Complement in Kidney Diseases
Complement in Kidney Diseases
Complement in Kidney Diseases
1038/s41581-023-00766-1
Abstract Sections
The complement cascade comprises soluble and cell surface proteins Introduction
and is an important arm of the innate immune system. Once activated, The complement system
the complement system rapidly generates large quantities of protein Complement-mediated kidney
fragments that are potent mediators of inflammatory, vasoactive and disease
metabolic responses. Although complement is crucial to host defence Complement as a downstream
and homeostasis, its inappropriate or uncontrolled activation can also mechanism of kidney injury
drive tissue injury. For example, the complement system has been known Susceptibility of the kidney to
complement-mediated injury
for more than 50 years to be activated by glomerular immune complexes
and to contribute to autoimmune kidney disease. Notably, the latest Therapeutic options for
targeting the complement
research shows that complement is also activated in kidney diseases cascade
that are not traditionally thought of as immune-mediated, including
Complement biomarkers
haemolytic–uraemic syndrome, diabetic kidney disease and focal
Conclusions
segmental glomerulosclerosis. Several complement-targeted drugs
have been approved for the treatment of kidney disease, and additional
anti-complement agents are being investigated in clinical trials. These
drugs are categorically different from other immunosuppressive agents
and target pathological processes that are not effectively inhibited
by other classes of immunosuppressants. The development of these
new drugs might therefore have considerable benefits in the treatment
of kidney disease.
1
Institute for Clinical and Experimental Medicine, Prague, Czech Republic. 2University of Colorado Anschutz
Medical Campus, Aurora, Colorado, USA. e-mail: joshua.thurman@cuanschutz.edu
C4Bp FH,FI
C3 C5
C1q
Podocyte Endothelial IgG or IgM C5a
cell Antigen
C5b–9
FH CD59 GBM C5
C4d C4bC2a C3b convertase
CD46
CD55 GPI anchor
Slit diaphragm
d AAV
c DKD and FSGS
C2, C4 Lectin Alternative Terminal Neutrophil PR3 and/or MPO
pathway pathway pathway
C3 C5
Loss of c5aR
MBL regulators C3b C5a C5a
Saccharide (+) Vascular
C5 damage ANCA
↑ Alternative
C4bC2a convertase
pathway activity
O2
O2
e
C3 C3G aHUS C3
Alternative C3bBb Alternative C3b Regulators
pathway pathway
C3bBb
C3a C5
Fig. 1 | Mechanisms of complement activation in glomerular disease. enhanced activation within the glomerulus. d, Anti-neutrophil cytoplasmic
Activation of the classical, lectin and alternative pathways leads to the deposition antibody (ANCA)-associated vasculitis (AAV). Various stimuli might prime
of C3b on glomerular capillaries. a, In healthy kidneys, C3b is efficiently neutrophils to expose proteinase 3 (PR3) or myeloperoxidase (MPO), which are
inactivated by soluble and membrane-bound complement regulators. recognized by ANCA. Binding of ANCA leads to neutrophil activation, including
b, Immune complex-mediated diseases. IgG or IgM bind to deposited or in situ induction of an oxidative burst and degranulation. Furthermore, activated
antigens and generate the classical pathway C3 convertase (C4bC2a). Deposited neutrophils might release factors that activate the alternative pathway. C5a induces
C4b is converted to C4d, which can serve as a biomarker of classical pathway upregulation of PR3 and MPO on neutrophils, propagating this process. e, C3
activation. The C3 convertase cleaves C3, and this process is amplified through glomerulopathy (C3G) and atypical haemolytic–uraemic syndrome (aHUS). In
the alternative pathway. Activation also generates C5 convertases (C4b2a3b C3G, impaired regulation of the alternative pathway leads to overactivity in the
and C3bBbC3b), which cleave C5 and activate the terminal pathway. c, Diabetic fluid phase, and soluble fragments might contribute to kidney injury84. However,
kidney disease (DKD) and focal segmental glomerulosclerosis (FSGS). Pattern abundant clinical evidence suggests that alternative pathway activation also
recognition molecules, such as mannose binding lectin (MBL) and natural occurs directly on the capillary wall58. In aHUS, although the alternative pathway
IgM might recognize glycated proteins and activate the complement system. is regulated in the fluid phase, regulation on the surface of endothelial cells is
Complement regulation can also become impaired in these diseases, promoting impaired. GBM, glomerular basement membrane.
lectin pathways, and C3b is a component of the alternative pathway can also express complement proteins29,30, and this is often detected
C3 convertase (C3bBb). Although these proteins are synthesized as by RNA technologies31,32. Importantly, complement gene expression
soluble plasma proteins, their attachment on target surfaces focuses is not synonymous with activation per se, but local production of the
complement activation at these sites, including the kidney. Kidney cells substrate proteins might increase the degree of activation in the kidney,
and also contributes to the systemic pool of complement proteins33. or lysis of target cells38. Within the kidney, full activation of the comple-
Recent work has also shown that complement proteins are involved ment cascade, which generates all of these fragments, has profound
in physiological processes that occur within cells (‘intracellular com- local and systemic effects.
plement’)34. The synthesis of complement proteins by cells within the
kidney, might therefore also have a role in non-canonical functions Complement regulation
beyond those discussed here. As outlined above, multiple mechanisms of complement activation
exist, each of which elicits downstream responses. To protect itself from
Complement effects uncontrolled complement activation, the body expresses regulatory
Several protein fragments generated during complement activation cell-surface and soluble proteins39 (Fig. 3). These regulators act at several
mediate physiological responses (Fig. 2). C3 goes through a sequential levels within the complement cascade. Some regulators control the
cleavage process, yielding C3b, iC3b and, eventually, C3d fragments. convertases — either by shortening the half-life of these complexes (decay
C3b is an active component of the C3 and C5 convertases, whereas acceleration) or by serving as cofactors for the cleavage of C3b or C4b by
iC3b and C3d are no longer enzymatically active. Leukocyte recep- the protease factor I (cofactor activity). Several circulating proteins block
tors can bind each of these C3 fragments specifically and mediate a activation of the classical and lectin pathways specifically. C4-binding
variety of downstream functions35. C3a and C5a (also known as ana- protein (C4bp), for example, controls the classical pathway C3 convertase
phylatoxins) are small soluble peptides released following the cleavage through decay acceleration and cofactor activity40. MAp44 (also known
of C3 and C5, respectively. Leukocytes and parenchymal cells express as MAP-1) is a specific inhibitor of the lectin pathway41. In addition, ana-
seven-transmembrane G protein coupled receptors for C3a and C5a. phylatoxins are rapidly cleaved and inactivated by carboxypeptidase N42,
C3a and C5a have potent pro-inflammatory effects but can also attenu- whereas CD59 prevents C5b–9 formation39.
ate inflammation in some settings, and can promote recovery from Factor H is a soluble glycoprotein that acts through decay accelera-
injury36,37. The larger fragment generated by C5 cleavage (C5b) is the initial tion and cofactor activity to control the alternative pathway in solution
component of a multimeric complex (C5b–9; also known as the mem- and on host tissues43. Although all kidney cells express more than one
brane attack complex), which can form pores in plasma membranes, type of complement regulator on their surface, factor H is particu-
allowing passage of ions across the membrane and causing activation larly important for controlling alternative pathway activation in the
Neutrophil B lymphocyte
CR3
3
CR2
iC3b C3d BCR
MBL and
Lectin saccharide
1 C3 C5
pathway
C2, C4 C3 C5
C1q and convertase convertase
Classical IgM or IgG C4bC2a C4bC2aC3b
pathway or C3bBb C3b or C3bBbC3b C5b
4
C2, C4 C6, C7, C8,
Amplification multiple C9
Alternative loop Platelet
pathway
C3a
C3, FB, FD
2
C5a
Endothelial
cells
C3aR 5
Mesangial C5b–9
cell C5aR
GBM
Podocytes
Slit diaphragm
Fig. 2 | Complement fragments mediate multiple downstream effects. The a receptor for C3d (complement receptor 2 (CR2)). Addition of a C3b molecule to
C3 convertase of the classical and lectin pathways — the C4bC2a complex — and the C3-convertases also creates a C5-convertase (4). The C5-convertases cleave
the C3-convertase of the alternative pathways — the C3bBb complex — cleave C5 into the C5a and C5b fragments. The biological functions of C5a, including
C3 into C3a and C3b fragments (1). C3a exerts many biological functions via its vasoactive and pro-inflammatory effects, are exerted through its receptor (C5aR).
receptor (C3aR), which is expressed on mesangial cells, podocytes, platelets and C5aR is expressed on neutrophils, mesangial cells and endothelial cells, among
leukocytes, among other cell types (2). As a component of the alternative pathway other cell types. C5b associates with C6, C7, C8 and multiple C9 molecules to
C3-convertase, C3b feeds into the amplification loop. C3b also undergoes further form C5b–9, also referred to as the membrane attack complex (MAC). C5b–9
cleavage, yielding iC3b and C3d (3). Myeloid cells express a receptor for iC3b creates a pore in cell membranes that can induce cell activation or lysis (5).
(complement receptor 3 (CR3)), and B cells and follicular dendritic cells express GBM, glomerular basement membrane; MBL, mannose binding lectin.
glomerulus (Box 1). For example, congenital and acquired defects in FH-related
proteins FH
factor H function are strong risk factors for atypical haemolytic– Glycocalyx ?
CD55
uraemic syndrome (aHUS) and C3 glomerulopathy (C3G)44,45. The CD46
CD59
factor H-related proteins (FHRs) are a group of five structurally related
proteins that arose through duplication of the factor H gene (CFH)43.
