Skip to main content
Erschienen in: Seminars in Immunopathology 1/2018

Open Access 11.01.2018 | Review

Diseases of complement dysregulation—an overview

verfasst von: Edwin K. S. Wong, David Kavanagh

Erschienen in: Seminars in Immunopathology | Ausgabe 1/2018

Abstract

Atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy (C3G), and paroxysmal nocturnal hemoglobinuria (PNH) are prototypical disorders of complement dysregulation. Although complement overactivation is common to all, cell surface alternative pathway dysregulation (aHUS), fluid phase alternative pathway dysregulation (C3G), or terminal pathway dysregulation (PNH) predominates resulting in the very different phenotypes seen in these diseases. The mechanism underlying the dysregulation also varies with predominant acquired autoimmune (C3G), somatic mutations (PNH), or inherited germline mutations (aHUS) predisposing to disease. Eculizumab has revolutionized the treatment of PNH and aHUS although has been less successful in C3G. With the next generation of complement therapeutic in late stage development, these archetypal complement diseases will provide the initial targets.
Hinweise
This article is a contribution to the special issue on Complement in Health and Disease: Novel Aspects and Insights - Guest Editors: Paul Morgan and David Kavanagh

Introduction

Atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathy (C3G), and paroxysmal nocturnal hemoglobinuria (PNH) serve as exemplars of the mechanisms by which complement dysregulation may cause disease. In this review, we shall compare and contrast the underlying pathophysiological mechanisms and the response to treatments.

Atypical HUS

Classification of atypical hemolytic uremic syndrome

The last 20 years has seen striking advances in our understanding of the molecular mechanisms underlying thrombotic microangiopathies (TMAs) and with this has come a complex and rapidly evolving nomenclature [1]. Historically, TMAs were categorized on clinical findings: HUS for renal dominant disease, thrombotic thrombocytopenic purpura (TTP) for predominant neurological involvement. Subsequently, TTP was defined by severe ADAMTS13 deficiency; HUS caused by shiga toxin-producing Escherichia coli (STEC) defined as STEC-HUS, with aHUS broadly used for all other causes of TMA. With the discovery of genetic and acquired complement dysregulation in a proportion of patients with aHUS, the term complement-mediated aHUS was used to refer to this subgroup. When reviewing historical literature, “aHUS” may refer specifically to complement-mediated TMA, or be more loosely applied to any TMA that is not TTP or STEC-HUS (reviewed [1]). In this review, we use the term complement-mediated aHUS when the etiology is defined as such, and use aHUS where etiology is ill defined. Current classifications describe acquired primary TMAs, inherited primary TMAs, secondary TMAs, and infection-associated TMAs (Table 1) although it should be borne in mind that underlying complement genetic predispositions often require a secondary trigger for TMA to manifest. The role of complement in secondary TMAs and infection associated TMA is yet to be defined (Fig. 1).
Table 1
Classification of thrombotic microangiopathies
Primary TMA: hereditary
 aHUS with complement gene mutation
  (CFH; CFI; CFB; C3; CD46; CFHR1 hybrid)
 TTP with ADAMTS13 mutation
 MMACHC TMA
 DGKE TMA
Primary TMA: hereditary
 aHUS with complement autoantibodies
  (anti-FH; anti-FI)
 TTP with ADAMTS13 autoantibody
Secondary TMAs
 TMA with glomerular disease
  (FSGS; IgAN, C3G/MPGN, MN, AAV)
 Malignancy associated TMA
 Drug induced TMA
  Direct toxicity (interferon B; bevacizumab)
  Immune mediated damage (e.g., quinine)
 TMA with autoimmune conditions
  (SLE, SRC, CAPS)
 De novo TMA after solid organ transplant
 HELLP
Infection associated TMA
 STEC-HUS
 Pneumococcal HUS
 HIV associated aHUS
 Other
AAV ANCA (anti-neutrophil cytoplasmic antibody) associated vasculitis; ADAMTS13 a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13; aHUS atypical hemolytic uremic syndrome; C3G C3 glomerulopathy; CAPS catastrophic antiphospholipid syndrome; MMACHC Methylmalonic aciduria and homocystinuria, cblC type; DGKE gene encoding diacylglycerol kinase Ɛ; FH factor H; FI factor I, FSGS focal segmental glomerulosclerosis; HELLP syndrome of hemolysis, elevated liver enzymes, and low platelets; HIV human immunodeficiency virus; HUS hemolytic uraemic syndrome; IgAN IgA nephropathy; MN membranous nephropathy; MPGN membranoproliferative glomerulonephritis; SLE systemic lupus erythematosus; SRC scleroderma renal crisis; STEC, shiga toxin-producing Escherichia coli; TMA thrombotic microangiopathy; TTP thrombotic thrombocytopenic purpura

Pathology

The pathological findings seen in complement-mediated aHUS reflect tissue responses to endothelial injury: endothelial swelling and mesangiolysis in active lesions, double contours of the basement membrane in chronic lesions (reviewed [2]). The absence of overt platelet fibrin thrombosis from renal biopsies of TMA has recently led to a suggested reclassification to microangiopathy +/− thrombosis [2].

Inherited primary complement-mediated aHUS

First described in 1998 by Warwicker et al. [3], mutations in factor H (CFH) are the commonest cause of inherited complement-mediated aHUS, accounting for around 25% of cases [4]. Factor H (FH) is the most important fluid-phase regulator of the alternative pathway (AP) of complement. FH is composed of 20 complement control protein modules (CCPs), also known as short consensus repeats (SCRs). The four N-terminal CCPs (CCPs 1–4) mediate the complement regulatory functions of the protein by competing with factor B for C3b binding, accelerating the decay of the C3 convertase into its components, and acting as a cofactor for factor I-mediated proteolytic inactivation of C3b [5, 6]. The vast majority of CFH mutations seen in complement-mediated aHUS do not occur in this region, but instead in the C terminal domains (CCP 19–20) [4]. It is this region which mediates FH self-surface binding via its interaction with C3b, sialic acid, and glycosaminoglycans [7, 8]. In complement-mediated aHUS, the mutations are usually heterozygous, do not result in a quantitative deficiency of FH but instead have variable consequences on binding to GAGs, sialic acid, and C3b which impairs cell surface complement regulation [9, 10] (reviewed4).
In addition to point mutations, its location in the RCA cluster makes CFH particularly prone to genomic rearrangements. This is an area of the genome that arose from several large genomic duplications, and these low copy repeats can cause genome instability in this region. The CFH mutations S1191L, V1197A, and combined S1191L/V1197A arose through gene conversion between CFHR1 and CFH [11]. A hybrid (fusion) gene comprising the 21 N-terminal exons of CFH and the 2 C terminal exons of CFHR1 was demonstrated to have arisen through nonallelic homologous recombination and resulted in complement-mediated aHUS [12]. More recently, several other hybrid genes consisting of the N-terminal exons of CFH and the 5 C-terminal exons of CFHR3 have been reported [13, 14]. As with C-terminal point mutations in CFH, these hybrid genes also result in loss of cell surface complement regulation.

Membrane cofactor protein

Membrane cofactor protein (MCP; CD46) is a surface-bound complement regulatory protein that acts as a cofactor for the factor I (FI) mediated cleavage of C3b and C4b that are deposited on host cells [15]. Mutations in CD46 are the second commonest cause of complement-mediated aHUS accounting for around 15% of patients.
The majority of mutations are found in the extracellular domains of CD46 that are responsible for C3b and C4b binding. Unlike CFH, most CD46 mutations result in a quantitative defect in CD46 (~ 75%) [4].

Complement factor I

Complement factor I is a serum serine protease, which functions as a critical mediator of complement regulation by cleaving C3b and C4b in the presence of its cofactors (FH for C3b; C4BP (C4b binding protein) for C4b, CD46 and CR1 (complement receptor 1) for both [4].
Around 10% of complement-mediated aHUS is predisposed to by mutations in the FI gene (CFI) [16]. The CFI mutations described in complement-mediated aHUS are all heterozygous.

Complement C3

C3 is the central component of the complement cascade. C3 is cleaved to form the anaphylatoxin C3a and C3b, which is highly reactive and can bind to cell surfaces via its reactive thioester. C3b can then interact with factor B (FB) in the presence of factor D to form the alternative pathway convertase introducing a positive-feedback loop.
Mutations in C3 account for around 2–10% of complement-mediated aHUS. The mutations in C3 linked to complement-mediated aHUS result in complement over activation by either (1) preventing complement regulators binding to C3 and inactivating it or by binding to FB with greater affinity [1719]. These mutations result in increased complement activation on platelets and glomerular endothelium.

Complement factor B

Factor B carries the catalytic site of the complement AP convertase (C3bBb). Mutations in CFB are very rare in complement-mediated aHUS. As with C3 mutations, complement over activation occurs by impaired complement regulation or increased convertase formation [20]. These mutations have been demonstrated to increase complement deposition on endothelial cells.

