Elsevier

Molecular Immunology

Volume 45, Issue 5, March 2008, Pages 1199-1207
Molecular Immunology

Review
Role of complement and complement regulators in the removal of apoptotic cells

https://doi.org/10.1016/j.molimm.2007.09.008Get rights and content

Abstract

Apoptosis, followed by rapid phagocytic clearance, is the primary mechanism by which organisms dispose of unwanted cells. The intracellular and extracellular composition of an apoptotic cell changes to decrease immunogenicity and enhance its uptake. By changing their extracellular composition, apoptotic cells acquire the capacity to bind complement initiation molecules such as C1q and MBL. Binding of these molecules can lead to complement activation. Membrane bound complement inhibitors are down-regulated during apoptosis, which would leave the cell less protected against complement activation; however, recent data show that fluid-phase complement inhibitors may compensate for this loss of regulation. Importantly, binding of complement is a process that mainly takes place during the late stages of apoptosis. Most cells will be cleared before that stage under steady state conditions, but during overwhelming apoptosis or impaired phagocytosis, apoptotic cells may remain in tissues for a longer time and acquire complement proteins. Based on the data from deficiencies of early complement components and the development of systemic lupus erythematosus with accumulation of dead cells, it is clear that, under certain conditions, apoptotic cells persist, becoming necrotic and overloading the scavenging capacities of the complement system. Although the complement system is also involved in inducing apoptosis in target cells, this review will focus on the role of complement in the clearance of apoptotic cells.

Introduction

Tissue homeostasis is achieved by replacing old cells with new when needed, which requires the efficient and non-inflammatory removal of the old cells. Most cells are removed from the body via a process that involves apoptosis and efficient phagocytosis of the dead cells (Henson and Hume, 2006). Host-derived dead cells are a valuable source of self-antigens that can be used to instruct the adaptive immune system to distinguish between self and non-self. However, autoimmunity could arise if these self-antigens were presented in a pro-inflammatory context. In many clinical conditions apoptosis and clearance of apoptotic cells has been suggested to play an important role. Understanding how the complement system is involved in this process will be valuable for intervention in autoimmune diseases, inflammatory conditions, neurological pathologies and malignancies.

The body has devised redundant mechanisms that ensure that the process of cell removal and replacement is kept in an anti-inflammatory state (Gardai et al., 2006). This is mediated by careful induction of cell death via apoptosis: an active process of cell death that involves activation of several intracellular pathways that make sure the intracellular content, including the nucleus, is condensed, fragmented and stored in ‘safe’ vesicles. Several changes also take place at the cell surface that makes the cell recognizable to the surrounding cells and phagocytes as being dead and ready to be removed. One of the best known changes is the loss of lipid bilayer asymmetry (or membrane flip-flop), exposing high levels of phosphatidyl-serine, which by itself already constitutes a pro-phagocytic ‘eat me’ signal (Fadok et al., 1998). Conversely, several membrane proteins normally present on the cell surface are either internalized or shed (Elward and Gasque, 2003, Elward et al., 2005, Gardai et al., 2005). Down-regulation of several molecules, such as CD47, CD200 and sialic acid (Hoek et al., 2000, Oldenborg et al., 2000, Wright et al., 2000), may already initiate recognition because they serve as ‘don’t-eat me’ signals. Other molecules that are normally only found in the intracellular compartment may acquire access to the cell surface and enhance recognition via receptors on neighboring cells or on phagocytes (Gardai et al., 2005, Gardai et al., 2006). In addition to changes from within, the cell also acquires proteins from the extracellular environment that bind to the apoptotic cell surface and may serve as opsonins (Krysko et al., 2006). An array of non-complement molecules have been reported to do so, including β2 glycoprotein, milk fat globule protein (MFG-E8), protein S, growth arrest specific gene number 6 (Gas-6), thrombospondin, IgM, pentraxin 3 (PTX3), serum amyloid P component (SAP), C-reactive protein (CRP), and several components of the complement system such as C1q, MBL, ficolins and C3 also bind. It is currently unknown whether this process is equally important for cells that are present in a protein rich environment, such as circulating blood cells, as compared to cells within tissues. Numerous in vitro studies have shown that binding of these plasma proteins enhanced uptake by phagocytes; however, it is interesting to note that other studies have reported inhibition of uptake for cells coated with PTX3 (van Rossum et al., 2004) and C4b-binding protein (C4BP)–protein S complex (Kask et al., 2004). The relative binding affinities of these different proteins and the ligands involved are less well characterized. However, differences in experimental protocol, cell types studied and precisely how investigators define early and late apoptosis have led to big differences between studies regarding the time point at which dying cells acquire complement components as opsonin. Although some findings were reported to show binding during early apoptosis, it is now becoming increasingly clear that binding of complement initiation molecules and complement activation are events that mainly take place on late apoptotic cells (Gaipl et al., 2001, Trouw et al., 2007). This is in contrast to some of the earlier apoptotic events such as membrane flip-flop, down-regulation of ‘don’t-eat me’ signals and binding of other plasma proteins, which initiate phagocytosis long before complement comes into play.