Endothelial
Rather than inhibit complement activation, the FHRs are thought to cells
be positive regulators of the alternative pathway. Genetic variants GBM
of the CFHR genes are associated with the same kidney diseases as
Podocytes
factor H, possibly because the mutant proteins antagonize factor H
binding to kidney surfaces. Wild-type FHR proteins also seem to affect Slit
complement regulation or dysregulation within the kidney46,47. diaphragm
downstream fragments contribute to injury. Consequently, even in at the level of C3 or C5, such as selective C5a blockade, should leave
this well-understood disease, the optimal approach to therapeutic the early classical pathway intact and might circumvent this problem.
complement inhibition remains to be determined. Preclinical studies also suggest that selective blockade of the alterna-
tive pathway maintains the protective effects of the classical pathway
Lupus nephritis. In lupus nephritis, autoantibodies bind to intrinsic while preventing the generation of downstream pro-inflammatory
kidney antigens as well as deposited antigens. Intra-renal complement fragments92,93.
activation is probably due to activation of the classical pathway by
deposited immune complexes. Paradoxically, the classical pathway Anti-glomerular basement membrane disease. Anti-GBM disease is
also protects the host from developing autoimmunity, and deficien- caused by autoantibodies against the non-collagenous-1 (NC1) domain
cies of classical pathway proteins (C1q, C1r, C1s, C2 or C4) are strong of type IV collagen in the GBM94. Patients with IgG1 autoantibodies
monogenic risk factors for developing systemic lupus erythematosus85. (complement activating) tend to develop kidney disease, whereas
Mechanistically, activation of the classical pathway facilitates rapid those with IgG4 autoantibodies (non-complement activating) do not95.
clearance of apoptotic cells, which prevents the development of auto- Although plasma C3 and C4 levels are not usually decreased, C1q and C4d
immunity against nuclear antigens86. However, once autoimmunity are deposited in glomeruli, consistent with classical pathway activation96.
develops, activation of the classical pathway within the glomerulus Complement-inhibiting agents are protective in preclinical mod-
triggers downstream inflammatory responses. els of anti-GBM disease, demonstrating that activation of complement
The clinical evidence that complement is activated in the kidneys on the GBM contributes to glomerular injury97. Therapy currently
of patients with active lupus nephritis is abundant. Decreases in plasma focuses on elimination of the pathogenic B cells and auto-antibodies
C4 levels can predict flares, which seem to coincide with a fall in but, even with treatment, many patients progress to kidney failure98.
C3 levels87. By contrast, complement activation fragments (such as iC3b Of note, even if plasmapheresis removes autoantibodies from the cir-
and C4d) are increased in the plasma of patients with systemic lupus culation, any antibodies already bound within the capillary wall might
erythematosus compared with healthy controls, and might be more continue to cause glomerular injury. Therefore, complement-inhibiting
sensitive and specific for active kidney disease than the level of intact drugs might, in theory, prevent glomerular injury while waiting for
C3 or C4, respectively88,89. Deposits of immune complexes, and of C1q, other interventions to reduce autoantibody levels. One case report
C4 and C3 fragments (also known as a ‘full house’ immunofluorescence indicated that C5 blockade was protective in two patients who had
pattern) are almost always seen in biopsy samples from patients with not responded to other treatments (specifically, glucocorticoids,
active kidney disease, confirming that activation occurs within the cyclophosphamide, rituximab and plasmapheresis), which supports
glomerulus72,90. a potential role for complement blockade in this disease99.
Although the complement system is activated in lupus nephri-
tis, whether complement blockade has a therapeutic role remains Other antibody-mediated glomerular diseases
uncertain. Complement inhibitors might reduce inflammatory injury of In some glomerular diseases, autoantibodies do not activate the clas-
the kidney whereas suppression of autoantibody production is not yet sical pathway within the glomerulus. However, evidence suggests that
controlled with other immunosuppressive agents. However, the role of the complement system still mediates kidney injury.
the classical pathway in preventing the development of autoimmunity
raises questions as to whether complement blockade might worsen IgA nephropathy. Although IgA does not activate the classical pathway,
the underlying disease91. Drugs that target the complement cascade C3 fragments are co-deposited with IgA in almost all biopsy samples of
Associated infectious disease Shiga toxin-producing Escherichia coli infection associated with Post-infectious glomerulonephritis
haemolytic–uraemic syndrome
Complement activation phase Endothelial cell and/or glycocalyx Fluid phase and/or glomerular basement membrane
Kidney failure 60–70% without complement inhibition; 10–15% with complement 50% at 10 years67
inhibition203,209
Extrarenal manifestations Systemic thrombotic microangiopathy; retinal drusen are rare Partial lipodystrophy211, retinal drusen212
Cytokine Macrophage
GBM Slit diaphragm Podocyte
release Haematuria
Endothelial cells Podocyte damage
Proteinuria
Fig. 4 | Complement-inhibiting drugs for the treatment of kidney diseases. activation of the adaptive immune response; pro-inflammatory CD4 T cells,
Complement activation generates C3a, C3b,iC3b,C3d, C5a and C5b–9. cytotoxic T cells, macrophages and cytokines all contribute to tissue injury.
These fragments have multiple pro-inflammatory effects and are sufficient to Consequently, complement inhibitors might only block a subset of downstream
cause glomerular injury. a, Complement fragments are the primary drivers of mediators of injury and would probably need to be used in combination with
disease in atypical haemolytic–uraemic syndrome and C3 glomerulopathy. In drugs that block the primary autoimmune response. However, standard immuno
these diseases, complement-inhibiting drugs block the pathological downstream suppressive drugs can take weeks or months to reduce kidney inflammation,
effects of complement activation. b, In antibody-mediated kidney diseases, and complement-inhibiting drugs might rapidly suppress kidney injury in
loss of tolerance enables the production of self-reactive antibodies. These the period before those drugs take effect. In addition, complement inhibition
autoantibodies can form immune complexes and generate pro-inflammatory might also modulate the primary adaptive immune response that causes
complement fragments. These inflammatory mediators can contribute to the autoimmunity.
patients with IgA nephropathy (IgAN)100. Furthermore, most patients The detection of glomerular C3 deposits in the absence of C4d in
do not have hypocomplementaemia, but those with decreased levels of many cases of IgAN, and the detection of Bb and properdin, also sug-
C3 have worse outcomes than patients with C3 levels within the normal gest an important role for the alternative pathway110,111. Additionally,
range101. The mechanism by which complement is activated in IgAN is a large genome-wide association study found that a relatively com-
unclear, but IgA1 can activate the alternative pathway directly in vitro102. mon deletion of genes for two of the FHRs (CFHR3 and CHFR1)112 is
Analysis of the circulating and deposited IgA1 has also revealed that protective against development of IgAN; this correlation was con-
the O-linked glycans in the hinge region of the molecule are galac- firmed in follow-up studies113,114. FHR1 and FHR3 can antagonize factor
tose deficient103. These modified glycans are targeted by IgA and IgG H-dependent regulation of the alternative pathway. Absence of these
autoantibodies104, and glomerular immune deposits contain a mixture proteins might therefore improve control of the alternative pathway
of galactose-deficient IgA1 as well as autoantibodies105. Complement and protect against disease development. Conversely, FHR5 deposits
activation could therefore be caused by the IgA1 deposited in the are seen in the glomeruli of patients with IgAN115,116, and higher plasma
glomeruli and/or the concomitant IgG autoantibodies. levels of FHR1 and FHR5 are associated with a worse prognosis47,117.
Histological evidence suggests that activation of the classical CFHR5 mutations are also associated with the risk of developing IgAN,
and lectin pathways occurs in subsets of patients with IgAN. C4d is possibly because the variants competitively inhibit the binding of
seen in >30% of biopsies106, whereas C1q deposits are less frequent107. factor H118. Together, these findings suggest a crucial disease-causing
Glomerular MBL is also seen in ~25% of cases and correlates with C4d role for the alternative pathway in IgAN, even if complement activa-
deposition19. The detection of these proteins in the glomeruli cor- tion is initially triggered through the classical or lectin pathways in
relates with a worse prognosis, supporting a pathogenic role for some patients.
complement19,106–109. Of note, although proteins from the classical
and lectin pathways are only detected in subsets of patients with IgAN, Anti-neutrophil cytoplasmic antibody-associated vasculitis.
antigen retrieval might reveal a greater prevalence for these proteins, Anti-neutrophil cytoplasmic antibody (ANCA)-associated vasculi-
as shown in MN20. tis (AAV) is an immunoglobulin-mediated disease that commonly
MBL can bind to galactose-deficient IgA1, similar to what was affects the glomerular capillaries. However, immunoglobulin and
shown for galactose-deficient IgG4 autoantibodies in MN79. MBL complement are only sparsely deposited in the glomeruli of affected
might also bind to neoepitopes displayed on injured glomerular cells. patients119. Experiments using a mouse model of AAV showed that mice
with a genetic deletion of Cfb (necessary for activation of the alter- Glomerular diseases not associated with antibodies
native pathway) were protected from injury, whereas deletion of C4 Kidney biopsy samples are frequently immunostained for C3 and evi-
(necessary for activation of the classical and lectin pathways) was not dence suggests that complement is activated in glomerular diseases
protective120. These preclinical studies were surprising because the dis- not traditionally thought of as immune mediated.
ease is antibody-mediated, which would suggest complement activa-
tion through the classical pathway. Furthermore, complement deposits Diabetic kidney disease. Patients with diabetes might have kidney dis-
are prominent neither in the glomeruli of affected patients nor in the ease of any aetiology, but diabetic kidney disease (DKD) is the leading
mouse model120. C5-deficient mice were also protected from injury, cause of CKD and kidney failure worldwide. Deposition of complement
implicating either C5a or C5b–9 in disease pathogenesis 120. proteins in the glomeruli of patients with DKD is associated with worse
A subsequent study showed that deficiency or pharmacological inhi- outcomes130,131, but how complement is activated or whether it has a
bition of C5aR1 was protective, whereas mice deficient in C6, and pathological role remains unknown. Two mechanisms linking comple-
thus unable to form C5b–9, were not protected121. Collectively, these ment and DKD have been proposed — activation of the lectin pathway
preclinical studies indicate that complement activation in AAV pri- by glycated proteins and deregulation of the complement system
marily involves the alternative pathway, and that glomerular injury is owing to a glycation-induced impairment of complement regulators.
mediated by C5a. MBL binds to altered glycans and activates complement in vitro132
Although AAV is often described as ‘pauci-immune’, one study and might therefore bind to glycoproteins altered by the hypergly-
noted that immunoglobulin and complement proteins can be caemic state (Fig. 1). Several animal133–135 and human136,137 studies
detected in most biopsy samples122. Furthermore, pathogenic comple- also demonstrated increased activity of the lectin pathway in DKD.
ment activation might occur outside of the kidney. The original study Collectin-11, which is a lectin pathway activator that is important
using the AAV mouse model demonstrated that binding of ANCA in acute kidney injury (AKI; see below) is also elevated in plasma of
(either anti-proteinase 3 (PR3) or anti-myeloperoxidase antibodies) to patients with diabetes and microalbuminuria compared with plasma
activated neutrophils caused the cells to release an unidentified fac- levels of patients with diabetes and normoalbuminuria138. These obser-
tor that triggers activation of complement in serum120. Complement vations potentially represent downstream mechanisms of activation
activation might therefore occur on or near the neutrophils, rather of the lectin pathway in affected kidneys.
than directly on the glomerular capillary wall. Binding of C5a to C5aR1 Interesting experiments showed that hyperglycaemia can impair
on neutrophils induces the translocation of PR3 and myeloperoxidase the function of complement regulatory proteins directly. Chronic
to the neutrophil surface, thereby increasing the expression of ANCA hyperglycaemia correlates with urine levels of glycated soluble CD59,
ligands on the cells. C5a might therefore potentiate the action of which is functionally less effective at controlling C5b–9 formation
ANCA, which might, in turn, lead to complement activation through than the non-glycated form139,140. This effect might mechanistically link
their effects on neutrophils (Fig. 1). AAV is not usually associated with hyperglycaemia with acquired impairments of complement regulation.