Thrombomodulin

Thrombomodulin (THBD) plays a key role in regulating clot formation by the activation of protein C by thrombin and enhancing thrombin-mediated activation of plasma procarboxypeptidase B (CPB2), an inhibitor of fibrinolysis. Procarboxypeptidase B also inactivates complement-derived anaphylatoxins C3a and C5a. THBD has also been suggested to have a role in the regulation of the AP by accelerating FI-mediated inactivation of C3b [21].
Rare genetic variants have been described in some aHUS cohorts but their causality remains to be established [21].

Common genetic susceptibility factors

In addition to rare mutations, a number of common single nucleotide polymorphisms (SNPs) in CD46 and CFH have been associated with complement-mediated aHUS. A CD46 haplotype (CD46ggaac) block has been associated with a two- to threefold increased risk of complement-mediated aHUS [2224]. This encompasses 2 SNPs in the promoter region of CD46, and reporter gene assays have suggested that this haplotype reduces transcriptional activity by 25% albeit without decreased CD46 cell surface expression in vivo [24, 25]. A CFH haplotype (CFH-H3; tgtgt) has been shown to increase the risk of complement-mediated aHUS two- to fourfold [22, 26]. This risk haplotype contains a SNP CFH-Val62 which has a subtle decrease in cofactor activity compared to the protective variant [27, 28].

Acquired primary complement-mediated aHUS

Acquired defects in complement regulation have been seen in the form of autoantibodies to FH. First reported in 2005, these account for ~ 10% of complement-mediated aHUS [29]. It predominantly presents in childhood, frequently with a gastrointestinal prodrome [30].
There is a strong association with a homozygous deletion of CFHR3 and CFHR1, which encodes complement factor H-related proteins (FHR) 3 and 1 [31]. Subsequently, FHR1 deficiency resulting from point mutations in CFHR1 or from a deletion incorporating CFHR1 and CFHR4 has been reported in individuals with FH autoantibody-mediated aHUS [32]. Although this suggests a key role in the deficiency of FHR1 in the generation of FH autoantibodies, the mechanism remains obscure and several patients have been reported with factor H autoantibodies in the absence of the CFHR1/CFHR3 deletion.
The majority of autoantibodies bind to the C-terminal domain of FH, thus mimicking the defects see in the inherited form. The antibodies have been shown to reduce binding to C3b and other C3 fragments [33, 34]. They perturb FH-mediated cell surface protection and in some individuals the autoantibodies also impair cofactor activity or decay accelerating activity [33, 34].
Autoantibodies against FI have also been reported, but are rare and their functional relevance remains to be established [35].

Clinical presentation and outcome

The incidence of aHUS is ~ 0.4/million population [36]. Hemolysis and ischemic organ injury, predominantly in the kidney, define the clinical presentation of aHUS [1]. Extra-renal manifestations (cardiac, ocular, pancreatic) are reported although it is not known whether they are a direct consequence of the TMA, a direct effect of complement activation, or relate to the associated severe hypertension and uremia [4].
Historically, the prognosis for patients with aHUS was poor and in the pre-eculizumab era at 3–5 years after onset, 36–48% [22, 37] of children and 64–67%, [37] of adults had died or reached ESRD. The underlying genetic cause predicted the outcome of disease with those carrying CD46 mutations having the best prognosis (3 year renal survival 94%). In individuals with CFH mutations, up to 77% of patients had developed ESRD or had died at 3–5 years. Only 30–40% of individuals with CFI and C3 mutations will be alive with native kidney function at 3–5 years, [4, 37]. In those with FH-autoantibodies, 36.5–63% die or reach ESRD over a similar timescale.
The outcome following renal transplantation was also poor and again the outcome was predicted largely by the underlying genetic abnormality. The overall recurrence was 68% and 5-year death-censored graft survival 51% with highest risk associated with CFH, CFB, and C3 mutations and the lowest with CD46 mutations. [38, 39]

Disease penetrance

The genetic mutations seen in complement-mediated aHUS are not causative but are instead predisposing, suggesting that additional genetic and environmental modifiers are important. Penetrance of disease is age related and has been reported to be as high as 64% by the age of 70 for individuals carrying a single genetic mutation [2]. A small proportion of aHUS patients (~ 3%) will have more than one mutation with increased penetrance per additional mutation [40]. Risk haplotypes have also been shown to increase disease penetrance. Even where an individual has multiple genetic risk factors, a trigger is frequently required (e.g., infections [41], pregnancy [42]) to unmask a latent complement defect. These triggers usually activate complement: complement activation is the normal physiological response to infection and occurs in the placenta in normal pregnancy.

Eculizumab in aHUS

The elucidation of the role of complement in disease provided the rationale for the use of eculizumab in complement-mediated aHUS. Eculizumab, a recombinant humanized monoclonal antibody directed against C5, blocks the cleavage of C5 into its effector components C5a and C5b. Landmark studies in primary aHUS published in 2013 demonstrated its efficacy which have been replicated in subsequent extension studies [43], prospective (non-randomized) studies [44, 45], and cohort analysis [36]. The role of eculizumab in secondary aHUS remains to be established (reviewed [46]).
In individuals requiring a renal transplant with a diagnosis of primary complement-mediated aHUS, the high recurrence rate following transplantation necessities pre-emptive eculizumab [2, 39].
The optimal length of treatment with eculizumab for individuals presenting with complement-mediated aHUS is unclear although the current license for eculizumab is for lifelong treatment; eculizumab withdrawal has been reported in a large series of aHUS patients with relapse reported in around one third of patients, all carrying complement mutations [47]. In those patients experiencing a relapse post withdrawal, rapid reintroduction of complement inhibition normalized the renal function. This suggests that a disease-driven intermittent regime could be used although prospective trials are required.

Eculizumab non-responsive aHUS

There is no biomarker currently that will confirm the diagnosis of a primary complement-mediated aHUS in the acute setting, and the diagnosis is therefore one of exclusion. As early initiation of eculizumab has been shown to lead to better outcomes, treatment is often commenced in patients with suspected primary complement-mediated aHUS, and discontinued if an alternative etiology is subsequently identified. With the increasing use of eculizumab in clinical practice, it has become apparent that there are subgroups of aHUS that do not respond to eculizumab. In the recent pediatric trial, Greenbaum et al. highlighted that for those with a rare genetic variant in the complement system or an autoantibody to FH, all had an improvement in estimated glomerular filtration rate (eGFR), while 27% of individuals without an identified complement abnormality failed to show an improvement [45]. It is not clear whether this represents late presentation of disease or true non-response.
More recently, individuals presenting with a TMA with failure to respond to eculizumab have been demonstrated to have genetic variants in the non-complement genes DGKE [48], INF2 [49], and MMACHC [50].

DGKE

Diacylglycerol kinase epsilon (DGKE) is a lipid kinase that catalyzes the phosphorylation of diacylglycerol substrates (DAGs) to phosphatidic acid. Recessive mutations causing aHUS were first reported in 2013 [48]. The exact mechanisms resulting in the TMA are yet to be fully elucidated; however, loss of DGKE function results in enhanced signaling through arachidonic acid-containing DAGs (AADAGs) and enhanced activation of PKC (protein kinase C). In the endothelium, PKC activation results in upregulation of prothrombotic factors and the downregulation of VEGFR2 signaling and these may play a role.
Only a small number of cases have been published, but it appears to present aged < 1 year and commonly results in progressive chronic kidney disease (CKD) and ESRF [37]. There is insufficient evidence to determine optimal management; there are reports of both response and non-response to eculizumab. Concomitant mutations in complement genes have been reported. Genetic pleiotropy is seen: DGKE mutations have also been associated with mesangioproliferative glomerulonephritis (MPGN) [51] .

Inverted formin 2

Mutations in inverted formin 2 (INF2) have recently been reported in families with TMAs which was non-responsive to eculizumab [49]. INF2 is a ubiquitously expressed formin protein which accelerates actin polymerization and depolymerization, thus regulating a range of cytoskeleton dependent cellular functions including the secretory pathway. As with DGKE, genetic pleiotropism is also seen with most individuals with INF2 mutations presenting with focal segmental glomerulosclerosis (FSGS) and nephrotic syndrome. It remains to be seen whether this is a primary aHUS or secondary phenomenon in association with FSGS.

Methylmalonic aciduria and homocystinuria, cobalamin C (cblC) type

Homozygous or compound heterozygous mutations in the MMACHC gene result in a disorder of cobalamin (cbl; vitamin B12) metabolism that causes aHUS. Although the pathophysiologic mechanisms that result in endothelial damage are unclear, metabolic therapy with hydroxycobalamin is very effective at preventing disease. It appears that MMACHC-mediated aHUS is complement independent as the small number of published reports of eculizumab use describe non-response [50].

Polymorphisms in C5 and the use of Coversin to treat TMA

A rare polymorphism in C5 (p.R885H) has been reported in the Japanese population which prevented eculizumab binding [52]. More recently, a European with a functionally significant CFH mutation (p.D1119G) and a TMA post-bone marrow transplant was shown to carry this SNP preventing the use of eculizumab [53]. In this case, an alternative C5 inhibitor, Coversin, was used. This is a recombinant protein derived from the tick, Ornithodoros moubata. As this binds to a different epitope on C5, this completely blocked the terminal pathway and there appeared to be a clinical response although there was a limited supply of the drug and the patient died [53].

