You are on page 1of 13

DOI: 10.1002/JLB.

3MIR0220-270R

SLB MEMBER INSIGHT REVIEW

Complement: Bridging the innate and adaptive immune


systems in sterile inflammation

Martin W. Lo Trent M. Woodruff

School of Biomedical Sciences, Faculty of


Medicine, The University of Queensland, Abstract
St Lucia, Brisbane, Queensland, Australia The complement system is a collection of soluble and membrane-bound proteins that together act
Correspondence as a powerful amplifier of the innate and adaptive immune systems. Although its role in infection is
Prof Trent Woodruff, Laboratory for
well established, complement is becoming increasingly recognized as a key contributor to sterile
Neuroinflammation, School of Biomedical
Sciences, The University of Queensland, St Lucia inflammation, a chronic inflammatory process often associated with noncommunicable diseases.
4072, Australia. In this context, damaged tissues release danger signals and trigger complement, which acts on a
Email: t.woodruff@uq.edu.au range of leukocytes to augment and bridge the innate and adaptive immune systems. Given the
detrimental effect of chronic inflammation, the complement system is therefore well placed as an
anti-inflammatory drug target. In this review, we provide a general outline of the sterile activators,
effectors, and targets of the complement system and a series of examples (i.e., hypertension, can-
cer, allograft transplant rejection, and neuroinflammation) that highlight complement’s ability to
bridge the 2 arms of the immune system.

KEYWORDS
C1q, alloimmunity, hypertension, cancer, anaphylatoxins, neuroinflammation

1 INTRODUCTION 1.2 Danger signals


Sterile inflammation can be defined as inflammation in response to
1.1 Sterile inflammation
necrotic tissue, which releases danger signals that recruit and acti-
Noncommunicable diseases (NCDs) account for 70% of all deaths vate the immune system.4 These danger signals or damage-associated
worldwide and are a group of chronic conditions that include cardio- molecular patterns (DAMPs) are ubiquitously expressed molecules
vascular disease, cancer, and neurologic disorders.1 Many are asso- such as ATP, high mobility group box 1 (HMGB1), and hyaluronan,
ciated with powerful sterile inflammatory responses that strive to which under homeostatic conditions are hidden intracellularly or in
remove, contain, and repair dying tissue and are driven by a num- hydrophobic regions of the extracellular matrix.3 However, tissue dam-
ber of key biologic processes including necrosis, danger signaling, and age exposes these DAMPs, which are detected by a number of widely
complement activation.2,3 Paradoxically, in the event of excess or pro- expressed receptor classes including: Toll-like, NOD-like, RIG-I-like,
longed inflammation, this well-intentioned response can cause collat- C-type lectin and absence in melanoma 2-like receptors.3 Through
eral damage and initiate a positive feedback loop that exacerbates the these receptors, DAMPs stimulate monocytes, macrophages, den-
underlying pathology.3 Thus, sterile inflammation remains an attrac- dritic cells (DCs), neutrophils, mast cells, NK cells, and eosinophils
tive target for therapeutic intervention in NCDs. to release proinflammatory mediators, which in turn leads to the
recruitment of inflammatory cells and the activation of the adap-
tive immune response5 Thus, DAMPs are the universal initiators of
sterile inflammation.
Abbreviations: BBB, blood–brain barrier; DAMPs, damage associated molecular patterns;
DCs, dendritic cells; EAE, experimental autoimmune encephalomyelitis; HMGB1, high
mobility group box 1; IR, ischemia-reperfusion; KO, knockout; MAC, membrane attack
complex; MASPs, MBL-associated serine proteases; MBL, mannose-binding lectin; MDSCs,
1.3 Complement system: Activation pathways
myeloid-derived suppressor cells; MS, multiple sclerosis; NCDs, noncommunicable diseases;
ROS, reactive oxygen species; RRMS, relapsing-remitting multiple sclerosis; TILs, tumor In concert with DAMPs, the complement system is a powerful collec-
infiltrating lymphocytes; Treg, T regulatory cell; WT, wild-type. tion of over 40 serine proteases, receptors, and regulators that amplify

Received: 5 November 2019 Revised: 7 February 2020 Accepted: 19 February 2020


J Leukoc Biol. 2020;1–13. www.jleukbio.org ○
c 2020 Society for Leukocyte Biology 1
2 LO AND WOODRUFF

C2/C4 MBL/Ficolin/ Prekallikrein


C1q/C1r/C1s Collectin/MASP
FXII
Renin
Classical Mannose- Kallikrien
binding Extrinsic
lectin Thrombin/FXIa/FXa/
C3
Extrinsic FXIa/Plasmin
C2bC4b (C3b)Bd
FXII
Alternative
C3a Altered surface/
C3b Factor B/Factor D/
Properdin
Alternative

C5
C2bC4bC3b (C3b)2Bd

Thrombin/FIXa/FXa/
FXIa/Plasmin

Extrinsic

C5a C5b-9

F I G U R E 1 Complement activation pathways. Inactive molecules and cleavage products at tail and heads of arrows respectively; intersecting
lines denote proteolytic cleavage; pathways are given in bold; and DAMP detection mechanisms are shown in orange

danger signals and augment the inflammatory response. We provide upstream pathways, and in itself can be triggered by “altered” sur-
here a short summary of complement activation and regulation, how- faces such as damaged or foreign tissue.16 Unlike the other systems,
ever detailed information can be found elsewhere.6-8 These serine pro- the alternative pathway undergoes autoactivation in which C3 spon-
teases are inactive in their native state, but once triggered, undergo taneously hydrolyses to form C3(H2O) and binds to factor B.17 Fac-
proteolytic cleavage and take part in a number of chain reactions that tor D is then able to cleave factor B into Ba and Bb, of which the
are organized into 3 major pathways (Fig. 1), of which the classical and latter remains associated with the complex (“initiation” C3 conver-
mannose-binding lectin (MBL) pathways play a key role in danger signal tase). Subsequent cleavage of C3 into C3a and C3b allows the forma-
detection. The classical pathway is canonically triggered by IgM or IgG tion of C3bBb (“amplification” C3 convertase) and C3bBb and C2bC4b
immune complexes, which bind to C1q of the C1 complex (C1qC1r2 s2 ) from the upstream pathway can incorporate additional C3b to adopt
to promote cleavage of C1r2 s2 . This complex then goes on to cleave C5 convertase activity. Conversion of C5 to C5a and C5b then facili-
C2 and C4 to form the C3 convertase, C2bC4b,9,10 and may also be tates the formation of C5b9 or the membrane attack complex (MAC).18
triggered by a diverse set of molecules including the DAMP HMGB1, C3a, C3b, C5a, and MAC are potent inflammatory mediators and thus
C-reactive protein, pentraxin 3, and apoptotic cell membranes.11 the pathways downstream of C3 are tightly regulated by a range of
Homologous to the classical pathway, the MBL pathway is activated by inhibitors including factor I, carboxypeptidase N, and C4BP19 ; which
carbohydrate moieties found on apoptotic and necrotic cells.12 These interestingly can activate B cells and promote IgE isotype switching.20
sugars are detected by a number of proteins including MBL, collectin- Damaged tissues often lack these inhibitors and can thus trigger the
10/11, and ficolin H/L/M,13 which go on to activate the 3 MBL- alternative pathway, a process further promoted by properdin, which
associated serine proteases (MASPs). MASP-1 is a C2 convertase that stabilizes C3bBb and is secreted by activated neutrophils.17,21 Super-
can also cleave MASP-2, which is a C2 and C4 convertase, and MASP-3 imposed over these 3 canonically defined cascades, the extrinsic path-
can cleave factor D to activate the downstream alternative pathway.13 way (Fig. 1) is a more recently recognized collection of proteases that
The MBL pathway can also be triggered by the DAMPs HMGB1,14 have isolated convertase activity. Many of these are derived from
ATP,15 and S100a914 ; C-reactive protein; and pentraxin 3.13 Thus, the the cardiovascular system and include factor IXa, Xa, XIa, throm-
classical and MBL pathways are powerful sensors for danger signals, bin, and plasmin, which are C3 and C5 convertases22 ; renin and
dying cells and acute phase reactants. kallikrein, which cleave C323,24 ; and Hageman factor which cleaves
Further downstream, the alternative pathway is an amplification the C1 complex.25 Thus, a host of sterile stimuli can trigger the
loop that can contribute up to 80% of complement activation from complement system.
LO AND WOODRUFF 3

A C1q–antigen signalling B C1q–antigen–apoptotic body signalling


C1q–antigen complex C1q–antigen–apoptotic body
complex
cC1qR–
SIGNR1
IL-12 IL-12R IL-10 IL-10R1/2
? ?
STAT3/4 ?
NF-KB MAPK Th1 Th1/17
cC1qR–LRP
effector (CD91) effector
PI3K
status status
IL-12 & TNFa IL-10
TRAFs
TNFa TNFRs

Dendritic cell Naive T cell Dendritic cell Naive T cell

C C1q–antigen–apoptotic body signalling D B cell CR2 signalling


B cell receptor
IL-27 IL-27R
? PI3K
STAT1 CR3 (CD11b+CD18)-
Vav Memory B/
cC1qR–LRP iC3b-antigen
Plasma cell
(CD91) Th1/17
status
effector
status
IL-27
B cell
coreceptor
CR2 (CD21)-CD19