low C3 and C4 levels, but one study found that patients with active Increased levels of glycated soluble CD59 in plasma were also reported
disease had elevated plasma and urine C5a levels compared with as a sensitive and specific biomarker for gestational diabetes141.
patients in remission and healthy individuals123. Analysis of plasma Notably, CD59 is ordinarily linked to the surface of cells through a
and urine from patients with active AAV also revealed that levels of Bb glycosylphosphatidylinositol anchor, but the significance of increased
(indicative of activation of the alternative pathway) were higher plasma levels of the protein is unclear. Moreover, in a mouse model of
in patients with active disease than in those in remission124,125. The DKD, tissue-specific deletion of CD55 (another cell surface complement
same study reported deposition of Bb in the glomeruli, indicating regulatory protein) in podocytes was associated with exacerbated kid-
that some activation of the alternative pathway might occur within ney disease142, highlighting the importance of adequate complement
the kidney124. regulation for protecting the diabetic glomerulus. Genetic variants in
The C5aR1 antagonist avacopan has now been tested in several factor H are also risk factors for the development of kidney and cardio-
clinical trials of patients with AAV126,127. Standard treatment of AAV vascular complications in patients with diabetes, presumably owing to
involves either cyclophosphamide or rituximab in combination with differences in the inhibitory function of the variants143.
glucocorticoids128,129. In a phase III study, patients were randomly
assigned to cyclophosphamide or rituximab in combination with Focal segmental glomerulosclerosis. Focal segmental glomeru-
either prednisone (tapered over 21 weeks) or avacopan (continued losclerosis (FSGS) is a podocytopathy144. Inflammatory changes are
for 52 weeks)126. Compared with standard treatment plus pred- not usually seen in the glomeruli by light microscopy nor are immune
nisone, cyclophosphamide or rituximab combined with avacopan complexes typically detected through electron microscopy. Although
was non-inferior in inducing remission by 26 weeks and sustained immunostaining often reveals glomerular deposits of IgM and C3,
remission in the avacopan group was greater at 52 weeks. Importantly, especially in sclerotic areas145,146, these findings have been ascribed to
patients treated with C5a blockade received significantly fewer glu- passive trapping of plasma IgM and C3 in the damaged tissue. However,
cocorticoids over the course of the study and, accordingly, had fewer preclinical and clinical studies showed that the deposited IgM activates
glucocorticoid-associated toxicities than those receiving standard the classical pathway146,147. Accordingly, glomerular C4d positivity was
treatment. Based on these results, avacopan has been approved in the reported in 73% of kidney tissue samples from patients with FSGS com-
USA, Canada and Europe as an adjunct therapy in AAV. This approval pared with only 10% of samples from healthy individuals148. Glomerular
is a success story for the use of animal models to identify and validate C4d colocalized with C1q but not MBL, indicating that activation likely
new treatments for human disease. Nevertheless, the mechanisms occurs through the classical pathway148. Complement activation frag-
by which C5a contributes to kidney injury are incompletely under- ments, including C4a and C4d, are also increased in urine and plasma
stood. For example, whether C5a acts within the kidney or directly on of patients with FSGS compared with healthy individuals and disease
neutrophils remains unclear. controls, providing further evidence of activation of the classical
pathway149,150. Moreover, CD55 expression is lower in glomeruli of can also be detected within the cysts and in the urine of patients
patients with FSGS than in those of healthy controls142, which might with PKD, suggesting that similar processes might occur in human
potentiate complement activation, as discussed above for DKD (Fig. 1). disease164. Currently, no active clinical trials are testing the effect of
Immunoglobulin deposition and complement activation complement-inhibiting drugs in PKD.
are probably downstream events in FSGS that occur after injury
neoepitopes have been generated and complement regulation Susceptibility of the kidney to complement-
has become impaired. The glomerular immune deposits observed mediated injury
in FSGS might therefore be markers, rather than causes, of injury. As outlined above, the complement system is activated in kidney dis-
Nevertheless, complement-deficient mice are protected in a model of eases of widely differing aetiologies, suggesting that the balance of
adriamycin-induced podocyte injury151,152 and glomerular complement complement regulation or activation is easily disrupted in this organ.
deposits are associated with worse outcomes in patients with FSGS145. However, many differences exist between the microenvironment in
Therapeutic complement inhibition might therefore be an effective the glomerulus and that of the tubulointerstitium.
method of attenuating disease. Glomeruli are ‘inward-facing’ and are continually exposed to
high concentrations of complement proteins and immune complexes
C1q nephropathy. C1q nephropathy is defined by intense glomerular that are generated elsewhere in the body. Consequently, adequate
C1q staining. However, unlike in C3G, a defined cut-off of minimal complement regulation is probably crucial at this location to protect
intensity has not been specified and co-deposition of IgM, IgG or C3 is the kidney from the constant influx of these proteins. All cell-surface
frequent153,154. C1q deposits do not always correlate with immunoglobu- complement regulators are expressed on resident glomerular cells165
lin or C3 levels153, and whether these C1q deposits are an indicator of (Fig. 3). Furthermore, expression of these regulators increases in some
activation of the classical pathway remains unclear. By light micros- glomerular diseases, possibly as a protective response to complement
copy, the glomeruli usually have patterns of injury that are character- activation166,167. Nevertheless, complement regulation in the glomeru-
istic of minimal change disease or FSGS, although some patients have lus is clearly inadequate to prevent its activation in many glomerular
proliferative glomerulonephritis153,154. One study of children with mini- diseases. Perhaps a threshold of immune complex abundance exists,
mal change disease reported that C1q positivity was associated with beyond which these protective mechanisms are overwhelmed, or
more frequent relapses but found no differences in kidney outcomes155. immune complexes might become more pathogenic when a ‘second hit’
impairs local complement regulation. In contrast to glomerular cells,
Tubulointerstitial kidney diseases the GBM is fully dependent on soluble complement regulators, such as
The complement system is activated in several tubulointerstitial dis- factor H58. Consequently, although the GBM is susceptible to congenital
eases. The mechanisms and localization of complement activation in or acquired defects in factor H function, the availability of factor H to
these diseases suggests that activation in the tubulointerstitium occurs protect the GBM is probably not affected by glomerular cell injury.
independently of that in the glomerulus. In contrast to the microenvironment in the glomerulus, tubular
epithelial cells actively promote complement activation in the setting
Acute kidney injury. Much of the mechanistic data about complement of injury. CD46 is the main regulator expressed on tubular epithelial
in AKI comes from animal models of ischaemia–reperfusion injury. cells165, but several types of injury are associated with a reduction in
Studies in mice showed that the alternative pathway is activated in the complement regulation on the surface of these cells157,168. Moreover,
post-ischaemic tubulointerstitium156. Local production of complement tubular epithelial cells can synthesize C3 protein and lectin pathway
proteins increases after ischaemia, accompanied by an acquired loss of components (collectin-11 and l-fucose) that amplify local activation
complement regulation in the injured tubules157. A 2016 study reported a further169,170. Given that tubular epithelial cells are a ‘barrier’ epithelium,
mechanism by which the lectin pathway is activated in the kidney follow- these mechanisms that facilitate local complement activation might
ing ischaemia158. Both collectin-11 and its ligand l-fucose are produced have evolved as a defence against urinary pathogens.
by stressed tubular epithelial cells. Secreted collectin-11 can bind l-
fucose displayed on the outer cell membrane and trigger activation Therapeutic options for targeting the complement
of the lectin pathway. The same mechanism of lectin pathway activa- cascade
tion occurs on tubular epithelial cells injured by myoglobin159. Thus, Eculizumab was the first anti-complement therapeutic to be approved
this process might be a common pathway by which complement is by the FDA for the treatment of a kidney disease (specifically, aHUS) in
activated following tubular injury. 2011. Additional anti-complement drugs have since been approved for
Biomarker evidence also suggests that complement activation non-kidney diseases or are in the late stages of clinical development171–173
occurs in patients with AKI. Urinary factor B levels prior to surgery pre- (Box 2 and Table 2). Multiple anti-complement drugs with potential
dict the development of AKI in patients undergoing cardiac surgery160. applications in nephrology might therefore soon become available.
After surgery, urinary levels of the Ba fragment predict the develop- These drugs are categorically different from other types of immunosup-
ment of AKI161. Of note, C3d is deposited in the tubulointerstitium of pressive drugs and are likely to be beneficial in some kidney diseases
patients with AKI in a pattern similar to that seen in AKI rodent models2. for which other immunomodulatory drugs are ineffective (Fig. 4).
A clinical trial of ravulizumab (an anti-C5 antibody) for prevention of Complement inhibitors might also be useful in combination with
AKI in patients undergoing non-urgent cardiac surgery is ongoing immunosuppressive drugs for antibody-mediated diseases, providing a
(ClinicalTrials.gov NCT05746559). rapid anti-inflammatory effect that is not achieved by cytotoxic agents.
Interestingly, despite kidney transplant recipients being maintained
Polycystic kidney disease. In a mouse model of autosomal-recessive on multiple immunosuppressive medications, complement activation
polycystic kidney disease (PKD), C3 was strongly upregulated in the still contributes to kidney allograft injury in many settings, including
kidneys162 and C3−/− mice had attenuated cystogenesis163. C3 fragments ischaemia–reperfusion injury and antibody-mediated rejection174.