C3 Glomerulopathy

Introduction

C3 glomerulopathy (C3G) is a recently identified pathological entity describing a group of diseases in which uncontrolled complement activation can lead to complement deposition within the glomerulus [54]. Historically, uncontrolled complement activation has been associated with the disease, membranoproliferative glomerulonephritis (MPGN) [55, 56]. These diseases are ultra-rare, affecting ~ 1 per million population [5759].

Classification and pathology of C3G

The classification of diseases in uncontrolled complement activation resulting in glomerulopathy continues to evolve. The term C3G was introduced to alert the clinician to the possibility of an underlying abnormality of the complement system [54]. C3G can be further classified by the pattern of dense deposits on electron microscopy. Dense deposit disease (DDD) is a specific form of C3G that is classified by the presence of dense osmiophilic intramembranous deposits seen on electron microscopy. C3 glomerulonephritis (C3GN) are forms of C3G in which deposits on electron microscopy may be light dense, amorphous mesangial, paramesangial, subendothelial, or subepithelial.
A diagnosis of C3G requires a renal biopsy. The presence of C3 dominant staining on immunofluorescence (IF), with an intensity of at least two orders of magnitude greater than any other immunoreactant (IgG, IgM, IgA, and C1q) provides the best sensitivity and specificity for C3G [60]. This current definition of C3G captures about 90% of cases of DDD, and possibly fewer cases of C3GN [60]. Once C3G has been diagnosed on renal biopsy, light microscopy then identifies diverse patterns of glomerular injury that include MPGN [54, 60], and detects additional features such as crescentic disease or markers of chronic disease such as interstitial fibrosis and tubular atrophy.
MPGN is a pattern of glomerular injury characterized by the presence of mesangial expansion, cellular proliferation, and double-contouring of the glomerular basement membrane [61]. The classification of MPGN was historically based upon the position of electron dense deposits relative to the glomerular basement membrane. Three types of MPGN were defined in this manner, type 1 (subendothelial), type 2 or DDD (intramembranous), and type 3 (subendothelial and subepithelial) [62]. Type 1 and type 3 MPGN typically stained positive for immunoglobulins and C3 on IF whereas type 2 MPGN typically stained for C3 only. MPGN of all types were previously associated with uncontrolled complement activation.

Overlap of C3G and MPGN

The classifications of C3G and MPGN overlap [63]. The new classification of C3G includes cases of MPGN that were previously classified as type 1, 2, or 3. Furthermore, the definition of C3G identifies cases without MPGN, in which uncontrolled complement activation might not have been recognized [64]. Evidence of uncontrolled complement activation continues to be found in cases of MPGN that do not meet the diagnostic criteria of C3G [6567]. Currently, these cases are termed immune-complex MPGN. Therefore, suspicion of uncontrolled complement activation should include the current definition of C3G and an overlap with cases of immune-complex MPGN (Fig. 2).

Immune-mediated glomerulonephritis

Although uncontrolled complement activation may result in immune-complex MPGN, other secondary causes of MPGN should also be considered. These causes are summarized [68]. Treatment can then be directed at the secondary cause.

Monoclonal gammopathy of renal significance

The absence of immunoglobulin on renal biopsy (in the setting of C3 deposition) is the hallmark of a diagnosis of C3G, usually indicative of an underlying disorder of complement regulation. However, it is increasingly recognized that deposition of monotypic immunoglobulin may be masked [69], and only identified following pronase digest. Upon identification of monoclonal deposits on the renal biopsy, the possibility of a monoclonal gammopathy needs to be considered. Treatment of these conditions is usually directed at the cause of the monoclonal gammopathy [70].

Complement derangement in C3G and MPGN

A range of acquired and inherited abnormalities have been described as a cause of complement dysregulation in cases of C3G and MPGN. Unlike in aHUS, these abnormalities tend to result in fluid phase dysregulation of AP and acquired abnormalities predominate.

FH deficiency

FH deficiency leads to uncontrolled activation of AP in the fluid phase [71, 72] and causes MPGN and C3G [56, 64, 73]. Genetic causes of FH deficiency are summarized in Table 2. Briefly, FH deficiency can be quantitative and due to mutations in CFH, inherited in homozygosity or heterozygosity, resulting in complete or partial FH deficiency, respectively. Other pathogenic mutations in CFH lead to a functional deficiency of FH due to the expression of proteins that have defective binding to C3b resulting in impairment of fluid phase complement regulation [80, 81, 83]. Overall, the prevalence of rare genetic variants in these cohorts ranges from 4 to 16.2% of patients [65, 66, 87]. The functional significance of a number of other rare genetic variants reported in these case series of MPGN or C3G is not known (Table 2). [96]
Table 2
Rare genetic variants in CFH, CFI, CD46, C3, and CFB reported in C3G and MPGN
Gene
Effect
Variant
Gene
Effect
Variant
CFH
Complete FH deficiency (homozygous)
P88T*
[66, 74]
CFH
VUS (normal FH levels)
P26S
£
R127L*
[73]
ΔG122-E128
[65]
C431S
[65, 73]
D130N
[65]
C597R
[65]
A161S
[65]
P621Y
[75]
IVS11 + 5
[65]
C673S
[73]
G334A
£
Y899X
[76]
G650V
[64], £
Y1008X
[66]
F717L
[65]
W1096R*
£
H878Y
2
Partial FH deficiency
P76X
[64]
A892V
3
L77X
[65]
R1210V
[65]
V143I
[65]
VUS (FH levels not known)
R127C
[66]
I216T
[77]
S199G/E1172X
£
R232X
[65], £
C431S
£
C673R
[65]
N516K
[78]
K768X
[76]
V609I
£
C1043X
[65]
M725X
[79]
Functional FH deficiency (homozygous)
R78G
[66]
V837I/E1145D
[78]
ΔK224*
[80]
Q950H
[66]
R53C
[65, 81]
T956M
[78] [82] [66]
Functional FH deficiency
R83S*
[83]
C3
Gain of function
Δ923-924DG*
[84]
R1210C
[64] [66]
I756T*
[85]
R53C
[65] [86]
R161W
[87]
CD46
VUS
K66N
[66, 88]
VUS
R148Q
[87]
V181M
[65]
A443S
[87]
CFI
FI deficiency
G119R
[65, 8991]
L1100P
[87]
A240G
[65, 92, 93]
L1318R
[87]
C309R
[65]
V86I
[66]
C327R
[65]
R505C
[66]
VUS
c.1-4C > T
[66]
V619M
[66]
G57D
[66]
G637R
[66]
None
G261D
[65, 79, 91, 94]
R1042Q
[66]
I306S
[65]
S1063N
[66]
CFB
Gain of function
I242L
[87]
R1303H
[66]
VUS
D279E
[87]
R1320Q
[66]
S367R*
[95]
D1362N
[66]
G161R
[66]
C1518R
[66]
H451R
[66]
D1625H
[66]
R679W
[66]
None
K1051M
[66]
All variants heterozygous except where indicated
VUS variant of uncertain significance, FH complement factor H, FI complement factor I. * reported in familial disease, £ unpublished, Δ amino acid deletion

FHR proteins

Recent studies suggest an important role of FHR proteins in complement regulation. FHR proteins may compete with FH for C3b binding and prevent the regulatory activities of FH on surface-bound C3b [97]. SCR1 and 2 of FHR1, 2, and 5 have a very high degree of sequence homology (Fig. 3) and share a dimerization motif. These studies show that these FHR proteins exist in dimeric form. FHR proteins do not have complement regulatory domains and as a result, dimeric forms of FHRs with high avidity to C3b act as competitive antagonists, preventing the normal regulatory function of FH and therefore “deregulating FH” [97].

Genomic abnormalities in C3G

First identified in a large Cypriot pedigree [98], a number of genomic abnormalities in the RCA cluster have been described in familial cases of C3G (Table 3). In all cases, the genomic abnormality resulted in the formation of a larger CFHR gene resulting in FHR proteins with additional SCRs [97105]. Functional study of dimeric forms of abnormal FHR proteins demonstrated an enhanced ability to compete with FH resulting in enhanced deregulation of FH as a possible mechanism of disease [97, 102]. In the case of the FHR2-FHR5 hybrid protein reported by Chen et al., functional studies showed evidence of stabilization of C3bBb in the presence of the hybrid FHR2-FHR5 protein resulting in complement activation in the fluid phase [99] and a possible role in binding properdin [100].
Table 3
Abnormal FHR proteins described in C3G
Abnormal FHR
protein
Phenotype
Effect
Reference
FHR212FHR51–9
DDD
Stabilizes C3bBb
[99, 100]
FHR512FHR51–9
CFHR5 nephropathy
De-regulates FH
[97, 98]
FHR512FHR21–4
C3GN
Not known
[101]
FHR11234FHR11–5
Low C3
De-regulates FH
[102]
FHR312FHR11–5
C3GN
De-regulates FH
[97, 103]
FHR1123FHR51–9
DDD/C3GN overlap
De-regulates FH
[104]
FHR512FHR21–4
C3GN
De-regulates FH
[105]
Abnormal FHR protein—the subscript indicates the SCR of each FHR protein that form the abnormal FHR protein
FHR factor H-related, DDD dense deposit disease, C3GN C3 glomerulonephritis, C3bBb C3 convertase of the alternative pathway, FH factor H
Rare genetic variants in CFHR5 have also been reported in C3GN and include C269R [106] and N216P [79]. The functional significance of both variants has yet to be determined.