Macrophage Naive T cell Follicular Naive B cell


dendritic cell

E Autocrine and paracrine C3a/C5a signalling


Proteolysis C3 C5 Proteolysis
TLR FB FD C3a
C5a
C3b
C3a
C3 C5
C5a C3 C3 FB FD
C3b FB FD

T effector
NF-KB
status
Cytokine Lysosome
expression
Nucleus
MHC-TCR and
B7-CD28
complex

Dendritic cell Naive T cell

F I G U R E 2 Mechanisms by which complement bridges the innate and adaptive immune system. (A) C1q-opsonized antigen activates an
unknown pathway in DCs, potentially via cC1qR–SIGNR1, that triggers NF-𝜅B nuclear translocation and up-regulates proinflammatory cytokine
IL-12 and TNF𝛼 production. These cytokines then stimulate their respective receptors on T cells to promote Th1 polarization. (B) C1q-opsonized
antigen with apoptotic cells are phagocytosed by DCs, potentially via the cC1qR–LRP receptor complex. This phagocytosis then promotes immuno-
suppressive cytokine IL-10 production through currently unknown mechanisms and IL-10 then down-regulates Th1/17 induction in naive T cells.
(C) Similarly, phagocytosis of C1q-opsonized antigen with apoptotic cells in macrophages promotes the production of IL-27, which inhibits Th1/17
polarization in naïve T cells. (D) CR2 signaling at the APC-B cell immune synapse lowers the threshold for B cell activation and promotes germinal
center formation. (E) APC-T cell interaction triggers auto- and paracrine C3a/C3b/C5a signaling that drives effector T cell differentiation and DC
cytokine production. Stimulation is represented by normal arrowheads and inhibition represented by blunt arrowheads

1.4 Complement system: Effector molecules phagocytes to tailor the inflammatory response to its antigen.19 When
bound to “regular” antigen, it acts as a proinflammatory mediator and
The major effector fragments of the complement system including
encourages DCs to present antigen, migrate to lymphoid tissue and
C1q, C3a, C3b, C5a, and C5b9/MAC are potent molecules that coor-
secrete Th1 polarizing cytokines such as IL-12 and TNF𝛼 26-29 (Fig. 2A);
dinate and link the innate and adaptive immune systems (Fig. 2). C1q
but when bound to apoptotic cells, C1q stimulates DCs to produce IL-
is an opsonin that is thought to signal through C1qR and SIGNR1 on
4 LO AND WOODRUFF

10, which suppresses Th1 and Th17 proliferation as a means of lim- activate the immune system and affects both the vascular system and
iting inflammation during apoptosis (Fig. 2B), a phenomenon that is target organs.52 Cytokines produced during inflammation promote
mirrored in macrophages (Fig. 2C).30-33 C3b and its proteolytic prod- the formation of a neointima (an intimal thickening), which increases
ucts iC3b, C3c, C3dg, and C3d are also opsonins of antigens and vascular resistance and blood pressure, and may also increase renal
apoptotic cells, but bind to CR1/CD35 (C3b and iC3b), CR2/CD21 tubular production of angiotensin and salt and water retention, which
(iC3b and C3dg), CR3/CD11b+CD18 (iC3b, C3dg, and C3d), and both increase blood volume and intravascular pressure.53 Additionally,
CR4/CD11c+CD18 (iC3b); which are differentially expressed amongst matrix metalloproteinases secreted by a variety of cells can degrade
leukocytes but are all considered to be involved in phagocytosis and the extracellular matrix of the vessel wall and allow immune cells to
antigen presentation.19,34 In particular, CR2 has a unique role in infiltrate affected organs and inflict damage by promoting apoptosis
humoral immunity (Fig. 2D). As part of the B cell coreceptor complex, and enhancing collagen synthesis and matrix deposition.53
it can lower the threshold for B cell activation, prolong antigen dis- Regulating these processes, Treg cells are a specialized subpop-
play in germinal centers, and facilitate firm contact between follicular ulation of T cells that suppress the immune response to maintain
DCs and B cells to initiate germinal center reactions and the forma- homeostasis and self-tolerance. In an angiotensin II (Ang II)-induced
tion of plasma and memory B cells.35,36 By contrast, CR3 acts in a sim- hypertensive mouse model, FOXP3+ Tregs that are known to express
ilar fashion to C1qR in that its ligation by opsonized apoptotic bodies the Ang II receptor type 1 exhibited increased C3aR and C5aR1
can reduce DC immunostimulatory function by reducing cytokine pro- expression,54,55 which would imply that Ang II is able to prime CD4+
duction (e.g., IL-12).37,38 Thus, complement factors from the upstream T cells for differentiation away from a FOXP3+ Treg phenotype.56 In
pathways act as dynamic opsonins that influence both the innate and support of this, C3aR–C5aR1 double knockout (DKO) mice that under-
adaptive immune systems. went Ang II hypertensive treatment had reduced CD4+ and CD8+
Further downstream, C3a and C5a are potent and versatile T cells and a mitigated decrease in FOXP3+ Tregs in their blood and
complement peptides or “anaphylatoxins” that signal through C3aR kidneys compared to wild-type (WT) control.54 DKO mice that under-
and C5aR1/2, respectively.39,40 In innate immunity, both have well went Ang II hypertensive treatment also had reduced systolic and
defined roles and multiple leukocyte targets in which 1 or both can diastolic blood pressure compared to WT mice and depletion of Tregs
promote or inhibit chemotaxis, granulocyte degranulation, phago- abolished the protective effects against Ang II-induced hypertension
cytosis, respiratory burst formation, inflammasome activation, and and target organ damage in DKO mice.54 Thus, at least in mouse
cytokine and eicosanoid production,41-44 though the function of models, C3aR and C5aR exert a hypertensive effect in Ang II-induced
C5aR2 is controversial and has been reviewed elsewhere.39 In addi- hypertension mediated by a down-regulation of the protective effects
tion, C3a and C5a have been increasingly recognized as regula- conferred by Tregs (Fig. 3A). In addition, hypertension in humans is
tors of the adaptive immune response. Both are produced at the associated with elevated plasma C3a and C5a concentrations and
APC-T cell interface to promote T cell activation and APC mat- reduced FOXP3+ Treg counts54,57 and thus, an analogous mechanism
uration (Fig. 2E) and production from intracellular stores known may occur in humans. However, although this antihypertensive effect
as the “complosome” with C3b signaling through CD46 are nec- is linked to anti-inflammatory cytokine production (e.g., IL-10 and TGF-
essary for T cell survival, activation, and differentiation, in which 𝛽) and reduced blood vessel infiltration by macrophages, its full nature
the anaphylatoxins and C3b cause polarization away from and is incompletely understood.58,59 This and other emerging studies sug-
towards the T regulatory (Treg) phenotype respectively.45 Lastly, gest complement is a key regulator of the innate and adaptive immune
C5b-9/MAC is a toroidal pore that depending on its form can system in hypertension.57,60–62
induce cell lysis and apoptosis, cytokine production,46 inflammasome
activation,47 and gene expression.48 Thus, the complement system is
a diverse group of molecules, each with a repertoire of innate and 3 CANCER
adaptive capabilities.
Cancer is expected to rank as the leading cause of death and the sin-
gle most important barrier to increasing life expectancy in the world
2 HYPERTENSION in the 21st century.63 Though there are over 100 distinct types of
cancer, all tumors have defects in regulatory circuits that govern nor-
Hypertension is the leading global burden of disease risk factor and can mal cell proliferation and homeostasis and to varying degrees are
be defined as systolic and/or diastolic blood pressure above 130 and characterized by the 8 hallmarks: (1) evasion of apoptosis, (2) self-
80 mmHg, respectively.49,50 sufficiency in growth signals, (3) insensitivity to anti-growth signals,
Mostly an idiopathic phenomenon, hypertension is driven by renal (4) sustained angiogenesis, (5) limitless replicative potential, (6) tis-
sodium dyshomeostasis and reduced arterial compliance.50 The for- sue invasion and metastasis, (7) reprogramming of energy metabolism,
mer is often caused by lifestyle factors and comorbidities, whereas the and (8) evading immune destruction.64,65 In particular, cancer is associ-
latter is due to endothelial dysfunction, blood vessel fibrosis, and vas- ated with a distinct form of chronic inflammation that both predisposes
cular smooth muscle cell hyperplasia.51 Sterile inflammation in hyper- to cancer development (15–20% of all cancer cases) and advances
tension is elicited by hypertensive stimuli that directly or indirectly established malignancies.66,67 Cancer-associated inflammation has a
LO AND WOODRUFF 5

C3a IL-10
C3aR
A Hypertension B Tumor
CD8+ TIL

ANG C3a IL-10R


TGF-B Tumour
Cytolytic 1/2
production
AT1R C5a Tumor function
of C5a IL-10
Treg cell
effector
FOXP3
status C5aR1 (CD88) C5a
IL-10
C5a
C5a
C5a
Macrophage C5a
TAM Treg
vascular wall infiltration
effector
status
MDSC
Endothelial-dependent MDSC MDSC CD4+ TIL
vascular smooth muscle relaxation chemotaxis differentiation IL-10R1/2
into TAM and
TGF-BR
IL-10 and TGF-B
C Allograft transplant
Tumour evasion of
immunosurveillance
Host APC
Host APC migration in
donor antigen
afferent lymphatics Host APC
uptake