Alternative pathway
Factor B Iptacopan aHUS NCT04889430, NCT05795140
C3G and/or post-Tx recurrence of C3G NCT03955445, NCT03832114c, NCT04817618
IC-MPGN NCT05755386
LN NCT05268289
IgAN NCT03373461c, NCT04557462, NCT04578834
IONI-FB-LRx IgAN NCT04014335
Factor D Danicopan (ALXN2040) C3G, IC-MPGN NCT03124368c, NCT03369236c, NCT03459443c
Vemircopan (ALXN2050) IgAN, LN NCT05097989
C3 Pegcetacoplan TA-TMA NCT05148299
C3G, IC-MPGN NCT05067127, NCT05809531
Post-Tx recurrence of C3G or IC-MPGN NCT04572854
Classical pathway
C1s Sutimlimab (BIVV009)a Cold agglutinin disease NCT05132127c, NCT03347396c, NCT03347422c
C1INH Cinryze, Berinert, Ruconest AMR NCT02547220c, NCT03221842c, NCT01147302c,
NCT01134510c
Ischaemia–reperfusion injury, delayed graft function in NCT02134314c, NCT04696146
kidney transplantation
Lectin pathway
MASP2 Narsoplimab (OMS721) TA-TMA NCT05855083
C3G, IgAN, LN, MN NCT02682407
IgAN NCT03608033
TMA including aHUS NCT02222545c, NCT03205995
Terminal pathway
C5 Eculizumab AMR NCT01399593c
STEC-HUS NCT01410916c
Delayed graft function NCT02145182c
aHUS NCT01193348c, NCT05726916
Ravulizumab (ALXN1210) TMA NCT04743804
TA-TMA NCT04543591, NCT04557735
IgAN, LN NCT04564339
aHUS NCT03131219c
Acute kidney injury following cardiac surgery NCT05746559
Nomacopan (rVA576) b
TA-TMA NCT04784455
IgAN NCT03841448
C5a Vilobelimab ANCA-associated vasculitis NCT03895801c
C5aR1 Avacopan C3G NCT03301467c
ANCA-associated vasculitis NCT02994927c, NCT01363388c, NCT02222155c
IgAN NCT02384317c
aHUS, atypical haemolytic uraemic syndrome; AMR, antibody-mediated rejection; ANCA, antineutrophil cytoplasmic antibody; C3G, C3 glomerulopathy; IC-MPGN, immune complex
membranoproliferative glomerulonephritis; IgAN, immunoglobulin A nephropathy; LN, lupus nephritis; MN, membranous nephropathy; STEC-HUS, Shiga toxin-producing Escherichia coli
infection-associated haemolytic–uraemic syndrome; TA-TMA, transplant-associated thrombotic microangiopathy; TMA, thrombotic microangiopathy; Tx, transplantation. aSutimlimab has not
yet been tested in kidney diseases. bNomacopan inhibits both C5 and leukotriene B4. cCompleted or with reported results.
In addition to mediating inflammation within the kidney directly, and T cell polarization184,185. Given this crosstalk between the comple-
complement activation fragments also have several immunomodula- ment system and the adaptive immune system, anti-complement
tory effects. C3 fragments serve as adjuvants, lower the threshold for therapies might not only affect the innate immune pathways but also
B cell activation183 and affect the activation of T cells by dendritic cells attenuate the upstream autoimmune response that is the primary cause
of some diseases. Nevertheless, the effect of complement inhibitors on for persistent production of C5b–9 in these patients while they were
autoimmunity, particularly in the context of other immunosuppressive receiving eculizumab is unknown.
drugs, remains to be confirmed in clinical studies.
Immunostaining of kidney biopsies for complement proteins
The risks of complement blockade Plasma complement biomarkers are not specific indicators of com-
The primary risk of complement-inhibiting drugs is probably the plement activation in the kidney. However, immunostaining for
increased susceptibility to infection. The overall risk of complement tissue-bound C3 and C4 fragments can provide evidence of comple-
blockade depends on where in the cascade the drug works, the dura- ment activation within the kidney. Moreover, comparison of C3 and C4
tion of treatment, and other clinical characteristics that might predis- deposits indicates which activation pathways are involved. In kidney
pose a patient to infection186. The risk of infection will presumably be transplant recipients, detection of C4d in the peritubular capillaries
even greater when complement inhibitors are combined with other is one of the criteria for diagnosing antibody-mediated rejection4.
immunosuppressive drugs. Of note, complement proteins are fre- Although not covalently bound within the kidney, many other com-
quently consumed in autoimmune disease, which already increases plement proteins can also be detected within glomerular deposits,
the risk of infection in affected patients and might offset the additive including C1q. In an immune complex-mediated disease, deposited
risk of a complement inhibitor187. These considerations must also immunoglobulin activates the classical pathway and is co-deposited with
be weighed against the overall prognosis of the underlying disease C1q, C4d and C3 fragments68,193,194. By contrast, the detection of C3
and the availability of other treatments. Vaccination and the use of deposits in the relative absence of immunoglobulin or C4d suggests
prophylactic antibiotics will probably be essential to mitigate the risk activation of the alternative pathway and is the basis of the C3G
of infection. Moreover, as mentioned above, congenital deficiency of diagnosis48,194. Of note, although detection of complement proteins
classical pathway proteins increases the risk of developing autoim- in the kidney provides useful clinical information, the first two kidney
mune disease and whether therapeutic blockade poses the same risk diseases for which complement inhibitors have been approved — aHUS
remains unknown. As new anti-complement drugs enter the clinic, and AAV — are not characterized by prominent complement deposition
it will be important to determine whether they cause or exacerbate in kidney biopsy samples.
autoimmunity.
Evidence for complement dysregulation
Complement biomarkers Given that the complement cascade requires a delicate balance between
Complement system assessment is currently part of the standard clini- activating and regulatory molecules, acquired or genetic defects that
cal evaluation for many kidney diseases188. Complement proteins and disrupt this balance are important drivers of disease. Molecular studies
fragments are measured in plasma, and kidney biopsy samples are have shown that variants in genes involved in the alternative pathway
routinely immunostained for C3c, C4d and C1q. These analyses help to are strong risk factors for the development of aHUS and C3G45,56,70,195,196.
establish the diagnoses of some kidney diseases and provide informa- Several clinical laboratories now provide genetic analyses for patients
tion about disease activity. Unfortunately, testing is not yet capable of with kidney disease. Genetic studies are not necessary for the diagnosis
accurately identifying which patients will benefit from treatment with of aHUS, but identification of a complement gene variant can provide
complement-inhibiting drugs188. useful prognostic information and aid in clinical decision making192,197.
Acquired causes of complement dysregulation include production
Measurement of complement proteins and fragments of autoantibodies that block the function of factor H51, or production of
Several kidney diseases are associated with decreased levels of C3 and endogenous proteins that antagonize factor H198. Nonetheless, even in
C4, including lupus nephritis, post-infectious glomerulonephritis, patients with these molecular impairments, disease is often triggered
cryoglobulinaemia, C3G and IC-MPGN52,87,189. Decreased levels of C3 by an infection199. This observation suggests that an additional ‘hit’ is
and C4 are interpreted as indicators of disease activity. However, in needed. Infections might increase production of complement proteins
other diseases, complement activation in the kidney causes damage by the liver as they are acute phase reactants. Infections might also alter
without depleting circulating levels of complement proteins. Even in the expression of complement regulatory proteins by injured cells, or
lupus nephritis, which is well known for its association with hypocom- induce the release of complement-activating extracellular vesicles200.
plementaemia, these serological tests do not accurately reflect disease Notably, heterozygous defects in factor H are also strong risk factors
activity within the kidney87. for aHUS. Moreover, once a disease flare has been triggered, plasma
Levels of the complement protein fragments that are released exchange is often insufficient to stop the pathological process49. These
as complement proteins are cleaved and consumed might be a more acquired imbalances in activation or regulation might therefore be
sensitive indicator of complement activation than assessment of C3 irreversible and cannot be attenuated by restoring normal levels of
and C4, and complement fragments have been explored as biomarkers functioning factor H.
of kidney disease88. In patients with MN, urinary C5b–9 levels seem to
correlate with ongoing immune complex formation in the glomerulus Conclusions
and might therefore serve as markers of immunological activity190,191. The kidney is uniquely susceptible to pathological complement acti-
The measurement of complement activation fragments is perhaps vation and abundant clinical evidence suggests that complement is
most useful in aHUS, where tests to monitor complement activity could pathogenic in diseases of widely different aetiologies. Uncontrolled
be used to guide treatment with complement inhibitors. Plasma C5b–9 activation of the alternative pathway is the primary driver of aHUS
levels might be particularly useful for monitoring the use of C5 inhibi- and C3G. The complement system is also activated by immunoglobu-
tors, which block C5b–9 formation. For example, in a study of patients lin and certain pattern recognition molecules, including MBL. These
with aHUS in whom eculizumab was discontinued, elevated C5b–9 complement-activating proteins bind to targets expressed in the
levels at the time of discontinuation predicted relapse192. The reason injured kidney. Importantly, complement regulation is frequently
impaired in the damaged kidney, potentially allowing secondary dis- 18. Garred, P. et al. A journey through the lectin pathway of complement-MBL and beyond.
Immunol. Rev. 274, 74–97 (2016).
ease progression in common diseases such as DKD and FSGS. Given the 19. Roos, A. et al. Glomerular activation of the lectin pathway of complement in IgA
frequent involvement of complement activation in kidney disease, nephropathy is associated with more severe renal disease. J. Am. Soc. Nephrol. 17,
the introduction of anti-complement drugs in the clinic is an important 1724–1734 (2006).
20. Seifert, L. et al. The classical pathway triggers pathogenic complement activation
advance in nephrology. in membranous nephropathy. Nat. Commun. 14, 473 (2023).
Complement inhibitory drugs have been approved for aHUS and 21. Harboe, M., Ulvund, G., Vien, L., Fung, M. & Mollnes, T. E. The quantitative role of
AAV, and are currently being tested in many additional kidney diseases. alternative pathway amplification in classical pathway induced terminal complement
activation. Clin. Exp. Immunol. 138, 439–446 (2004).
These studies have already demonstrated the efficacy and safety of 22. de Boer, E. C. et al. The contribution of the alternative pathway in complement activation
complement inhibition in patients with reduced kidney function. How- on cell surfaces depends on the strength of classical pathway initiation. Clin. Transl.
ever, much about the full potential and limitations of anti-complement Immunol. 12, e1436 (2023).
23. Lachmann, P. J., Lay, E. & Seilly, D. J. Experimental confirmation of the C3 tickover
drugs for the treatment of kidney disease remains unknown. Unfor- hypothesis by studies with an Ab (S77) that inhibits tickover in whole serum. FASEB J. 32,
tunately, many challenges complicate the clinical trials required to 123–129 (2018).
24. Jean, D. et al. A cysteine proteinase, which cleaves human C3, the third component
clarify these open questions, as many of these complement-associated
of complement, is involved in tumorigenicity and metastasis of human melanoma.
kidney diseases are rare and progress slowly. As studies are conducted, Cancer Res. 56, 254–258 (1996).
careful analysis of the results will be required to delineate the effects of 25. Volanakis, J. E., Barnum, S. R., Giddens, M. & Galla, J. H. Renal filtration and catabolism
of complement protein D. N. Engl. J. Med. 312, 395–399 (1985).
anti-complement drugs on kidney inflammation, as well as their effects
26. Zhang, Y. et al. C3(H2O) prevents rescue of complement-mediated C3 glomerulopathy
on the systemic immune response and the risk of infection. In addition, in Cfh−/− Cfd−/− mice. JCI Insight https://doi.org/10.1172/jci.insight.135758 (2020).
the development of predictive biomarkers that can identify patients 27. Jalal, D. et al. Endothelial microparticles and systemic complement activation in patients
with chronic kidney disease. J. Am. Heart Assoc. https://doi.org/10.1161/JAHA.117.007818
most likely to respond to complement inhibition would be invaluable.
