You are on page 1of 17

REVIEW ARTICLE

Self-nonself discrimination by the complement system


Seppo Meri1,2,3
1 Immunobiology, Research Programs Unit, Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Finland
2 HUSLAB, Helsinki University Hospital, Finland
3 Humanitas University, Milan, Italy

Correspondence The alternative pathway (AP) of complement can recognize nonself structures
S. Meri, Immunobiology, Research by only two molecules, C3b and factor H. The AP deposits C3b covalently
Programs Unit, Department of Bacteriology
on nonself structures via an amplification system. The actual discrimination is
and Immunology, Haartman Institute,
performed by factor H, which has binding sites for polyanions (sialic acids,
University of Helsinki, Helsinki 00014,
Finland glycosaminoglycans, phospholipids). This robust recognition of ‘self’ protects
Fax: +358 9 191 26382 our own intact viable cells and tissues, while activating structures are recog-
Tel: +358 50 5812462 nized by default. Foreign targets are opsonized for phagocytosis or killed.
E-mail: seppo.meri@helsinki.fi Mutations in factor H predispose to severe diseases. In hemolytic uremic syn-
drome, they promote complement attack against blood cells and vascular
(Received 20 June 2016, revised 29 June
endothelial cells and lead, for example, to kidney and brain damage. Even
2016, accepted 6 July 2016, available online
pathogens can exploit factor H. In fact, the ability to bind factor H discrimi-
19 July 2016)
nates most pathogenic microbes from nonpathogenic ones.
doi:10.1002/1873-3468.12284
Keywords: escape; hemolytic uremic syndrome; sialic acid
Edited by Wilhelm Just

Distinguishing structures of ‘self’ from ‘nonself’ is one considered as nonself and will be labeled for destruc-
of the very fundamental processes in biology and a tion (opsonophagocytosis or direct killing). Molecu-
property of the immune system. Sensor molecules for larly, this type of active self-recognition requires far
fine structures, like antibodies in the classical pathway less effort than maintaining a large repertoire of speci-
of complement, usually recognize nonself. This fic recognition molecules. A similar strategy is used in
requires a large repertoire of different molecules. For the actions of natural killer cells (NK cells), where for-
more robust recognition of targets, certain types of eign structures are recognized according to the so
pattern-recognition molecules are used, for example, called ‘missing self’ concept. Thereby, NK cells recog-
scavenger receptors, Toll-like receptors, Nod-like nize cells that lack or have lost host-specific major his-
receptors, and C-type lectins, which are molecules that tocompatibility complex (MHC) molecules [3].
bind sugars in a calcium-dependent manner. The latter Subsequently, NK cell recognition has been observed
include, for example, mannan-binding lectin (MBL), to depend on both the presence and absence of specific
ficolins, and DC-SIGN [1,2]. molecular markers [4].
The alternative pathway (AP) of complement can The discrimination between host and nonhost (or
recognize a broad spectrum of nonself structures by ‘self and nonself’) structures by the immune system is
default. Anything not actively recognized as self is also essential for the maintenance of the integrity of

Abbreviations
aHUS, atypical hemolytic uremic syndrome; AMD, age-related macular degeneration; AP, alternative pathway; CCP, complement control pro-
tein; DAF, decay-accelerating factor; DDD, dense deposit disease; DMBT1, deleted in malignant brain tumor 1; FHBP, factor H binding pro-
tein; FHL-1, factor H-like protein 1; FHRs, factor H-related proteins; MBL, mannan-binding lectin; MCP, membrane cofactor protein; NK,
natural killer; PNH, paroxysmal nocturnal hemoglobinuria; SAG, salivary agglutinin; SALSA, salivary scavenger and agglutinin; TEP1, thioester
protein 1.

2418 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

our body. In addition to surveillance against microbes, development of new vaccines or ways to overcome
complement plays an important role also as a general microbial resistance to complement [11].
clearance mechanism [5]. Even in the absence of As is evident from the above, it is very important to
microbes an inability to distinguish borders of host know in detail the mechanisms how complement can
structures or an inappropriate loss of protection distinguish between self and nonself, what happens
against complement underlies many human diseases. A when errors occur and how do tumor cells and
disturbance in self-recognition is involved in distinct microbes manage to avoid being recognized by com-
syndromes like atypical hemolytic uremic syndrome plement. Both in malignancies and in invasive infec-
(aHUS), dense deposit disease (DDD), age-related tions, there is a need to neutralize the target’s ability
macular degeneration (AMD), and paroxysmal noctur- to block complement activation to allow utilization of
nal hemoglobinuria (PNH) [6]. Complement activation complement for attack against the unwanted cells. In
against self-structures may also occur in pregnancy contrast, the opposite situations, where complement
complications often involving endothelial cell dysfunc- unnecessarily attacks self (autoreactivity, autoimmu-
tion [7]. nity), require means to correct the errors or to provide
While the human complement system distinguishes therapy for the diseases they cause. For any interven-
between human cell surfaces in general (‘self’) and exter- tions, it is essential to understand in detail the molecu-
nal foreign molecular structures (‘nonself’), another lar mechanisms how complement recognizes or does
level of ‘self-nonself’ distinction occurs between individ- not recognize its targets.
uals. In pregnancy, as well as in transplantation, a con-
tact between the complement system from one person
The complement system–a key innate
(future mother, transplant recipient) with cells from
immune recognition system
another person may lead to incompatibility problems.
This may become reflected as a pregnancy disorder or The parenteral spaces of the normal healthy human
rejection of the transplant organ or cells. The problems body are kept free of microbes and other foreign mate-
currently are thought of being due to adaptive immune rial by innate immune defense mechanisms. As a cen-
responses (antibody or T cell-mediated rejection) but tral component of innate immunity, complement is
part of the problem could also be related to polymor- capable of destroying and invading microbes, generat-
phic variations in complement activating and regulating ing inflammation, and acting as a ‘waste-disposal’ sys-
components leading to incompatibility between self and tem [12]. Altogether the complement system consists of
nonself [8]. With the emerging easier and cheaper ways up to 50 different molecules in plasma and on cell
for efficient DNA sequencing, it may become possible membranes (Fig. 1). Each have their specific roles by
to analyze the role of these incompatibilities, for exam- acting as activating components, regulators or recep-
ple, in organ rejection. tors in the system.
Malignant tumor cells and pathogenic microbes Complement can become activated through three
cause different kinds of problems but common to both distinct pathways. In the classical pathway, foreign
is that they usually manage to escape complement materials are recognized by IgM or IgG antibodies,
attack. Thus, they resemble ‘self’. For tumor cells this whereafter the first component C1q binds and initiates
is natural, because most molecules that they carry on the complement activation cascade. Some structures,
the surfaces are present also on normal nonmalignant like bacterial lipopolysaccharides [13], C-reactive pro-
cells. It is, however evident, that this is not the whole tein [14], and degradation products of cells [15] can
truth. Some tumors have additional mechanisms to bind C1q independently of antibodies. In the lectin
protect themselves against complement attack, like pathway, sensor molecules recognize structures con-
secretion of complement inhibitors or production of taining carbohydrates, like mannose or N-acetylgluco-
additional protective layers of carbohydrates (like samine (MBL), for example, on yeast cell surfaces, or
mucins) on their surfaces. There is an urgent need to acetylated sugars by ficolins (ficolins 1, 2, and 3)
obtain new information about the complement evasion [1,16,17]. The alternative pathway (AP) is able, in a
mechanisms so that monoclonal antibody-based tumor robust manner, to discriminate normal human cells
immunotherapy could be improved [9,10]. from microbes (e.g., bacteria, fungi, parasites) and
A feature of pathogenic microbes is that they exploit from damaged or transformed human cells (e.g., virus-
different mechanisms to escape complement attack and infected or apoptotic cells) in an antibody-independent
thereby avoid being opsonized and phagocytosed. The fashion. Another key feature of the AP is that, it can
information on microbial complement resistance mech- amplify its own activation regardless of which pathway
anisms is valuable because it can be used for the it was initiated [18].

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2419
Self-nonself discrimination by the complement system S. Meri

Fig. 1. The different pathways, components and regulators of the complement system. For simplicity, no receptors (except for CR1, the
C3b/C4b receptor, which also acts as a regulator) or factor H-related proteins (FHR1-5) are shown in the figure.

Recognition of targets by the classical C1) [22] or receptors for phagocytosis (C1q receptors)
and lectin pathways [23]. Because there are multiple globular recognition
domains, an average of 6 9 3 in C1q, the molecules
The classical and lectin pathways use sensor molecules can bind to targets with high avidities.
for the recognition of distinct patterns on target sur- C1q has multiple targets. Of these, the traditional
faces [19]. The C1q family of proteins includes C1q, ones are IgG (primarily IgG1 and IgG3 subclasses)
MBL, ficolins 1, 2, and 3 (M, L, and H, respectively), and IgM class antibodies. Antibody-independent bind-
and surfactant proteins SpA and SpD [20]. In addi- ing of C1q occurs, for example, to exposed phospho-
tion, molecules like adiponectin and a family of C1q lipids on apoptotic or injured cells and to various
and TNF-like receptor proteins with 15 members intracellular structures, like mitochondria, chromatin,
(CTRP 1–15) show similarities to C1q [21]. In their and cytoskeletal filaments, exposed upon cell damage.
structures, separate polypeptide chains (A, B, and C in This contributes to the clearance of injured tissue
C1q) form collagenous triple helices. At the C-terminal materials by the reticuloendothelial system. Macro-
end of each polypeptide, there is another part that phages that have phagocytosed materials opsonized by
forms a globular domain that often acts as a carbohy- C1q and other complement activation components,
drate-recognizing lectin. Hence, these proteins are particularly iC3b, secrete immunosuppressive cytoki-
often collectively called as collectins. Individual helices nes, like TGF-beta and IL-10, with the apparent aim
with a collagenous tail and globular end domains in of carrying out homeostatic cleaning without causing
turn assemble into bundles with a variable number of unnecessary inflammation or immune activation [24].
subunits. The final protein product looks like a bunch Suppression of immunoinflammatory responses is
of flowers. The recognition part is located in the glob- mediated by the integrin-type iC3b receptor CD11b/18
ular domains, whereas the collagenous tail interacts or complement receptor type 3 (CR3) [25]. C1q also
with effector molecules (C1r and C1s in complement binds directly to many types of lipopolysaccharides