Immune synapse with


autocrine and paracrine
C3a and C5a
T cell migration in signalling
efferent blood vessel

Cytokine
production
Effector T cell
CD4+/CD8+ differentiation
T cell and proliferation
alloimmunity
Transplant tissue Lymph node

D Multiple sclerosis
CR4
CR3 (CD11c+CD18)
(CD11b+CD18)
?
Neuron
T cell T cell, B cell,
activation microglia
T cell autoimmunity

T cell CD46
receptor

Treg
effector
Immunosupression Oligodendrocyte
status

Autoimmune attack
T cells

F I G U R E 3 Complement in sterile inflammatory diseases. (A) In hypertension, C5aR1 and C3aR signalling inhibits Treg polarization and thus
down-regulates their anti-inflammatory and vasodilatory effects, of which only known mechanisms are shown above. (B) In tumorigenesis, C5a
acts as a chemokine for myeloid-derived suppressor cells (MDSCs), which differentiate into tumor-associated macrophages (TAMs) that produce
immunosuppressive and Treg polarizing cytokines. Additionally, autocrine C3a signaling in CD8+ tumor-infiltrating lymphocytes (TILs) inhibits
autocrine IL-10 signaling, which normally promotes cytolytic activity. (C) In allograft transplant rejection, indirect allorecognition is mediated by
host APCs that travel to the lymphatic system to interact with naive T cells. These T cells then form immune synapses and with the assistance of
autocrine and paracrine C3a and C5a signaling, proliferate and differentiate into effector T cells that return to the allograft to promote inflam-
mation and chronic rejection. (D) In MS, CR3 and CR4 drive T cell activation and autoimmune attack and defective CD46 O-glycosylation and
cosignaling with the T cell receptor impairs Treg effector status and IL-10 production
6 LO AND WOODRUFF

wide range of stimuli including gene mutations that promote the pri- direct effects on tumor progression and is a promising target for
mary production of cytokines,68 infective agents (e.g., human papil- therapeutic intervention.82
loma virus in cervical cancer), and cell death secondary to chemo-
/radiotherapy, hypoxia, metabolic stress, or mutagenic insult.69 More-
over, unlike inflammation linked to infection, in which sterilizing immu- 4 ALLOGRAFT TRANSPLANT REJECTION
nity is able to eradicate a pathogen and promote tissue repair, cancer-
associated inflammation is usually unable to completely remove all Allograft organ transplants are transplantations to a recipient from a
malignant cells, which is in no small part due to their ability to evade the genetically nonidentical donor of the same species and has emerged
immune system. This results in the continued release of danger signals as a viable therapeutic modality for a variety of ailments.83 Organs
and positive feedback amplification that leads to the accumulation of including kidneys, livers, and hearts can be transplanted with impres-
growth-promoting cytokines.69 Thus, inflammation in cancer is a com- sive short-term survival rates, but allograft rejection of which there are
plex process in which malignant cells seek to evade and exploit the 3 classes (hyperacute, acute, and chronic) remains as a limiting factor to
immune system. long-term survival.84 Hyperacute rejection occurs in the first few min-
Complement has a diverse set of functions in the context of utes following transplantation and is triggered by recipient antidonor
cancer including directly promoting proliferation and metastasis, antibodies that via the complement and coagulation cascades cause
stimulating angiogenesis, and regulating the immunosuppressive vessel thrombosis and secondary infarction. In modern times, this type
microenvironment.70-72 In the latter, the complement system acts as of rejection is a rarity as a result of ABO compatibility checks and
a bridge between innate and adaptive immune systems and plays a key human leukocyte antigen crossmatching.85 By contrast, acute rejec-
role in recruiting myeloid-derived suppressor cells (MDSCs) and mod- tion occurs between 1 week and several months after transplantation
ulating T-cell-mediated antitumor immunity. and arises as a result of intense cellular and humoral attack, but with
Myeloid-derived suppressor cells are a diverse group of imma- current immunosuppressive regimens occurs in less than 15% of non-
ture, immunosuppressive, and pathologically active leukocytes that are sensitized transplant patients. Chronic rejection however occurs after
the result of persistent myelopoiesis during chronic inflammation.73 months to years and is mediated by insidious cellular and humoral pro-
To varying degrees, these cells secrete immunosuppressive cytokines cesses that cause obliterative vasculopathy, parenchymal fibrosis, and
and eicosanoids, produce reactive oxygen species (ROS), facilitate tertiary lymphoid organ formation.85,86 This type of rejection responds
Treg recruitment, and rapidly differentiate into tumor-associated poorly to current immunosuppressive regimens and is now the leading
macrophages that persist in the tumor microenvironment.74 Com- cause of graft rejection.87,88 Transplant organs are scarce and valuable
plement peptide C5a acts as a chemokine for MDSCs and is pro- and thus development of novel therapeutics against acute and chronic
duced by some cancers to facilitate their recruitment (Fig. 3B).71,75–77 rejection is needed to maximize their utility.89
Non-small cell lung cancer patients have elevated plasma C5a lev- Whether a transplant undergoes acute or chronic rejection is
els and lung cancer cell lines are able to produce C5a in serum free thought to depend in part on the nature of the recipient’s immune
conditions.76 Moreover, C5aR1 antagonism and/or genetic knockout response to donor antigens. It is hypothesized that host exposure to
(KO) in a lung cancer mouse model reduced tumor growth, MDSC intact MHC alloantigen displayed on donor APCs (direct allorecog-
recruitment, and tumor expression of the immunomodulators ARG1, nition) results in acute rejection because of substantial expansion of
CTLA-4, IL-6, IL-10, LAG3, and PDL1, which are associated with Treg T cells of multiple specificities.86 By contrast, host exposure to donor
induction.76,78 Similarly, in a preclinical mouse model for breast can- alloantigen processed and presented by host APCs (indirect allorecog-
cer, C5a suppressed CD8+ and CD4+ T cell responses in lung tis- nition) leads to activation of a limited repertoire of T cells with a
sue to create a metastatic niche.79 In this instance, pharmacologic restricted ability to recognize graft targets, but importantly act as
blockade and genetic ablation of C5aR1 was sufficient to reduce helper T cells for B cells that produce alloantibodies, and thus causes
lung metastases with a concomitant increase in CD4+ and CD8+ chronic rejection instead.90-93 Other factors are also likely to have
T cell recruitment and a decrease in MDSC infiltration and IL-10 a role in determining the nature of rejection and include ischemia-
and TGF-𝛽 cytokine production.79 This suppression of T cell immuno- reperfusion (IR) injury, which enhances effector T cell accumulation in
surveillance is mirrored in an autocrine complement mechanism, in allografts.86 Thus, the APC-T cell relationship is not only a vital link
which autocrine C3a suppresses the expression of IL-10, a potent between the innate and adaptive immune system but also a key deter-
activator of CD8+ tumor-infiltrating lymphocytes (TILs) (Fig. 3B). In minant of transplant alloimmunity.
this context, C3-deficient mice are resistant to tumor development In the context of an allograft transplant, the complement system
in a T cell- and IL-10-dependent manner and produce CD8+ TILs is activated by IR injury and acts as a bridge between the innate
that have enhanced effector function.80 By contrast, C3aR KO mice and adaptive immune systems. During the transplantation procedure,
with B16F0 melanoma had reduced CD4+ T lymphocyte infiltration interim cold ischemic storage causes hypoxia, ATP depletion and mito-
compared with their WT littermates due to a neutrophil-dependent chondrial damage within the graft and subsequent reperfusion para-
mechanism, which highlights the complex relationship between com- doxically causes the formation of ROS, which further damages the
plement and cancer immunity.81 Thus, the complement system is graft and promotes graft dysfunction in a process known as IR injury.
a regulator of both the innate and adaptive immune systems with Injured tissue then releases neoantigens and DAMPs that activate the
LO AND WOODRUFF 7