Complement factor I and membrane cofactor

Rare genetic variants in other complement regulators (CFI and CD46) are infrequently identified in C3G and MPGN (Table 2). Reported variants have all been inherited in heterozygosity. A few (resulting in low FI levels [65, 89, 90]) are pathogenic but their importance in disease pathogenesis has not been established.

Components of the AP C3 convertase—C3 and FB

Detailed functional studies of several familial rare genetic variants in C3 in C3G have been described. In a case of familial DDD, the variant Δ923-924DG does not undergo conformational change to C3b but does form a C3 convertase that is resistant to decay by FH [84]. In a report of familial C3GN, the variant I756T results in defective C3b inactivation by FI in setting of cofactors CR1 and FH [85].
Rare genetic variants in C3 and CFB have also been described in cohorts of C3G and MPGN [87] [66] and includes the S367R variant in CFB in one familial case of C3GN [95]. Most of these variants have not been functionally studied (Table 2).

Polymorphisms

Common genetic susceptibility factors have been reported in MPGN and C3G. In DDD, the Y402H polymorphism in CFH [65, 107, 108] associates with an increased risk of disease. Conversely, the V62I polymorphism was shown to be protective against DDD [26, 66].
Risk haplotypes in CFH also associate with disease. The H1 haplotype that carries the at-risk Y402H SNP in DDD associates with an increased risk of DDD [26, 107]. The H2 haplotype that carries the protective SNP V62I was shown to be protective in DDD [26].
Common SNPs and haplotypes in CD46 have been studied in cohorts of C3G and MPGN. The intronic SNP c.-652G was protective in MPGN and C3GN. This association was also observed in cases of MPGN type 1 and C3GN, in which the haplotype CD46AAGGT was observed more frequently, while the haplotype CD46GAGGT was observed less frequently [65]. These findings were not observed in a later study [66]. In this later study, the SNPs c.-366A>G and c. *783 T>C were observed more frequently in immune-complex MPGN than controls [66]. In the same later study, the SNP c.-366A was observed more frequently in DDD compared to controls [66].
Several common SNPs in C3 and CFB have been studied in C3G and MPGN. The SNPs, R102G [108, 109], and P314L in C3 are associated with DDD [108]. The SNPs in CFB, R32W, and R32Q were not associated with C3G or MPGN [66].

Acquired abnormalities in C3G and MPGN

The first acquired abnormality discovered in MPGN was a circulating factor in serum that was found to increase cell lysis in the fluid phase [55]. This was later discovered to be IgG that stabilized C3bBb by 10-fold [110, 111] now known as C3 nephritic factor. There is a strong association of C3 nephritic factor with all forms of C3G and MPGN, and is prevalent in up to 80% of DDD and 50% of MPGN and C3GN [65, 78]. C3 nephritic factors are not specific to MPGN and C3G and have been observed in acquired partial lipodystrophy [112] and in normal individuals [113].
Autoantibodies to individual complement components and their regulators have been reported in cases of C3G and MPGN. Autoantibodies to FB were first described in DDD [78, 114]. In one patient, these were shown to stabilize C3bBb, causing C3 consumption and terminal pathway activation [114]. No functional data are available in a further report in which three DDD patients with autoantibodies to FB are described [78]. Autoantibodies to C3b and FB in the same patient were described in two cases of DDD. These patients lacked C3 nephritic factor but the antibodies enhanced C3bBb activity [115]. Autoantibodies in FB and C3b have recently been reported in a cohort of C3G and MPGN [67]. Functional studies of purified IgG from patients in this cohort also result in AP activation.
Autoantibodies to FH have also been described in patients with C3G and MPGN. These bind predominantly to the N-terminal domain of FH and impair the regulatory activity of FH [78, 116]. The prevalence of autoantibodies to FH was 11% in a MPGN/C3G cohort [117]. In this cohort, patients with autoantibodies to FH did not associate with homozygous deletion of CFHR3/1. These autoantibodies associated with C3 nephritic factor in children and monoclonal gammopathy in adults [117]. The association of autoantibodies to FH and monoclonal gammopathy had been previously described in case reports [118, 119].

Clinical presentation and outcome of C3G and MPGN

MPGN and C3G, especially the subset of patients with DDD, are typically diseases of childhood and young adulthood [57, 65]. They present with similar clinical features that include proteinuria, hematuria, and renal failure [57, 65]. Renal failure is progressive and 40% of patients develop end-stage renal disease (ESRD) at 10 years [57, 65, 120]. Recurrence in transplantation is common in all types, ranging from 30 to 40% in MPGN type 1 to 80–90% in DDD [121]. Plasma C3 levels are often low [57, 65, 66, 122]. Individual complement abnormalities have not been associated with a greater risk of adverse outcomes [57, 65] although the absence of a C3 nephritic factor or a rare genetic variant did have a higher risk of progression to ESRD in an Italian cohort [66].
There is an association of C3G and MPGN with extra-renal manifestations: acquired partial lipodystrophy [123, 124] and drusen [125].
The clinical features of patients with FHR5 nephropathy, a subset of patients with C3G, differ from other forms of C3G. First reported in large pedigrees with Cypriot ancestry, these patients often have Synpharyngitic hematuria. Prognosis is often worse in males with FHR5 nephropathy [98].

Treatments in C3G and MPGN

There are no universally effective treatments for C3G or MPGN. The only double blind randomized control trial in this group of patients was performed in 1992 [126]. Eighty children (with MPGN types 1, 2, and 3) were randomized to receive 40 mg/m2 of prednisolone on alternate days. In this study, long-term treatment with prednisolone did appear to improve the outcome of patients with MPGN. Other studies suggest some benefit from the use of cyclophosphamide [127], mycophenolate mofetil (MMF) [128, 129] and the combination of aspirin and dipyridamole [130132]. The use of rituximab has been described in case reports only [133135].

Complement inhibition

Unlike in aHUS, the role of complement inhibition in C3G and MPGN is not clear, despite the role of uncontrolled complement activation in disease. To date, there is one clinical trial of eculizumab in MPGN that is currently underway [136]. Nonetheless, eculizumab use has been reported in a growing number of cases of C3G and MPGN. Current reports suggest a more beneficial role for eculizumab in cases where prominent terminal pathway activity is seen [137141].
Other reported therapies that modulate complement include the replacement of FH in patients with a functional deficiency of FH using plasma exchange [142] and soluble CR1 [143]. Further studies are required to determine whether complement inhibition in C3G can be effective.

Paroxysmal nocturnal hemoglobinuria

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hemolytic anemia first described in 1882 [144]. In addition to hemolysis, thrombosis, muscle dystonias, chronic kidney disease, and bone marrow failure may occur [145]. The incidence of PNH is ~ 1–1.5 cases per million individuals worldwide [146]. Most patients present between the ages of 30–59 [146] and it is rare in children [147].

Genetics

Unlike the germline mutations described commonly in aHUS and less commonly in C3G, PNH is caused by somatic mutations in the phosphatidylinositol glycan anchor biosynthesis class A gene (PIGA) in one or more long-lasting hematopoietic stem cell (HSC) clones [148150]. The majority of mutations seen are indels resulting in frameshifts [151, 152]. PIGA’s location on the X chromosome accounts for the ability of one somatic mutation to cause PNH as only one allele is functional in men and women.
The expansion of PIGA-deficient HSC clones is central to the clinical phenotype of PNH; however, the PIGA mutations do not themselves confer a growth advantage [153, 154]. Both intrinsic (additive genetic and epigenetic variations [155]) and extrinsic (bone marrow failure [156]) clonal mechanisms have been suggested to account for the clonal expansion (reviewed [145]).
Although there are multiple genes involved in GPI synthesis, there is only one case of PNH reported where PIGA mutations were not seen. In this case, one somatic mutation in addition to a germline mutation in PIGT caused lack of GPI anchored cells and the PNH phenotype [157, 158].

Pathogenesis

PIGA encodes for a glycosyl transferase that is required in the biosynthetic pathway for the synthesis of glycosyl phosphatidylinositol (GPI) [159]. PIGA mutations lead to a deficiency of GPI-anchored proteins including CD14, CD16b, CD48, and the complement inhibitor proteins CD55 (decay accelerating factor; DAF) and CD59.
CD55 accelerates the decay of the alternative and classical pathway C3 and C5 convertases while CD59 is a terminal pathway regulator which binds C8 preventing C9 recruitment and formation of the membrane attack complex. Deficiency of these complement regulators is critical to PNH erythrocytes being susceptible to complement-mediated attack.
In addition to the dominant hemolytic clinical feature of PNH, thrombosis is commonly seen. The exact mechanisms of thrombosis in PNH are unclear (reviewed by Hill et al. [160]) although the interplay of the complement and coagulation cascades, particularly C5 receptor signaling pathways, activation of platelets and intravascular hemolysis have been suggested.

