAb treatment prevents the onset of arthritis in mice and isalso highly effective in ameliorating established disease [73]. Theseresults demonstrated an important role for activated terminalcomplement components not only in the induction but also in theprogression of disease in this model of immune-mediated jointinflammation.

3. Complement and psoriatic arthritis

Psoriatic arthritis (PsA) is a chronic, inflammatory arthritiscommonly associated with psoriasis. Ten to 30% of the patients withpsoriasis develop PsA. Skin involvement precedes joint symptoms inmost cases, however, articular involvement at disease onset withoutskin lesions occurs in 15% of PsA patients [74]. PsA pathogenesis is stillincompletely understood, but a role for innate immunity has beenrecognized [75].

Data on the role of complement in PsA are poor in the literature.Early studies on psoriatic patients have shown increased plasmaconcentrations of iC3b, C4d, and Bb fragments, especially in patientswith erythrodermic pustular psoriasis and in patients with PsA [76].

Partsch et al. have shown that synovial fluid (SF) from patientswith PsA exhibit a relatively low percentage of the C3c cleavage

621E. Ballanti et al. / Autoimmunity Reviews 10 (2011) 617–623

product, in similar amounts than in patients with osteoarthritis (OA)[77]. The same authors showed that the PsA SF displays the highest C3concentration when compared with RA and OA, and it might also behelpful for establishing the differential diagnosis of PSA vs RA on thebasis of the C3 level of the SF in those patients where an elevated levelof C3 is present [77].

A possible implication of complement activation in the pathogen-esis of PsA is supported by the finding of impaired expression ofcomplement regulators in these patients. Triolo et al. demonstrated alow expression of erythrocyte membrane-anchored CD59, which is animportant membrane inhibitor of MAC [78]. Consistently with thisobservation, increased SC5b-9 was seen in the plasma of patients withactive disease and an inverse correlation was also found betweenplasma SC5b-9 and the CD59 expression levels [78]. Consideringerythrocyte CD59 levels as representative of the CD59 tissuedistribution, the authors supposed that reduced CD59 expressioncould facilitate the activation of complement pathway and exacerbatesynovial cell injury [78].

In another interesting study published in 1994, the complementreceptor type 1 (CRl) levels on erythrocyte membranes from 23patients with PsAwas determined using ELISA [79]. The authors founda statistically significant decrease of CR1 density in PsA patients withpoliarthritis compared with controls. Moreover, the CRl concentrationhad an inverse correlation with the articular index of patients (but nocorrelation was found with CRP and ESR levels), which indicated thatthe decreased concentration of CRl in these patients could be relatedto the severity of disease [79].

Our group has recently conducted a study evaluating thecomplement system in PsA treated with anti-TNF, in which wedemonstrated that PsA patientswithmoderate to severe disease showhigher plasma levels of C3 and C4, compared with healthy subjects(unpublished results). In addition, we evaluated the release of splitproducts of C3 and B, to verify complement activation. These cleavagefragments were not detected suggesting that complement proteinsmay contribute to the acute phase response. In this view, in our studyit is not surprising to find elevated baseline C3 and C4 levels inpatients with a moderate to severe disease activity (unpublishedresults).

4. Complement and anti-TNFα agents

TNFα is a potent proinflammatory cytokine with pleiotropiceffects and represents one of the major players in the inflammatorycascade [80]. In the last decade anti-TNF agents have shown to beeffective for treatment of both RA [81–84] and PsA [85–88]. Thesefindings support the involvement of TNF in the pathogenesis of theserheumatic diseases. Thus, there is a strong evidence for an importantrole of both complement system and TNFα in the pathophysiology ofRA and PsA. However, whether there is any interaction between thesepathogenetic elements still is a controversial issue. In previous studiesTNFα has been reported to interact with complement system, byincreasing the synthesis of factor B and C3 in human hepatoma celllines [89].

Moreover Brodeur et al. reported that the mean levels of TNF in RAsynovial fluids were over 2-fold higher than those in degenerativejoint diseases and that TNF levels correlate positively with comple-ment activation markers C3a, SCSb-9, and Bb [28].