complement system and initiate a sterile inflammatory response.94-98 ing AMPA and NMDA receptors, the latter of which facilitates Ca2+
This initial insult is then also propagated by complement, which acts entry into the cytoplasm and drives cell death by activating proteases
as a potent modulator of cellular and humoral immunity. In the former, (e.g., calpains) and lipases and increasing the production of ROS and
autocrine and paracrine C3aR and C5aR1 signaling at the APC-T cell arachidonic acid.121,122 In severe cases, this can result in necrosis,
interface amplifies T cell effector responses and in numerous mouse which triggers a self-perpetuating cycle of glutamate release and exci-
models has been shown to accelerate rejection (Fig. 3C).99 For exam- totoxicity , as seen in the expanding umbra of an ischemic stroke.123
ple, anti-C5,100 C5aR1 antagonism,101 and C3aR1 KO grafts102 reduce From a drug development standpoint, these pathologic phenomena
T cell priming and improve allograft survival in cardiac and renal trans- necessitate a drug target that is ubiquitous, promptly expressed, and
plant mouse models, whereas genetic ablation of decay accelerating highly efficacious; characteristics that are found in neuroinflammation.
factor, which normally inhibits C3a and C5a production,103 augments Neuroinflammation is a broad term that encompasses a range of
T cell immunity and accelerates rejection in murine cardiac grafts.104 In context-driven inflammatory processes within the brain and spinal
addition, C3aR and C5aR1 signaling inhibits Treg polarization,56,105,106 cord that are distinct from “classic” sterile inflammation. These
and thus C5aR2, which in T cells does not interact with 𝛽-arrestin or processes can be grouped into 2 distinct categories, denoted here
inhibit ERK1/2 signaling, can regulate T cell effector function by act- as “primary” and “secondary” inflammation.124 “Primary” neuroin-
ing as a decoy receptor.107 Indeed, in a Treg-dependent cardiac trans- flammation refers to that in autoimmune diseases (e.g., multiple
plant mouse model, absence of recipient C5aR2 accelerated rejection, sclerosis [MS]) and is associated with significant peripheral leukocyte
whereas C5aR2 overexpression prolonged graft survival.107 Interest- invasion and a strong adaptive immune response characterized by
ingly, like in cancer, complement also has an immunosuppressive effect heightened MHC antigen presentation and T cell co-stimulation.124
mediated by MDSCs, as seen in myeloid cell-specific C5aR1 condi- In contrast, “secondary” neuroinflammation refers to that in response
tional KO mice with cardiac allografts, which have increased donor to primary neuropathology (e.g., neurodegeneration, cancer, and
T cell immunity, decreased Treg expansion and accelerated graft rejec- epilepsy) and is a glial-driven process with comparatively low
tion despite costimulatory blockade.108 Additionally, complement has chemokine production and T cell stimulatory capacity.124 Despite
an important role in humoral transplant alloimmunity, in which CR2 is these differences, neuroinflammation can be defined by a number of
expressed on B and follicular DCs and recognizes the opsonins iC3b, standard features including macrophage/microglial activation and a
C3dg, and C3d.19 When activated, this receptor lowers the thresh- suite of cytokines (IL-1𝛽, IL-6, and TNF𝛼), chemokines (CCL2, CCL5,
old for B cell activation and promotes antigen retention by follicular and CXCL1), secondary messengers (NO and prostaglandins), and
DCs,109,110 a process that is reflected in C3-deficient mice that fail to ROS.125 These immune mediators may be neuroprotective (e.g., in
produce high affinity IgG responses against major histocompatibility synaptic pruning, immune surveillance, neurotrophin production, and
antigens in skin grafts.111 Thus, the complement system is a power- euflammation) or neurotoxic (e.g., via cytotoxicity and excitotoxicity)
ful amplifier of sterile inflammation in allograft transplantation and is and the balance of their functions ultimately drives the overarching
currently being pursued in phase II/III clinical trials as a drug target for effect of the inflammatory response.125,126 In line with this, neuroin-
acute transplant rejection.82 flammation commonly has a biphasic nature characterized by an initial
neuroprotective reactive gliosis that may either resolve, or become
overwhelmed and transition into a self-perpetuating proinflammatory
5 NEUROINFLAMMATION reaction,127 the latter of which reflects the tendency of microglia to
become hyperactive in response to repeated immune stimulation.125
Neurological disorders are the leading cause of disability-adjusted Many of these processes have been correlated with disease progres-
life years and the second-leading-cause of death.112 Bar those con- sion and their genetic and pharmacologic modulation have been able
ditions with a relatively clear and isolated etiology and pathogen- to improve outcomes in animal models.43,128–139 Thus, neuroinflam-
esis (e.g., extradural hematoma), this diverse group of conditions is mation has become an attractive drug target for neurologic disease
typically challenging from an interventional and drug development and is currently being pursued in phase I/II/III clinical trials.140-142
standpoint, not least because of their profound clinicopathologic vari- The complement system is a key driver of neuroinflammation and
ability. Though typically defined by a small number of key features, is a promising therapeutic target given its universality, potency, and
neurologic diseases tend to each contain distinct subgroups or a wide timely reactivity. In neurologic conditions, the nature of complement
spectrum of patients with varying symptoms, progressions, etiolo- activation is heavily dependent on the permeability of the blood–brain
gies, and pathologic processes.113-117 Moreover, the CNS is particu- barrier (BBB). In response to minor insults, the BBB generally remains
larly sensitive to insult. As a hierarchical organ,118 the topology of its intact and complement may be synthesized locally by astrocytes (C1,
neuronal connections is critical to its overall function and recapitu- C2, C3, C4, C5, C6, C7, C8, C9, FB),143,144 microglia (C1, C3, C4),143
lating this during a repair process remains difficult.119 Lesions in the neurons (C5),145 and oligodendrocytes (C1, C2, C3, C4, C5, C6, C7, C8
CNS also tend to be propagated by excitotoxicity, an intrinsic neuronal C9)146 at relatively low (nanomolar) concentrations over a period of
property mediated by the neurotransmitter glutamate.120 Found at days, weeks and even months.147 By contrast, in response to severe
60–70% of all synapses in the CNS, this excitatory neurochemical is (e.g., neurotrauma) or chronic insults, the BBB may lose its integrity
released in excess upon neuronal breakdown and stimulates neighbor- and permit systemic and leukocyte-synthesized complement to enter
8 LO AND WOODRUFF

the CNS and reach relatively high (micromolar) concentrations in a rel- 6 CONCLUSION
atively short amount of time (24 h).147 In addition, the complement sys-
tem has a range of functions specific to the nervous system. In both The rates of NCDs continue to increase globally and with it a greater
development and disease, complement opsonins (i.e., C1q and C3b) need for disease modifying treatments. In these conditions, a variety of
facilitate microglia-mediated synaptic pruning and phagocytosis of cel- danger signals trigger a sterile inflammatory response that chronically
lular debris148-151 and C1q can affect microglial cytokine production exacerbates the underlying pathology, in which the complement sys-
in a similar manner to that in DCs whereby apoptotic blebs modulate tem acts as a potent amplifier with a wide range of innate and adaptive
the response, although whether or not microglia interact with T cells leukocyte targets. In hypertension, studies indicate an intimate rela-
in this manner is not known.152,153 Likewise, anaphylatoxins (i.e., C3a tionship between the cardiovascular and immune systems, in which
and C5a) have a number of canonical effects on glial cells including angiotensin II may act directly on T cells. Here, complement acts as
driving chemotaxis,154,155 promoting cytokine production,156,157 and an autocrine T cell activator that increases blood pressure, promotes
activating the inflammasome,158,159 but may also have a number of leukocyte tissue invasion and exacerbates end organ damage. By con-
direct effects on neurons including limiting excitotoxity,160 promot- trast, cancer cells exploit complement as a means of avoiding immuno-
ing neurogenesis,161 and driving neuronal stem cell proliferation in surveillance, in which C5a is a salient chemokine for MDSC recruit-
brain cancer.162 Such diverse capabilities are also exhibited by MAC, ment to malignancies. These dysregulated leukocytes secrete anti-
which has canonical cytolytic effects that are driven in autoimmune inflammatory and Treg polarizing cytokines that allow for unchecked
conditions by autoantibody activation of the classical pathway163-166 neoplastic proliferation. Likewise, this cellular immune response is
and immunomodulatory and protective effects on glia.167,168 These seen in transplant alloimmunity, in which complement activates T lym-
immune pathways occur in an environment with relatively low comple- phocytes at the APC–T cell interface and promotes the production
ment regulator expression and thus complement is a powerful driver of of alloreactive antibodies, and in MS, in which CR3/4 and dysregu-
neuroinflammation.169-172 lated CD46 signaling influence T cell activation. Thus, complement is
Unlike its effect on the innate immune response, complement’s a bridge between the 2 arms of the immune system and at this nexus is
effect on the adaptive immune response in neuroinflammation is rel- well placed for drug development against chronic inflammation.
atively under-investigated. Nevertheless, emerging studies give some
insight into its potential role. MS is an autoimmune neurologic dis- AUTHORSHIP
order characterized by the intermittent and progressive presentation
of a wide range of neurologic deficits.173 The condition is tradition- M.W.L. and T.M.W. coconceived the review. M.W.L. drafted the
ally viewed as a 2-stage disease with early inflammation responsible manuscript and created the figures, with T.M.W. contributing to edit-
for relapsing-remitting disease (RRMS) and delayed neurodegenera- ing and revisions.
tion causing nonrelapsing progression, that is, secondary and primary
progressive MS.173 In the both instances, patients develop inflamma- ACKNOWLEDGEMENTS
tory plaques that cause demyelination and eventually axonal damage.
T.M.W. acknowledges support from the National Health and Medical
However, in RRMS, the “classical active lesion” predominates and is
Research Council (NHMRC fellowship APP1105420).
associated with profound T cell (mainly MHC class I restricted CD8+
T cells), B cell, and plasma cell infiltrates, whereas in progressive dis-
ease, lesions tend to have an inactive core surrounded by a narrow DISCLOSURES
rim of activated microglia and macrophages.173,174 In relation to the
The authors declare no conflicts of interests.
former, CR3 and CR4 appear to play a critical role in T cell-mediated
disease. In the experimental autoimmune encephalomyelitis (EAE) MS
ORCID
mouse model, CR3 and CR4 KO mice exhibited reduced spinal cord
T cell infiltration and IFN𝛾 production by these cells (Fig. 3D).175,176 Martin W. Lo https://orcid.org/0000-0002-2736-5507
Additionally, CR3 and CR4 KO T cells have an unusual cytokine pro- Trent M. Woodruff https://orcid.org/0000-0003-1382-911X
file and limited EAE-inducing capacity when transferred to control
mice,175,176 though the signaling pathways that underlie these phe-
REFERENCES
nomena are incompletely understood.177 The impact of complement
on MS adaptive immunity can also be seen in Tregs in which isolated 1. Frumkin H, Haines A. Global environmental change and noncommu-
nicable disease risks. Annu Rev Public Health. 2019;40:261-282.
CD46 stimulation of these cells results in impaired IL-10 production
2. Camps J. Oxidative Stress and Inflammation in Non-communicable Dis-
and reduced immunosuppression.178 Indeed, MS T cells have defec- eases - Molecular Mechanisms and Perspectives in Therapeutics. Imprint:
tive T cell receptors that are unable to glycosylate CD46 for recruit- Springer, Cham: Springer International Publishing; 2014.
ment to the immune synapse, which is thought to drive Treg induction 3. Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to
damage. Nat Rev Immunol. 2010;10:826-837.
(Fig. 3D).179 These findings suggest that complement plays a key role in
4. Martin SJ. Cell death and inflammation: the case for IL-1 family
bridging the innate and adaptive immune system in neuroinflammation cytokines as the canonical DAMPs of the immune system. FEBS J.
and flag this area for future research. 2016;283:2599-2615.
LO AND WOODRUFF 9

5. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in 27. Yoo JK, Cho JH, Lee SW, Sung YC. IL-12 provides proliferation and
sterile inflammation and inflammatory diseases. Nat Rev Immunol. survival signals to murine CD4 T cells through phosphatidylinositol
2019;20:95-112. 3-kinase/Akt signaling pathway. J Immunol. 2002;169:3637-3643.
6. Ricklin D, Reis ES, Lambris JD. Complement in disease: a defence sys- 28. Jacobson NG, Szabo SJ, Weber-Nordt RM, et al. Interleukin 12 sig-
tem turning offensive. Nat Rev Immunol. 2016;12:383-401. naling in T helper type 1 (Th1) cells involves tyrosine phosphorylation
7. Noris M, Remuzzi G. Overview of complement activation and regula- of signal transducer and activator of transcription (Stat)3 and Stat4.
tion. Semin Nephrol. 2013;33:479-492. J Exp Med. 1995;181:1755-1762.
8. Bajic G, Degn SE, Thiel S, Andersen GR. Complement activation, reg- 29. Ward-Kavanagh LK, Lin WW, Sedy JR, Ware CF. The TNF receptor
ulation, and molecular basis for complement-related diseases. EMBO superfamily in co-stimulating and co-inhibitory responses. Immunity.
J. 2015;34:2735-2757. 2016;44:1005-1019.
9. Mortensen SA, Sander B, Jensen RK, et al. Structure and activation 30. Clarke EV, Weist BM, Walsh CM, Tenner AJ. Complement protein
of C1, the complex initiating the classical pathway of the complement C1q bound to apoptotic cells suppresses human macrophage and
cascade. Proc Natl Acad Sci USA. 2017;114:986-991. dendritic cell-mediated Th17 and Th1 T cell subset proliferation.
10. Bohlson SS, Garred P, Kemper C, Tenner AJ. Complement J Leukoc Biol. 2015;97:147-160.
nomenclature-deconvoluted. Front Immunol. 2019;10:1308. 31. Benoit ME, Clarke EV, Morgado P, Fraser DA, Tenner AJ. Comple-
11. Merle NS, Church SE, Fremeaux-Bacchi V, Roumenina LT. Comple- ment protein C1q directs macrophage polarization and limits inflam-
ment system part i - molecular mechanisms of activation and regu- masome activity during the uptake of apoptotic cells. J Immunol.
lation. Front Immunol. 2015;6:262. 2012;188:5682-5693.
12. Nauta AJ, Raaschou-Jensen N, Roos A, et al. Mannose-binding lectin 32. Peters A, Fowler KD, Chalmin F, Merkler D, Kuchroo VK, Pot C. IL-
engagement with late apoptotic and necrotic cells. Eur J Immunol. 27 induces Th17 differentiation in the absence of STAT1 signaling.
2003;33:2853-2863. J Immunol. 2015;195:4144-4153.
13. Garred P, Genster N, Pilely K, et al. A journey through the lectin 33. Bohlson SS, O’Conner SD, Hulsebus HJ, Ho M-M, Fraser DA. Comple-
pathway of complement-MBL and beyond. Immunol Rev. 2016;274: ment, C1q, and C1q-related molecules regulate macrophage polar-
74-97. ization. Front Immunol. 2014;5.
14. Ratajczak MZ, Adamiak M, Thapa A, Bujko K, Brzezniakiewicz-Janus 34. Reis ES, Mastellos DC, Hajishengallis G, Lambris JD. New insights into
K, Lenkiewicz AM. NLRP3 inflammasome couples purinergic signal- the immune functions of complement. Nat Rev Immunol. 2019.
ing with activation of the complement cascade for the optimal release 35. Mak TW, Saunders ME. The Immune Response: Basic and Clinical Princi-
of cells from bone marrow. Leukemia. 2019;33:815-825. ples. Burlington, MA: Elsevier Academic; 2006.
15. Ratajczak MZ, Adamiak M, Plonka M, Abdel-Latif A, Ratajczak 36. Heesters BA, Myers RC, Carroll MC. Follicular dendritic cells:
J. Mobilization of hematopoietic stem cells as a result of innate dynamic antigen libraries. Nat Rev Immunol. 2014;14:495.
immunity-mediated sterile inflammation in the bone marrow 37. Behrens EM, Sriram U, Shivers DK, et al. Complement receptor
microenvironment—the involvement of extracellular nucleotides 3 ligation of dendritic cells suppresses their stimulatory capacity.
and purinergic signaling. Leukemia. 2018;32:1116-1123. J Immunol. 2007;178:6268-6279.
16. Harboe M, Mollnes TE. The alternative complement pathway revis- 38. Skoberne M, Somersan S, Almodovar W, et al. The apoptotic-cell
ited. J Cell Mol Med. 2008;12:1074-1084. receptor CR3, but not alphavbeta5, is a regulator of human dendritic-
17. Thurman JM, Holers VM. The central role of the alternative cell immunostimulatory function. Blood. 2006;108:947-955.
complement pathway in human disease. J Immunol. 2006;176: 39. Li XX, Lee JD, Kemper C, Woodruff TM. The complement recep-
1305-1310. tor C5aR2: a powerful modulator of innate and adaptive immunity.
18. Łukawska E, Polcyn-Adamczak M, Niemir ZI. The role of the alterna- J Immunol. 2019;202:3339-3348.
tive pathway of complement activation in glomerular diseases. Clin 40. Coulthard LG, Woodruff TM. Is the complement activation product
Exp Med. 2018;18:297-318. C3a a proinflammatory molecule? Re-evaluating the evidence and
19. Zipfel PF, Skerka C. Complement regulators and inhibitory proteins. the myth. J Immunol. 2015;194:3542-3548.
Nat Rev Immunol. 2009;9:729-740. 41. Guo RF, Ward PA. Role of C5a in inflammatory responses. Annu Rev
20. Brodeur SR, Angelini F, Bacharier LB, et al. C4b-Binding protein Immunol. 2005;23:821-852.
(C4BP) activates B cells through the CD40 receptor. Immunity. 42. Ricklin D, Reis ES, Mastellos DC, Gros P, Lambris JD. Complement
2003;18:837-848. component C3—the “Swiss Army Knife” of innate immunity and host
21. Kemper C, Atkinson JP, Hourcade DE. Properdin: emerging roles defense. Immunol Rev. 2016;274:33-58.
of a pattern-recognition molecule. Annu Rev Immunol. 2010;28: 43. Brennan FH, Jogia T, Gillespie ER, et al. Complement receptor C3aR1
131-155. controls neutrophil mobilization following spinal cord injury through
22. Amara U, Flierl MA, Rittirsch D, et al. Molecular intercommunica- physiological antagonism of CXCR2. JCI Insight. 2019;4.
tion between the complement and coagulation systems. J Immunol. 44. Wu MC, Brennan FH, Lynch JP, et al. The receptor for comple-
2010;185:5628-5636. ment component C3a mediates protection from intestinal ischemia-
23. Békássy ZD, Kristoffersson A-C, Rebetz J, Tati R, Olin AI, Karpman D. reperfusion injuries by inhibiting neutrophil mobilization. Proc Natl
Aliskiren inhibits renin-mediated complement activation. Kidney Int. Acad Sci USA. 2013;110:9439-9444.
2018;94:689-700. 45. West EE, Kolev M, Kemper C. Complement and the regulation of
24. Irmscher S, Döring N, Halder LD, et al. Kallikrein cleaves C3 and acti- T cell responses. Annu Rev Immunol. 2018;36:309-338.
vates complement. J Innate Immun. 2018;10:94-105. 46. Triantafilou M, Hughes TR, Morgan BP, Triantafilou K. Complement-
25. Ghebrehiwet B, Randazzo BP, Dunn JT, Silverberg M, Kaplan ing the inflammasome. Immunology. 2016;147:152-164.
AP. Mechanisms of activation of the classical pathway of com- 47. Triantafilou K, Hughes TR, Triantafilou M, Morgan BP. The com-
plement by Hageman factor fragment. J Clin Invest. 1983;71: plement membrane attack complex triggers intracellular Ca2+
1450-1456. fluxes leading to NLRP3 inflammasome activation. J Cell Sci.
26. Csomor E, Bajtay Z, Sandor N, et al. Complement protein C1q 2013;126:2903-2913.
induces maturation of human dendritic cells. Mol Immunol. 2007;44: 48. Vlaicu SI, Tatomir A, Rus V, et al. The role of complement activation in
3389-3397. atherogenesis: the first 40 years. Immunol Res. 2016;64:1-13.
10 LO AND WOODRUFF

