Elsevier

Journal of Autoimmunity

Volume 28, Issues 2–3, March–May 2007, Pages 160-164
Journal of Autoimmunity

Review
Complement activation in anti-phospholipid syndrome: A clue for an inflammatory process?

https://doi.org/10.1016/j.jaut.2007.02.013Get rights and content

Abstract

Anti-phospholipid syndrome (APS) is defined by recurrent arterial/venous thrombosis and/or fetal losses in the persistent presence of anti-phospholipid antibodies (aPL). In in vivo experimental models aPL thrombogenic activity is associated with a pro-inflammatory endothelial phenotype (increased adhesion molecule [ADM] expression and leukocyte adhesion) in addition to a pro-coagulant one (tissue factor [TF] expression). This is in line with the in vitro aPL ability to trigger intracellular signalling and to up-regulate ADM, TF and pro-inflammatory cytokine/chemokine expression at the mRNA and protein level in endothelial cells. Comparable effects were also reported in monocytes in vitro. In addition, complement activation is required by aPL to display their thrombogenic activity in in vivo models. Interestingly, complement activation blocking as well as Tumor Necrosis Factor alpha neutralization protect animals from aPL-induced fetal losses. Altogether these findings speak in favour for a role of inflammation in APS in spite of the absence of a clear inflammatory signature in the patients. We could not find any complement (C3c and C4d) deposition in the placentas from 2 late abortions (20 weeks of gestation) in APS women. Further studies are necessary to investigate whether complement activation and inflammatory processes found in animal models are taking place in APS patients.

Introduction

Anti-phospholipid syndrome (APS) is a clinical disorder, characterised by recurrent thrombosis, fetal losses and circulating anti-phospholipid antibodies (aPL), reacting to PL-binding plasma proteins (namely β2glycoprotein I [β2GPI] and prothrombin) [1].

It is widely accepted that β2GPI-dependent aPL play a pathogenic role. In fact, several in vivo studies showed a direct effect of these autoantibodies in inducing fetal loss [2], [3], [4], [5], endothelial activation [6] and in enhancing the pro-coagulant effect of a trigger factor [6], [7], [8].

The thrombophilic state in APS has been associated with a vasculopathy rather than with a true inflammatory vasculitic process. Actually, even the recent classification criteria state that inflammatory signs should be absent in the thrombotic manifestations of APS [9]. Such a statement is apparently in clash with the evidence that aPL, and in particular anti-β2GPI antibodies, are able to induce an inflammatory phenotype in both monocytes and endothelial cells (EC) in in vitro experimental models [10]. Actually these cell types may up-regulate the synthesis and secretion of pro-inflammatory cytokines, chemokines and adhesion molecules (ADM) when incubated in the presence of β2GPI-dependent aPL [11], [12], [13], [14], [15]. Animals deficient for ADMs or treated with anti-VCAM-1 blocking antibodies were shown to be protected from the thrombogenic effect of human aPL IgG and VCAM-1 up-regulation was shown in the endothelium of carotid arteries in animals injected with human aPL IgG fractions (rev. in ref. [40]). As a whole these findings do suggest that an endothelial pro-inflammatory phenotype may be present also in in vivo APS animal models.

The obstetrical counterpart of APS is represented by the occurrence of pregnancy losses and pregnancy complications such as early pre-eclampsia. Both histopathological studies and experimental evidence of a direct effect of aPL on placental tissues strongly suggest that thrombotic phenomena alone cannot explain the obstetrical manifestations of the syndrome [16], [17]. A local inflammatory process has been suggested to play a role in inducing a defective placentation as a cause of the recurrent fetal losses associated to aPL. Actually local Tumor Necrosis Factor alpha (TNFα) production, neutrophil recruitment and complement activation/deposition have been described in experimental animal models of aPL-mediated fetal loss [18], [19], [20], [21]. In line with these experimental findings, histological evaluation of terminal placental tissues of APS women reported an increased infiltration of monocyte-macrophage lineage cells [22].

Section snippets

Potential role of complement in vascular thrombosis

Particular attention has been paid on the endothelial perturbation mediated by β2GPI-dependent aPL in APS pathogenesis [10], [23]. In this regard, it is important to underline the close relationship between the complement system and endothelial functions [24]. In fact, EC could synthesize different complement proteins, such as C3 and C4 and late components in response to pro-inflammatory cytokines [25], [26], [27]. In addition, complement system – through membrane attack complex activation –

APS thrombosis and complement

Notwithstanding, the mechanisms of pro-coagulant effects of anti-PL are not fully understood. In fact, a complex interaction between these autoantibodies, EC, platelets and leukocytes has been suggested to eventually trigger the coagulation process in combination with a second hit [10], [23]. In this “scenario” the complement system has been recently reported to play a role in mediating the aPL-induced clotting in experimental animal models.

In order to reproduce the thrombosis occurring in APS

APS recurrent fetal loss and complement

As mentioned before, complement activation has been suggested to play a pathogenic role also in aPL-associated pregnancy loss.