2420 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

and other bacterial components to help in the phago- interactions with complement need to be comprehen-
cytosis of microbes by blood or tissue leukocytes, sively worked out. This would be an important task
neutrophils, and macrophages. because of the potential role of these molecules in the
MBL binds to surfaces coated with carbohydrates development of atherosclerosis or metabolic distur-
containing mannose and/or N-acetyl-glucosamine [26]. bances, diseases of major importance.
These sugar assemblies, collectively termed as mannan,
are typically found on the surfaces of yeasts, like the
Activation mechanisms of the
common human pathogen Candida albicans. Also,
alternative pathway
some bacteria and their specific serotypes (like Sal-
monella enterica O:6,7 serotype) can have mannose (te- The AP is an ancient system, and as such very impor-
tramannose repeats) in their lipopolysaccharides, tant in protection against microbial infections. In addi-
although this is not common [27]. Ficolins have their tion to all vertebrates, some invertebrates, like
own specificity repertoires [28]. Structures that usually ascidians and chordates have an AP-like system [37].
are recognized contain acetylated monosaccharides. A C3 homolog, called thioester protein 1 (TEP1), has
Additional molecules in the family include collectin-10 been found in mosquitoes [38] and probably exists in
and collectin-11 (CL-K1) that bind to fucose or man- other insects, as well. The functions of TEP1 and C3
nose. Interestingly, mutation in the collectin-11 gene are only partially similar. Both are involved in antimi-
(colec11), as well as in MASP-3 have been found to crobial defense, but they are activated and regulated
associate with the rare 3MC syndrome with develop- by different mechanisms. In fact, TEP1 is more similar
mental defects [29]. Whether the association is related to a membrane-associated thioester protein, CD109,
to complement activity or something else is not yet than to C3. The function of CD109 is still unknown.
clear. Because of efficient amplification the AP complement
C1q, MBL, and ficolins operate primarily in par- activation can lead to inflammation and tissue damage.
enteral spaces. The surfactant proteins, SpA and SpD, Therefore, the AP needs to be strictly targeted only to
instead, are found on mucosal surfaces like in the res- structures that need to be eliminated. The mechanism
piratory tract and in the intestines. Mucosal surfaces of self-nonself (or host-nonhost) discrimination by the
contain also a soluble scavenger family protein called alternative pathway of complement has been studied
variously gp340, deleted in malignant brain tumor 1 originally using purified proteins. The actual activation
(DMBT1), salivary agglutinin (SAG) or salivary scav- system is composed of six main components: C3, B,
enger, and agglutinin (SALSA) [30,31]. We named the D, factor H, factor I (C3b inactivator), and P (prop-
protein as SALSA, as it was found to bind to erdin) [39].
microbes, recruit MBL, the surfactant proteins, SpA C3 is the central molecule in AP activation. It is
and SpA, and to regulate complement activation [32]. abundant in blood plasma with an average concentra-
SALSA is thus yet another sensor protein for the com- tion ranging from 0.5 to 1.2 gL 1. Because of its role
plement system, but operating principally in the in continuous surveillance for potential invaders or
intestines, respiratory tract and on other mucosal sur- damaged tissue structures as much as 50% of its
faces [33,34]. When bound to targets it can activate plasma pool may be regenerated every 1–2 days [40].
complement, but when in solution it inhibits the lectin C3 is spontaneously activated by slow hydrolysis to
pathway [32,35]. generate a form C3(H2O), where an internal thioester
Adiponectin and the C1q and TNF-related proteins bond (formed by side chains of cysteine 988 and glu-
(CTRPs) are an interesting family of collectins, tamine 991) has become disrupted. C3(H2O) resembles
because they have been indicated in the metabolism of C3b except that the C3a part has not become dissoci-
lipids and carbohydrates [21]. Adiponectin is an antidi- ated from the molecule. Functionally, C3(H2O) is able
abetic, antiatherogenic and antiinflammatory molecule. to act in manner similar to C3b in that it can bind fac-
As such it has been considered to have hormone-like tor B and generate an initial C3 convertase, C3(H2O)
properties. Its structural similarity to other collectins, Bb, where factor B has become cleaved and activated
however, points more toward a clearance function, by factor D [41,42]. The C3(H2O)Bb convertase will
possibly in the removal of potentially harmful lipids or activate native C3 molecules to C3b by binding factor
glycolipid particles. The functions of the 15 CTRPs B in the presence of magnesium ions will generate the
are still partially unknown. They have been implicated, actual AP C3 convertase, C3bBb, once factor B is
for example, in increasing glucose uptake, decreasing cleaved by factor D. The inherent half-life of the
fat mass or enhancing insulin sensitivity [36]. The C3bBb convertase is only 1.5 min [43]. The displaced
recognition specificities of these molecules and possible Bb can no longer bind back to C3b. Instead, factor H

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2421
Self-nonself discrimination by the complement system S. Meri

binds to C3b and promotes its inactivation by factor I. certain density can be extrapolated from the elongated
Factor H and the membrane protein CD55 (decay- structure of factor H. Once two C3b:s on a surface are
accelerating factor, DAF) can shorten the half-life of sufficiently close to each other, a single factor H could
the convertase by rapidly dissociating Bb [44]. bind simultaneously to two C3b molecules via its N-
terminal binding site [in complement control protein
(CCP) domains 2–5] and the C-terminal site (in
Control of AP activation
domains 19–20). Alternatively, in case, factor H forms
If the target surface supports AP activation, C3 activa- dimers under physiological conditions, they could rec-
tion will become amplified in cycles and the target ognize two C3b molecules when they are sufficiently
coated by C3b molecules within minutes. In contrast, close to each other. In fact, early studies could demon-
if the surface is a nonactivating one, the generated strate a ‘density effect’, a relative increase in binding
C3b molecules will become inactivated by factors H of factor H (but not of factor B), when the density of
and I, and activation stops. The fate of C3b is deter- C3b molecules reached a distinct threshold value [50].
mined only by relatively minor differences in the affin- On a self cell surface, also the membrane cofactor
ity of factor H for C3b depending on whether C3b is protein (MCP, CD46) on most cells (excluding ery-
in the fluid phase, bound to an activating surface or to throcytes) or complement receptor type 1 (CR1,
a nonactivating surface [45,46]. Soluble C3b and non- CD35) on all blood cells and, for example, glomerular
activating surface-bound C3b molecules have similar podocytes, can replace factor H as cofactors [51].
affinities for factor H, exceeding the affinity for factor These molecules reside on host cells and thereby pro-
B. This leads to C3b inactivation to iC3b, followed by vide additional protection against attack by autolo-
further degradation to C3c and C3dg. gous complement. The protection is limited to cell
On an activating surface, the affinity of factor H for membranes, and mostly only to those cells that express
C3b is relatively low. Thus, factor B binds to C3b them. An exception is CR1 that can reach the surfaces
rather than factor H and participates in forming of neighboring cells to provide a helping hand in C3b
C3bBb, a convertase that can be further stabilized by inactivation. Additional regulators operating at the C3
properdin (P). It is important to note that the differ- step of AP (decay-accelerating factor, DAF/CD55)
ence in affinity between nonactivator and activator and terminal pathway activation (protectin, CD59)
bound C3b for factor H is crucial. Factor H binding further guarantee the safety of viable self cells against
depends on the context of the structure to which C3b
has become bound. It is not only an interaction with
Random target “Self”
C3b but simultaneous binding to C3b and the surface
in the immediate neighborhood of covalently bound X NA
C3b H, I iC3b
C3b. C3b itself does not seem to undergo a conforma- C3b iC3b
tional change for increasing its affinity for factor H. C3b C3b iC3b iC3b
Instead, a tripartite complex is formed of the surface,
C3, B, D, Mg++
C3b and factor H. In solution, C3b apparently can
accommodate factor H binding via multiple sites to A C3b clusters C5 A
C6
have high enough affinity to prevent factor B binding C7 MAC
“Nonself” MAC
[47,48]. C8
AP activation progresses as individual C3b mole- C9 MAC

cules deposit randomly on the target surface. After the


X = any surface, A = activator, NA = nonactivator
formation of a C3 convertase via the activation of C3b
bound factor B by factor D, more C3 molecules can Fig. 2. Discrimination between activating (nonself) and
be activated. These new C3b:s will generate a cluster nonactivating (self) surfaces by the alternative pathway of
of C3b:s around the initial C3b molecules (Fig. 2). complement. Initial binding of C3b to any surface (depicted X)
Although C3b can bind covalently to another C3b occurs randomly. If the surface supports binding of factor H (self,
molecule to generate C3b dimers, multiple layers of nonactivator) the C3b molecules become proteolytically inactivated
C3b:s do not become deposited on the target. C3b has by factor I and activation stops. If the surface does not bind factor
H, factor B forms a C3bBb convertase enzyme with the deposited
more preference to bind to C4b or IgG than to C3b
C3b and cleaves further C3 molecules to a cluster of C3b:s. These
[49]. Also, the susceptibilities of the two individual in turn can generate new C3 convertases, whereby activation
C3b molecules in the C3b dimer to inactivation by fac- becomes amplified. As an end result, the target becomes
tors H and I differ [49]. A natural mechanism to opsonized by C3b molecules and also membrane attack complexes
restrict excessive C3b deposition and to limit it to a (MAC) are formed.