49. Bromfield S, Muntner P. High blood pressure: the leading global bur- 73. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol
den of disease risk factor and the need for worldwide prevention pro- Res. 2017;5:3-8.
grams. Curr Hypertens Rep. 2013;15:134-136. 74. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells
50. Bakris G, Ali W, Parati G. ACC/AHA versus ESC/ESH on hypertension coming of age. Nat Immunol. 2018;19:108-119.
guidelines. J Am Coll Cardiol. 2019;73:3018-3026. 75. Yoneda M, Imamura R, Nitta H, et al. Enhancement of cancer invasion
51. Oparil S, Acelajado MC, Bakris GL, et al. Hypertension. Nat Rev Dis and growth via the C5a-C5a receptor system: implications for can-
Primers. 2018;4:18014. cer promotion by autoimmune diseases and association with cervical
52. Norlander AE, Madhur MS, Harrison DG. The immunology of hyper- cancer invasion. Oncol Lett. 2019;17:913-920.
tension. J Exp Med. 2018;215:21-33. 76. Corrales L, Ajona D, Rafail S, et al. Anaphylatoxin C5a creates a
53. Bakris GL, Sorrentino MJ. Hypertension: A Companion to Braunwald’s favorable microenvironment for lung cancer progression. J Immunol.
Heart Disease. Philadelphia, PA: Elsevier; 2018. 2012;189:4674-4683.
54. Chen X-H, Ruan C-C, Ge Q, et al. Deficiency of complement C3a and 77. Cho MS, Vasquez HG, Rupaimoole R, et al. Autocrine effects of
C5a receptors prevents angiotensin II-induced hypertension via reg- tumor-derived complement. Cell Rep. 2014;6:1085-1095.
ulatory T cells. Circ Res. 2018;122:970-983. 78. Ajona D, Ortiz-Espinosa S, Moreno H, et al. A combined PD-1/C5a
55. Zhang J, Crowley SD. The role of type 1 angiotensin receptors on T blockade synergistically protects against lung cancer growth and
lymphocytes in cardiovascular and renal diseases. Curr Hypertens Rep. metastasis. Cancer Discov. 2017;7:694-703.
2013;15:39-46. 79. Vadrevu SK, Chintala NK, Sharma SK, et al. Complement c5a recep-
56. Strainic MG, Shevach EM, An F, Lin F, Medof ME. Absence of signal- tor facilitates cancer metastasis by altering T-cell responses in the
ing into CD4+ cells via C3aR and C5aR enables autoinductive TGF- metastatic niche. Cancer Res. 2014;74:3454-3465.
𝛽1 signaling and induction of Foxp3+ regulatory T cells. Nat Immunol. 80. Wang Y, Sun SN, Liu Q, et al. Autocrine complement inhibits IL10-
2013;14:162-171. dependent t-cell-mediated antitumor immunity to promote tumor
57. Zhang C, Li Y, Wang C, et al. Complement 5a receptor mediates progression. Cancer Discov. 2016;6:1022-1035.
angiotensin II-induced cardiac inflammation and remodeling. Arte- 81. Nabizadeh JA, Manthey HD, Steyn FJ, et al. The complement C3a
rioscler Thromb Vasc Biol. 2014;34:1240-1248. receptor contributes to melanoma tumorigenesis by inhibiting neu-
58. Ren J, Crowley SD. Role of T-cell activation in salt-sensitive hyperten- trophil and CD4+ T cell responses. J Immunol. 2016;196:4783-4792.
sion. Am J Physiol Heart Circul Physiol. 2019;316:H1345-H1353. 82. Mastellos DC, Ricklin D, Lambris JD. Clinical promise of next-
59. Kassan M, Wecker A, Kadowitz P, Trebak M, Matrougui K. generation complement therapeutics. Nat Rev Drug Discov.
CD4+CD25+Foxp3 regulatory T cells and vascular dysfunction 2019;18:707-729.
in hypertension. J Hypertens. 2013;31:1939-1943. 83. Ingulli E. Mechanism of cellular rejection in transplantation. Pediatr
60. Weiss S, Rosendahl A, Czesla D, et al. The complement recep- Nephrol. 2010;25:61-74.
tor C5aR1 contributes to renal damage but protects the heart 84. Rana A, Godfrey EL. Outcomes in solid-organ transplantation: suc-
in angiotensin II-induced hypertension. Am J Physiol Renal Physiol. cess and stagnation. Tex Heart Inst J. 2019;46:75-76.
2016;310:F1356-F1365. 85. Moreau A, Varey E, Anegon I, Cuturi MC. Effector mechanisms of
61. Ruan C-C, Gao P-J. Role of complement-related inflammation rejection. Cold Spring Harb Perspect Med. 2013;3.
and vascular dysfunction in hypertension. Hypertension. 2019;73: 86. Chalasani G, Li Q, Konieczny BT, et al. The allograft defines the type of
965-971. rejection (acute versus chronic) in the face of an established effector
62. Sumida T, Naito AT, Nomura S, et al. Complement C1q-induced acti- immune response. J Immunol. 2004;172:7813-7820.
vation of beta-catenin signalling causes hypertensive arterial remod- 87. Port FK, Dykstra DM, Merion RM, Wolfe RA. Organ donation
elling. Nat Commun. 2015;6:6241. and transplantation trends in the USA, 2003. Am J Transplant.
63. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global 2004;4(Suppl 9):7-12.
cancer statistics 2018: gLOBOCAN estimates of incidence and mor- 88. Kloc M, Ghobrial RM. Chronic allograft rejection: a significant hurdle
tality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. to transplant success. Burns Trauma. 2014;2:3-10.
2018;68:394-424. 89. Chapman JR, Kanellis J. Kidney donation and transplantation in
64. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. Australia: more than a supply and demand equation. Med J Aust.
2000;100:57-70. 2018;209:242-243.
65. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. 90. Vella JP, Spadafora-Ferreira M, Murphy B, et al. Indirect allorecogni-
Cell. 2011;144:646-674. tion of major histocompatibility complex allopeptides in human renal
66. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and can- transplant recipients with chronic graft dysfunction. Transplantation.
cer. Cell. 2010;140:883-899. 1997;64:795-800.
67. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflamma- 91. Ciubotariu R, Liu Z, Colovai AI, et al. Persistent allopeptide reactivity
tion. Nature. 2008;454:436-444. and epitope spreading in chronic rejection of organ allografts. J Clin
68. Schwitalla S, Ziegler PK, Horst D, et al. Loss of p53 in enterocytes Invest. 1998;101:398-405.
generates an inflammatory microenvironment enabling invasion and 92. Baker RJ, Hernandez-Fuentes MP, Brookes PA, Chaudhry AN, Cook
lymph node metastasis of carcinogen-induced colorectal tumors. HT, Lechler RI. Loss of direct and maintenance of indirect allore-
Cancer Cell. 2013;23:93-106. sponses in renal allograft recipients: implications for the patho-
69. Greten FR, Grivennikov SI. Inflammation and cancer: triggers, mech- genesis of chronic allograft nephropathy. Eur J Immunol. 2001;167:
anisms, and consequences. Immunity. 2019;51:27-41. 7199-7206.
70. Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT. Can- 93. Najafian N, Salama AD, Fedoseyeva EV, Benichou G, Sayegh MH.
cer and the complement cascade. Mol Cancer Res. 2010;8: Enzyme-linked immunosorbent spot assay analysis of peripheral
1453-1465. blood lymphocyte reactivity to donor HLA-DR peptides: potential
71. Markiewski MM, DeAngelis RA, Benencia F, et al. Modulation novel assay for prediction of outcomes for renal transplant recipients.
of the antitumor immune response by complement. Nat Immunol. J Am Soc Nephrol. 2002;13:252-259.
2008;9:1225-1235. 94. Zhang M, Takahashi K, Alicot EM, et al. Activation of the lectin path-
72. Afshar-Kharghan V. The role of the complement system in cancer. way by natural IgM in a model of ischemia/reperfusion injury. Eur J
J Clin Invest. 2017;127:780-789. Immunol. 2006;177:4727-4734.
LO AND WOODRUFF 11