Effect of TNF-inhibitors on the complement profile could behelpful to clarify these aspects.

Recently, Mitoma et al. performed an in vitro study concerning thecytotoxic effects of several anti-TNF agents. They reported that bothadalimumab and infliximab showed a potent complement-dependentcytotoxicity (CDC), while etanercept did not. This could explain whyetanercept (in contrast with adalimumab and infliximab) is notclinically effective for granulomatous diseases, such as Crohn's diseaseand Wegener's granulomatosis [90]. This finding was not confirmed

by Kaymakcalan et al. who suggested that the different clinicalefficacy profiles of these biologic agents are not explained bydifferences in complement-dependent lysis, because none of thethree drugs was able to induce CDC in activated human mononuclearcells [91].

Few studies in vivo have evaluated complement activation so far.Familian et al. investigated the effect of treatment with the anti-TNFmonoclonal antibody infliximab on overall complement activation, aswell as on CRP levels and CRP mediated complement activation, inpatients with RA [92]. Plasma levels of C3b/c and C4b/c, reflectingactivation of C3 and C4, respectively, before the start of infliximabtreatment were significantly higher than levels in healthy volunteers.Two weeks after the start of infliximab treatment all patients wereresponders according to change of the DAS28 and showed signifi-cantly lower levels of C3b/c and C4b/c than before treatment.Furthermore, plasma levels of CRP and CRP–complement complexessignificantly decreased in the patients during treatment and thedecrease of CRP and C3b/c levels wasmore pronounced in the patientswith a good response [92]. Considering these data together, theauthors supposed that cytokines released during inflammation triggercomplement activation, presumably through mechanisms involvingCRP and that reduction of this activation contributes to the anti-inflammatory effects of anti-TNF agents [92].

Wouters et al. reported a decrease (although not statisticallysignificant) of C1q–C4 complexes in ten RA patients treated withinfliximab after 22 weeks of therapy, in parallel with DAS28improvement [64].

To the best of our knowledge, no data on the modifications of thecomplement system in PsA patients treated with anti-TNF have beenreported.

As above mentioned, we performed the evaluation of complementsystem in patients with RA and PsA treated with the anti-TNF agentsadalimumab or etanercept (unpublished results). We observed astatistically significant decrease in C3 and C4 levels, independentlyfrom the anti-TNF used, both in RA and in PsA. A higher reduction incomplement C3 levels was observed in patients with good response aswell as patients with baseline lower C3 levels had a better response.Taking these findings together, we may consider reduction ofcomplement native components as an improvement of a pre-existingpro-inflammatory status reverted by anti-TNF drugs (unpublishedresults). It could be suppose that persistently elevated C3 levels mayrepresent as negative predictive factor influencing the outcome ofanti-TNF therapy in RA and PsA. As a consequence, C3 dosage mayprovide an additional tool in monitoring disease activity duringtreatment with anti-TNF (unpublished results). Our preliminary dataneed to be further confirmed.

5. Potential therapies targeting complement cascade ininflammatory arthritides

The complement system is increasingly recognized to be causallyinvolved in tissue injury during ischemic, inflammatory, and autoim-mune diseases. Because of this, complement is an attractivetherapeutic target for a wide range of diseases, such as connectivetissue diseases, glomerulonephritis, myocarditis, multiple sclerosis,type I diabetes mellitus, asthma, myocardial infarction, paroxysmalnocturnal hemoglobinuria, vasculitis and many others [9,12,93–95].

Up to date, several compounds interfering with complementcascade were studied in experimental models of arthritis, such assoluble CR1 [96] (which suppress complement activation at the maingathering point C3), C3a [97] and C5a [98] receptor antagonists (thatcan used to control the anaphylatoxins C3a and C5a), recombinantCD59 [99] (that inhibits the formation ofMAC). An important problemwith the modulation of complement activity is that a long-termsystemic suppression may cause side-effects due to diminished

622 E. Ballanti et al. / Autoimmunity Reviews 10 (2011) 617–623

complement activity, such as increased susceptibility to bacterialinfections [10].