2422 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

complement [52]. These two regulators are attached to C3b


CRP
C3c C3d
PTX3
cell membrane phospholipids via a glycophosphoinosi-
tol-linkage. An acquired defect in the anchoring sys- Factor H 1 4 7 8 11 12 13 14 19 20

tem and consequent lack of DAF and CD59 causes a ++ ++ ++


DDD AMD HUS
disease called paroxysmal nocturnal hemoglobinuria
(MPGN2)
(PNH) [53,54]. In PNH, descendants of the mutated
cell clone become susceptible to complement attack Fig. 3. Schematic structure of factor H. Binding sites for C3b,
[55,56]. In this disease, attacks may be precipitated, for polyanions (marked with ++) and pentraxins CRP and PTX-3 are
example, by infections that put the AP activation distributed along the chain of 20 complement control protein
ongoing. The night-time has been suggested to pro- domains (CCP). Locations of sites, whose disturbances because of
mutations or autoantibodies predispose to diseases. DDD, dense
mote disease attacks, because of low pH that has been
deposit disease (membranoproliferative glomerulonephritis type 2,
suggested to increase AP activity. In reality, attacks
MPGN2); AMD, age-related macular degeneration; HUS, hemolytic
are not restricted to night-time and wide variation in uremic syndrome.
the disease spectrum exists. Clinical problems in PNH
are related to a tendency for thrombosis in blood ves-
sels [57]. This indicates that the complement and coag- Furthermore, ligands for factor H include the pen-
ulation systems are closely interconnected. traxins C-reactive protein and PTX-3. They have a
major binding site on domain 7 of factor H [69–71]
and a secondary site on domains 8–11 [69] or 19–20
Binding of factor H to surface-
[72]. Thereby, factor H can wrap itself around CRP
associated C3b–key to discrimination
and bind to a different site than C1q [70]. The pro-
The first clue to the possible mechanism for protecting posed function for the pentraxin-factor H interaction
self cells from complement attack came when it was may be related to labeling an area of tissue damage,
shown that human and sheep red blood cells used sia- where the pentraxins bind to injured tissue compo-
lic acid for their protection against AP attack [58,59]. nents and activate the classical complement pathway
Subsequently, it was shown that factor H in the AP via C1q. To avoid excessive inflammation and to
was the factor that directly interacted with the surface delineate the area of damage for clearance, the
to mediate the discrimination between activators and pentraxins would recruit factor H, which can inhibit
nonactivators [45]. Factor H recognized sialic acids AP activation and promote inactivation of C3b to
and other polyanions, like heparan sulfate on surfaces, iC3b, which acts as a high affinity ligand for the
where prior deposition of C3b had occurred. Further- macrophage receptor CR3 (CD11b/18). Phagocytic
more, factor H showed specificity in its binding to clearance mediated by CR3 occurs in a noninflamma-
polyanions, the mere negative charge was not sufficient tory fashion, because upon engagement of this inte-
for the interaction [60]. Binding can occur to gly- grin receptor macrophages are signaled to produce
cosaminoglycans (e.g., hyaluronan, heparan sulfate, immunosuppressive cytokines TGF-beta and inter-
chondroitin sulfate, dermatan sulfate) or proteoglycans leukin 10 [24].
(e.g., versican, syndecan, decorin) [60] Later studies The N terminus of factor H (domains 1–4) was
were able to demonstrate that there were actually mul- found to bind to C3b and contain the functional activ-
tiple binding sites for polyanions in factor H [61–63]. ities, both the cofactor activity for C3b inactivation
Factor H has three binding sites for the main comple- and the decay-accelerating activity for the C3bBb con-
ment factor C3b and two to three for polyanions vertase [73]. Additional binding sites for C3b are
[48,64] (Fig. 3). One heparin site is located on domain apparently employed for allowing more dynamic inter-
7 [61]. Using site-directed mutagenesis, the endothelial actions with soluble or surface-associated C3b in the
cell, C3d- and heparin-binding sites were fine-mapped context of different kinds of surfaces [65,66]. An alter-
to domains 19–20 of factor H [62,64–66]. Studies on natively spliced product of the factor H gene, factor
the lipo-oligosaccharide sialic acid of Neisseria gonor- H-like protein 1 (FHL-1), contains the first seven
rhoeae [67] suggested that the region containing the domains of factor H and four unique amino acids [73].
domain 20 is more selective for sialic acid than the site Thereby, it contains the major functional domains of
at domain 7. Sialylated molecules include proteins, car- the N terminus but lacks the most critical self-recogni-
bohydrates, and lipids like gangliosides and glycosph- tion domains of the C terminus. It is a form of factor
ingolipids [68]. Although less studied, additional H that seems to lack the activator-nonactivator dis-
protection may be provided by negatively charged criminating capacity, despite the presence of the hep-
phospholipids. arin-binding site on domain 7.

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2423
Self-nonself discrimination by the complement system S. Meri

The ability to restrict AP activation is not limited to The atomic resolution structure of a complex
healthy normal cells but also malignant tumor cells between factor H C terminus and complement C3d
have means to protect themselves. Tumor cells usually has been solved by two groups [80,81]. These studies
express sialic acids, mucins, and sulfated polyanions at revealed the structural basis, whereby the AP can dis-
least as much as normal cells. In addition, for exam- criminate between host and nonhost structures
ple, glioblastoma and ovarian tumor cells can produce (schematically depicted in Fig. 4). This occurs by a
FHL-1 in higher relative amounts than normally joint dual recognition of distinct negatively charged
observed in blood [74,75]. Strikingly, factor H has host structures and the C3d domain of C3b by factor
been found to be the tumor antigen that is recognized H. If the target is not recognized as ‘self’, it will
by the BTA-STAT and BTA-TRAK tests that are rapidly be attacked by the alternative AP amplification
used for the detection of urinary bladder cancer [76]. system. Both groups found a C3d-binding site on fac-
Also tumors originating from skin have been found to tor H domain 19, whereas Kajander et al. demon-
produce factor H [77]. Thus, while most tumor cells strated that there is an additional binding site for C3d
bind the soluble complement inhibitors FHL-1 and also on domain 20. Domain 19 is apparently the pre-
factor H, some are even capable of synthesizing them ferred site for C3d because in domain 20 the surface
into their immediate vicinity. The local production of polyanion recognition site overlaps with that of C3d.
factor H and FHL-1 may create ‘a complement-free In the absence of polyanions, however, binding of fac-
zone’ around the tumors. Increased angiogenesis may tor H to a layer of C3d molecules could signal a satu-
bring more blood and complement to such sites, ration of the surface with deposited C3b/d to prevent
whereas altered metabolism and relative ischemia may unnecessary further C3b deposition. Surface-associated
increase complement activity at such sites, for example, C3d could thus compensate for the lack of polyanions
by lowering pH or providing targets for complement by attracting factor H through domain 20. Thereby, it
attack. FHL-1 does not recognize its targets on the is possible that once a sufficient layer of C3d, and pos-
basis of sialic acid expression but rather by sulfated sibly of C3b molecules, as well, has been deposited,
molecules like glycosaminoglycans. Factor H/FHL-1 AP activation no longer continues and stops. In solu-
production and binding to tumor cells should be kept tion, C3b is fully accessible to factor H and becomes
in mind when designing complement-activating mono- rapidly inactivated by factor I. Interestingly, when
clonal antibodies for therapy. C3b is bound covalently to an activating surface or,
for example, to IgG, C4b or another C3b, the access
Structural analyses of the recognition
process
Most recently, the AP recognition system has been
explored with the help of structural analyses, princi-
pally by X-ray crystallography. These have shed light
on the actual interacting amino acids in the different
factor H domains, target polyanions and C3b and
other details of the recognition process. Crucial for all
studies on complement was the elucidation of the X-
ray crystal structures of C3 and C3b by the group of
Piet Gros [78,79]. C3 has 13 domains that undergo
changes upon conversion into C3b. The biggest
changes are the release of C3a (a chemotaxin and an
anaphylatoxin) and transfer of the C3d part (the thi-
olester-containing domain, TED) to another region of
the molecule to allow its access to the surface where it
can bind covalently either to exposed hydroxyl (pre-
ferred) or amino groups [79]. Otherwise, C3b does not Fig. 4. Discrimination between self and nonself on cell surfaces by
factor H. On nonself surfaces like microbes (above), complement
seem to have any special surface recognition domains,
activation proceeds by default. On self surfaces, factor H binds to
which is in line with previous studies showing that the C3b-polyanion complex and inhibits complement activation by
recognition and discrimination is dependent on a sub- three different mechanisms: (1) inhibition of factor B binding, (2)
sequent interaction of factor H with the C3b-target decay dissociation of Bb from the C3 convertase, C3bBb and (3)
complex [45,46]. promotion of inactivation of C3b by factor I.