Clinical presentation and outcome

In PNH-affected erythrocytes, the constant activation of the AP causes a chronic low-level hemolysis and at time of infections or other complement triggering events there may be a hemolytic attack [161]. The most common cause of mortality of PNH is thromboembolism, with venous thrombosis more common than arterial thrombosis [160]. Smooth muscle dystonias including back pain, abdominal pain, erectile dysfunction, and dysphagia are seen [162]. Chronic kidney disease can occur with hemosiderin deposition leading to tubulointerstitial inflammation [163, 164]. Bone marrow failure is commonly seen in PNH but is not a consequence of the somatic mutations in PIGA [145].
Historically, the median survival of patients with PNH was approximately 10 years [165167]; however, since the introduction of eculizumab, in those without bone marrow failure, a normal lifespan can be expected [146, 168].

Eculizumab in PNH

Eculizumab is currently the only licensed therapy for PNH. Its introduction resulted in improvement in intravascular hemolysis [169, 170], thrombosis [171], renal function [172], and survival [168]. Variable response to treatment is seen with some patients presenting with residual hemolysis and requiring red blood cell transfusions. Low-level extravascular hemolysis is seen in most PNH patients on eculizumab. In this setting, C3 fragments opsonizing erythrocytes are recognized by macrophages in the spleen and liver resulting in their destruction [173]. It has recently been shown that a polymorphism in Complement receptor 1 (CR1) alters the level of CR1 on erythrocytes and consequently the level of C3 opsonization of erythrocytes [174]. This increases the clearance of the erythrocytes.
As seen in aHUS, a polymorphism in C5 can limit eculizumab’s effectiveness and complete blockage should be confirmed (AH50/CH50/sC5b-9) [52].

Novel complement inhibitors in PNH

Several novel complement inhibitors are in various stages of development for the treatment of PNH including the C5 inhibitors ALXN1210 (Alexion; NCT02946463, NCT03056040), RA101495 (RaPharma NCT03030183, NCT03078582), ALNCC5 (NCT02352493 Alnylam), Coversin (NCT02591862; Akari), the C3 inhibitor compstatin analogue APL-2 (NCT02588833; Apellis), and the factor D inhibitor ACH-4471 (Achillion). These are reviewed in more detail in this issue by Harris [175].