The initial stages of apoptosis can be divided into membrane flip-flop (as characterized by increased surface annexin-V binding) and exposure of nucleosomes from the nucleus displayed at the cell surface (Radic et al., 2004), followed by a gradual increase in membrane permeability (frequently measured as loss of propidium iodide exclusion). It is important to note that these are only changes to the cell that were easy to determine experimentally and do not per se represent essential stages in the process of cell death.

Binding of complement initiation molecules does not generally take place on early apoptotic cells despite the fact that the membrane has undergone flip-flop. In fact, flip-flop of membranes also takes place, in a reversible fashion, for example, on activated B cells (Dillon et al., 2001) and subsets of monocytes (Appelt et al., 2005). Although C1q can, under some circumstances, detect early changes on apoptotic cells (possibly involving nucleic acid exposure at the cell surface), several key complement inhibitors will act to control the activation of the complement cascade (Elward et al., 2005). Several lines of evidence suggest that the cell surface needs to be sufficiently remodeled for the initial complement molecules to be able to bind (Trouw et al., 2007). Purified C1q has been observed to bind directly to apoptotic cells, but it is likely that adaptor molecules, such as IgM, will also be involved in binding from whole serum. This again underscores the importance of determining which proteins and ligands are involved in this process in vivo. Due to the redundant nature of the apoptotic process, this may not be easy, but a more complete picture is now emerging for which complement pathways are activated by dying cells and which proteins associate with dying cells, making possible a clarification of the overall picture by systematic investigation of each protein. In addition, it is important to carefully consider the concentrations of reagents used for these studies. Initial studies have focused on classical pathway activation using relatively low concentrations of human serum that exclude, or underestimate, the contribution of the alternative pathway of complement.

As mentioned above, apoptosis is an important mechanism in tissue homeostasis, but it is important to realize that it also occurs in places with active inflammation. The environment in which apoptosis take place will have impact on complement activation, regulation and on the pro- or anti-inflammatory context of phagocytosis. Also the anatomical location of apoptosis and the contribution of complement on the phagocytosis of such cells will be important; an apoptotic cell present in blood will be interacting differently with complement compared to a cell present in the brain. These two aspects have not been addressed in great detail and would be relevant in the context of the potential use of complement inhibition for inflammatory conditions.

Section snippets

C1q, the canonical immune recognition molecule for apoptotic cells

Among an array of plasma proteins (histidine rich protein, IgG, IgM, SAP, CRP, PTX3) binding apoptotic cells, complement components bind directly or as a consequence of binding by other plasma proteins. C1q is the most studied complement component bound by apoptotic cells, and purified C1q can bind apoptotic cells directly as well as secondary to IgG and IgM binding (Ciurana et al., 2004, Quartier et al., 2005, Zwart et al., 2004). The interest in C1q in relation to apoptosis stems from the

Fluid-phase complement regulators and apoptosis

The complement system is very aggressive because of its enzymatic cascade and amplification nature and needs to be tightly regulated to prevent organ damage, but also to prevent systemic depletion of complement. For this purpose, cells are equipped with membrane bound complement inhibitors, in addition to the fluid-phase inhibitors in plasma, to provide constant control of complement activation. This allows complement activation on foreign particles, but limits spontaneous complement activation