2424 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

of factor H to C3b is restricted [49]. This makes the activity of factor H (in the N terminus) is disturbed or
bound C3b more resistant to inactivation and subjects the activity of the C3bBb convertase is enhanced. The
to become a subunit of the C3/C5 convertase leading underlying reasons can be mutations in factor H [98]
to AP activation, amplification, and initiation of the or autoantibodies against C3bBb (C3 nephritic factor
terminal pathway. Thus, the AP has elegant mecha- [99]; or factor H N terminus [92]. Reduced factor H
nisms to (a) promote activation on nonself surfaces control function or enhanced C3bBb activity can lead
and (b) stop it when sufficient and to (c) block activa- to overactivation of the AP in the fluid phase, binding
tion on self surfaces and (d) by soluble C3b to prevent and accumulation of C3b molecules and the membrane
self tissue damage and excess inflammation. Further- attack complex at sites that are not protected by the
more, the acute phase protein CRP can regulate and membrane-associated complement inhibitors. Typi-
direct the inhibitory activity of factor H to areas of tis- cally, this occurs at the glomerular basement mem-
sue damage during acute inflammation [69]. The other branes (GBM) of kidneys. Eventually, obstruction and
pentraxins, like PTX-3, may have this activity, as well thickening of the GBM leads to glomerular damage
[71]. and loss of kidney function.
While the pathogenetic processes in aHUS and
DDD are now relatively well understood, that of
Errors in discrimination–the basis for
AMD still remains unclear. A predisposing factor to
hemolytic uremic syndrome
both major forms of AMD, geographic atrophy (dry
The importance of the AP recognition and amplifica- AMD) or neovascular, wet AMD, is a polymorphic
tion mechanism is apparent in the disease atypical variant that changes amino acid tyrosine on position
hemolytic uremic syndrome (aHUS). aHUS is an 402 to histidine in domain 7 of factor H [94–97]. This
experiment of nature, where single amino acid muta- influences the binding of factor H to C-reactive protein
tions in the recognition domains of FH or in C3d [70,72,100] and to glycosaminoglycans [101]. In princi-
predispose vascular endothelial cells, blood cells, and ple, factor H function could be disturbed in the retinal
platelets to damage [66,82–84]. Factor H mutation- epithelial cells (RPE), the Bruch’s membrane beneath
associated aHUS is a severe disease that even in the RPE, the lipofuscin particles themselves or in the
heterozygous form can lead to end stage renal failure, small capillaries behind the epithelial cell layer. The
brain edema, and death of the patient. Factor H high turnover of retinal pigment into lipofuscin and
mutations underlie approximately 50% of aHUS other degradation products requires an efficient clear-
cases that have a genetic background. Other causes ance system. In AMD, the clearance is compromised
for aHUS are mutations in factor I, MCP/CD46, C3, and deposits of lipofuscin accumulate in the retina
factor B or thrombomodulin [85–88]. Also, autoanti- gradually obstructing the vision of the patients. Factor
bodies against the C terminus of factor H can cause H abnormality could predispose to inflammation or
a similar yet somewhat milder form of aHUS [89]. In lack of clearance of the retinal pigment remnants or
about a third of cases, the underlying reason is still both.
unknown. aHUS is distinct from the more common
‘typical’ form of HUS that is caused by Shiga-like
Role for sialic acid
toxins produced by certain serotypes of enterohemor-
rhagic E. coli (EHEC, e.g., O157 or O104 serotypes) The factor H-mediated protection of glomerular and
or Shigella bacteria. The role of complement in this other types of endothelial cells, blood cells (erythro-
‘eHUS’ is still uncertain [90]. Common to all forms cytes and leukocytes) as well as platelets against com-
of HUS, however, is endothelial, blood cell and plate- plement was for a long time assumed to be due to
let damage, which predisposes to thrombosis, an glycosaminoglycans, especially heparin or heparan sul-
entity collectively referred to as thrombotic microan- fate. Heparin was also used as a model polyanion in
giopathy (TMA). Other forms of TMA can occur in studies addressing self-nonself discrimination and self-
thrombotic thrombocytopenic purpura (TTP), PNH, protection by the AP. It was, however, puzzling that
secondary to transplantation, infection, drug treat- some mutations in the FH C terminus associated with
ment, malignancy or as a complication of pregnancy aHUS (like L1189R and E1198A) did not disrupt
[87]. binding of factor H to heparin but rather increased it
Errors (mutations, polymorphisms) in factor H or [102] . Recently, we found that these mutations inter-
autoantibodies against it can predispose also to dense fere most specifically with the ability of factor H to
deposit disease [91–93] and age-related macular degen- bind to sialic acid [103]. Removal of sialic acid from
eration (AMD) [94–97]. In DDD, the functional platelets and endothelial cells was found to reduce

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2425
Self-nonself discrimination by the complement system S. Meri

their ability to bind factor H and be protected against variable. Usually, the FHR proteins have relatively
complement attack. Thus, the fundamental defect in low plasma concentrations, that of FHR1 being the
aHUS associated with factor H mutations seems to highest (approximately 50 µgmL 1). At local sites and
be an inability of the mutated factor H C terminus to during inflammation, like in the middle ear fluid dur-
recognize sialic acid on the surfaces of cells in contact ing otitis media, the concentrations may, however, be
with the plasma complement system. Sialic acids are a higher [107].
group of carbohydrates that are regulated dynamically Strikingly, while the FHRs lack the N-terminal
at different times, ages, and conditions [68]. The functional domains, they all have homologs of the two
GM3 type ganglioside seemed to be the preferred sia- most C-terminal CCP domains of factor H [108].
lic acid recognized by factor H [104]. Some viruses, Thus, they may have an ability to distinguish between
like the influenza viruses, have sialic acids both as AP activators and nonactivators. But, why would they
their receptors on host respiratory tracts as well as do this if they do not have the actual functional
targets for their major enzymes, neuraminidases. Also, domains? The answer to this puzzling question may be
some non-shigatoxin-producing bacteria associated related to an ability to ‘fine-tune’ and regulate the
with aHUS, like pneumococcus, have neuraminidases. actual factor H-mediated control of complement acti-
The finely tuned self-recognition mediated by factor vation [107]. At times, factor H may unnecessarily
H can thus have implications beyond the typically strongly inhibit complement attack against a target.
complement-associated kidney diseases. By implica- FHRs have an ability to compete out the C terminus
tion, the maintenance of self-barriers and ability to of factor H, whereby the factor H-mediated control is
discriminate between self and nonself by complement released and complement activation is temporarily
may play a role in other common diseases, where sub- enhanced. The dimerization and heterodimerization of
tle changes on cell surfaces or other host structures FHRs would further make this effect stronger [109].
may occur. This may be a way to enhance attack, for example,
against microbes that have an ability to bind factor H
via the C terminus [110]. FHRs could then act as ‘de-
Factor H-like protein 1 and factor
coys’ to prevent factor H binding and allow comple-
H-related proteins
ment attack against the invader. The function of
As indicated above, factor H has the ability to distin- FHRs could thus represent a rational evolutionary
guish between activators and nonactivators by the step for fine-tuning the regulatory activity of factor H.
self-recognition domains in the C terminus of the Abnormalities in FHRs have been described, and
molecule (domains 19–20), while the actual functional some of them are related to distinct disease conditions.
activities (cofactor and decay-accelerating activity) Because of many similar sequence areas, the RCA
reside in the N-terminal domains 1–4 [73]. The alter- cluster in chromosome 1 is prone to genomic rear-
natively spliced product from the factor H gene rangements through gene conversion and nonhomolo-
FHL-1 is always active in the fluid phase but lacks gous recombination [106]. The deletion of FHR1-3
the discriminatory ability on surfaces. Its blood con- gene is relatively common leading to a deficiency of
centration is usually only about 10% of that of factor both FHR1 and FHR3 [111]. Interestingly, a large
H but at local sites it can be present at higher con- majority of individuals, who develop anti-factor H
centrations. Also, some tumor cells produce equal antibodies and aHUS have this deletion. The anti-fac-
amounts of FH and FHL-1. FHL-1 may thus have tor H antibodies appear to partially disrupt the self-
different clearance kinetics than FH and it may also protective ability of factor H. The reason for this con-
be produced in different quantities at different sites nection is not entirely clear, but it is possible that
or conditions [105]. The tumor cells may use FHL-1 FHRs 1 and 3 somehow induce or maintain tolerance
for their own protection in the local microenviron- to factor H. Alternatively, the interaction of factor H
ment where it is being produced [74]. in the absence of competing FHRs with microbial fac-
In addition to factor H/FHL-1, five different factor tor H-binding proteins (FHBPs) could generate
H-related proteins (FHRs 1–5) are encoded by their neoepitopes against which antibodies develop, among
own genes in chromosome 1 (1q31-32) in the regula- those also anti-factor H antibodies [112]. The emerging
tors of complement activation (RCA) gene cluster, anti-factor H antibodies are primarily directed against
where genes for most other complement regulators are the C terminus of factor and thus prevent its ability to
located, as well [106]. The FHRs vary in length and control complement activation on blood cells and
number of CCP domains. Each may have their own endothelial cells. This leads to aHUS, which–luckily–in
properties, but reports on their functional activities are most cases is less severe than in cases related to FH

2426 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

mutations. Also, removal of antibodies with plasma- Microbial escape of complement by