Interesting results have been obtained in vivo with PMX53, a C5amimetic compound that binds the C5a receptor, without causingagonist effects. Rats treated with PMX53 showed a significantimprovement of arthritis [98]. Unfortunately, the results of itsapplication in human disease were not so promising. Vergunst et al.performed a double-blind, placebo controlled trial in 21 patients withactive RA [100]. They reported that RA patients didn't show a clinicalimprovement when compared with patients who received placebo[100]. Other studies are needed to clarify the potential use ofcomplement as a therapeutic target in inflammatory arthritides.

6. Conclusions

Extensive scientific evidence is now available supporting a role ofcomplement system in the pathogenesis of RA and, to a less extent,the contribution of the complement cascade has been demonstratedin PsA too. TNFα could interact with complement system byincreasing the synthesis of complement proteins in hepatic cells,during acute phase response [89], and this explain the finding ofincreased complement levels in sera of patients with inflammatoryarthritides, reverted by anti-TNF administration. On the other hand,TNFα seems to activate complement system, as demonstrated by areduction of complement cleavage products in sera of patients treatedwith anti-TNF agents [92]. A mechanism proposed is that anti-TNFαdecrease plasma levels of CRP, which can activate complementcascade through classical pathway. Therefore, the reduction ofcomplement activation could contribute to the anti-inflammatoryeffects of anti-TNFα agents [92].

To conclude, specific modulation and inhibition of local comple-ment production could be an attractive target for RA and PsA.Potential compounds interfering with the complement cascade havebeen tested, but further studies are needed to find complement-modulating agents able to act on the pathogenetic mechanisms ofthese diseases without interfering with essential, beneficial comple-ment functions.

Take-home messages

• Activation of the complement system can have opposing effects: onthe one hand complement proteins have many protective functions,contributing to host defense against invading pathogens, on theother hand can contribute to inflammation and tissue injury.

• There is evidence of a role of the complement system in thepathogenesis of RA and, to a less extent, in PsA and this support apossible effectiveness of anti-complement agents for the treatmentof both these diseases.

• Reduction of the complement activation is, probably, one of themechanism by which TNFα-inhibitors exert their effectiveness ininflammatory arthritides.

References

[1] Rus H, Cudrici C, Niculescu F. The role of the complement system in innateimmunity. Immunol Res 2005;33:103–12.

[2] Brown EJ. Complement receptors and phagocytosis. Curr Opin Immunol 1991;3:76–82.

[3] Moffitt MC, Frank MM. Complement resistance in microbes. Springer SeminImmunopathol 1994;15:327–44.

[4] Morgan BP, Marchbank KJ, Longhi MP, Harris CL, Gallimore AM. Complement:central to innate immunity and bridging to adaptive responses. Immunol Lett2005;97:171–9.

[5] Walport MJ. Complement. First of two parts. N Engl J Med 2001;344:1058–66.[6] Walport MJ. Complement. Second of two parts. N Engl J Med 2001;344:1140–4.[7] Nauta AJ, Castellano G, Xu W, Woltman AM, Borrias MC, Daha MR, et al.

Opsonization with C1q and mannose-binding lectin targets apoptotic cells todendritic cells. J Immunol 2004;173:3044–50.

[8] Gál P, Dobó J, Závodszky P, Sim RB. Early complement proteases: C1r, C1s andMASPs. A structural insight into activation and functions. Mol Immunol 2009;46:2745–52.

[9] Chen M, Daha MR, Kallenberg CG. The complement system in systemicautoimmune disease. J Autoimmun 2010;34:J276–86.

[10] Mizuno M. A review of current knowledge of the complement system and thetherapeutic opportunities in inflammatory arthritis. Curr Med Chem 2006;13:1707–17.

[11] Nauta AJ, Roos A, Daha MR. A regulatory role for complement in innate immunityand autoimmunity. Int Arch Allergy Immunol 2004;134:310–23.

[12] Théroux P, Martel C. Complement activity and pharmacological inhibition incardiovascular disease. Can J Cardiol 2006;22(Suppl B):18B–24B.