Membrane bound complement regulators as don’t-eat me signals

Host cells use a wide armamentarium of membrane-bound complement inhibitors, which inhibit assembly of either the C3-cleaving enzymes or the formation of the cytotoxic and cytolytic membrane attack complex (MAC) (for review Morgan, 1995, Nicholson-Weller and Wang, 1994, Riley-Vargas et al., 2004). Decay accelerating factor (DAF, CD55) binds to and dissociates the classical and alternative C3/C5 convertase enzyme complexes. Membrane cofactor protein (MCP, CD46) acts as a cofactor for factor I,

Autoantibody formation

Many of the autoantigens present on apoptotic cells are targeted by autoantibodies in autoimmune diseases like SLE (Denny et al., 2006, Frisoni et al., 2005, Vay et al., 2006). It is not only the endogenous antigens of the dying cell that are targeted, but also the adaptor molecules that bind to the apoptotic cells to enhance their clearance (Kravitz et al., 2005, Kravitz and Shoenfeld, 2006, Salcido-Ochoa et al., 2002). For example, anti-C1q and anti-MBL autoantibodies have been observed in

Conclusion

In conclusion, the role of complement in the clearance of apoptotic cells has been described in many papers. However, many differences have been reported for the relative importance of complement activation at each stage of cell death and the importance of complement in clearance of apoptotic cells, likely due to variations in experimental set-up and stage of apoptosis of the cells. What is clear, however, is that a functioning complement system with appropriate activation and inhibition is

Acknowledgements

LT is a recipient of a VENI grant from the Netherlands Organisation for Scientific Research. PG and his team are supported by University and Conseil Regional of la Reunion, French Overseas Ministry (MOM) and PG is a fellow of INSERM (University-INSERM partnership). AB and her team are supported by Swedish Foundation for Strategic Research and Swedish Research Council.

References (95)

  • S.J. Gardai et al.

    Cell-surface calreticulin initiates clearance of viable or apoptotic cells through trans-activation of LRP on the phagocyte

    Cell

    (2005)
  • P. Gasque

    Complement: a unique innate immune sensor for danger signals

    Mol. Immunol.

    (2004)
  • J. Hakulinen et al.

    ADAM10-mediated release of complement membrane cofactor protein during apoptosis of epithelial cells

    J. Biol. Chem.

    (2006)
  • P.M. Henson et al.

    Apoptotic cell removal in development and tissue homeostasis

    Trends Immunol.

    (2006)
  • A. Hillarp et al.

    Protein S binding in relation to the subunit composition of human C4b-binding protein

    FEBS Lett.

    (1989)
  • M.L. Jensen et al.

    Ficolin-2 recognizes DNA and participates in the clearance of dying host cells

    Mol. Immunol.

    (2007)
  • H. Jiang et al.

    DNA binds and activates complement via residues 14–26 of the human C1q A chain

    J. Biol. Chem.

    (1992)
  • L. Kask et al.

    The C4b-binding protein–protein S complex inhibits the phagocytosis of apoptotic cells

    J. Biol. Chem.

    (2004)
  • M. Kuraya et al.

    Specific binding of L-ficolin and H-ficolin to apoptotic cells leads to complement activation

    Immunobiology

    (2005)
  • B.P. Morgan

    Complement regulatory molecules: application to therapy and transplantation

    Immunol. Today

    (1995)
  • B.P. Morgan et al.

    Expression of complement in the brain: role in health and disease

    Immunol. Today

    (1996)
  • N. Palaniyar et al.

    Nucleic acid is a novel ligand for innate, immune pattern recognition collectins surfactant proteins A and D and mannose-binding lectin

    J. Biol. Chem.

    (2004)
  • M.C. Pickering et al.

    Ultraviolet-radiation-induced keratinocyte apoptosis in C1q-deficient mice

    J. Invest. Dermatol.

    (2001)
  • R.C. Riley-Vargas et al.

    CD46: expanding beyond complement regulation

    Trends Immunol.

    (2004)
  • M. Skoberne et al.

    The apoptotic-cell receptor CR3, but not alphavbeta5, is a regulator of human dendritic-cell immunostimulatory function

    Blood

    (2006)
  • F. Takizawa et al.

    Enhancement of macrophage phagocytosis upon iC3b deposition on apoptotic cells

    FEBS Lett.

    (1996)
  • L.A. Trouw et al.

    Autoantibodies to complement components

    Mol. Immunol.

    (2001)
  • A. Tsutsumi et al.

    Mannose binding lectin: genetics and autoimmune disease

    Autoimmun. Rev.