(2018).
One final point worth mentioning is the high cost of anti-complement 28. Merle, N. S., Church, S. E., Fremeaux-Bacchi, V. & Roumenina, L. T. Complement system
drugs. The expense of these drugs places a heavy burden on the health part I — molecular mechanisms of activation and regulation. Front. Immunol. 6, 262
(2015).
care system, and they are not available in all countries201. Accurate
29. Brown, K. M. et al. Influence of donor C3 allotype on late renal-transplantation outcome.
complement biomarkers could help to ensure that these drugs are N. Engl. J. Med. 354, 2014–2023 (2006).
only used when needed, and the dose of inhibition could be tailored 30. Pratt, J. R., Basheer, S. A. & Sacks, S. H. Local synthesis of complement component C3
regulates acute renal transplant rejection. Nat. Med. 8, 582–587 (2002).
to achieve full inhibition at the lowest necessary dose202. 31. Lake, B. B. et al. A single-nucleus RNA-sequencing pipeline to decipher the molecular
anatomy and pathophysiology of human kidneys. Nat. Commun. 10, 2832 (2019).
Published online: xx xx xxxx 32. Sircar, M. et al. Complement 7 is up-regulated in human early diabetic kidney disease.
Am. J. Pathol. 188, 2147–2154 (2018).
References 33. Tang, S., Zhou, W., Sheerin, N. S., Vaughan, R. W. & Sacks, S. H. Contribution of renal
1. Ehrlich, P. Collected Studies on Immunity (John Wiley & Sons, 1906). secreted complement C3 to the circulating pool in humans. J. Immunol. 162, 4336–4341
2. Thurman, J. M., Lucia, M. S., Ljubanovic, D. & Holers, V. M. Acute tubular necrosis is (1999).
characterized by activation of the alternative pathway of complement. Kidney Int. 67, 34. West, E. E. & Kemper, C. Complosome — the intracellular complement system. Nat. Rev.
524–530 (2005). Nephrol. 19, 426–439 (2023).
3. Noris, M. et al. Hypocomplementemia discloses genetic predisposition to hemolytic 35. Vandendriessche, S., Cambier, S., Proost, P. & Marques, P. E. Complement receptors and
uremic syndrome and thrombotic thrombocytopenic purpura: role of factor H their role in leukocyte recruitment and phagocytosis. Front. Cell. Dev. Biol. 9, 624025
abnormalities. Italian Registry of Familial and Recurrent Hemolytic Uremic Syndrome/ (2021).
Thrombotic Thrombocytopenic Purpura. J. Am. Soc. Nephrol. 10, 281–293 (1999). 36. Stokowska, A. et al. Complement C3a treatment accelerates recovery after stroke via
4. Loupy, A. et al. The Banff 2019 Kidney Meeting Report (I): updates on and clarification modulation of astrocyte reactivity and cortical connectivity. J. Clin. Invest. https://doi.org/
of criteria for T cell- and antibody-mediated rejection. Am. J. Transplant. 20, 2318–2331 10.1172/JCI162253 (2023).
(2020). 37. Strey, C. W. et al. The proinflammatory mediators C3a and C5a are essential for liver
5. Reis, E. S., Mastellos, D. C., Hajishengallis, G. & Lambris, J. D. New insights into the regeneration. J. Exp. Med. 198, 913–923 (2003).
immune functions of complement. Nat. Rev. Immunol. 19, 503–516 (2019). 38. Serna, M., Giles, J. L., Morgan, B. P. & Bubeck, D. Structural basis of complement
6. Stephan, A. H., Barres, B. A. & Stevens, B. The complement system: an unexpected role membrane attack complex formation. Nat. Commun. 7, 10587 (2016).
in synaptic pruning during development and disease. Annu. Rev. Neurosci. 35, 369–389 39. Zipfel, P. F. & Skerka, C. Complement regulators and inhibitory proteins. Nat. Rev.
(2012). Immunol. 9, 729–740 (2009).
7. Arbore, G. et al. T helper 1 immunity requires complement-driven NLRP3 inflammasome 40. Blom, A. M., Webb, J., Villoutreix, B. O. & Dahlback, B. A cluster of positively charged
activity in CD4+ T cells. Science 352, aad1210 (2016). amino acids in the C4BP α-chain is crucial for C4b binding and factor I cofactor function.
8. Zhang, X. et al. Regulation of Toll-like receptor-mediated inflammatory response J. Biol. Chem. 274, 19237–19245 (1999).
by complement in vivo. Blood 110, 228–236 (2007). 41. Degn, S. E. et al. MAp44, a human protein associated with pattern recognition molecules
9. Huber-Lang, M. et al. Generation of C5a in the absence of C3: a new complement of the complement system and regulating the lectin pathway of complement activation.
activation pathway. Nat. Med. 12, 682–687 (2006). J. Immunol. 183, 7371–7378 (2009).
10. Holers, V. M. Contributions of animal models to mechanistic understandings of 42. Mueller-Ortiz, S. L. et al. Targeted disruption of the gene encoding the murine
antibody-dependent disease and roles of the amplification loop. Immunol. Rev. 313, small subunit of carboxypeptidase N (CPN1) causes susceptibility to C5a
181–193 (2023). anaphylatoxin-mediated shock. J. Immunol. 182, 6533–6539 (2009).
11. Pouw, R. B., Vredevoogd, D. W., Kuijpers, T. W. & Wouters, D. Of mice and men: the factor 43. Lucientes-Continente, L., Marquez-Tirado, B. & Goicoechea de Jorge, E. The factor H
H protein family and complement regulation. Mol. Immunol. 67, 12–20 (2015). protein family: the switchers of the complement alternative pathway. Immunol. Rev. 313,
12. Ebanks, R. O. & Isenman, D. E. Mouse complement component C4 is devoid of classical 25–45 (2023).
pathway C5 convertase subunit activity. Mol. Immunol. 33, 297–309 (1996). 44. Martin Merinero, H. et al. Functional characterization of 105 factor H variants associated
13. Brooimans, R. A. et al. Interleukin 2 mediates stimulation of complement C3 biosynthesis with aHUS: lessons for variant classification. Blood 138, 2185–2201 (2021).
in human proximal tubular epithelial cells. J. Clin. Invest. 88, 379–384 (1991). 45. Servais, A. et al. Primary glomerulonephritis with isolated C3 deposits: a new entity
14. Welch, T. R., Beischel, L. S., Frenzke, M. & Witte, D. Regulated expression of complement which shares common genetic risk factors with haemolytic uraemic syndrome.
factor B in the human kidney. Kidney Int. 50, 521–525 (1996). J. Med. Genet. 44, 193–199 (2007).
15. Zwirner, J., Felber, E., Herzog, V., Riethmuller, G. & Feucht, H. E. Classical pathway 46. Medjeral-Thomas, N. R. et al. Glomerular complement factor H-related protein 5
of complement activation in normal and diseased human glomeruli. Kidney Int. 36, (FHR5) is highly prevalent in C3 glomerulopathy and associated with renal impairment.
1069–1077 (1989). Kidney Int. Rep. 4, 1387–1400 (2019).
16. Dodds, A. W. & Matsushita, M. The phylogeny of the complement system and the origins 47. Medjeral-Thomas, N. R. et al. Circulating complement factor H-related proteins 1 and
of the classical pathway. Immunobiology 212, 233–243 (2007). 5 correlate with disease activity in IgA nephropathy. Kidney Int. 92, 942–952 (2017).
17. Matsushita, M. et al. Origin of the classical complement pathway: Lamprey 48. Goodship, T. H. et al. Atypical hemolytic uremic syndrome and C3 glomerulopathy:
orthologue of mammalian C1q acts as a lectin. Proc. Natl Acad. Sci. USA 101, 10127–10131 conclusions from a “Kidney Disease: Improving Global Outcomes” (KDIGO)
(2004). Controversies Conference. Kidney Int. 91, 539–551 (2017).
49. Noris, M. & Remuzzi, G. Atypical hemolytic-uremic syndrome. N. Engl. J. Med. 361, 83. Cunningham, P. N. & Quigg, R. J. Contrasting roles of complement activation and its
1676–1687 (2009). regulation in membranous nephropathy. J. Am. Soc. Nephrol. 16, 1214–1222 (2005).
50. Savige, J. et al. Retinal disease in the C3 glomerulopathies and the risk of impaired vision. 84. Gao, S., Cui, Z. & Zhao, M. H. Complement C3a and C3a receptor activation mediates
Ophthalmic Genet. 37, 369–376 (2016). podocyte injuries in the mechanism of primary membranous nephropathy. J. Am. Soc.
51. Durey, M. A., Sinha, A., Togarsimalemath, S. K. & Bagga, A. Anti-complement-factor Nephrol. 33, 1742–1756 (2022).
H-associated glomerulopathies. Nat. Rev. Nephrol. 12, 563–578 (2016). 85. Pickering, M. C., Botto, M., Taylor, P. R., Lachmann, P. J. & Walport, M. J. Systemic lupus
52. Donadelli, R. et al. Unraveling the molecular mechanisms underlying complement erythematosus, complement deficiency, and apoptosis. Adv. Immunol. 76, 227–324 (2000).
dysregulation by nephritic factors in C3G and IC-MPGN. Front. Immunol. 9, 2329 (2018). 86. Taylor, P. R. et al. A hierarchical role for classical pathway complement proteins in the
53. Sethi, S. et al. Dense deposit disease associated with monoclonal gammopathy of clearance of apoptotic cells in vivo. J. Exp. Med. 192, 359–366 (2000).
undetermined significance. Am. J. Kidney Dis. 56, 977–982 (2010). 87. Birmingham, D. J. et al. The complex nature of serum C3 and C4 as biomarkers of lupus
54. Schaefer, F. et al. Clinical and genetic predictors of atypical hemolytic uremic syndrome renal flare. Lupus 19, 1272–1280 (2010).
phenotype and outcome. Kidney Int. 94, 408–418 (2018). 88. Kim, A. H. J. et al. Association of blood concentrations of complement split product iC3b
55. Blanc, C. et al. Overall neutralization of complement factor H by autoantibodies in and serum C3 with systemic lupus erythematosus disease activity. Arthritis Rheumatol.
the acute phase of the autoimmune form of atypical hemolytic uremic syndrome. 71, 420–430 (2019).