[13] Sjöholm AG, Jönsson G, Braconier JH, Sturfelt G, Truedsson L. Complementdeficiency and disease: an update. Mol Immunol 2006;43:78–85.

[14] Bussone G, Mouthon L. Autoimmune manifestations in primary immunedeficiencies. Autoimmun Rev 2009;8:332–6.

[15] Boackle SA. Complement and autoimmunity. Biomed Pharmacother 2003;57:269–73.

[16] Carroll MC. The role of complement in B cell activation and tolerance. AdvImmunol 2000;74:61–88.

[17] Truedsson L, Bengtsson AA, Sturfelt G. Complement deficiencies and systemiclupus erythematosus. Autoimmunity 2007;40:560–6.

[18] Muñoz LE, Janko C, Schulze C, Schorn C, Sarter K, Schett G, et al. Autoimmunityand chronic inflammation—two clearance-related steps in the etiopathogenesisof SLE. Autoimmun Rev 2010;10:38–42.

[19] Speth C, Wurzner R, Stoiber H, Dierich MP. The complement system:pathophysiology and clinical relevance. Wien Klin Wochenschr 1999;111:378–91.

[20] Collard CD, Lekowski R, Jordan JE, Agah A, Stahl GL. Complement activationfollowing oxidative stress. Mol Immunol 1999;36:941–8.

[21] Morgan BP. The complement system: an overview. Methods Mol Biol 2000;150:1–13.

[22] D'Cruz D, Taylor J, Ahmed T, Asherson R, Khamashta M, Hughes GR. Complementfactor 2 deficiency: a clinical and serological family study. Ann Rheum Dis1992;51:1254–6.

[23] Kilpatrick DC. Mannan-binding lectin: clinical significance and applications.Biochim Biophys Acta 2002;1572:401–13.

[24] Miura N, Prentice HL, Scheider PM, Perlmutter DH. Synthesis and regulation ofthe two human complement C4 genes in stable transfected mouse fibroblasts. JBiol Chem 1987;262:7298–305.

[25] Andus T, Heinrich PC, Bauer J, Trau-Thi TA, Decker K, Manuel D, et al.Discrimination of hepatocyte stimulating activity from human recombinanttumour necrosis factor a. Eur J Immunol 1988;17:1193–7.

[26] Anthony R, EL-Omar E, Lappin DF, MacSween RNM, Whaley K. Regulation ofhepatic synthesis of C3 and C4 during acute-phase response in the rat. Eur JImmunol 1989;19:1405–12.

[27] Ramadori G, Van Daume J, Riedert H, Mayer Zum Buschenfelde KH. Interleukin-6,the third mediator of acute-phase reaction, modulates hepatic protein synthesisin human and mouse comparison as with interleukin-1β and tumour necrosisfaclor-α. Eur J Immunol 1988;18:1259–64.

[28] Brodeur JP, Ruddy S, Schwartz LB, Moxley G. Synovial fluid levels of complementSC5b-9 and fragment Bb are elevated in patients with rheumatoid arthritis.Arthritis Rheum 1991;34:1531–7.

[29] Ruddy S, Colten HR. Rheumatoid arthritis. Biosynthesis of complement proteinsby synovial tissues. N Engl J Med 1974;290:1284–8.

[30] Moffat GJ, Lappin D, Birnie GD, Whaley K. Complement biosythesis in humansynovial tissue. Clin Exp Immunol 1989;78:54–60.

[31] Cohen HR. Biosynthesis of complement. Adv lmmunol 1976;22:67–118.[32] Cook KS, Min HY, Johnson D, Chaplinsky RJ, Frier JS, Hunt CR, et al. Adipsin: a

circulating serine protease homology secreted by adipose tissue and sciaticnerve. Science 1987;237:402–8.

[33] Morris KM, Aden DP, Knowles BB, Colten HR. Complement biosynthesis by thehuman hepatoma-derived cell line HepG2. J Clin Invest 1982;70:906–13.

[34] Whaley K, Guc D, Gulati P, Lappin D. Synthesis of complement components bysynovial membrane. Immunopharmacology 1992;24:83–9.