Summary

Together, these classical diseases of complement dysregulation provide a window on the vastly different phenotypes that can result from the subtle variations in complement regulation. These diseases will provide the test bed for the next generation of complement inhibitory agents.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Goodship TH, Cook HT, Fakhouri F, Fervenza FC, Fremeaux-Bacchi V, Kavanagh D et al (2017) Atypical hemolytic uremic syndrome and C3 glomerulopathy: conclusions from a “Kidney Disease: Improving Global Outcomes” (KDIGO) Controversies Conference. Kidney Int 91:539–551PubMedCrossRef Goodship TH, Cook HT, Fakhouri F, Fervenza FC, Fremeaux-Bacchi V, Kavanagh D et al (2017) Atypical hemolytic uremic syndrome and C3 glomerulopathy: conclusions from a “Kidney Disease: Improving Global Outcomes” (KDIGO) Controversies Conference. Kidney Int 91:539–551PubMedCrossRef
3.
Zurück zum Zitat Warwicker P, Goodship TH, Donne RL, Pirson Y, Nicholls A, Ward RM et al (1998) Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int 53:836–844PubMedCrossRef Warwicker P, Goodship TH, Donne RL, Pirson Y, Nicholls A, Ward RM et al (1998) Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int 53:836–844PubMedCrossRef
5.
Zurück zum Zitat Richards A, Kavanagh D, Atkinson JP (2007) Inherited complement regulatory protein deficiency predisposes to human disease in acute injury and chronic inflammatory states the examples of vascular damage in atypical hemolytic uremic syndrome and debris accumulation in age-related macular degeneration. Adv Immunol 96:141–177PubMedCrossRef Richards A, Kavanagh D, Atkinson JP (2007) Inherited complement regulatory protein deficiency predisposes to human disease in acute injury and chronic inflammatory states the examples of vascular damage in atypical hemolytic uremic syndrome and debris accumulation in age-related macular degeneration. Adv Immunol 96:141–177PubMedCrossRef
6.
Zurück zum Zitat Schmidt CQ, Herbert AP, Kavanagh D, Gandy C, Fenton CJ, Blaum BS et al (2008) A new map of glycosaminoglycan and C3b binding sites on factor H. J Immunol 181:2610–2619PubMedCrossRef Schmidt CQ, Herbert AP, Kavanagh D, Gandy C, Fenton CJ, Blaum BS et al (2008) A new map of glycosaminoglycan and C3b binding sites on factor H. J Immunol 181:2610–2619PubMedCrossRef
7.
Zurück zum Zitat Blaum BS, Hannan JP, Herbert AP, Kavanagh D, Uhrin D, Stehle T (2015) Structural basis for sialic acid-mediated self-recognition by complement factor H. Nat Chem Biol 11:77–82PubMedCrossRef Blaum BS, Hannan JP, Herbert AP, Kavanagh D, Uhrin D, Stehle T (2015) Structural basis for sialic acid-mediated self-recognition by complement factor H. Nat Chem Biol 11:77–82PubMedCrossRef
8.
Zurück zum Zitat Morgan HP, Schmidt CQ, Guariento M, Blaum BS, Gillespie D, Herbert AP et al (2011) Structural basis for engagement by complement factor H of C3b on a self surface. Nat Struct Mol Biol 18:463–470PubMedPubMedCentralCrossRef Morgan HP, Schmidt CQ, Guariento M, Blaum BS, Gillespie D, Herbert AP et al (2011) Structural basis for engagement by complement factor H of C3b on a self surface. Nat Struct Mol Biol 18:463–470PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Ferreira VP, Herbert AP, Cortes C, McKee KA, Blaum BS, Esswein ST et al (2009) The binding of factor H to a complex of physiological polyanions and C3b on cells is impaired in atypical hemolytic uremic syndrome. J Immunol 182:7009–7018PubMedPubMedCentralCrossRef Ferreira VP, Herbert AP, Cortes C, McKee KA, Blaum BS, Esswein ST et al (2009) The binding of factor H to a complex of physiological polyanions and C3b on cells is impaired in atypical hemolytic uremic syndrome. J Immunol 182:7009–7018PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Lehtinen MJ, Rops AL, Isenman DE, van der Vlag J, Jokiranta TS (2009) Mutations of factor H impair regulation of surface-bound C3b by three mechanisms in atypical hemolytic uremic syndrome. J Biol Chem 284:15650–15658PubMedPubMedCentralCrossRef Lehtinen MJ, Rops AL, Isenman DE, van der Vlag J, Jokiranta TS (2009) Mutations of factor H impair regulation of surface-bound C3b by three mechanisms in atypical hemolytic uremic syndrome. J Biol Chem 284:15650–15658PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Heinen S, Sanchez-Corral P, Jackson MS, Strain L, Goodship JA, Kemp EJ et al (2006) De novo gene conversion in the RCA gene cluster (1q32) causes mutations in complement factor H associated with atypical hemolytic uremic syndrome. Hum Mutat 27:292–293PubMedCrossRef Heinen S, Sanchez-Corral P, Jackson MS, Strain L, Goodship JA, Kemp EJ et al (2006) De novo gene conversion in the RCA gene cluster (1q32) causes mutations in complement factor H associated with atypical hemolytic uremic syndrome. Hum Mutat 27:292–293PubMedCrossRef
12.
Zurück zum Zitat Venables JP, Strain L, Routledge D, Bourn D, Powell HM, Warwicker P et al (2006) Atypical haemolytic uraemic syndrome associated with a hybrid complement gene. PLoS Med 3:e431PubMedPubMedCentralCrossRef Venables JP, Strain L, Routledge D, Bourn D, Powell HM, Warwicker P et al (2006) Atypical haemolytic uraemic syndrome associated with a hybrid complement gene. PLoS Med 3:e431PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Francis NJ, McNicholas B, Awan A, Waldron M, Reddan D, Sadlier D et al (2012) A novel hybrid CFH/CFHR3 gene generated by a microhomology-mediated deletion in familial atypical hemolytic uremic syndrome. Blood 119:591–601PubMedCrossRef Francis NJ, McNicholas B, Awan A, Waldron M, Reddan D, Sadlier D et al (2012) A novel hybrid CFH/CFHR3 gene generated by a microhomology-mediated deletion in familial atypical hemolytic uremic syndrome. Blood 119:591–601PubMedCrossRef
14.
Zurück zum Zitat Challis RC, Araujo GS, Wong EK, Anderson HE, Awan A, Dorman AM et al (2016) A de novo deletion in the regulators of complement activation cluster producing a hybrid complement factor H/complement factor H-related 3 gene in atypical hemolytic uremic syndrome. J Am Soc Nephrol 27:1617–1624PubMedCrossRef Challis RC, Araujo GS, Wong EK, Anderson HE, Awan A, Dorman AM et al (2016) A de novo deletion in the regulators of complement activation cluster producing a hybrid complement factor H/complement factor H-related 3 gene in atypical hemolytic uremic syndrome. J Am Soc Nephrol 27:1617–1624PubMedCrossRef
15.
Zurück zum Zitat Richards A, Kathryn Liszewski M, Kavanagh D, Fang CJ, Moulton E, Fremeaux-Bacchi V et al (2007) Implications of the initial mutations in membrane cofactor protein (MCP; CD46) leading to atypical hemolytic uremic syndrome. Mol Immunol 44:111–122PubMedCrossRef Richards A, Kathryn Liszewski M, Kavanagh D, Fang CJ, Moulton E, Fremeaux-Bacchi V et al (2007) Implications of the initial mutations in membrane cofactor protein (MCP; CD46) leading to atypical hemolytic uremic syndrome. Mol Immunol 44:111–122PubMedCrossRef
16.
Zurück zum Zitat Kavanagh D, Kemp EJ, Mayland E, Winney RJ, Duffield JS, Warwick G et al (2005) Mutations in complement factor I predispose to development of atypical hemolytic uremic syndrome. J Am Soc Nephrol 16:2150–2155PubMedCrossRef Kavanagh D, Kemp EJ, Mayland E, Winney RJ, Duffield JS, Warwick G et al (2005) Mutations in complement factor I predispose to development of atypical hemolytic uremic syndrome. J Am Soc Nephrol 16:2150–2155PubMedCrossRef
17.
Zurück zum Zitat Fremeaux-Bacchi V, Miller EC, Liszewski MK, Strain L, Blouin J, Brown AL et al (2008) Mutations in complement C3 predispose to development of atypical hemolytic uremic syndrome. Blood 112:4948–4952PubMedPubMedCentralCrossRef Fremeaux-Bacchi V, Miller EC, Liszewski MK, Strain L, Blouin J, Brown AL et al (2008) Mutations in complement C3 predispose to development of atypical hemolytic uremic syndrome. Blood 112:4948–4952PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Sartz L, Olin AI, Kristoffersson AC, Stahl AL, Johansson ME, Westman K et al (2012) A novel C3 mutation causing increased formation of the C3 convertase in familial atypical hemolytic uremic syndrome. J Immunol 188:2030–2037PubMedCrossRef Sartz L, Olin AI, Kristoffersson AC, Stahl AL, Johansson ME, Westman K et al (2012) A novel C3 mutation causing increased formation of the C3 convertase in familial atypical hemolytic uremic syndrome. J Immunol 188:2030–2037PubMedCrossRef
19.
Zurück zum Zitat Roumenina LT, Jablonski M, Hue C, Blouin J, Dimitrov JD, Dragon-Durey MA et al (2009) Hyperfunctional C3 convertase leads to complement deposition on endothelial cells and contributes to atypical hemolytic uremic syndrome. Blood 114:2837–2845PubMedCrossRef Roumenina LT, Jablonski M, Hue C, Blouin J, Dimitrov JD, Dragon-Durey MA et al (2009) Hyperfunctional C3 convertase leads to complement deposition on endothelial cells and contributes to atypical hemolytic uremic syndrome. Blood 114:2837–2845PubMedCrossRef
20.
Zurück zum Zitat Goicoechea de Jorge E, Harris CL, Esparza-Gordillo J, Carreras L, Arranz EA, Garrido CA et al (2007) Gain-of-function mutations in complement factor B are associated with atypical hemolytic uremic syndrome. Proc Natl Acad Sci U S A 104:240–245PubMedCrossRef Goicoechea de Jorge E, Harris CL, Esparza-Gordillo J, Carreras L, Arranz EA, Garrido CA et al (2007) Gain-of-function mutations in complement factor B are associated with atypical hemolytic uremic syndrome. Proc Natl Acad Sci U S A 104:240–245PubMedCrossRef
21.