95. Atkinson C, Qiao F, Yang X, et al. Targeting pathogenic postischemic 114. Tippett LJ, Hogg VM. All in the family: huntington’s disease, variabil-
self-recognition by natural IgM to protect against posttransplanta- ity and challenges for clinical neuropsychology. In: Macniven J A B, ed.
tion cardiac reperfusion injury. Circulation. 2015;131:1171-1180. Neuropsychological Formulation: A Clinical Casebook. Cham: Springer
96. Chun N, Fairchild RL, Li Y, et al. Complement dependence of murine International Publishing; 2016:47-70.
costimulatory blockade-resistant cellular cardiac allograft rejection. 115. van Munster CE, Uitdehaag BM. Outcome measures in clinical trials
Am J Transplant. 2017;17:2810-2819. for multiple sclerosis. CNS Drugs. 2017;31:217-236.
97. Farrar CA, Tran D, Li K, et al. Collectin-11 detects stress-induced 116. Helbig I, Swinkels MEM, Aten E, et al. Structural genomic variation
L-fucose pattern to trigger renal epithelial injury. J Clin Invest. in childhood epilepsies with complex phenotypes. Eur J Hum Genet.
2016;126:1911-1925. 2014;22:896-901.
98. Sommer W, Tudorache I, Kuhn C, et al. C1-esterase-inhibitor for 117. Slattery CF, Crutch SJ, Schott JM. Phenotypical variation in
primary graft dysfunction in lung transplantation. Transplantation. Alzheimer’s disease: insights from posterior cortical atrophy.
2014;97:1185-1191. Pract Neurol (Fort Wash Pa). 2015;15:2-4.
99. Raedler H, Heeger PS. Complement regulation of T-cell alloimmunity. 118. Wiest G. Neural and mental hierarchies. Front Psychol. 2012;3:516.
Curr Opin Organ Transplant. 2011;16:54-60. 119. Xu X, Warrington AE, Bieber AJ, Rodriguez M. Enhancing CNS repair
100. Raedler H, Vieyra MB, Leisman S, et al. Anti-complement com- in neurological disease: challenges arising from neurodegeneration
ponent C5 mAb synergizes with CTLA4Ig to inhibit alloreactive and rewiring of the network. CNS Drugs. 2011;25:555-573.
T cells and prolong cardiac allograft survival in mice. Am J Transplant. 120. Choi DW. Excitotoxic cell death. J Neurobiol. 1992;23:1261-1276.
2011;11:1397-1406. 121. Nadler J. Plasticitiy of glutamate synpatic mechanisms. In: Noebels
101. Gueler F, Rong S, Gwinner W, et al. Complement 5a recep- JA, Rogawski M A, eds. Jasper’s Basic Mechanisms of the Epilepsies
tor inhibition improves renal allograft survival. J Am Soc Nephrol. [Internet]. Bethesda, MD: National Center for Biotechnology Informa-
2008;19:2302-2312. tion (US); 2012.
102. Mathern DR, J KH, Heeger PS. Absence of recipient C3aR1 signal- 122. Rang HP, Dale MM, Flower RJ, Henderson G. Mechanisms of Neuronal
ing limits expansion and differentiation of alloreactive CD8+ T cell Death. Rang and Dale’s Pharmacology. England: Elsevier Churchill Liv-
immunity and prolongs murine cardiac allograft survival. Am J Trans- ingstone; 2016.
plant. 2019;19:1628-1640. 123. Won SJ, Kim DY, Gwag BJ. Cellular and molecular pathways of
103. Lublin DM, Atkinson JP. Decay-accelerating factor: biochemistry, ischemic neuronal death. J Biochem Mol Biol. 2002;35:67-86.
molecular biology, and function. Annu Rev Immunol. 1989;7:35-58. 124. Filiou MD, Arefin AS, Moscato P, Graeber MB. ‘Neuroinflamma-
104. Pavlov V, Raedler H, Yuan S, et al. Donor deficiency of decay- tion’ differs categorically from inflammation: transcriptomes of
accelerating factor accelerates murine T cell-mediated cardiac allo- Alzheimer’s disease, Parkinson’s disease, schizophrenia and inflam-
graft rejection. J Immunol. 2008;181:4580-4589. matory diseases compared. Neurogenetics. 2014;15:201-212.
105. Kwan WH, van der Touw W, Paz-Artal E, Li MO, Heeger PS. Sig- 125. DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in
naling through C5a receptor and C3a receptor diminishes func- the details. J Neurochem. 2016;139(Suppl 2):136-153.
tion of murine natural regulatory T cells. J Exp Med. 2013;210: 126. Sochocka M, Diniz BS, Leszek J. Inflammatory response in the CNS:
257-268. friend or Foe? Mol Neurobiol. 2017;54:8071-8089.
106. van der Touw W, Cravedi P, Kwan WH, Paz-Artal E, Merad M, 127. Schwartz M, Baruch K. The resolution of neuroinflammation in neu-
Heeger PS. Cutting edge: receptors for C3a and C5a modulate sta- rodegeneration: leukocyte recruitment via the choroid plexus. EMBO
bility of alloantigen-reactive induced regulatory T cells. J Immunol. J. 2014;33:7-22.
2013;190:5921-5925. 128. Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ.
107. Verghese DA, Demir M, Chun N, et al. T cell expression of C5a Inflammatory cytokine levels correlate with amyloid load in trans-
receptor 2 augments murine regulatory T cell (TREG) genera- genic mouse models of Alzheimer’s disease. J Neuroinflammation.
tion and TREG-dependent cardiac allograft survival. J Immunol. 2005;2:9.
2018;200:2186-2198. 129. Passamonti L, Tsvetanov KA, Jones PS, et al. Neuroinflammation and
108. Llaudo I, Fribourg M, Medof ME, Conde P, Ochando J, Heeger PS. functional connectivity in Alzheimer’s disease: interactive influences
C5aR1 regulates migration of suppressive myeloid cells required for on cognitive performance. J Neurosci Nur. 2019;39:7218-7226.
costimulatory blockade-induced murine allograft survival. Am J Trans- 130. Henkel JS, Beers DR, Wen S, et al. Regulatory T-lymphocytes medi-
plant. 2019;19:633-645. ate amyotrophic lateral sclerosis progression and survival. EMBO Mol
109. Dempsey PW, Allison ME, Akkaraju S, Goodnow CC, Fearon DT. C3d Med. 2013;5:64-79.
of complement as a molecular adjuvant: bridging innate and acquired 131. Zhao W, Beers DR, Hooten KG, et al. Characterization of gene expres-
immunity. Science (New York, NY). 1996;271:348-350. sion phenotype in amyotrophic lateral sclerosis monocytes. JAMA
110. Fang Y, Xu C, Fu YX, Holers VM, Molina H. Expression of comple- Neurol. 2017;74:677-685.
ment receptors 1 and 2 on follicular dendritic cells is necessary for 132. Weiss A, Träger U, Wild EJ, et al. Mutant huntingtin fragmentation
the generation of a strong antigen-specific IgG response. J Immunol. in immune cells tracks Huntington’s disease progression. J Clin Invest.
1998;160:5273-5279. 2012;122:3731-3736.
111. Marsh JE, Farmer CK, Jurcevic S, Wang Y, Carroll MC, Sacks 133. Fan Z, Pan Y-T, Zhang Z-Y, et al. Systemic activation of NLRP3
SH. The allogeneic T and B cell response is strongly dependent inflammasome and plasma 𝛼-synuclein levels are correlated with
on complement components C3 and C4. Transplantation. 2001;72: motor severity and progression in Parkinson’s disease. J Neuroin-
1310-1318. flamm. 2020;17:11.
112. GBD 2015 DALYs and HALE Collaborators 2016, Global, regional, 134. Bitsch A, Schuchardt J, Bunkowski S, Kuhlmann T, Bruck W. Acute
and national disability-adjusted life-years (DALYs) for 315 diseases axonal injury in multiple sclerosis. correlation with demyelination and
and injuries and healthy life expectancy (HALE), 1990–2015: a sys- inflammation. Brain. 2000;123(Pt 6):1174-1183.
tematic analysis for the Global Burden of Disease Study 2015. Lancet 135. Lee JD, Kumar V, Fung JN, Ruitenberg MJ, Noakes PG, Woodruff
North Am Ed 388, 1603-1658. TM. Pharmacological inhibition of complement C5a-C5a1 recep-
113. Takeda T, Kitagawa K, Arai K. Phenotypic variability and its tor signalling ameliorates disease pathology in the hSOD1(G93A)
pathological basis in amyotrophic lateral sclerosis. Neuropathology. mouse model of amyotrophic lateral sclerosis. Br J Pharmacol.
2020;40:40-56. 2017;174:689-699.
12 LO AND WOODRUFF