[35] Neumann E, Barnum SR, Tarner IH, Echols J, Fleck M, Judex M, et al. Localproduction of complement proteins in rheumatoid arthritis synovium. ArthritisRheum 2002;46:934–45.

[36] Guc D, Gulati P, Lemercier C, Lappin D, Birnie GD, Whaley K. Expression of thecomponents and regulatory proteins of the alternative complement pathway andthe membrane attack complex in normal and diseased synovium. Rheumatol Int1993;13:139–46.

[37] Makinde VA, Senaldi G, Jawad AS, Berry H, Vergani D. Reflection of diseaseactivity in rheumatoid arthritis by indices of activation of the classicalcomplement pathway. Ann Rheum Dis 1989;48:302–6.

[38] Hietala MA, Nandakumar KS, Persson L, Fahlen S, Holmdahl R, Pekna M.Complement activation by both classical and alternative pathways is critical forthe effector phase of arthritis. Eur J Immunol 2004;34:1208–16.

[39] Petersen NE, Elmgreen J, Teisner B, Svehag SE. Activation of classical pathwaycomplement in chronic inflammation: elevated levels of circulating C3d and C4dsplit products in rheumatoid arthritis and Crohn's disease. Acta Med Scand1988;223:557–60.

[40] Zubler RH, Nydegger U, Perrin LH, Fehr K, McCormick J, Lambert PH, et al.Circulating and intra-articular immune complexes in patients with rheumatoidarthritis: correlation of 125IClq binding activity with clinical and biologicalfeatures of the disease. J Clin Invest 1976;57:1308–19.

623E. Ballanti et al. / Autoimmunity Reviews 10 (2011) 617–623

[41] Hay FC, Nineham LJ, Perumal R, Roitt IM. Intra-articular and circulating immunecomplexes and antiglobulins (IgG and IgM) in rheumatoid arthritis: correlationwith clinical features. Ann Rheum Dis 1979;38:1–7.

[42] Robbins DL, Fiegal Jr DW, Leek JC, Shapiro R, Wiesner K. Complement activationby 19S IgM rheumatoid factor: relationship to disease activity in rheumatoidarthritis. J Rheumatol 1986;13:33–8.

[43] Sato Y, Sato R, Watanabe H, Kogure A, Watanabe K, Nishimaki T, et al.Complement activating properties of monoreactive and polyreactive IgMrheumatoid factors. Ann Rheum Dis 1993;52:795–800.

[44] Sato Y, Watanabe H, Kogure A, Miyata M, Watanabe K, Nishimaki T, et al.Complement-activating properties of IgM rheumatoid factors reacting with IgGsubclasses. Clin Rheumatol 1995;14:425–8.

[45] Tanimoto K, Cooper NR, Johnson JS, Vaughan JH. Complement fixation byrheumatoid factor. J Clin Invest 1975;55:437–45.

[46] Sabharwal UK, Vaughan JH, Fong S, Bennett PH, Carson DA, Curd JG. Activation ofthe classical pathway of complement by rheumatoid factors: assessment byradioimmunoassay for C4. Arthritis Rheum 1982;25:161–7.

[47] Trouw LA, Haisma EM, Levarht EW, van der Woude D, Ioan-Facsinay A, Daha MR,et al. Anti-cyclic citrullinated peptide antibodies from rheumatoid arthritispatients activate complement via both the classical and alternative pathways.Arthritis Rheum 2009;60:1923–31.

[48] Volonakis JE. Complement activation by C-reactive protein complexes. Ann NYAcad Sci 1982;389:235–49.

[49] Wolbink GJ, Brouwer MC, Buysman S, ten Berge IJM, Hack CE. CRP-mediatedactivation of complement in vivo: assessment by measuring circulatingcomplement-C-reactive protein complexes. J Immunol 1996;157:473–9.

[50] Lagrand WK, Niessen HWM, Wolbink GJ, Jaspers LH, Visser CA, Verheugt FWA,et al. C-reactive protein co-localizes with complement in human hearts duringacute myocardial infarction. Circulation 1997;95:97–103.