Zurück zum Zitat Delvaeye M, Noris M, De Vriese A, Esmon CT, Esmon NL, Ferrell G et al (2009) Thrombomodulin mutations in atypical hemolytic-uremic syndrome. N Engl J Med 361:345–357PubMedPubMedCentralCrossRef Delvaeye M, Noris M, De Vriese A, Esmon CT, Esmon NL, Ferrell G et al (2009) Thrombomodulin mutations in atypical hemolytic-uremic syndrome. N Engl J Med 361:345–357PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Fremeaux-Bacchi V, Fakhouri F, Garnier A, Bienaime F, Dragon-Durey MA, Ngo S et al (2013) Genetics and outcome of atypical hemolytic uremic syndrome: a nationwide French series comparing children and adults. Clin J Am Soc Nephrol 8:554–562PubMedPubMedCentralCrossRef Fremeaux-Bacchi V, Fakhouri F, Garnier A, Bienaime F, Dragon-Durey MA, Ngo S et al (2013) Genetics and outcome of atypical hemolytic uremic syndrome: a nationwide French series comparing children and adults. Clin J Am Soc Nephrol 8:554–562PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Fremeaux-Bacchi V, Kemp EJ, Goodship JA, Dragon-Durey MA, Strain L, Loirat C et al (2005) The development of atypical haemolytic-uraemic syndrome is influenced by susceptibility factors in factor H and membrane cofactor protein: evidence from two independent cohorts. J Med Genet 42:852–856PubMedPubMedCentralCrossRef Fremeaux-Bacchi V, Kemp EJ, Goodship JA, Dragon-Durey MA, Strain L, Loirat C et al (2005) The development of atypical haemolytic-uraemic syndrome is influenced by susceptibility factors in factor H and membrane cofactor protein: evidence from two independent cohorts. J Med Genet 42:852–856PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Esparza-Gordillo J, Goicoechea de Jorge E, Buil A, Carreras Berges L, Lopez-Trascasa M, Sanchez-Corral P et al (2005) Predisposition to atypical hemolytic uremic syndrome involves the concurrence of different susceptibility alleles in the regulators of complement activation gene cluster in 1q32. Hum Mol Genet 14:703–712PubMedCrossRef Esparza-Gordillo J, Goicoechea de Jorge E, Buil A, Carreras Berges L, Lopez-Trascasa M, Sanchez-Corral P et al (2005) Predisposition to atypical hemolytic uremic syndrome involves the concurrence of different susceptibility alleles in the regulators of complement activation gene cluster in 1q32. Hum Mol Genet 14:703–712PubMedCrossRef
25.
Zurück zum Zitat Frimat M, Roumenina L, Tabarin F, Halbwachs-Mecarelli L, Fremeaux-Bacchi V (2012) Membrane cofactor protein (MCP) haplotype, which predisposes to atypical hemolytic and uremic syndrome, has no consequence on neutrophils and endothelial cells MCP levels or on HUVECs ability to activate complement. Immunobiology 217:1187–1188CrossRef Frimat M, Roumenina L, Tabarin F, Halbwachs-Mecarelli L, Fremeaux-Bacchi V (2012) Membrane cofactor protein (MCP) haplotype, which predisposes to atypical hemolytic and uremic syndrome, has no consequence on neutrophils and endothelial cells MCP levels or on HUVECs ability to activate complement. Immunobiology 217:1187–1188CrossRef
26.
Zurück zum Zitat Pickering MC, de Jorge EG, Martinez-Barricarte R, Recalde S, Garcia-Layana A, Rose KL et al (2007) Spontaneous hemolytic uremic syndrome triggered by complement factor H lacking surface recognition domains. J Exp Med 204:1249–1256PubMedPubMedCentralCrossRef Pickering MC, de Jorge EG, Martinez-Barricarte R, Recalde S, Garcia-Layana A, Rose KL et al (2007) Spontaneous hemolytic uremic syndrome triggered by complement factor H lacking surface recognition domains. J Exp Med 204:1249–1256PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Pechtl IC, Kavanagh D, McIntosh N, Harris CL, Barlow PN (2011) Disease-associated N-terminal complement factor H mutations perturb cofactor and decay-accelerating activities. J Biol Chem 286:11082–11090PubMedPubMedCentralCrossRef Pechtl IC, Kavanagh D, McIntosh N, Harris CL, Barlow PN (2011) Disease-associated N-terminal complement factor H mutations perturb cofactor and decay-accelerating activities. J Biol Chem 286:11082–11090PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Tortajada A, Montes T, Martinez-Barricarte R, Morgan BP, Harris CL, de Cordoba SR (2009) The disease-protective complement factor H allotypic variant Ile62 shows increased binding affinity for C3b and enhanced cofactor activity. Hum Mol Genet 18:3452–3461PubMedPubMedCentralCrossRef Tortajada A, Montes T, Martinez-Barricarte R, Morgan BP, Harris CL, de Cordoba SR (2009) The disease-protective complement factor H allotypic variant Ile62 shows increased binding affinity for C3b and enhanced cofactor activity. Hum Mol Genet 18:3452–3461PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Dragon-Durey MA, Loirat C, Cloarec S, Macher MA, Blouin J, Nivet H et al (2005) Anti-factor H autoantibodies associated with atypical hemolytic uremic syndrome. J Am Soc Nephrol: JASN 16:555–563PubMedCrossRef Dragon-Durey MA, Loirat C, Cloarec S, Macher MA, Blouin J, Nivet H et al (2005) Anti-factor H autoantibodies associated with atypical hemolytic uremic syndrome. J Am Soc Nephrol: JASN 16:555–563PubMedCrossRef
30.
Zurück zum Zitat Brocklebank V, Johnson S, Sheerin TP, Marks SD, Gilbert RD, Tyerman K et al (2017) Factor H autoantibody is associated with atypical hemolytic uremic syndrome in children in the United Kingdom and Ireland. Kidney Int 92:1261–1271PubMedPubMedCentralCrossRef Brocklebank V, Johnson S, Sheerin TP, Marks SD, Gilbert RD, Tyerman K et al (2017) Factor H autoantibody is associated with atypical hemolytic uremic syndrome in children in the United Kingdom and Ireland. Kidney Int 92:1261–1271PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Jozsi M, Licht C, Strobel S, Zipfel SL, Richter H, Heinen S et al (2008) Factor H autoantibodies in atypical hemolytic uremic syndrome correlate with CFHR1/CFHR3 deficiency. Blood 111:1512–1514PubMedCrossRef Jozsi M, Licht C, Strobel S, Zipfel SL, Richter H, Heinen S et al (2008) Factor H autoantibodies in atypical hemolytic uremic syndrome correlate with CFHR1/CFHR3 deficiency. Blood 111:1512–1514PubMedCrossRef
32.
Zurück zum Zitat Moore I, Strain L, Pappworth I, Kavanagh D, Barlow PN, Herbert AP et al (2010) Association of factor H autoantibodies with deletions of CFHR1, CFHR3, CFHR4, and with mutations in CFH, CFI, CD46, and C3 in patients with atypical hemolytic uremic syndrome. Blood 115:379–387PubMedPubMedCentralCrossRef Moore I, Strain L, Pappworth I, Kavanagh D, Barlow PN, Herbert AP et al (2010) Association of factor H autoantibodies with deletions of CFHR1, CFHR3, CFHR4, and with mutations in CFH, CFI, CD46, and C3 in patients with atypical hemolytic uremic syndrome. Blood 115:379–387PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Jozsi M, Strobel S, Dahse HM, Liu WS, Hoyer PF, Oppermann M et al (2007) Anti factor H autoantibodies block C-terminal recognition function of factor H in hemolytic uremic syndrome. Blood 110:1516–1518PubMedCrossRef Jozsi M, Strobel S, Dahse HM, Liu WS, Hoyer PF, Oppermann M et al (2007) Anti factor H autoantibodies block C-terminal recognition function of factor H in hemolytic uremic syndrome. Blood 110:1516–1518PubMedCrossRef
34.
Zurück zum Zitat Blanc C, Roumenina LT, Ashraf Y, Hyvarinen S, Sethi SK, Ranchin B et al (2012) Overall neutralization of complement factor H by autoantibodies in the acute phase of the autoimmune form of atypical hemolytic uremic syndrome. J Immunol 189:3528–3537PubMedCrossRef Blanc C, Roumenina LT, Ashraf Y, Hyvarinen S, Sethi SK, Ranchin B et al (2012) Overall neutralization of complement factor H by autoantibodies in the acute phase of the autoimmune form of atypical hemolytic uremic syndrome. J Immunol 189:3528–3537PubMedCrossRef
35.
Zurück zum Zitat Kavanagh D, Pappworth IY, Anderson H, Hayes CM, Moore I, Hunze EM et al (2012) Factor I autoantibodies in patients with atypical hemolytic uremic syndrome: disease-associated or an epiphenomenon? Clin J Am Soc Nephrol 7:417–426PubMedPubMedCentralCrossRef Kavanagh D, Pappworth IY, Anderson H, Hayes CM, Moore I, Hunze EM et al (2012) Factor I autoantibodies in patients with atypical hemolytic uremic syndrome: disease-associated or an epiphenomenon? Clin J Am Soc Nephrol 7:417–426PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Sheerin NS, Kavanagh D, Goodship TH, Johnson S (2016) A national specialized service in England for atypical haemolytic uraemic syndrome-the first year’s experience. QJM 109:27–33PubMedCrossRef Sheerin NS, Kavanagh D, Goodship TH, Johnson S (2016) A national specialized service in England for atypical haemolytic uraemic syndrome-the first year’s experience. QJM 109:27–33PubMedCrossRef
37.
Zurück zum Zitat Noris M, Caprioli J, Bresin E, Mossali C, Pianetti G, Gamba S et al (2010) Relative role of genetic complement abnormalities in sporadic and familial aHUS and their impact on clinical phenotype. Clin J Am Soc Nephrol 5:1844–1859PubMedPubMedCentralCrossRef Noris M, Caprioli J, Bresin E, Mossali C, Pianetti G, Gamba S et al (2010) Relative role of genetic complement abnormalities in sporadic and familial aHUS and their impact on clinical phenotype. Clin J Am Soc Nephrol 5:1844–1859PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Kavanagh D, Richards A, Goodship T, Jalanko H (2010) Transplantation in atypical hemolytic uremic syndrome. Semin Thromb Hemost 36:653–659PubMedCrossRef Kavanagh D, Richards A, Goodship T, Jalanko H (2010) Transplantation in atypical hemolytic uremic syndrome. Semin Thromb Hemost 36:653–659PubMedCrossRef
39.
Zurück zum Zitat Le Quintrec M, Zuber J, Moulin B, Kamar N, Jablonski M, Lionet A et al (2013) Complement genes strongly predict recurrence and graft outcome in adult renal transplant recipients with atypical hemolytic and uremic syndrome. Am J Transplant 13:663–675PubMedCrossRef Le Quintrec M, Zuber J, Moulin B, Kamar N, Jablonski M, Lionet A et al (2013) Complement genes strongly predict recurrence and graft outcome in adult renal transplant recipients with atypical hemolytic and uremic syndrome. Am J Transplant 13:663–675PubMedCrossRef
40.