136. Woodruff TM, Lee JD, Noakes PG. Role for terminal complement acti- 157. An XQ, Xi W, Gu CY, Huang X. Complement protein C5a enhances
vation in amyotrophic lateral sclerosis disease progression. Proc Natl the beta-amyloid-induced neuro-inflammatory response in microglia
Acad Sci USA. 2014;111:E3-4. in Alzheimer’s disease. Med Sci (Paris) 34 Focus issue F1. 2018:
137. Woodruff TM, Crane JW, Proctor LM, et al. Therapeutic activity of 116-120.
C5a receptor antagonists in a rat model of neurodegeneration. FASEB 158. Gordon R, Albornoz EA, Kumar V, et al. Complement C5a activates
J. 2006;20:1407-1417. microglial NLRP3 inflammasomes and drives neurodegeneration in
138. Nath N, Khan M, Paintlia MK, Singh I, Hoda MN, Giri S. Metformin Parkinson’s disease through C5aR1. Immunobiology. 2016;221:1190.
attenuated the autoimmune disease of the central nervous system in 159. Yao ST, Cao F, Chen JL, et al. NLRP3 is required for complement-
animal models of multiple sclerosis. J Immunol. 2009;182:8005-8014. mediated caspase-1 and IL-1beta activation in ICH. J Mol Neurosci.
139. Brennan FH, Gordon R, Lao HW, et al. The complement receptor 2017;61:385-395.
C5aR controls acute inflammation and astrogliosis following spinal 160. Mukherjee P, Thomas S, Pasinetti GM. Complement anaphylatoxin
cord injury. J Neurosci. 2015;35:6517-6531. C5a neuroprotects through regulation of glutamate receptor subunit
140. Dong Y, Li X, Cheng J, Hou L. Drug development for Alzheimer’s dis- 2 in vitro and in vivo. J Neuroinflamm. 2008;5:5.
ease: microglia induced neuroinflammation as a target? Int J Mol Sci. 161. Rahpeymai Y, Hietala MA, Wilhelmsson U, et al. Complement: a
2019;20. novel factor in basal and ischemia-induced neurogenesis. EMBO J.
141. Khalid SI, Ampie L, Kelly R, Ladha SS, Dardis C. Immune modulation 2006;25:1364-1374.
in the treatment of amyotrophic lateral sclerosis: a review of clinical 162. Bouwens van der Vlis TAM, Kros JM, Mustafa DAM, et al. The com-
trials. Front Neurol. 2017;8:486. plement system in glioblastoma multiforme. Acta Neuropathol Com-
142. Kolahdouzan M, Futhey NC, Kieran NW, Healy LM. Novel molecular mun. 2018;6:91.
leads for the prevention of damage and the promotion of repair in 163. Duan T, Smith AJ, Verkman AS. Complement-independent bystander
neuroimmunological disease. Front Immunol. 2019;10. injury in AQP4-IgG seropositive neuromyelitis optica produced by
143. Gasque P, Fontaine M, Morgan BP. Complement expression in human antibody-dependent cellular cytotoxicity. Acta Neuropathol Commun.
brain. Biosynthesis of terminal pathway components and regulators 2019;7:112.
in human glial cells and cell lines. J Immunol. 1995;154:4726-4733. 164. Willison HJ, Halstead SK, Beveridge E, et al. The role of comple-
144. Lévi-Strauss M, Mallat M. Primary cultures of murine astrocytes pro- ment and complement regulators in mediating motor nerve terminal
duce C3 and factor B, two components of the alternative pathway of injury in murine models of Guillain-Barre syndrome. J Neuroimmunol.
complement activation. J Immunol. 1987;139:2361-2366. 2008;201–202:172-182.
145. Pavlovski D, Thundyil J, Monk PN, Wetsel RA, Taylor SM, Woodruff 165. Alawieh A, Langley EF, Weber S, Adkins D, Tomlinson S. Identifying
TM. Generation of complement component C5a by ischemic neurons the role of complement in triggering neuroinflammation after trau-
promotes neuronal apoptosis. FASEB J. 2012;26:3680-3690. matic brain injury. J Neurosci. 2018;38:2519-2532.
146. Hosokawa M, Klegeris A, Maguire J, McGeer PL. Expression of com- 166. Michailidou I, Jongejan A, Vreijling JP, et al. Systemic inhibition of the
plement messenger RNAs and proteins by human oligodendroglial membrane attack complex impedes neuroinflammation in chronic
cells. Glia. 2003;42:417-423. relapsing experimental autoimmune encephalomyelitis. Acta Neu-
147. Peterson SL, Anderson AJ. Complement and spinal cord injury: tra- ropathol Commun. 2018;6:36.
ditional and non-traditional aspects of complement cascade function 167. Rus H, Cudrici C, Niculescu F. C5b-9 complement complex in autoim-
in the injured spinal cord microenvironment. Exp Neurol. 2014;258: mune demyelination and multiple sclerosis: dual role in neuroinflam-
35-47. mation and neuroprotection. Ann Med. 2005;37:97-104.
148. Luchena C, Zuazo-Ibarra J, Alberdi E, Matute C, Capetillo-Zarate 168. Yang C, Yang L, Liu Y. Soluble complement complex C5b-9 promotes
E. Contribution of neurons and glial cells to complement-mediated microglia activation. J Neuroimmunol. 2014;267:16-19.
synapse removal during development, aging and in Alzheimer’s dis- 169. Singhrao SK, Neal JW, Rushmere NK, Morgan BP, Gasque P. Sponta-
ease. Mediat Inflamm. 2018;2018:2530414. neous classical pathway activation and deficiency of membrane reg-
149. Bodea LG, Wang Y, Linnartz-Gerlach B, et al. Neurodegeneration by ulators render human neurons susceptible to complement lysis. Am J
activation of the microglial complement-phagosome pathway. J Neu- Pathol. 2000;157:905-918.
rosci. 2014;34:8546-8556. 170. Orsini F, De Blasio D, Zangari R, Zanier ER, De Simoni MG. Versatility
150. Krukowski K, Chou A, Feng X, et al. Traumatic brain injury in of the complement system in neuroinflammation, neurodegeneration
aged mice induces chronic microglia activation, synapse loss, and and brain homeostasis. Front Cell Neurosci. 2014;8:380.
complement-dependent memory deficits. Int J Mol Sci. 2018;19. 171. Shen Y, Meri S. Yin and Yang: complement activation and regulation
151. Michailidou I, Willems JG, Kooi EJ, et al. Complement C1q-C3- in Alzheimer’s disease. Prog Neurobiol. 2003;70:463-472.
associated synaptic changes in multiple sclerosis hippocampus. Ann 172. Alexander JJ, Anderson AJ, Barnum SR, Stevens B, Tenner AJ.
Neurol. 2015;77:1007-1026. The complement cascade: yin-Yang in neuroinflammation–neuro-
152. Fraser DA, Pisalyaput K, Tenner AJ. C1q enhances microglial protection and -degeneration. J Neurochem. 2008;107:1169-1187.
clearance of apoptotic neurons and neuronal blebs, and modu- 173. Dobson R, Giovannoni G. Multiple sclerosis - a review. Eur J Neurol.
lates subsequent inflammatory cytokine production. J Neurochem. 2019;26:27-40.
2010;112:733-743. 174. Prineas JW, Kwon EE, Cho ES, et al. Immunopathology of secondary-
153. Schetters STT, Gomez-Nicola D, Garcia-Vallejo JJ, Van Kooyk Y. progressive multiple sclerosis. Ann Neurol. 2001;50:646-657.
Neuroinflammation: microglia and T cells get ready to tango. Front 175. Bullard DC, Hu X, Schoeb TR, Axtell RC, Raman C, Barnum SR. Crit-
Immunol. 2017;8:1905. ical requirement of CD11b (Mac-1) on T cells and accessory cells
154. Armstrong RC, Harvath L, Dubois-Dalcq ME. Type 1 astrocytes and for development of experimental autoimmune encephalomyelitis.
oligodendrocyte-type 2 astrocyte glial progenitors migrate toward J Immunol. 2005;175:6327-6333.
distinct molecules. J Neurosci Res. 1990;27:400-407. 176. Bullard DC, Hu X, Adams JE, Schoeb TR, Barnum SR. p150/95
155. Yao J, Harvath L, Gilbert DL, Colton CA. Chemotaxis by a CNS (CD11c/CD18) expression is required for the development of exper-
macrophage, the microglia. J Neurosci Res. 1990;27:36-42. imental autoimmune encephalomyelitis. Am J Pathol. 2007;170:
156. Ingersoll SA, Martin CB, Barnum SR, Martin BK. CNS-specific expres- 2001-2008.
sion of C3a and C5a exacerbate demyelination severity in the cupri- 177. Wagner C, Hänsch GM, Stegmaier S, Denefleh B, Hug F, Schoels M.
zone model. Mol Immunol. 2010;48:219-230. The complement receptor 3, CR3 (CD11b/CD18), on T lymphocytes:
LO AND WOODRUFF 13

activation-dependent up-regulation and regulatory function. Eur J


Immunol. 2001;31:1173-1180. How to cite this article: Lo MW, Woodruff TM. Comple-
178. Astier AL, Meiffren G, Freeman S, Hafler DA. Alterations in CD46-
ment: Bridging the innate and adaptive immune systems in
mediated Tr1 regulatory T cells in patients with multiple sclerosis.
J Clin Invest. 2006;116:3252-3257.
sterile inflammation. J Leukoc Biol. 2020;1–13. https://doi.org/
179. Ni Choileain S, Hay J, Thomas J, et al. TCR-stimulated changes in cell 10.1002/JLB.3MIR0220-270R
surface CD46 expression generate type 1 regulatory T cells. Sci Signal.
2017;10.

You might also like