pheresis provides an additional method of therapy. mimicry of self
The role of FHRs in regulating factor H is sup-
ported by the recent observations of abnormal oligo- Despite the potent antimicrobial effects of the comple-
mers or hybrids of the FHR molecules associated with ment system many microbes are resistant to
C3 glomerulopathy [113,114]. C3 glomerulopathy opsonophagocytosis or direct complement-mediated
patients have C3 deposits in their kidney glomeruli in killing. Often, the complement resistant microbes are
the absence of antibody deposits. DDD is considered pathogens for humans. The reasons why certain
as one form of C3 glomerulopathy. C3 glomeru- microbes are virulent pathogens and others, often even
lonephritis (C3GN) is another one, but without dense close relatives, are not has remained obscure in many
deposits. This form is associated with FHR abnormali- cases. Apparently, part of the species specificity and
ties (oligomers, hybrids, mutations, deletions). FHR5 virulence spectra of microbes is dependent on resis-
mutation has been found to cause a glomerular dis- tance to complement [118,119]. Many common patho-
ease, FHR5 nephropathy, which is endemic in Cypri- gens escape complement attack using specific proteins
ots [113]. FHR-1 mutation and abnormal FHR and mechanisms. For example, we identified a new
oligomerization have been found in connection with family of complement inhibitors from the Lyme dis-
C3 glomerulopathy [114]. Some forms may even be ease Borrelia burgdorferi spirochetes [120,121]. The
related to C3 mutations. Common to all these disor- proteins belong to the OspE family of outer membrane
ders is that they disturb AP regulation and predispose proteins that have the ability to hijack factor H from
to glomerular disease. the human or animal host [118]. Subsequently, other
groups made similar observations and another FHBP
family, CspA was also found [122–126]. Factor H
Protecting self by membrane binding molecules have since been found from many
regulators of complement different kinds of bacteria, including different types of
Discrimination between self and nonself by the com- pathogenic streptococci [127] and Gram-negative bac-
plement system is not limited to factor H, although it teria [128–130].
has the unique ability to ‘sense’ a variety of targets Group B meningococcus, during recent years, has
and operate from the fluid phase. On human cell mem- been the most important causative agent of meningitis
branes, complement activation is inhibited by comple- and meningococcal sepsis in the Western world. The
ment receptor type 1 (CR1, CD35), membrane development of vaccines against this bacterium has
cofactor protein (MCP, CD46), and decay-accelerating been delayed because its polysialic acid capsule is non-
factor (DAF, CD55). Their task is to limit comple- immunogenic and potentially cross-reactive with
ment activation at the C3 step [52]. Another regulator, human brain [131]. In addition to having an antiop-
protectin or CD59, inhibits formation of the mem- sonic capsule, group B meningococcus was found to
brane attack complex by binding to intermediate C5b- bind the complement inhibitor C4 bp by porin A
8 and C5b-9 terminal complement complexes and pre- [132]. This could prevent both antibody-mediated clas-
venting incorporation and polymerization of C9 sical and lectin pathway activation. Using whole gen-
[115,116]. These molecules specifically protect ‘self’. ome screening methodology and systematic analysis,
They are to some extent species selective, that is, tuned so called reverse vaccinology, proteins with potential,
principally to protect against complement of homolo- as vaccines, were found from meningococcus [133].
gous species. While mixing cells from one species and One of the most promising vaccine candidates turned
blood plasma from another is an artificial situation, out to be a FHBP [128]. Subsequently, the FHBP was
attempts have been made to transplant organs across included in a new multicomponent vaccine against
species, for example, from pigs to humans. This usu- group B meningococcal infections [129]. This vaccine
ally fails because there may be natural antibodies (Bexsero, Novartis) has now been approved for use in
(antigalactose, for example) against the other species’ several countries. Another vaccine containing two out
cells and because the complement regulators in the of the three main variants of FHBP (Trumenba,
transplanted organ may not protect against the recipi- Pfizer) has also been developed [130].
ent complement. Attempts to overcome this problem In retrospect, a concept that was developed earlier
by genetic engineering to remove antigen targets or to from functional hypothesis-driven studies [121,134] led
add transgenic human DAF or MCP to pigs have been to the same conclusions as unbiased genome-wide
under development, but so far the approach has not searches for vaccine candidates [133]. The most elegant
been adopted to general use [117]. feature of the FHBP-based vaccines is that antibodies

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2427
Self-nonself discrimination by the complement system S. Meri

developed against the proteins not only recognize the markers of ‘self’ provides protection to our own
target microbe but they also neutralize an important intact viable cells and tissues. In contrast, activating
virulence property, that is, recruitment of factor H by structures are recognized by default: anything not
the microbe. Vaccines against group B meningococcus positively recognized by factor H is being attacked
represent a breakthrough, because they complement upon. This fundamental mechanism leads to
the existing vaccines against other capsule types of opsonization and phagocytosis or direct killing of
meningococci. It is particularly important for use in microbes and other targets. Even senescent, ischemic,
individuals who lack AP or terminal pathway comple- injured or apoptotic ‘self’ cells can be recognized in
ment components or who are treated with eculizumab this way for removal by macrophages. An indication
(anti-C5) antibody therapy. Otherwise, complement of the importance of the recognition system is muta-
deficiency in these situations would lead to a signifi- tions in factor H that predispose to severe diseases,
cantly increased (around 1000-fold) risk for meningo- where complement attacks autologous cells. In aHUS,
coccal meningitis and sepsis. alterations in the recognition system predispose to
From the self-nonself discrimination point of view, complement attack against blood cells and vascular
microbes that bind factor H can be considered to endothelial cells causing thrombotic microangiopathy
mimic ‘self’. A comparison of microbial FHBPs has and damage to many tissues (e.g., kidneys and brain).
shown that they fall into two major structural cate- Most specifically, the factor H mutations affect sialic
gories: alpha-helical coiled-coil structures (such as yer- acid recognition. Pathogenic microbes have also
sinial YadA, group A streptococcal M-protein and learned to use factor H to escape C attack. Thus,
group B streptococcal Bac-protein) or beta-barrel-type many bacteria express specific proteins with a similar
membrane proteins (Rck of salmonella, Ail of Yersi- ability to exploit factor H. The ability to bind factor
nia, and OspE of borrelia) [134]. The binding sites of H is so far the best described feature that discrimi-
the proteins on factor H are usually located in the nates pathogenic microbes from nonpathogenic ones.
heparin-binding domains 7 or 20. The site on domain
7 is used, for example, by the meningococcal FHBP.
Acknowledgements
The domain 20 is used by the borrelial OspE and
many other microbial proteins. A survey of the finger- Matti Laine is kindly acknowledged for drawing Fig. 1.
prints of several different bacterial proteins and All current and previous members of my research
microbes on the domain 20 of factor H (using a panel group and collaborators are gratefully acknowledged.
of mutants) showed that the microbes use, not only Especially, the following have contributed to studies
the same domain, but to a large extent also the same presented in this review: Sakari Jokiranta, Hanna
amino acid side chains for the interaction [135]. It is Jarva, Martin Reichhardt, Matti Laine, Sami Jun-
astonishing that microbes as distant as borrelia, strep- nikkala, Taru Meri, Livija Deban, Jens Hellwage,
tococcus, haemophilus, pseudomonas, and even the Derek Ho, Satu Hyv€ arinen, Adrian Goldman, Veli-
yeast Candida albicans use a common site on factor H Pekka Jaakola, Tommi Kajander. Especially I want to
to recruit this inhibitor for their protection against thank prof Michael Pangburn, University of Texas
complement attack. Health Science Center. The author has received finan-
cial support from the Academy of Finland, The Sigrid
Juselius Foundation, The Stockmann Foundation,
Conclusion
Signe and Ane Gyllenberg Foundation, Helsinki
The classical and lectin pathways of complement use University Hospital Grants (EVO), Finland and
antibodies and other sensor molecules for specific Humanitas University, Milan, Italy.
recognition of targets. Somewhat miraculously, how-
ever, the alternative pathway (AP) relies only on two
References
molecules, C3b and factor H, to distinguish between
a wide variety of nonself structures from self. The 1 Endo Y, Matsushita M and Fujita T (2015) New
AP deposits covalently bound C3b on activating insights into the role of ficolins in the lectin pathway
(nonself) structures via a positive feedback loop of innate immunity. Int Rev Cell Mol Biol 316, 49–110.
amplification system. The actual discrimination is per- 2 Garcia-Vallejo JJ and van Kooyk Y (2013) The
formed by the key regulator factor H, which has physiological role of DC-SIGN: a tale of mice and
binding sites for cell surface polyanions (sialic acids, men. Trends Immunol 34, 482–486.
glycosaminoglycans, phospholipids) in addition to 3 Karre K (2008) Natural killer cell recognition of
those for C3b and C3d. This robust recognition of missing self. Nat Immunol 9, 477–480.

2428 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

4 Carrillo-Bustamante P, Kesmir C and de Boer RJ 18 Lachmann PJ (2009) The amplification loop of the
(2016) The evolution of natural killer cell receptors. complement pathways. Adv Immunol 104, 115–149.
Immunogenetics 68, 3–18. 19 Kohl J (2006) Self, non-self, and danger: a
5 Walport MJ (2001) Complement. First of two parts. N complementary view. Adv Exp Med Biol 586, 71–94.
Engl J Med 344, 1058–1066. 20 Holmskov U, Thiel S and Jensenius JC (2003)
6 Meri S (2007) Loss of self-control in the complement Collections and ficolins: humoral lectins of the innate
system and innate autoreactivity. Ann N Y Acad Sci immune defense. Annu Rev Immunol 21, 547–578.
1109, 93–105. 21 Schaffler A and Buechler C (2012) CTRP family:
7 Girardi G, Yarilin D, Thurman JM, Holers VM and linking immunity to metabolism. Trends Endocrinol
Salmon JE (2006) Complement activation induces Metab 23, 194–204.
dysregulation of angiogenic factors and causes fetal 22 Gaboriaud C, Ling WL, Thielens NM, Bally I and
rejection and growth restriction. J Exp Med 203, 2165– Rossi V (2014) Deciphering the fine details of c1
2175. assembly and activation mechanisms: “mission
8 Lokki AI, Heikkinen-Eloranta J, Jarva H, Saisto T, impossible”? Front Immunol 5, 565.
Lokki ML, Laivuori H and Meri S (2014) 23 Ghebrehiwet B, Hosszu KK, Valentino A, Ji Y and
Complement activation and regulation in preeclamptic Peerschke EI (2014) Monocyte expressed
placenta. Front Immunol 5, 312. macromolecular C1 and C1q receptors as molecular
9 Jurianz K, Ziegler S, Garcia-Schuler H, Kraus S, sensors of danger: implications in SLE. Front Immunol
Bohana-Kashtan O, Fishelson Z and Kirschfink M 5, 278.
(1999) Complement resistance of tumor cells: basal and 24 Amarilyo G, Verbovetski I, Atallah M, Grau A, Wiser
induced mechanisms. Mol Immunol 36, 929–939. G, Gil O, Ben-Neriah Y and Mevorach D (2010)
10 Gorter A and Meri S (1999) Immune evasion of tumor iC3b-opsonized apoptotic cells mediate a distinct anti-
cells using membrane-bound complement regulatory inflammatory response and transcriptional NF-
proteins. Immunol Today 20, 576–582. kappaB-dependent blockade. Eur J Immunol 40, 699–
11 Serruto D, Rappuoli R, Scarselli M, Gros P and van 709.
Strijp JA (2010) Molecular mechanisms of complement 25 Ross GD (2000) Regulation of the adhesion versus
evasion: learning from staphylococci and cytotoxic functions of the Mac-1/CR3/alphaMbeta2-
meningococci. Nat Rev Microbiol 8, 393–399. integrin glycoprotein. Crit Rev Immunol 20, 197–222.
12 Ricklin D, Hajishengallis G, Yang K and Lambris JD 26 Jack DL, Klein NJ and Turner MW (2001) Mannose-
(2010) Complement: a key system for immune binding lectin: targeting the microbial world for
surveillance and homeostasis. Nat Immunol 11, 785–797. complement attack and opsonophagocytosis. Immunol
13 Clas F, Schmidt G and Loos M (1985) The role of the Rev 180, 86–99.
classical pathway for the bactericidal effect of normal 27 Saxen H, Reima I and Makela PH (1987) Alternative
sera against gram-negative bacteria. Curr Top complement pathway activation by Salmonella O
Microbiol Immunol 121, 19–72. polysaccharide as a virulence determinant in the
14 Richards RL, Gewurz H, Osmand AP and Alving CR mouse. Microb Pathog 2, 15–28.
(1977) Interactions of C-reactive protein and 28 Matsushita M, Kilpatrick D, Shiraki H, Liu Y,
complement with liposomes. Proc Natl Acad Sci USA Tateishi K, Tsujimura M, Endo Y and Fujita T (2014)
74, 5672–5676. Purification, measurement of concentration, and
15 Rossen RD, Michael LH, Kagiyama A, Savage HE, functional complement assay of human ficolins.
Hanson G, Reisberg MA, Moake JN, Kim SH, Self D Methods Mol Biol 1100, 141–159.
and Weakley S (1988) Mechanism of complement 29 Rooryck C, Diaz-Font A, Osborn DP, Chabchoub E,
activation after coronary artery occlusion: evidence Hernandez-Hernandez V, Shamseldin H, Kenny J,
that myocardial ischemia in dogs causes release of Waters A, Jenkins D, Kaissi AA et al. (2011)
constituents of myocardial subcellular origin that Mutations in lectin complement pathway genes
complex with human C1q in vivo. Circ Res 62, 572– COLEC11 and MASP1 cause 3MC syndrome. Nat
584. Genet 43, 197–203.
16 Thiel S and Gadjeva M (2009) Humoral pattern 30 Madsen J, Mollenhauer J and Holmskov U (2010)
recognition molecules: mannan-binding lectin and Review: Gp-340/DMBT1 in mucosal innate immunity.
ficolins. Adv Exp Med Biol 653, 58–73. Innate immunity 16, 160–167.
17 Garred P, Honore C, Ma YJ, Munthe-Fog L and 31 Ligtenberg AJ, Veerman EC, Nieuw Amerongen AV
Hummelshoj T (2009) MBL2, FCN1, FCN2 and and Mollenhauer J (2007) Salivary agglutinin/
FCN3 – the genes behind the initiation of the lectin glycoprotein-340/DMBT1: a single molecule with
pathway of complement. Mol Immunol 46, 2737–2744. variable composition and with different functions in