[51] Wolbink GJ, Bossink AWJ, Groeneveld ABJ, de Groot MCM, Thijs LG, Hack CE.Complement activation in patients with sepsis is in part mediated by C-reactiveprotein. J Infect Dis 1998;177:81–7.

[52] Molenaar ET, Voskuyl AE, Familian A, van Mierlo GJ, Dijkmans BA, Hack CE.Complement activation in patients with rheumatoid arthritis mediated in part byC-reactive protein. Arthritis Rheum 2001;44:997–1002.

[53] Sjöberg A, Onnerfjord P, Mörgelin M, Heinegård D, Blom AM. The extracellularmatrix and inflammation: fibromodulin activates the classical pathway ofcomplement by directly binding C1q. J Biol Chem 2005;280:32301–8.

[54] Groeneveld TW, Oroszlan M, Owens RT, Faber-Krol MC, Bakker AC, Arlaud GJ,et al. Interactions of the extracellular matrix proteoglycans decorin and biglycanwith C1q and collectins. J Immunol 2005;175:4715–23.

[55] Ruddy S, Austen KF. Activation of the complement and properdin systems inrheumatoid arthritis. Ann NY Acad Sci 1975;256:96–104.

[56] El-Ghobarey A, Whaley K. Alternative pathway complement activation inrheumatoid arthritis. J Rheumatol 1980;7:453–60.

[57] Malhotra R, Wormald MR, Rudd PM, Fischer PB, Dwek RA, Sim RB. Glycosylationchanges of IgG associated with rheumatoid arthritis can activate complement viathe mannose-binding protein. Nat Med 1995;1:237–43.

[58] Aggarwal A, Bhardwaj A, Alam S, Misra R. Evidence for activation of the alternatecomplement pathway in patients with juvenile rheumatoid arthritis. Rheuma-tology (Oxford) 2000;39:189–92.

[59] Doherty M, Richards N, Hornby J, Powell R. Relation between synovial fluid C3degradation products and local joint inflammation in rheumatoid arthritis,osteoarthritis, and crystal associated arthropathy. Ann Rheum Dis 1988;47:190–7.

[60] Oleesky DA, Daniels RH, Williams BD, Amos N, Morgan BP. Terminal complementcomplexes and C1/C1 inhibitor complexes in rheumatoid arthritis and otherarthritic conditions. Clin Exp Immunol 1991;84:250–5.

[61] Konttinen YT, Ceponis A, Meri S, Vuorikoski A, Kortekangas P, Sorsa T, et al.Complement in acute and chronic arthritides: assessment of C3c, C9, andprotectin (CD59) in synovial membrane. Ann Rheum Dis 1996;55:888–94.

[62] Jose PJ, Moss IK, Maini RN, Williams TJ. Measurement of the chemotacticcomplement fragment C5a in rheumatoid synovial fluids by radioimmunoassay:role of C5a in the acute inflammatory phase. Ann Rheum Dis 1990;49:747–52.

[63] Høgåsen K, Mollnes TE, Harboe M, Götze O, Hammer HB, Oppermann M.Terminal complement pathway activation and low lysis inhibitors in rheumatoidarthritis synovial fluid. J Rheumatol 1995;22:24–8.

[64] Wouters D, Voskuyl AE, Molenaar ET, Dijkmans BA, Hack CE. Evaluation of classicalcomplement pathway activation in rheumatoid arthritis: measurement of C1q–C4complexes as novel activation products. Arthritis Rheum 2006;54:1143–50.

[65] Mollnes TE, Lea T, Mellbye OJ, Pahle J, Grand O, HarboeM. Complement activationin rheumatoid arthritis evaluated by C3dg and the terminal complementcomplex. Arthritis Rheum 1986;29:715–21.

[66] Olmez U, Garred P, Mollnes TE, Harboe M, Berntzen HB, Munthe E. C3 activationproducts, C3 containing immune complexes, the terminal complement complex andnative C9 in patients with rheumatoid arthritis. Scand J Rheumatol 1991;20:183–9.

[67] Trentham DE, Townes AS, Kang AH. Autoimmunity to type II collagen anexperimental model of arthritis. J Exp Med 1977;146:857–68.