    (2005)
  • D. Vay et al.

    Anti-phospholipid antibodies associated with alcoholic liver disease target oxidized phosphatidylserine on apoptotic cell plasma membranes

    J. Hepatol.

    (2006)
  • G.J. Wright et al.

    Lymphoid/neuronal cell surface OX2 glycoprotein recognizes a novel receptor on macrophages implicated in the control of their function

    Immunity

    (2000)
  • H.A. Anderson et al.

    Serum-derived protein S binds to phosphatidylserine and stimulates the phagocytosis of apoptotic cells

    Nat. Immunol.

    (2003)
  • U. Appelt et al.

    Viable, apoptotic and necrotic monocytes expose phosphatidylserine: cooperative binding of the ligand Annexin V to dying but not viable cells and implications for PS-dependent clearance

    Cell Death Differ.

    (2005)
  • G. Attali et al.

    Increased sensitivity of early apoptotic cells to complement-mediated lysis

    Eur. J. Immunol.

    (2004)
  • S.R. Barnum et al.

    C4b-binding protein, a regulatory component of the classical pathway of complement, is an acute-phase protein and is elevated in systemic lupus erythematosus

    Complement Inflamm.

    (1990)
  • M. Botto et al.

    Homozygous C1q deficiency causes glomerulonephritis associated with multiple apoptotic bodies

    Nat. Genet.

    (1998)
  • I.E. Bultink et al.

    Deficiency of functional mannose-binding lectin is not associated with infections in patients with systemic lupus erythematosus

    Arthritis Res. Ther.

    (2006)
  • A.E. Bygrave et al.

    Spontaneous autoimmunity in 129 and C57BL/6 mice-implications for autoimmunity described in gene-targeted mice

    PLoS Biol.

    (2004)
  • C.L. Ciurana et al.

    Complement activation by necrotic cells in normal plasma environment compares to that by late apoptotic cells and involves predominantly IgM

    Eur. J. Immunol.

    (2004)
  • D.S. Cole et al.

    Beyond lysis: how complement influences cell fate

    Clin. Sci. (Lond.)

    (2003)
  • B. Dahlback et al.

    Visualization of human C4b-binding protein and its complexes with vitamin K-dependent protein S and complement protein C4b

    Proc. Natl. Acad. Sci. U.S.A.

    (1983)
  • M.F. Denny et al.

    Accelerated macrophage apoptosis induces autoantibody formation and organ damage in systemic lupus erythematosus

    J. Immunol.

    (2006)
  • S.R. Dillon et al.

    Annexin V binds to positively selected B cells

    J. Immunol.

    (2001)
  • V.A. Fadok et al.

    The role of phosphatidylserine in recognition of apoptotic cells by phagocytes

    Cell Death Differ.

    (1998)
  • L. Frisoni et al.

    Nuclear autoantigen translocation and autoantibody opsonization lead to increased dendritic cell phagocytosis and presentation of nuclear antigens: a novel pathogenic pathway for autoimmunity?

    J. Immunol.

    (2005)
  • U.S. Gaipl et al.

    Complement binding is an early feature of necrotic and a rather late event during apoptotic cell death

    Cell Death Differ.

    (2001)
  • U.S. Gaipl et al.

    Cooperation between C1q and DNase I in the clearance of necrotic cell-derived chromatin

    Arthritis Rheum.

    (2004)
  • F.P. Garcia de et al.

    Differential regulation of alpha and beta chains of C4b-binding protein during acute-phase response resulting in stable plasma levels of free anticoagulant protein S

    Blood

    (1994)
  • Cited by (152)

    • The role of properdin and Factor H in disease

      2022, Advances in Immunology
      Citation Excerpt :

      Complement activation on neutrophils results in increased release of C5a fragments and the MAC complex, which could further activate neutrophils and other cells in close contact with adherent neutrophils, further contributing to a pro-inflammatory microenvironment. The complement system removes dead/dying cells through opsonization and promotion of phagocytosis (Flierman & Daha, 2007; Trouw, Blom, & Gasque, 2008). Properdin released by phagocytes binds to apoptotic and necrotic cells (Kemper et al., 2008; Xu et al., 2008), and may contribute to their removal directly or through properdin-mediated complement activation.

    View all citing articles on Scopus
    View full text