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2429
Self-nonself discrimination by the complement system S. Meri

infection, inflammation and cancer. Biol Chem 388, 45 Meri S and Pangburn MK (1990) Discrimination
1275–1289. between activators and nonactivators of the alternative
32 Reichhardt MP, Loimaranta V, Thiel S, Finne J, Meri pathway of complement: regulation via a sialic acid/
S and Jarva H (2012) The salivary scavenger and polyanion binding site on factor H. Proc Natl Acad
agglutinin binds MBL and regulates the lectin pathway Sci USA 87, 3982–3986.
of complement in solution and on surfaces. Front 46 Pangburn MK, Pangburn KL, Koistinen V, Meri S
Immunol 3, 205. and Sharma AK (2000) Molecular mechanisms of
33 Reichhardt MP, Jarva H, de Been M, Rodriguez JM, target recognition in an innate immune system:
Jimenez Quintana E, Loimaranta V, de Vos WM and interactions among factor H, C3b, and target in the
Meri S (2014) The salivary scavenger and agglutinin in alternative pathway of human complement. J Immunol
early life: diverse roles in amniotic fluid and in the 164, 4742–4751.
infant intestine. J Immunol 193, 5240–5248. 47 Jokiranta TS, Zipfel PF, Hakulinen J, Kuhn S,
34 Reichhardt MP and Meri S (2016) SALSA: a regulator Pangburn MK, Tamerius JD and Meri S (1996)
of the early steps of complement activation on Analysis of the recognition mechanism of the
mucosal surfaces. Front Immunol 7, 85. alternative pathway of complement by monoclonal
35 Leito JT, Ligtenberg AJ, van Houdt M, van den Berg anti-factor H antibodies: evidence for multiple
TK and Wouters D (2011) The bacteria binding interactions between H and surface bound C3b. FEBS
glycoprotein salivary agglutinin (SAG/gp340) activates Lett 393, 297–302.
complement via the lectin pathway. Mol Immunol 49, 48 Jokiranta TS, Hellwage J, Koistinen V, Zipfel PF and
185–190. Meri S (2000) Each of the three binding sites on
36 Seldin MM, Tan SY and Wong GW (2014) Metabolic complement factor H interacts with a distinct site on
function of the CTRP family of hormones. Rev Endocr C3b. J Biol Chem 275, 27657–27662.
Metab Disord 15, 111–123. 49 Meri S and Pangburn MK (1990) A mechanism of
37 Nonaka M and Yoshizaki F (2004) Primitive activation of the alternative complement pathway by
complement system of invertebrates. Immunol Rev 198, the classical pathway: protection of C3b from
203–215. inactivation by covalent attachment to C4b. Eur J
38 Blandin SA, Marois E and Levashina EA (2008) Immunol 20, 2555–2561.
Antimalarial responses in Anopheles gambiae: from a 50 Koistinen V (1991) Effect of complement-protein-
complement-like protein to a complement-like C3b density on the binding of complement factor H
pathway. Cell Host Microbe 3, 364–374. to surface-bound C3b. Biochem J 280 (Pt 1), 255–
39 Pangburn MK and Muller-Eberhard HJ (1984) The 259.
alternative pathway of complement. Springer Semin 51 Hourcade D, Liszewski MK, Krych-Goldberg M and
Immunopathol 7, 163–192. Atkinson JP (2000) Functional domains, structural
40 Charlesworth JA, Williams DG, Sherington E, variations and pathogen interactions of MCP, DAF
Lachmann PJ and Peters DK (1974) Metabolic and CR1. Immunopharmacology 49, 103–116.
studies of the third component of complement 52 Morgan BP and Meri S (1994) Membrane proteins
and the glycine-rich beta glycoprotein in patients that protect against complement lysis. Springer Semin
with hypocomplementemia. J Clin Invest 53, 1578– Immunopathol 15, 369–396.
1587. 53 Nicholson-Weller A, March JP, Rosenfeld SI and
41 Pangburn MK and Muller-Eberhard HJ (1980) Austen KF (1983) Affected erythrocytes of patients
Relation of putative thioester bond in C3 to activation with paroxysmal nocturnal hemoglobinuria are
of the alternative pathway and the binding of C3b to deficient in the complement regulatory protein, decay
biological targets of complement. J Exp Med 152, accelerating factor. Proc Natl Acad Sci USA 80, 5066–
1102–1114. 5070.
42 Law SK and Dodds AW (1996) Catalysed hydrolysis – 54 Pangburn MK, Schreiber RD, Trombold JS and
the complement quickstep. Immunol Today 17, 105. Muller-Eberhard HJ (1983) Paroxysmal nocturnal
43 Pangburn MK and Muller-Eberhard HJ (1986) The hemoglobinuria: deficiency in factor H-like functions
C3 convertase of the alternative pathway of human of the abnormal erythrocytes. J Exp Med 157, 1971–
complement. Enzymic properties of the bimolecular 1980.
proteinase. Biochem J 235, 723–730. 55 Rosse WF and Dacie JV (1966) Immune lysis of
44 Fujita T, Inoue T, Ogawa K, Iida K and Tamura N normal human and paroxysmal nocturnal
(1987) The mechanism of action of decay-accelerating hemoglobinuria (PNH) red blood cells. II. The role of
factor (DAF). DAF inhibits the assembly of C3 complement components in the increased sensitivity of
convertases by dissociating C2a and Bb. J Exp Med PNH red cells to immune lysis. J Clin Invest 45, 749–
166, 1221–1228. 757.