[68] Courtenay JS, Dallman MJ, Dayan AD, Martin A, Mosedale B. Immunisation againstheterologous type II collagen induces arthritis in mice. Nature 1980;283:666–8.

[69] Cathcart ES, Hayes KC, Gonnerman WA, Lazzari AA, Franzblau C. Experimentalarthritis in a nonhuman primate. I. Induction by bovine type II collagen. LabInvest 1986;54:26–31.

[70] Yoo TJ, Kim SY, Stuart JM, Floyd RA,OlsonGA, CremerMA, et al. Induction of arthritisin monkeys by immunization with type II collagen. J Exp Med 1988;168:777–82.

[71] Myers LK, Rosloniec EF, Cremer MA, Kang AH. Collagen-induced arthritis, ananimal model of autoimmunity. Life Sci 1997;61:1861–78.

[72] Hietala MA, Jonsson IM, Tarkowski A, Kleinau S, PeknaM. Complement deficiencyameliorates collagen-induced arthritis in mice. J Immunol 2002;169:454–9.

[73] Wang Y, Rollins SA, Madri JA, Matis LA. Anti-C5 monoclonal antibody therapyprevents collagen-induced arthritis and ameliorates established disease. ProcNatl Acad Sci U S A 1995;92:8955–9.

[74] MyersWA, Gottlieb AB, Mease P. Psoriasis and psoriatic arthritis: clinical featuresand disease mechanisms. Clin Dermatol 2006;24:438–47.

[75] Hueber AJ, McInnes IB. Immune regulation in psoriasis and psoriatic arthritis—recent developments. Immunol Lett 2007;114:59–65.

[76] Rosenberg EW, Noah PW, Wyatt RJ, Jones RM, Kolb WP. Complement activationin psoriasis. Clin Exp Dermatol 1990;15:16–20.

[77] Partsch G, Bauer K, Bröll H, Petera P, Dunky A, Merétey K. Complement C3cleavage product in synovial fluids detected by immunofixation. Z Rheumatol1991;50:82–5.

[78] Triolo G, Accardo-Palumbo A, Sallì L, Ciccia F, Ferrante A, Tedesco L, et al. Impairedexpression of erythrocyte glycosyl-phosphatidylinositol-anchored membraneCD59 in patients with psoriatic arthritis. Relation to terminal complementpathway activation. Clin Exp Rheumatol 2003;21:225–8.

[79] Rivas D, Riestra-Noriega JL, Torre-Alonso JC, Rodriguez A, Gutiérrez C. Decrease indetectable complement receptor type 1 levels on erythrocytes from patients withpsoriatic polyarthritis. Br J Rheumatol 1994;33:626–30.

[80] Green S, Chiasson MA, Shah RG. Evidence for the presence of an antitumor factorin serum of normal animals. Cancer Lett 1979;6:235–40.

[81] Maini R, St Clair EW, Breedveld F, Furst D, Kalden J, Weisman M, et al. Infliximab(chimeric anti-tumour necrosis factor alpha monoclonal antibody) versusplacebo in rheumatoid arthritis patients receiving concomitant methotrexate:a randomised phase III trial. ATTRACT Study Group. Lancet 1999;354:1932–9.

[82] Lipsky PE, van der Heijde DM, St Clair EW, Furst DE, Breedveld FC, Kalden JR.Infliximab and methotrexate in the treatment of rheumatoid arthritis. Anti-Tumor Necrosis Factor Trial in Rheumatoid Arthritis with Concomitant TherapyStudy Group. N Engl J Med 2000;343:1594–602.

[83] Moreland LW, Schiff MH, Baumgartner SW, Tindall EA, Fleischmann RM, BulpittKJ, et al. Etanercept therapy in rheumatoid arthritis. A randomized, controlledtrial. Ann Intern Med 1999;130:478–86.

[84] Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA,et al. Adalimumab, a fully human anti-tumor necrosis factor alpha monoclonalantibody, for the treatment of rheumatoid arthritis in patients taking concom-itant methotrexate: the ARMADA trial. Arthritis Rheum 2003;48:35–45.