Several studies showed that passive transfer of human aPL IgG in pregnant mice can cause fetal loss and growth retardation [2], [3], [4], [5]. Although some experimental models highlight the role of thrombosis in the pathogenesis of fetal loss in APS, histological findings on the placenta tissues suggest that pro-inflammatory elements can co-operate to tissue damage [22]

Case report

In spite of the experimental evidence there is scarce information on complement activation in the course of APS. Plasma hypocomplementemia was originally reported in a small series of patients with Central Nervous System (CNS) involvement and low plasma complement levels were also described in APS patients by some groups [42], [43], [44], [45]. In addition, a relationship between anti-cardiolipin antibodies and null alleles of C4 was also reported [46].

However, no clear correlation between

References (48)

  • M. Blank et al.

    Prevention of experimental antiphopholipid syndrome and endothelial cell activation by synthetic peptides

    Proc Natl Acad Sci USA

    (1999)
  • A. Piona et al.

    Placental thrombosis and fetal loss after passive transfer of mouse lupus monoclonal or human polyclonal anti-cardiolipin antibodies in pregnant naive BALB/c mice

    Scand J Immunol

    (1995)
  • W. Ikematsu et al.

    Human anticardiolipin monoclonal autoantibodies cause placental necrosis and fetal loss in BALB/c mice

    Arthritis Rheum

    (1998)
  • S.S. Pierangeli et al.

    Anti-phospholipid syndrome patients activate endothelial cells in vitro and in vivo

    Circulation

    (1999)
  • S.S. Pierangeli et al.

    Experimental thrombosis and antiphospholipid antibodies: new insights

    J Autoimmun

    (2000)
  • M. Pier Luigi et al.

    Pathogenic mechanisms of antiphospholipid syndrome: a new autoimmune disease

    Drug Disc Today: Dis Mech

    (2004)
  • M. Bohgaki et al.

    The p38 mitogen-activated protein kinase (MAPK) pathway mediates induction of the tissue factor gene in monocytes stimulated with human monoclonal anti-beta2Glycoprotein I antibodies

    Int Immunol

    (2004)
  • R. Simantov et al.

    Activation of cultured vascular endothelial cells by antiphospholipid antibodies

    J Clin Invest

    (1995)
  • N. Del Papa et al.

    Relationship between anti-phospholipid and anti-endothelial cell antibodies III: beta 2 glycoprotein I mediates the antibody binding to endothelial membranes and induces the expression of adhesion molecules

    Clin Exp Rheumatol

    (1995)
  • P.L. Meroni et al.

    Statins prevent endothelial cell activation induced by antiphospholipid (anti-beta2-glycoprotein I) antibodies: effect on the proadhesive and proinflammatory phenotype

    Arthritis Rheum

    (2001)
  • C.S. Cho et al.

    Antiphospholipid antibodies induce monocyte chemoattractant protein-1 in endothelial cells

    J Immunol

    (2002)
  • P.L. Meroni et al.

    Antiphospholipid antibodies as cause of pregnancy loss

    Lupus

    (2004)
  • N. Di Simone et al.

    Pathogenic role of anti-{beta}2-glycoprotein I antibodies on human placenta: functional effects related to implantation and roles of heparin

    Hum Reprod Update

    (2006 Nov 11)
  • V.M. Holers et al.

    C3 activation is required for anti-phospholipid antibody-induced fetal loss

    J Exp Med

    (2002)
  • Cited by (76)

    • Histopathology in the placentae of women with antiphospholipid antibodies: A systematic review of the literature

      2015, Autoimmunity Reviews
      Citation Excerpt :

      A second study demonstrated that the staining for C4d on extravillous trophoblasts, on the apical and basal syncytiotrophoblast membranes and within the syncytiotrophoblast cytoplasm, was greater in the placentae of aPL-positive women compared to aPL-negative control women [82]. A third study observed no deposition of C4d in the placentae of aPL-positive women [74]. The cleavage of complement component C3, which occurs downstream of C4 cleavage [83], was investigated by four studies (Table 8).

    • Antiphospholipid Antibody Syndrome

      2014, Obstetrics and Gynecology Clinics of North America
    • Anti-phospholipid induced murine fetal loss: Novel protective effect of a peptide targeting the β2 glycoprotein I phospholipid-binding site. Implications for human fetal loss

      2012, Journal of Autoimmunity
      Citation Excerpt :

      For example, passive infusion of large amounts of IgG aPL in naive mice after embryo implantation may induce fetal loss via placental inflammation [7–12]. Again, these experimental observations do not find correspondence in immunohistological analysis of abortive material or term placentas from APS women which does not offer conclusive information on the pathogenic contribution of acute local inflammation and complement deposition [6,13–15]. Evidence has been collected for alternative aPL-mediated non-thrombotic/non-inflammatory mechanisms ending into a defective placentation [2,3,5,12,16].

    View all citing articles on Scopus
    View full text