2430 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

56 Bessler M, Mason PJ, Hillmen P, Miyata T, Yamada serum resistance of sialylated Neisseria gonorrhoeae. J
N, Takeda J, Luzzatto L and Kinoshita T (1994) Exp Med 187, 743–752.
Paroxysmal nocturnal haemoglobinuria (PNH) is 68 Varki A and Gagneux P (2012) Multifarious roles of
caused by somatic mutations in the PIG-A gene. sialic acids in immunity. Ann N Y Acad Sci 1253, 16–36.
EMBO J 13, 110–117. 69 Jarva H, Jokiranta TS, Hellwage J, Zipfel PF and
57 Hill A, Kelly RJ and Hillmen P (2013) Thrombosis in Meri S (1999) Regulation of complement activation by
paroxysmal nocturnal hemoglobinuria. Blood 121, C-reactive protein: targeting the complement inhibitory
4985–4996. quiz 5105. activity of factor H by an interaction with short
58 Kazatchkine MD, Fearon DT and Austen KF (1979) consensus repeat domains 7 and 8-11. J Immunol 163,
Human alternative complement pathway: membrane- 3957–3962.
associated sialic acid regulates the competition between 70 Laine M, Jarva H, Seitsonen S, Haapasalo K,
B and beta1 H for cell-bound C3b. J Immunol 122, Lehtinen MJ, Lindeman N, Anderson DH, Johnson
75–81. PT, Jarvela I, Jokiranta TS et al. (2007) Y402H
59 Pangburn MK and Muller-Eberhard HJ (1978) polymorphism of complement factor H affects binding
Complement C3 convertase: cell surface restriction of affinity to C-reactive protein. J Immunol 178, 3831–
beta1H control and generation of restriction on 3836.
neuraminidase-treated cells. Proc Natl Acad Sci USA 71 Deban L, Jarva H, Lehtinen MJ, Bottazzi B, Bastone
75, 2416–2420. A, Doni A, Jokiranta TS, Mantovani A and Meri S
60 Meri S and Pangburn MK (1994) Regulation of (2008) Binding of the long pentraxin PTX3 to
alternative pathway complement activation by factor H: interacting domains and function in the
glycosaminoglycans: specificity of the polyanion regulation of complement activation. J Immunol 181,
binding site on factor H. Biochem Biophys Res 8433–8440.
Commun 198, 52–59. 72 Okemefuna AI, Nan R, Miller A, Gor J and Perkins
61 Blackmore TK, Sadlon TA, Ward HM, Lublin DM SJ (2010) Complement factor H binds at two
and Gordon DL (1996) Identification of a heparin independent sites to C-reactive protein in acute phase
binding domain in the seventh short consensus repeat concentrations. J Biol Chem 285, 1053–1065.
of complement factor H. J Immunol 157, 5422–5427. 73 Kuhn S, Skerka C and Zipfel PF (1995) Mapping of
62 Blackmore TK, Hellwage J, Sadlon TA, Higgs N, the complement regulatory domains in the human
Zipfel PF, Ward HM and Gordon DL (1998) factor H-like protein 1 and in factor H1. J Immunol
Identification of the second heparin-binding domain in 155, 5663–5670.
human complement factor H. J Immunol 160, 3342– 74 Junnikkala S, Jokiranta TS, Friese MA, Jarva H,
3348. Zipfel PF and Meri S (2000) Exceptional resistance of
63 Pangburn MK, Atkinson MA and Meri S (1991) human H2 glioblastoma cells to complement-mediated
Localization of the heparin-binding site on killing by expression and utilization of factor H and
complement factor H. J Biol Chem 266, 16847–16853. factor H-like protein 1. J Immunol 164, 6075–6081.
64 Hellwage J, Jokiranta TS, Friese MA, Wolk TU, 75 Junnikkala S, Hakulinen J, Jarva H, Manuelian T,
Kampen E, Zipfel PF and Meri S (2002) Complement Bjorge L, Butzow R, Zipfel PF and Meri S (2002)
C3b/C3d and cell surface polyanions are recognized by Secretion of soluble complement inhibitors factor H
overlapping binding sites on the most carboxyl- and factor H-like protein (FHL-1) by ovarian tumour
terminal domain of complement factor H. J Immunol cells. Br J Cancer 87, 1119–1127.
169, 6935–6944. 76 Cheng ZZ, Corey MJ, Parepalo M, Majno S,
65 Jokiranta TS, Cheng ZZ, Seeberger H, Jozsi M, Hellwage J, Zipfel PF, Kinders RJ, Raitanen M, Meri
Heinen S, Noris M, Remuzzi G, Ormsby R, Gordon S and Jokiranta TS (2005) Complement factor H as a
DL, Meri S et al. (2005) Binding of complement factor marker for detection of bladder cancer. Clin Chem 51,
H to endothelial cells is mediated by the carboxy- 856–863.
terminal glycosaminoglycan binding site. Am J Pathol 77 Riihila PM, Nissinen LM, Ala-aho R, Kallajoki M,
167, 1173–1181. Grenman R, Meri S, Peltonen S, Peltonen J and
66 Jokiranta TS, Jaakola VP, Lehtinen MJ, Parepalo M, Kahari VM (2014) Complement factor H: a biomarker
Meri S and Goldman A (2006) Structure of for progression of cutaneous squamous cell carcinoma.
complement factor H carboxyl-terminus reveals J Invest Dermatol 134, 498–506.
molecular basis of atypical haemolytic uremic 78 Janssen BJ, Huizinga EG, Raaijmakers HC, Roos A,
syndrome. EMBO J 25, 1784–1794. Daha MR, Nilsson-Ekdahl K, Nilsson B and Gros P
67 Ram S, Sharma AK, Simpson SD, Gulati S, (2005) Structures of complement component C3
McQuillen DP, Pangburn MK and Rice PA (1998) A provide insights into the function and evolution of
novel sialic acid binding site on factor H mediates immunity. Nature 437, 505–511.

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2431
Self-nonself discrimination by the complement system S. Meri

79 Janssen BJ, Christodoulidou A, McCarthy A, Lambris 91 Levy M, Halbwachs-Mecarelli L, Gubler MC, Kohout
JD and Gros P (2006) Structure of C3b reveals G, Bensenouci A, Niaudet P, Hauptmann G and
conformational changes that underlie complement Lesavre P (1986) H deficiency in two brothers with
activity. Nature 444, 213–216. atypical dense intramembranous deposit disease.
80 Kajander T, Lehtinen MJ, Hyvarinen S, Bhattacharjee Kidney Int 30, 949–956.
A, Leung E, Isenman DE, Meri S, Goldman A and 92 Meri S, Koistinen V, Miettinen A, Tornroth T and
Jokiranta TS (2011) Dual interaction of factor H with Seppala IJ (1992) Activation of the alternative
C3d and glycosaminoglycans in host-nonhost pathway of complement by monoclonal lambda light
discrimination by complement. Proc Natl Acad Sci chains in membranoproliferative glomerulonephritis. J
USA 108, 2897–2902. Exp Med 175, 939–950.
81 Morgan HP, Schmidt CQ, Guariento M, Blaum BS, 93 Appel GB, Cook HT, Hageman G, Jennette JC,
Gillespie D, Herbert AP, Kavanagh D, Mertens HD, Kashgarian M, Kirschfink M, Lambris JD, Lanning L,
Svergun DI, Johansson CM et al. (2011) Structural Lutz HU, Meri S et al. (2005) Membranoproliferative
basis for engagement by complement factor H of glomerulonephritis type II (dense deposit disease): an
C3b on a self surface. Nat Struct Mol Biol 18, 463– update. J Am Soc Nephrol 16, 1392–1403.
470. 94 Edwards AO, Ritter R 3rd, Abel KJ, Manning A,
82 Thompson RA and Winterborn MH (1981) Panhuysen C and Farrer LA (2005) Complement
Hypocomplementaemia due to a genetic deficiency of factor H polymorphism and age-related macular
beta 1H globulin. Clin Exp Immunol 46, 110–119. degeneration. Science 308, 421–424.
83 Richards A, Buddles MR, Donne RL, Kaplan BS, 95 Hageman GS, Anderson DH, Johnson LV, Hancox
Kirk E, Venning MC, Tielemans CL, Goodship JA LS, Taiber AJ, Hardisty LI, Hageman JL, Stockman
and Goodship TH (2001) Factor H mutations in HA, Borchardt JD, Gehrs KM et al. (2005) A
hemolytic uremic syndrome cluster in exons 18-20, a common haplotype in the complement regulatory gene
domain important for host cell recognition. Am J Hum factor H (HF1/CFH) predisposes individuals to age-
Genet 68, 485–490. related macular degeneration. Proc Natl Acad Sci USA
84 Manuelian T, Hellwage J, Meri S, Caprioli J, Noris 102, 7227–7232.
M, Heinen S, Jozsi M, Neumann HP, Remuzzi G and 96 Haines JL, Hauser MA, Schmidt S, Scott WK, Olson
Zipfel PF (2003) Mutations in factor H reduce binding LM, Gallins P, Spencer KL, Kwan SY, Noureddine
affinity to C3b and heparin and surface attachment to M, Gilbert JR et al. (2005) Complement factor H
endothelial cells in hemolytic uremic syndrome. J Clin variant increases the risk of age-related macular
Invest 111, 1181–1190. degeneration. Science 308, 419–421.
85 Le Quintrec M, Roumenina L, Noris M and 97 Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS,
Fremeaux-Bacchi V (2010) Atypical hemolytic Haynes C, Henning AK, SanGiovanni JP, Mane SM,
uremic syndrome associated with mutations in Mayne ST et al. (2005) Complement factor H
complement regulator genes. Semin Thromb Hemost polymorphism in age-related macular degeneration.
36, 641–652. Science 308, 385–389.
86 Noris M, Mescia F and Remuzzi G (2012) STEC- 98 Jansen JH, Hogasen K and Grondahl AM (1995)
HUS, atypical HUS and TTP are all diseases of Porcine membranoproliferative glomerulonephritis type
complement activation. Nat Rev Nephrol 8, 622–633. II: an autosomal recessive deficiency of factor H. Vet
87 Meri S (2013) Complement activation in diseases Rec 137, 240–244.
presenting with thrombotic microangiopathy. Eur J 99 Daha MR, Fearon DT and Austen KF (1976) C3
Intern Med 24, 496–502. nephritic factor (C3NeF): stabilization of fluid phase
88 Rodriguez de Cordoba S, Hidalgo MS, Pinto S and and cell-bound alternative pathway convertase. J
Tortajada A (2014) Genetics of atypical hemolytic Immunol 116, 1–7.
uremic syndrome (aHUS). Semin Thromb Hemost 40, 100 Sjoberg AP, Trouw LA, Clark SJ, Sjolander J,
422–430. Heinegard D, Sim RB, Day AJ and Blom AM (2007)
89 Dragon-Durey MA, Blanc C, Garnier A, Hofer J, The factor H variant associated with age-related
Sethi SK and Zimmerhackl LB (2010) Anti-factor H macular degeneration (His-384) and the non-disease-
autoantibody-associated hemolytic uremic syndrome: associated form bind differentially to C-reactive
review of literature of the autoimmune form of HUS. protein, fibromodulin, DNA, and necrotic cells. J Biol
Semin Thromb Hemost 36, 633–640. Chem 282, 10894–10900.
90 Orth-Holler D and Wurzner R (2014) Role of 101 Clark SJ, Perveen R, Hakobyan S, Morgan BP, Sim
complement in enterohemorrhagic Escherichia coli- RB, Bishop PN and Day AJ (2010) Impaired binding
induced hemolytic uremic syndrome. Semin Thromb of the age-related macular degeneration-associated
Hemost 40, 503–507. complement factor H 402H allotype to Bruch’s