J. Immunol. 189, 3528–3537 (2012). 89. Martin, M. et al. Plasma C4d correlates with C4d deposition in kidneys and with
56. Martinez-Barricarte, R. et al. Human C3 mutation reveals a mechanism of dense deposit treatment response in lupus nephritis patients. Front. Immunol. 11, 582737 (2020).
disease pathogenesis and provides insights into complement activation and regulation. 90. Jennette, J. C. & Hipp, C. G. Immunohistopathologic evaluation of C1q in 800 renal
J. Clin. Invest. 120, 3702–3712 (2010). biopsy specimens. Am. J. Clin. Pathol. 83, 415–420 (1985).
57. Osborne, A. J. et al. Statistical validation of rare complement variants provides insights 91. Turley, A. J. et al. Spectrum and management of complement immunodeficiencies
into the molecular basis of atypical hemolytic uremic syndrome and C3 glomerulopathy. (excluding hereditary angioedema) across Europe. J. Clin. Immunol. 35, 199–205 (2015).
J. Immunol. 200, 2464–2478 (2018). 92. Elliott, M. K. et al. Effects of complement factor D deficiency on the renal disease of
58. Thurman, J. M. & Harrison, R. A. The susceptibility of the kidney to alternative pathway MRL/lpr mice. Kidney Int. 65, 129–138 (2004).
activation — a hypothesis. Immunol. Rev. https://doi.org/10.1111/imr.13168 (2022). 93. Watanabe, H. et al. Modulation of renal disease in MRL/lpr mice genetically deficient
59. Rondeau, E. et al. The long-acting C5 inhibitor, ravulizumab, is effective and safe in in the alternative complement pathway factor B. J. Immunol. 164, 786–794 (2000).
adult patients with atypical hemolytic uremic syndrome naive to complement inhibitor 94. Pedchenko, V. et al. Molecular architecture of the Goodpasture autoantigen in anti-GBM
treatment. Kidney Int. 97, 1287–1296 (2020). nephritis. N. Engl. J. Med. 363, 343–354 (2010).
60. Legendre, C. M. et al. Terminal complement inhibitor eculizumab in atypical 95. Bowman, C., Ambrus, K. & Lockwood, C. M. Restriction of human IgG subclass
hemolytic-uremic syndrome. N. Engl. J. Med. 368, 2169–2181 (2013). expression in the population of auto-antibodies to glomerular basement membrane.
61. Ruggenenti, P. et al. C5 Convertase blockade in membranoproliferative Clin. Exp. Immunol. 69, 341–349 (1987).
glomerulonephritis: a single-arm clinical trial. Am. J. Kidney Dis. 74, 224–238 (2019). 96. Ma, R. et al. The alternative pathway of complement activation may be involved in the
62. Nasr, S. H. et al. Proliferative glomerulonephritis with monoclonal IgG deposits: a distinct renal damage of human anti-glomerular basement membrane disease. PLoS One 9,
entity mimicking immune-complex glomerulonephritis. Kidney Int. 65, 85–96 (2004). e91250 (2014).
63. Nasr, S. H. et al. Proliferative glomerulonephritis with monoclonal IgG deposits. 97. Quigg, R. J. et al. Blockade of antibody-induced glomerulonephritis with Crry-Ig,
J. Am. Soc. Nephrol. 20, 2055–2064 (2009). a soluble murine complement inhibitor. J. Immunol. 160, 4553–4560 (1998).
64. Nasr, S. H. et al. Dysproteinemia, proteinuria, and glomerulonephritis. Kidney Int. 69, 98. van Daalen, E. E. et al. Predicting outcome in patients with anti-GBM glomerulonephritis.
772–775 (2006). Clin. J. Am. Soc. Nephrol. 13, 63–72 (2018).
65. Ravindran, A., Fervenza, F. C., Smith, R. J. H. & Sethi, S. C3 glomerulopathy associated 99. Nithagon, P. et al. Eculizumab and complement activation in anti-glomerular basement
with monoclonal Ig is a distinct subtype. Kidney Int. 94, 178–186 (2018). membrane disease. Kidney Int. Rep. 6, 2713–2717 (2021).
66. Meri, S., Koistinen, V., Miettinen, A., Tornroth, T. & Seppala, I. J. Activation of 100. Haas, M. Histology and immunohistology of IgA nephropathy. J. Nephrol. 18, 676–680
the alternative pathway of complement by monoclonal lambda light chains in (2005).
membranoproliferative glomerulonephritis. J. Exp. Med. 175, 939–950 (1992). 101. Kim, S. J. et al. Decreased circulating C3 levels and mesangial C3 deposition predict
67. Servais, A. et al. Acquired and genetic complement abnormalities play a critical role in renal outcome in patients with IgA nephropathy. PLoS One 7, e40495 (2012).
dense deposit disease and other C3 glomerulopathies. Kidney Int. 82, 454–464 (2012). 102. Hiemstra, P. S., Gorter, A., Stuurman, M. E., Van Es, L. A. & Daha, M. R. Activation of the
68. Sethi, S., Nasr, S. H., De Vriese, A. S. & Fervenza, F. C. C4d as a diagnostic tool in alternative pathway of complement by human serum IgA. Eur. J. Immunol. 17, 321–326
proliferative GN. J. Am. Soc. Nephrol. 26, 2852–2859 (2015). (1987).
69. Hou, J. et al. Toward a working definition of C3 glomerulopathy by immunofluorescence. 103. Suzuki, H. et al. IgA1-secreting cell lines from patients with IgA nephropathy produce
Kidney Int. 85, 450–456 (2014). aberrantly glycosylated IgA1. J. Clin. Invest. 118, 629–639 (2008).
70. Iatropoulos, P. et al. Complement gene variants determine the risk of immunoglobulin- 104. Suzuki, H. et al. Aberrantly glycosylated IgA1 in IgA nephropathy patients is recognized
associated MPGN and C3 glomerulopathy and predict long-term renal outcome. by IgG antibodies with restricted heterogeneity. J. Clin. Invest. 119, 1668–1677 (2009).
Mol. Immunol. 71, 131–142 (2016). 105. Rizk, D. V. et al. Glomerular immunodeposits of patients with IgA nephropathy are
71. Diebolder, C. A. et al. Complement is activated by IgG hexamers assembled at the cell enriched for IgG autoantibodies specific for galactose-deficient IgA1. J. Am. Soc.
surface. Science 343, 1260–1263 (2014). Nephrol. 30, 2017–2026 (2019).
72. Ding, Y. et al. The spectrum of C4d deposition in renal biopsies of lupus nephritis 106. Espinosa, M. et al. Association of C4d deposition with clinical outcomes in IgA
patients. Front. Immunol. 12, 654652 (2021). nephropathy. Clin. J. Am. Soc. Nephrol. 9, 897–904 (2014).
73. Verroust, P. J., Wilson, C. B., Cooper, N. R., Edgington, T. S. & Dixon, F. J. Glomerular 107. Tan, L. et al. A multicenter, prospective, observational study to determine association
complement components in human glomerulonephritis. J. Clin. Invest. 53, 77–84 of mesangial C1q deposition with renal outcomes in IgA nephropathy. Sci. Rep. 11, 5467
(1974). (2021).
74. Ma, H., Sandor, D. G. & Beck, L. H. Jr The role of complement in membranous 108. Jiang, Y. et al. Glomerular C4d deposition and kidney disease progression in IgA
nephropathy. Semin. Nephrol. 33, 531–542 (2013). nephropathy: a systematic review and meta-analysis. Kidney Med. 3, 1014–1021 (2021).
75. Tomas, N. M. et al. Autoantibodies against thrombospondin type 1 domain-containing 7A 109. Wu, L. et al. Immunofluorescence deposits in the mesangial area and glomerular
induce membranous nephropathy. J. Clin. Invest. 126, 2519–2532 (2016). capillary loops did not affect the prognosis of immunoglobulin a nephropathy except
76. Murtas, C. et al. Coexistence of different circulating anti-podocyte antibodies in C1q:a single-center retrospective study. BMC Nephrol. 22, 43 (2021).
membranous nephropathy. Clin. J. Am. Soc. Nephrol. 7, 1394–1400 (2012). 110. Evans, D. J. et al. Glomerular deposition of properdin in Henoch-Schonlein syndrome
77. van der Zee, J. S., van Swieten, P. & Aalberse, R. C. Inhibition of complement activation and idiopathic focal nephritis. Br. Med. J. 3, 326–328 (1973).
by IgG4 antibodies. Clin. Exp. Immunol. 64, 415–422 (1986). 111. Chiu, Y. L. et al. Alternative complement pathway is activated and associated with
78. Val-Bernal, J. F., Garijo, M. F., Val, D., Rodrigo, E. & Arias, M. C4d immunohistochemical galactose-deficient IgA1 antibody in IgA nephropathy patients. Front. Immunol. 12,
staining is a sensitive method to confirm immunoreactant deposition in formalin-fixed 638309 (2021).
paraffin-embedded tissue in membranous glomerulonephritis. Histol. Histopathol. 26, 112. Holmes, L. V. et al. Determining the population frequency of the CFHR3/CFHR1 deletion
1391–1397 (2011). at 1q32. PLoS One 8, e60352 (2013).
79. Haddad, G. et al. Altered glycosylation of IgG4 promotes lectin complement pathway 113. Xie, J. et al. Fine mapping implicates a deletion of CFHR1 and CFHR3 in protection from
activation in anti-PLA2R1-associated membranous nephropathy. J. Clin. Invest. 131, IgA nephropathy in Han Chinese. J. Am. Soc. Nephrol. 27, 3187–3194 (2016).
https://doi.org/10.1172/JCI140453 (2021). 114. Kiryluk, K. et al. Discovery of new risk loci for IgA nephropathy implicates genes involved
80. Bally, S. et al. Phospholipase A2 receptor-related membranous nephropathy and in immunity against intestinal pathogens. Nat. Genet. 46, 1187–1196 (2014).
mannan-binding lectin deficiency. J. Am. Soc. Nephrol. 27, 3539–3544 (2016). 115. Murphy, B., Georgiou, T., Machet, D., Hill, P. & McRae, J. Factor H-related protein-5: a novel
81. Lhotta, K., Wurzner, R., Rumpelt, H. J., Eder, P. & Mayer, G. Membranous nephropathy in a component of human glomerular immune deposits. Am. J. Kidney Dis. 39, 24–27 (2002).
patient with hereditary complete complement C4 deficiency. Nephrol. Dial. Transpl. 19, 116. Medjeral-Thomas, N. R. et al. Progressive IgA nephropathy is associated with low
990–993 (2004). circulating mannan-binding lectin-associated serine protease-3 (MASP-3) and increased
82. Baker, P. J. et al. Depletion of C6 prevents development of proteinuria in experimental glomerular factor H-related protein-5 (FHR5) deposition. Kidney Int. Rep. 3, 426–438
membranous nephropathy in rats. Am. J. Pathol. 135, 185–194 (1989). (2018).
117. Tortajada, A. et al. Elevated factor H-related protein 1 and factor H pathogenic 150. Huang, J. et al. Complement activation profile of patients with primary focal segmental
variants decrease complement regulation in IgA nephropathy. Kidney Int. 92, 953–963 glomerulosclerosis. PLoS One 15, e0234934 (2020).