[85] Gordon KB, Langley RG, Leonardi C, Toth D, Menter MA, Kang S, et al. Clinicalresponse to Adalimumab treatment in patients with moderate to severepsoriasis: double-blind, randomized controlled trial and open-label extensionstudy. J Am Acad Dermatol 2006;55:598–606.

[86] Mease PJ, Goffe BS, Metz J, VanderStoep A, Finck B, Burge DJ. Etanercept in thetreatment of psoriatic arthritis and psoriasis: a randomised trial. Lancet2000;356:385–90 12.

[87] Mease PJ. Etanercept, a TNF antagonist for treatment for psoriatic arthritis andpsoriasis. Skin Therapy Lett 2003;8:1–4.

[88] Antoni CE, Kavanaugh A, Kirkham B, Tutuncu Z, Burmester GR, Schneider U, et al.Sustained benefits of infliximab therapy for dermatologic and articularmanifestations of psoriatic arthritis: results from the infliximab multinationalpsoriatic arthritis controlled trial (IMPACT). Arthritis Rheum 2005;52:1227–36.

[89] Perlmutter DH, Dinarello CA, Punsal PI, Colten HR. Cachectin/tumor necrosis factorregulates hepatic acute-phase gene expression. J Clin Invest 1986;78:1349–54.

[90] Mitoma H, Horiuchi T, Tsukamoto H, Tamimoto Y, Kimoto Y, Uchino A, et al.Mechanisms for cytotoxic effects of anti-tumor necrosis factor agents ontransmembrane tumor necrosis factor alpha-expressing cells: comparison amonginfliximab, etanercept, and adalimumab. Arthritis Rheum 2008;58:1248–57.

[91] Kaymakcalan Z, Sakorafas P, Bose S, Scesney S, Xiong L, Hanzatian DK, et al.Comparisons of affinities, avidities, and complement activation of adalimumab,infliximab, and etanercept in binding to soluble and membrane tumor necrosisfactor. Clin Immunol 2009;131:308–16.

[92] Familian A, Voskuyl AE, van Mierlo GJ, Heijst HA, Twisk JW, Dijkmans BA, et al.Infliximab treatment reduces complement activation in patients with rheuma-toid arthritis. Ann Rheum Dis 2005;64:1003–8.

[93] Holers VM. The complement system as a therapeutic target in autoimmunity. ClinImmunol 2003;107:140–51.

[94] Makrides SC. Therapeutic inhibition of the complement system. Pharmacol Rev1998;50:59–87.

[95] Asghar SS, Pasch MC. Therapeutic inhibition of the complement system. Y2Kupdate. Front Biosci 2000;5:E63–81.

[96] Goodfellow RM, Williams AS, Levin JL, Williams BD, Morgan BP. Solublecomplement receptor one (sCR1) inhibits the development and progression ofrat collagen-induced arthritis. Clin Exp Immunol 2000;119:210–6.

[97] Ames RS, Lee D, Foley JJ, Jurewicz AJ, Tornetta MA, Bautsch W, et al. Identificationof a selective nonpeptide antagonist of the anaphylatoxin C3a receptor thatdemonstrates antiinflammatory activity in animal models. J Immunol 2001;166:6341–8.

[98] Woodruff TM, Strachan AJ, Dryburgh N, Shiels IA, Reid RC, Fairlie DP, et al.Antiarthritic activity of an orally active C5a receptor antagonist against antigen-induced monarticular arthritis in the rat. Arthritis Rheum 2002;46:2476–85.

[99] Fraser DA, Harris CL, Williams AS, Mizuno M, Gallagher S, Smith RA, et al.Generation of a recombinant, membrane-targeted form of the complementregulator CD59: activity in vitro and in vivo. J Biol Chem 2003;278:48921–7.

[100] Vergunst CE, Gerlag DM, Dinant H, Schulz L, Vinkenoog M, Smeets TJ, et al.Blocking the receptor for C5a in patients with rheumatoid arthritis does notreduce synovial inflammation. Rheumatology (Oxford) 2007;46:1773–8.