2432 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies
S. Meri Self-nonself discrimination by the complement system

membrane in human retina. J Biol Chem 285, 30192– hemolytic uremic syndrome is structurally different
30202. from its homologous site in factor H-related protein 1,
102 Lehtinen MJ, Rops AL, Isenman DE, van der Vlag J supporting a novel model for induction of
and Jokiranta TS (2009) Mutations of factor H impair autoimmunity in this disease. J Biol Chem 290, 9500–
regulation of surface-bound C3b by three mechanisms 9510.
in atypical hemolytic uremic syndrome. J Biol Chem 113 Gale DP, de Jorge EG, Cook HT, Martinez-Barricarte
284, 15650–15658. R, Hadjisavvas A, McLean AG, Pusey CD, Pierides
103 Hyvarinen S, Meri S and Jokiranta TS (2016) A, Kyriacou K, Athanasiou Y et al. (2010)
Disturbed sialic acid recognition on endothelial cells Identification of a mutation in complement factor H-
and platelets in complement attack causes atypical related protein 5 in patients of Cypriot origin with
hemolytic uremic syndrome. Blood 127, 2701–2710. glomerulonephritis. Lancet 376, 794–801.
104 Blaum BS, Hannan JP, Herbert AP, Kavanagh D, 114 Tortajada A, Yebenes H, Abarrategui-Garrido C,
Uhrin D and Stehle T (2015) Structural basis for sialic Anter J, Garcia-Fernandez JM, Martinez-Barricarte R,
acid-mediated self-recognition by complement factor Alba-Dominguez M, Malik TH, Bedoya R, Cabrera
H. Nat Chem Biol 11, 77–82. Perez R et al. (2013) C3 glomerulopathy-associated
105 Friese MA, Hellwage J, Jokiranta TS, Meri S, Peter CFHR1 mutation alters FHR oligomerization and
HH, Eibel H and Zipfel PF (1999) FHL-1/ complement regulation. J Clin Invest 123, 2434–2446.
reconectin and factor H: two human complement 115 Meri S, Morgan BP, Davies A, Daniels RH, Olavesen
regulators which are encoded by the same gene are MG, Waldmann H and Lachmann PJ (1990) Human
differently expressed and regulated. Mol Immunol 36, protectin (CD59), an 18,000–20,000 MW complement
809–818. lysis restricting factor, inhibits C5b–8 catalysed
106 Hourcade D, Holers VM and Atkinson JP (1989) The insertion of C9 into lipid bilayers. Immunology 71, 1–
regulators of complement activation (RCA) gene 9.
cluster. Adv Immunol 45, 381–416. 116 Rollins SA and Sims PJ (1990) The complement-
107 Narkio-Makela M, Hellwage J, Tahkokallio O and inhibitory activity of CD59 resides in its capacity to
Meri S (2001) Complement-regulator factor H and block incorporation of C9 into membrane C5b-9. J
related proteins in otitis media with effusion. Clin Immunol 144, 3478–3483.
Immunol 100, 118–126. 117 Cooper DK, Ezzelarab MB, Hara H, Iwase H, Lee W,
108 Zipfel PF, Skerka C, Hellwage J, Jokiranta ST, Meri Wijkstrom M and Bottino R (2016) The pathobiology
S, Brade V, Kraiczy P, Noris M and Remuzzi G of pig-to-primate xenotransplantation: a historical
(2002) Factor H family proteins: on complement, review. Xenotransplantation 23, 83–105.
microbes and human diseases. Biochem Soc Trans 30, 118 Alitalo A, Meri T, Lankinen H, Seppala I, Lahdenne
971–978. P, Hefty PS, Akins D and Meri S (2002) Complement
109 Goicoechea de Jorge E, Caesar JJ, Malik TH, Patel inhibitor factor H binding to Lyme disease spirochetes
M, Colledge M, Johnson S, Hakobyan S, Morgan BP, is mediated by inducible expression of multiple
Harris CL, Pickering MC et al. (2013) Dimerization of plasmid-encoded outer surface protein E paralogs.
complement factor H-related proteins modulates J Immunol 169, 3847–3853.
complement activation in vivo. Proc Natl Acad Sci 119 Jarva H, Janulczyk R, Hellwage J, Zipfel PF, Bjorck
USA 110, 4685–4690. L and Meri S (2002) Streptococcus pneumoniae evades
110 Caesar JJ, Lavender H, Ward PN, Exley RM, Eaton complement attack and opsonophagocytosis by
J, Chittock E, Malik TH, Goiecoechea De Jorge E, expressing the pspC locus-encoded Hic protein that
Pickering MC, Tang CM et al. (2014) Competition binds to short consensus repeats 8-11 of factor H. J
between antagonistic complement factors for a single Immunol 168, 1886–1894.
protein on N. meningitidis rules disease susceptibility. 120 Alitalo A, Meri T, Ramo L, Jokiranta TS, Heikkila T,
eLife 3, doi: 10.7554/eLife.04008. Seppala IJ, Oksi J, Viljanen M and Meri S (2001)
111 Zipfel PF, Edey M, Heinen S, Jozsi M, Richter H, Complement evasion by Borrelia burgdorferi: serum-
Misselwitz J, Hoppe B, Routledge D, Strain L, resistant strains promote C3b inactivation. Infect
Hughes AE et al. (2007) Deletion of complement Immun 69, 3685–3691.
factor H-related genes CFHR1 and CFHR3 is 121 Hellwage J, Meri T, Heikkila T, Alitalo A, Panelius J,
associated with atypical hemolytic uremic syndrome. Lahdenne P, Seppala IJ and Meri S (2001) The
PLoS Genet 3, e41. complement regulator factor H binds to the surface
112 Bhattacharjee A, Reuter S, Trojnar E, Kolodziejczyk protein OspE of Borrelia burgdorferi. J Biol Chem 276,
R, Seeberger H, Hyvarinen S, Uzonyi B, Szilagyi A, 8427–8435.
Prohaszka Z, Goldman A et al. (2015) The major 122 Kraiczy P, Skerka C, Kirschfink M, Brade V and
autoantibody epitope on factor H in atypical Zipfel PF (2001) Immune evasion of Borrelia

FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies 2433
Self-nonself discrimination by the complement system S. Meri

burgdorferi by acquisition of human complement meningococcal vaccine candidate GNA1870 binds the
regulators FHL-1/reconectin and factor H. Eur J complement regulatory protein factor H and enhances
Immunol 31, 1674–1684. serum resistance. J Immunol 177, 501–510.
123 Stevenson B, El-Hage N, Hines MA, Miller JC and 129 Scarselli M, Arico B, Brunelli B, Savino S, Di
Babb K (2002) Differential binding of host Marcello F, Palumbo E, Veggi D, Ciucchi L, Cartocci
complement inhibitor factor H by Borrelia burgdorferi E, Bottomley MJ et al. (2011) Rational design of a
Erp surface proteins: a possible mechanism underlying meningococcal antigen inducing broad protective
the expansive host range of Lyme disease spirochetes. immunity. Sci Transl Med 3, 91ra62.
Infect Immun 70, 491–497. 130 Green LR, Eiden J, Hao L, Jones T, Perez J, McNeil
124 McDowell JV, Wolfgang J, Tran E, Metts MS, LK, Jansen KU and Anderson AS (2016) Approach to
Hamilton D and Marconi RT (2003) Comprehensive the discovery, development, and evaluation of a novel
analysis of the factor h binding capabilities of Borrelia Neisseria meningitidis serogroup B vaccine. Methods
species associated with lyme disease: delineation of Mol Biol 1403, 445–469.
two distinct classes of factor h binding proteins. Infect 131 Finne J, Leinonen M and Makela PH (1983)
Immun 71, 3597–3602. Antigenic similarities between brain components and
125 Wallich R, Pattathu J, Kitiratschky V, Brenner C, bacteria causing meningitis. Implications for vaccine
Zipfel PF, Brade V, Simon MM and Kraiczy P (2005) development and pathogenesis. Lancet 2, 355–357.
Identification and functional characterization of 132 Jarva H, Ram S, Vogel U, Blom AM and Meri S
complement regulator-acquiring surface protein 1 of (2005) Binding of the complement inhibitor C4 bp to
the Lyme disease spirochetes Borrelia afzelii and serogroup B Neisseria meningitidis. J Immunol 174,
Borrelia garinii. Infect Immun 73, 2351–2359. 6299–6307.
126 Brooks CS, Vuppala SR, Jett AM, Alitalo A, Meri S 133 Giuliani MM, Adu-Bobie J, Comanducci M, Arico B,
and Akins DR (2005) Complement regulator-acquiring Savino S, Santini L, Brunelli B, Bambini S, Biolchi A,
surface protein 1 imparts resistance to human serum Capecchi B et al. (2006) A universal vaccine for
in Borrelia burgdorferi. J Immunol 175, 3299–3308. serogroup B meningococcus. Proc Natl Acad Sci USA
127 Jarva H, Hellwage J, Jokiranta TS, Lehtinen MJ, 103, 10834–10839.
Zipfel PF and Meri S (2004) The group B 134 Meri S, Jordens M and Jarva H (2008) Microbial
streptococcal beta and pneumococcal Hic proteins are complement inhibitors as vaccines. Vaccine 26 (Suppl
structurally related immune evasion molecules that 8), I113–I117.
bind the complement inhibitor factor H in an 135 Meri T, Amdahl H, Lehtinen MJ, Hyvarinen S,
analogous fashion. J Immunol 172, 3111–3118. McDowell JV, Bhattacharjee A, Meri S, Marconi R,
128 Madico G, Welsch JA, Lewis LA, McNaughton A, Goldman A and Jokiranta TS (2013) Microbes bind
Perlman DH, Costello CE, Ngampasutadol J, Vogel complement inhibitor factor H via a common site.
U, Granoff DM and Ram S (2006) The PLoS Pathog 9, e1003308.

2434 FEBS Letters 590 (2016) 2418–2434 ª 2016 Federation of European Biochemical Societies

You might also like