(2017). 151. Lenderink, A. M. et al. The alternative pathway of complement is activated in the
118. Zhai, Y. L. et al. Rare variants in the complement factor H-related protein 5 gene glomeruli and tubulointerstitium of mice with adriamycin nephropathy. Am. J. Physiol.
contribute to genetic susceptibility to IgA nephropathy. J. Am. Soc. Nephrol. 27, Renal Physiol. 293, F555–F564 (2007).
2894–2905 (2016). 152. Turnberg, D. et al. Complement activation contributes to both glomerular and
119. Harris, A. A., Falk, R. J. & Jennette, J. C. Crescentic glomerulonephritis with a paucity tubulointerstitial damage in adriamycin nephropathy in mice. J. Immunol. 177,
of glomerular immunoglobulin localization. Am. J. Kidney Dis. 32, 179–184 (1998). 4094–4102 (2006).
120. Xiao, H., Schreiber, A., Heeringa, P., Falk, R. J. & Jennette, J. C. Alternative complement 153. Hisano, S. et al. Clinicopathologic correlation and outcome of C1q nephropathy.
pathway in the pathogenesis of disease mediated by anti-neutrophil cytoplasmic Clin. J. Am. Soc. Nephrol. 3, 1637–1643 (2008).
autoantibodies. Am. J. Pathol. 170, 52–64 (2007). 154. Vizjak, A. et al. Pathology, clinical presentations, and outcomes of C1q nephropathy.
121. Xiao, H. et al. C5a receptor (CD88) blockade protects against MPO-ANCA GN. J. Am. Soc. Nephrol. 19, 2237–2244 (2008).
J. Am. Soc. Nephrol. 25, 225–231 (2014). 155. Gunasekara, V. N., Sebire, N. J. & Tullus, K. C1q nephropathy in children: clinical
122. Haas, M. & Eustace, J. A. Immune complex deposits in ANCA-associated crescentic characteristics and outcome. Pediatr. Nephrol. 29, 407–413 (2014).
glomerulonephritis: a study of 126 cases. Kidney Int. 65, 2145–2152 (2004). 156. Thurman, J. M., Ljubanovic, D., Edelstein, C. L., Gilkeson, G. S. & Holers, V. M. Lack of a
123. Yuan, J. et al. C5a and its receptors in human anti-neutrophil cytoplasmic antibody functional alternative complement pathway ameliorates ischemic acute renal failure in
(ANCA)-associated vasculitis. Arthritis Res. Ther. 14, R140 (2012). mice. J. Immunol. 170, 1517–1523 (2003).
124. Gou, S. J., Yuan, J., Wang, C., Zhao, M. H. & Chen, M. Alternative complement pathway 157. Thurman, J. M. et al. Altered renal tubular expression of the complement inhibitor Crry
activation products in urine and kidneys of patients with ANCA-associated GN. permits complement activation after ischemia/reperfusion. J. Clin. Invest. 116, 357–368
Clin. J. Am. Soc. Nephrol. 8, 1884–1891 (2013). (2006).
125. Gou, S. J., Yuan, J., Chen, M., Yu, F. & Zhao, M. H. Circulating complement activation in 158. Farrar, C. A. et al. Collectin-11 detects stress-induced L-fucose pattern to trigger renal
patients with anti-neutrophil cytoplasmic antibody-associated vasculitis. Kidney Int. 83, epithelial injury. J. Clin. Invest. 126, 1911–1925 (2016).
129–137 (2013). 159. Boudhabhay, I. et al. Complement activation is a crucial driver of acute kidney injury
126. Jayne, D. R. W., Merkel, P. A., Schall, T. J., Bekker, P. & Group, A. S. Avacopan for the in rhabdomyolysis. Kidney Int. 99, 581–597 (2021).
treatment of ANCA-associated vasculitis. N. Engl. J. Med. 384, 599–609 (2021). 160. Merchant, M. L., Brier, M. E., Slaughter, M. S., Klein, J. B. & McLeish, K. R. Biomarker
127. Jayne, D. R. W. et al. Randomized trial of C5a receptor inhibitor avacopan in enhanced risk prediction for development of AKI after cardiac surgery. BMC Nephrol.
ANCA-associated vasculitis. J. Am. Soc. Nephrol. 28, 2756–2767 (2017). 19, 102 (2018).
128. Stone, J. H. et al. Rituximab versus cyclophosphamide for ANCA-associated vasculitis. 161. Laskowski, J., Philbrook, H. T., Parikh, C. R. & Thurman, J. M. Urine complement
N. Engl. J. Med. 363, 221–232 (2010). activation fragments are increased in patients with kidney injury after cardiac surgery.
129. Jones, R. B. et al. Rituximab versus cyclophosphamide in ANCA-associated renal Am. J. Physiol. Renal Physiol. 317, F650–F657 (2019).
vasculitis. N. Engl. J. Med. 363, 211–220 (2010). 162. Mrug, M. et al. Overexpression of innate immune response genes in a model of recessive
130. Jiao, Y. et al. Activation of complement C1q and C3 in glomeruli might accelerate the polycystic kidney disease. Kidney Int. 73, 63–76 (2008).
progression of diabetic nephropathy: evidence from transcriptomic data and renal 163. Zhou, J. et al. Kidney injury accelerates cystogenesis via pathways modulated by heme
histopathology. J. Diabetes Investig. 13, 839–849 (2022). oxygenase and complement. J. Am. Soc. Nephrol. 23, 1161–1171 (2012).
131. Duan, S. et al. Association of glomerular complement C4c deposition with the 164. Mrug, M. et al. Complement C3 activation in cyst fluid and urine from autosomal
progression of diabetic kidney disease in patients with type 2 diabetes. Front. Immunol. dominant polycystic kidney disease patients. J. Intern. Med. 276, 539–540 (2014).
11, 2073 (2020). 165. Ichida, S., Yuzawa, Y., Okada, H., Yoshioka, K. & Matsuo, S. Localization of the
132. Fortpied, J., Vertommen, D. & Van Schaftingen, E. Binding of mannose-binding lectin complement regulatory proteins in the normal human kidney. Kidney Int. 46, 89–96
to fructosamines: a potential link between hyperglycaemia and complement activation (1994).
in diabetes. Diabetes Metab. Res. Rev. 26, 254–260 (2010). 166. Cosio, F. G., Sedmak, D. D., Mahan, J. D. & Nahman, N. S. Jr. Localization of decay
133. Ostergaard, J. et al. Mannose-binding lectin deficiency attenuates renal changes in a accelerating factor in normal and diseased kidneys. Kidney Int. 36, 100–107 (1989).
streptozotocin-induced model of type 1 diabetes in mice. Diabetologia 50, 1541–1549 167. Endoh, M. et al. Immunohistochemical demonstration of membrane cofactor protein
(2007). (MCP) of complement in normal and diseased kidney tissues. Clin. Exp. Immunol. 94,
134. Ostergaard, J. A. et al. Mannan-binding lectin in diabetic kidney disease: the impact 182–188 (1993).
of mouse genetics in a type 1 diabetes model. Exp. Diabetes Res. 2012, 678381 (2012). 168. Baker, P. J., Adler, S., Yang, Y. & Couser, W. G. Complement activation by heat-killed
135. Ostergaard, J. A. et al. Diabetes-induced changes in mannan-binding lectin levels and human kidney cells: formation, activity, and stabilization of cell-bound C3 convertases.
complement activation in a mouse model of type 1 diabetes. Scand. J. Immunol. 77, J. Immunol. 133, 877–881 (1984).
187–194 (2013). 169. Tang, S., Sheerin, N. S., Zhou, W., Brown, Z. & Sacks, S. H. Apical proteins stimulate
136. Saraheimo, M. et al. Increased levels of mannan-binding lectin in type 1 diabetic patients complement synthesis by cultured human proximal tubular epithelial cells. J. Am.
with incipient and overt nephropathy. Diabetologia 48, 198–202 (2005). Soc. Nephrol. 10, 69–76 (1999).
137. Hansen, T. K. et al. Association between mannose-binding lectin, high-sensitivity 170. Farrar, C. A., Zhou, W., Lin, T. & Sacks, S. H. Local extravascular pool of C3 is a
C-reactive protein and the progression of diabetic nephropathy in type 1 diabetes. determinant of postischemic acute renal failure. FASEB J. 20, 217–226 (2006).
Diabetologia 53, 1517–1524 (2010). 171. Roth, A. et al. Sutimlimab in cold agglutinin disease. N. Engl. J. Med. 384, 1323–1334
138. Lu, H., Deng, S., Zheng, M. & Hu, K. iTRAQ plasma proteomics analysis for candidate (2021).
biomarkers of type 2 incipient diabetic nephropathy. Clin. Proteom. 16, 33 (2019). 172. Hillmen, P. et al. Pegcetacoplan versus eculizumab in paroxysmal nocturnal
139. Qin, X. et al. Glycation inactivation of the complement regulatory protein CD59: hemoglobinuria. N. Engl. J. Med. 384, 1028–1037 (2021).
a possible role in the pathogenesis of the vascular complications of human diabetes. 173. Harris, C. L. Expanding horizons in complement drug discovery: challenges and
Diabetes 53, 2653–2661 (2004). emerging strategies. Semin. Immunopathol. 40, 125–140 (2018).
140. Acosta, J. et al. Molecular basis for a link between complement and the vascular 174. Stites, E., Le Quintrec, M. & Thurman, J. M. The complement system and
complications of diabetes. Proc. Natl Acad. Sci. USA 97, 5450–5455 (2000). antibody-mediated transplant rejection. J. Immunol. 195, 5525–5531 (2015).
141. Ghosh, P. et al. Plasma glycated CD59, a novel biomarker for detection of 175. Khaled, S. K. et al. Narsoplimab, a Mannan-binding lectin-associated serine protease-2
pregnancy-induced glucose intolerance. Diabetes Care 40, 981–984 (2017). inhibitor, for the treatment of adult hematopoietic stem-cell transplantation-associated
142. Angeletti, A. et al. Loss of decay-accelerating factor triggers podocyte injury and thrombotic microangiopathy. J. Clin. Oncol. 40, 2447–2457 (2022).
glomerulosclerosis. J. Exp. Med. https://doi.org/10.1084/jem.20191699 (2020). 176. Bomback, A. S. et al. Alternative complement pathway inhibition with iptacopan for the
143. Valoti, E. et al. Impact of a complement factor H gene variant on renal dysfunction, treatment of C3 glomerulopathy-study design of the APPEAR-C3G trial. Kidney Int. Rep.
cardiovascular events, and response to ACE inhibitor therapy in type 2 diabetes. 7, 2150–2159 (2022).
Front. Genet. 10, 681 (2019). 177. Hasturk, H. et al. Phase IIa clinical trial of complement C3 inhibitor AMY-101 in adults with
144. Kopp, J. B. et al. Podocytopathies. Nat. Rev. Dis. Prim. 6, 68 (2020). periodontal inflammation. J. Clin. Invest. https://doi.org/10.1172/JCI152973 (2021).
145. Zhang, Y. M. et al. Clinical significance of IgM and C3 glomerular deposition in primary 178. Li, C., Li, H., Wen, Y. B., Li, X. M. & Li, X. W. Analysis of predictive factors for
focal segmental glomerulosclerosis. Clin. J. Am. Soc. Nephrol. 11, 1582–1589 (2016). immunosuppressive response in anti-phospholipase A2 receptor antibody positive
146. Trachtman, H. et al. Natural antibody and complement activation characterize patients membranous nephropathy. BMC Nephrol. 19, 354 (2018).
with idiopathic nephrotic syndrome. Am. J. Physiol. Renal Physiol. 321, F505–F516 179. Bech, A. P., Hofstra, J. M., Brenchley, P. E. & Wetzels, J. F. Association of anti-PLA2R
(2021). antibodies with outcomes after immunosuppressive therapy in idiopathic membranous
147. Strassheim, D. et al. IgM contributes to glomerular injury in FSGS. J. Am. Soc. Nephrol. nephropathy. Clin. J. Am. Soc. Nephrol. 9, 1386–1392 (2014).
24, 393–406 (2013). 180. Robson, J. et al. Glucocorticoid treatment and damage in the anti-neutrophil cytoplasm
148. van de Lest, N. A. et al. Glomerular C4d deposition can precede the development antibody-associated vasculitides: long-term data from the European Vasculitis Study
of focal segmental glomerulosclerosis. Kidney Int. 96, 738–749 (2019). Group trials. Rheumatology 54, 471–481 (2015).
149. Thurman, J. M. et al. Complement activation in patients with focal segmental 181. Stojan, G. & Petri, M. The risk benefit ratio of glucocorticoids in SLE: have things changed
glomerulosclerosis. PLoS One 10, e0136558 (2015). over the past 40 years? Curr. Treatm. Opt. Rheumatol. 3, 164–172 (2017).
182. Morigi, M. et al. C3a receptor blockade protects podocytes from injury in diabetic 206. Smith, R. J. et al. New approaches to the treatment of dense deposit disease.
nephropathy. JCI Insight https://doi.org/10.1172/jci.insight.131849 (2020). J. Am. Soc. Nephrol. 18, 2447–2456 (2007).
183. Carter, R. H., Spycher, M. O., Ng, Y. C., Hoffman, R. & Fearon, D. T. Synergistic interaction 207. Kavanagh, D., Goodship, T. H. & Richards, A. Atypical hemolytic uremic syndrome.
between complement receptor type 2 and membrane IgM on B lymphocytes. Semin. Nephrol. 33, 508–530 (2013).
J. Immunol. 141, 457–463 (1988). 208. Nasr, S. H. et al. Dense deposit disease: clinicopathologic study of 32 pediatric and adult
184. Strainic, M. G., Shevach, E. M., An, F., Lin, F. & Medof, M. E. Absence of signaling into CD4+ patients. Clin. J. Am. Soc. Nephrol. 4, 22–32 (2009).
cells via C3aR and C5aR enables autoinductive TGF-β1 signaling and induction of Foxp3+ 209. Fakhouri, F. et al. Insights from the use in clinical practice of eculizumab in adult patients
regulatory T cells. Nat. Immunol. 14, 162–171 (2013). with atypical hemolytic uremic syndrome affecting the native kidneys: an analysis of
185. Heeger, P. S. et al. Decay-accelerating factor modulates induction of T cell immunity. 19 cases. Am. J. Kidney Dis. 63, 40–48 (2014).
J. Exp. Med. 201, 1523–1530 (2005). 210. Zuber, J. et al. New insights into postrenal transplant hemolytic uremic syndrome.
186. Bomback, A. S. et al. Improving clinical trials for anticomplement therapies in Nat. Rev. Nephrol. 7, 23–35 (2011).
complement-mediated glomerulopathies: report of a scientific workshop sponsored 211. Misra, A., Peethambaram, A. & Garg, A. Clinical features and metabolic and autoimmune
by the National Kidney Foundation. Am. J. Kidney Dis. 79, 570–581 (2022). derangements in acquired partial lipodystrophy: report of 35 cases and review of the
187. Skattum, L., Martensson, U. & Sjoholm, A. G. Hypocomplementaemia caused by literature. Medicine 83, 18–34 (2004).
C3 nephritic factors (C3 NeF): clinical findings and the coincidence of C3 NeF type II 212. Dalvin, L. A., Fervenza, F. C., Sethi, S. & Pulido, J. S. Shedding light on Fundus Drusen
with anti-C1q autoantibodies. J. Intern. Med. 242, 455–464 (1997). Associated with Membranoproliferative Glomerulonephritis: breaking stereotypes of
188. Thurman, J. M. & Fremeaux-Bacchi, V. Alternative pathway diagnostics. Immunol. Rev. types I, II, and III. Retin. Cases Brief. Rep. https://doi.org/10.1097/ICB.0000000000000164
313, 225–238 (2023). (2015).
189. Wyatt, R. J., Forristal, J., West, C. D., Sugimoto, S. & Curd, J. G. Complement profiles 213. Noris, M. et al. Dynamics of complement activation in aHUS and how to monitor
in acute post-streptococcal glomerulonephritis. Pediatr. Nephrol. 2, 219–223 (1988). eculizumab therapy. Blood 124, 1715–1726 (2014).
190. Brenchley, P. E. et al. Urinary C3dg and C5b-9 indicate active immune disease in human 214. Chauvet, S. et al. Results from a national-wide retrospective cohort measure the impact
membranous nephropathy. Kidney Int. 41, 933–937 (1992). of C3 and soluble C5b-9 levels on kidney outcomes in C3 glomerulopathy. Kidney Int.
191. Coupes, B. M., Kon, S. P., Brenchley, P. E., Short, C. D. & Mallick, N. P. The temporal https://doi.org/10.1016/j.kint.2022.05.027 (2022).
relationship between urinary C5b-9 and C3dg and clinical parameters in human 215. Zhang, Y. et al. Factor H autoantibodies and complement-mediated diseases.
membranous nephropathy. Nephrol. Dial. Transplant. 8, 397–401 (1993). Front. Immunol. 11, 607211 (2020).
192. Fakhouri, F. et al. Eculizumab discontinuation in children and adults with atypical 216. Corvillo, F. et al. Nephritic factors: an overview of classification, diagnostic tools and
hemolytic-uremic syndrome: a prospective multicenter study. Blood 137, 2438–2449 clinical associations. Front. Immunol. 10, 886 (2019).
(2021). 217. Chauvet, S. et al. Both monoclonal and polyclonal immunoglobulin contingents mediate
193. Hill, G. S., Hinglais, N., Tron, F. & Bach, J. F. Systemic lupus erythematosus. Morphologic complement activation in monoclonal gammopathy associated-C3 glomerulopathy.
correlations with immunologic and clinical data at the time of biopsy. Am. J. Med. 64, Front. Immunol. 9, 2260 (2018).
61–79 (1978). 218. Schubart, A. et al. Small-molecule factor B inhibitor for the treatment of
194. Drachenberg, C. B. et al. Epidemiology and pathophysiology of glomerular C4d staining complement-mediated diseases. Proc. Natl Acad. Sci. USA 116, 7926–7931 (2019).
in native kidney biopsies. Kidney Int. Rep. 4, 1555–1567 (2019). 219. Zelek, W. M., Xie, L., Morgan, B. P. & Harris, C. L. Compendium of current complement
195. Fremeaux-Bacchi, V. et al. Genetics and outcome of atypical hemolytic uremic therapeutics. Mol. Immunol. 114, 341–352 (2019).
syndrome: a nationwide French series comparing children and adults. Clin. J. Am. Soc.
Nephrol. 8, 554–562 (2013). Author contributions
196. Caprioli, J. et al. Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical All authors researched data for the article, made substantial contributions to discussions
presentation, response to treatment, and outcome. Blood 108, 1267–1279 (2006). of the content and wrote, reviewed or edited the manuscript before submission.
197. Le Quintrec, M. et al. Complement genes strongly predict recurrence and graft
outcome in adult renal transplant recipients with atypical hemolytic and uremic Competing interests
syndrome. Am. J. Transplant. 13, 663–675 (2013). J.M.T. is a consultant for Q32 Bio, Inc., which is a company developing complement inhibitors.
198. Renner, B. et al. Annexin A2 enhances complement activation by inhibiting factor H. He holds stock and will receive royalty income from Q32 Bio, Inc. He co-founded and
J. Immunol. 196, 1355–1365 (2016). holds stock in Compsit3, a company developing complement diagnostics. V.P. declares
199. Noris, M. et al. Relative role of genetic complement abnormalities in sporadic and no competing interests.
familial aHUS and their impact on clinical phenotype. Clin. J. Am. Soc. Nephrol. 5,
1844–1859 (2010). Additional information
200. Karasu, E., Eisenhardt, S. U., Harant, J. & Huber-Lang, M. Extracellular vesicles: packages Peer review information Nature Reviews Nephrology thanks A. Blom, R. Quigg and R. Taylor
sent with complement. Front. Immunol. 9, 721 (2018). for their contribution to the peer review of this work.
201. Sethi, S. K. et al. Eculizumab for atypical hemolytic-uremic syndrome in India: first report
from India and the challenges faced. Indian J. Nephrol. 27, 58–61 (2017). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims
202. Sridharan, M., Go, R. S. & Willrich, M. A. V. Clinical utility and potential cost savings in published maps and institutional affiliations.
of pharmacologic monitoring of eculizumab for complement-mediated thrombotic
microangiopathy. Mayo Clin. Proc. Innov. Qual. Outcomes 6, 458–464 (2022). Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this
203. Fakhouri, F., Zuber, J., Fremeaux-Bacchi, V. & Loirat, C. Haemolytic uraemic syndrome. article under a publishing agreement with the author(s) or other rightsholder(s); author
Lancet 390, 681–696 (2017). self-archiving of the accepted manuscript version of this article is solely governed by the
204. Smith, R. J. H. et al. C3 glomerulopathy — understanding a rare complement-driven renal terms of such publishing agreement and applicable law.
disease. Nat. Rev. Nephrol. 15, 129–143 (2019).
205. Yan, K., Desai, K., Gullapalli, L., Druyts, E. & Balijepalli, C. Epidemiology of atypical hemolytic © Springer Nature Limited 2023
uremic syndrome: a systematic literature review. Clin. Epidemiol. 12, 295–305 (2020).