Zurück zum Zitat Bresin E, Rurali E, Caprioli J, Sanchez-Corral P, Fremeaux-Bacchi V, Rodriguez de Cordoba S et al (2013) Combined complement gene mutations in atypical hemolytic uremic syndrome influence clinical phenotype. J Am Soc Nephrol: JASN 24:475–486PubMedPubMedCentralCrossRef Bresin E, Rurali E, Caprioli J, Sanchez-Corral P, Fremeaux-Bacchi V, Rodriguez de Cordoba S et al (2013) Combined complement gene mutations in atypical hemolytic uremic syndrome influence clinical phenotype. J Am Soc Nephrol: JASN 24:475–486PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Caprioli J, Noris M, Brioschi S, Pianetti G, Castelletti F, Bettinaglio P et al (2006) Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome. Blood 108:1267–1279PubMedPubMedCentralCrossRef Caprioli J, Noris M, Brioschi S, Pianetti G, Castelletti F, Bettinaglio P et al (2006) Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome. Blood 108:1267–1279PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Bruel A, Kavanagh D, Noris M, Delmas Y, Wong EKS, Bresin E et al (2017) Hemolytic uremic syndrome in pregnancy and postpartum. Clin J Am Soc Nephrol 12:1237–1247PubMedCrossRef Bruel A, Kavanagh D, Noris M, Delmas Y, Wong EKS, Bresin E et al (2017) Hemolytic uremic syndrome in pregnancy and postpartum. Clin J Am Soc Nephrol 12:1237–1247PubMedCrossRef
43.
Zurück zum Zitat Licht C, Greenbaum LA, Muus P, Babu S, Bedrosian CL, Cohen DJ et al (2015) Efficacy and safety of eculizumab in atypical hemolytic uremic syndrome from 2-year extensions of phase 2 studies. Kidney Int 87:1061–1073PubMedPubMedCentralCrossRef Licht C, Greenbaum LA, Muus P, Babu S, Bedrosian CL, Cohen DJ et al (2015) Efficacy and safety of eculizumab in atypical hemolytic uremic syndrome from 2-year extensions of phase 2 studies. Kidney Int 87:1061–1073PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Fakhouri F, Hourmant M, Campistol JM, Cataland SR, Espinosa M, Gaber AO et al (2016) Terminal complement inhibitor eculizumab in adult patients with atypical hemolytic uremic syndrome: a single-arm, open-label trial. Am J Kidney Dis 68:84–93PubMedCrossRef Fakhouri F, Hourmant M, Campistol JM, Cataland SR, Espinosa M, Gaber AO et al (2016) Terminal complement inhibitor eculizumab in adult patients with atypical hemolytic uremic syndrome: a single-arm, open-label trial. Am J Kidney Dis 68:84–93PubMedCrossRef
45.
Zurück zum Zitat Greenbaum LA, Fila M, Ardissino G, Al-Akash SI, Evans J, Henning P et al (2016) Eculizumab is a safe and effective treatment in pediatric patients with atypical hemolytic uremic syndrome. Kidney Int 89:701–711PubMedCrossRef Greenbaum LA, Fila M, Ardissino G, Al-Akash SI, Evans J, Henning P et al (2016) Eculizumab is a safe and effective treatment in pediatric patients with atypical hemolytic uremic syndrome. Kidney Int 89:701–711PubMedCrossRef
46.
Zurück zum Zitat Brocklebank V, Kavanagh D (2017) Complement C5 inhibiting therapy for the thrombotic microangiopathies: accumulating evidence, but not a panacea. Clin Kidney J 10:600–624PubMedPubMedCentralCrossRef Brocklebank V, Kavanagh D (2017) Complement C5 inhibiting therapy for the thrombotic microangiopathies: accumulating evidence, but not a panacea. Clin Kidney J 10:600–624PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Fakhouri F, Fila M, Provot F, Delmas Y, Barbet C, Chatelet V et al (2017) Pathogenic variants in complement genes and risk of atypical hemolytic uremic syndrome relapse after eculizumab discontinuation. Clin J Am Soc Nephrol 12:50–59PubMedCrossRef Fakhouri F, Fila M, Provot F, Delmas Y, Barbet C, Chatelet V et al (2017) Pathogenic variants in complement genes and risk of atypical hemolytic uremic syndrome relapse after eculizumab discontinuation. Clin J Am Soc Nephrol 12:50–59PubMedCrossRef
48.
Zurück zum Zitat Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi M, Tang WH, Le Quintrec M et al (2013) Recessive mutations in DGKE cause atypical hemolytic-uremic syndrome. Nat Genet 45:531–536PubMedPubMedCentralCrossRef Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi M, Tang WH, Le Quintrec M et al (2013) Recessive mutations in DGKE cause atypical hemolytic-uremic syndrome. Nat Genet 45:531–536PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Challis RC, Ring T, Xu Y, Wong EK, Flossmann O, Roberts IS et al (2017) Thrombotic microangiopathy in inverted formin 2-mediated renal disease. J Am Soc Nephrol 28:1084–1091PubMedCrossRef Challis RC, Ring T, Xu Y, Wong EK, Flossmann O, Roberts IS et al (2017) Thrombotic microangiopathy in inverted formin 2-mediated renal disease. J Am Soc Nephrol 28:1084–1091PubMedCrossRef
50.
Zurück zum Zitat Beck BB, van Spronsen F, Diepstra A, Berger RM, Komhoff M (2017) Renal thrombotic microangiopathy in patients with cblC defect: review of an under-recognized entity. Pediatr Nephrol 32:733–741PubMedCrossRef Beck BB, van Spronsen F, Diepstra A, Berger RM, Komhoff M (2017) Renal thrombotic microangiopathy in patients with cblC defect: review of an under-recognized entity. Pediatr Nephrol 32:733–741PubMedCrossRef
51.
Zurück zum Zitat Ozaltin F, Li B, Rauhauser A, An SW, Soylemezoglu O, Gonul II et al (2013) DGKE variants cause a glomerular microangiopathy that mimics membranoproliferative GN. J Am Soc Nephrol 24:377–384PubMedCrossRef Ozaltin F, Li B, Rauhauser A, An SW, Soylemezoglu O, Gonul II et al (2013) DGKE variants cause a glomerular microangiopathy that mimics membranoproliferative GN. J Am Soc Nephrol 24:377–384PubMedCrossRef
52.
Zurück zum Zitat Nishimura J, Yamamoto M, Hayashi S, Ohyashiki K, Ando K, Brodsky AL et al (2014) Genetic variants in C5 and poor response to eculizumab. N Engl J Med 370:632–639PubMedCrossRef Nishimura J, Yamamoto M, Hayashi S, Ohyashiki K, Ando K, Brodsky AL et al (2014) Genetic variants in C5 and poor response to eculizumab. N Engl J Med 370:632–639PubMedCrossRef
53.
Zurück zum Zitat Goodship T, Pinto F, Weston-Davies W, Silva J, Nishimura J, Nunn M et al (2017) Use of the complement inhibitor coversin to treat HSCT-associated TMA. Blood Adv 1:1254–1258PubMedPubMedCentralCrossRef Goodship T, Pinto F, Weston-Davies W, Silva J, Nishimura J, Nunn M et al (2017) Use of the complement inhibitor coversin to treat HSCT-associated TMA. Blood Adv 1:1254–1258PubMedPubMedCentralCrossRef
54.
55.
Zurück zum Zitat Spitzer RE, Vallota EH, Forristal J, Sudora E, Stitzel A, Davis NC et al (1969) Serum C’3 lytic system in patients with glomerulonephritis. Science 164:436–437PubMedCrossRef Spitzer RE, Vallota EH, Forristal J, Sudora E, Stitzel A, Davis NC et al (1969) Serum C’3 lytic system in patients with glomerulonephritis. Science 164:436–437PubMedCrossRef
56.
Zurück zum Zitat Levy M, Halbwachs-Mecarelli L, Gubler MC, Kohout G, Bensenouci A, Niaudet P et al (1986) H deficiency in two brothers with atypical dense intramembranous deposit disease. Kidney Int 30:949–956PubMedCrossRef Levy M, Halbwachs-Mecarelli L, Gubler MC, Kohout G, Bensenouci A, Niaudet P et al (1986) H deficiency in two brothers with atypical dense intramembranous deposit disease. Kidney Int 30:949–956PubMedCrossRef
57.
Zurück zum Zitat Medjeral-Thomas NR, O'Shaughnessy MM, O’Regan JA, Traynor C, Flanagan M, Wong L et al (2014) C3 glomerulopathy: clinicopathologic features and predictors of outcome. Clin J Am Soc Nephrol: CJASN 9:46–53PubMedCrossRef Medjeral-Thomas NR, O'Shaughnessy MM, O’Regan JA, Traynor C, Flanagan M, Wong L et al (2014) C3 glomerulopathy: clinicopathologic features and predictors of outcome. Clin J Am Soc Nephrol: CJASN 9:46–53PubMedCrossRef
58.
Zurück zum Zitat Briganti EM, Dowling J, Finlay M, Hill PA, Jones CL, Kincaid-Smith PS et al (2001) The incidence of biopsy-proven glomerulonephritis in Australia. Nephrol Dial Transplant 16:1364–1367PubMedCrossRef Briganti EM, Dowling J, Finlay M, Hill PA, Jones CL, Kincaid-Smith PS et al (2001) The incidence of biopsy-proven glomerulonephritis in Australia. Nephrol Dial Transplant 16:1364–1367PubMedCrossRef
59.
Zurück zum Zitat West CD (1992) Idiopathic membranoproliferative glomerulonephritis in childhood. Pediatr Nephrol 6:96–103PubMedCrossRef West CD (1992) Idiopathic membranoproliferative glomerulonephritis in childhood. Pediatr Nephrol 6:96–103PubMedCrossRef
60.
Zurück zum Zitat Hou J, Markowitz GS, Bomback AS, Appel GB, Herlitz LC, Barry Stokes M et al (2014) Toward a working definition of C3 glomerulopathy by immunofluorescence. Kidney Int 85:450–456PubMedCrossRef Hou J, Markowitz GS, Bomback AS, Appel GB, Herlitz LC, Barry Stokes M et al (2014) Toward a working definition of C3 glomerulopathy by immunofluorescence. Kidney Int 85:450–456PubMedCrossRef
61.
Zurück zum Zitat Sethi S, Fervenza FC (2012) Membranoproliferative glomerulonephritis—a new look at an old entity. N Engl J Med 366:1119–1131PubMedCrossRef Sethi S, Fervenza FC (2012) Membranoproliferative glomerulonephritis—a new look at an old entity. N Engl J Med 366:1119–1131PubMedCrossRef
62.
Zurück zum Zitat Anders D, Thoenes W (1975) Basement membrane-changes in membranoproliferative glomerulonephritis: a light and electron microscopic study. Virchows Arch A Pathol Anat Histol 369:87–109PubMedCrossRef Anders D, Thoenes W (1975) Basement membrane-changes in membranoproliferative glomerulonephritis: a light and electron microscopic study. Virchows Arch A Pathol Anat Histol 369:87–109PubMedCrossRef
63.
Zurück zum Zitat Sethi S, Nester CM, Smith RJ (2012) Membranoproliferative glomerulonephritis and C3 glomerulopathy: resolving the confusion. Kidney Int 81:434–441PubMedCrossRef Sethi S, Nester CM, Smith RJ (2012) Membranoproliferative glomerulonephritis and C3 glomerulopathy: resolving the confusion. Kidney Int 81:434–441PubMedCrossRef
64.
Zurück zum Zitat Servais A, Fremeaux-Bacchi V, Lequintrec M, Salomon R, Blouin J, Knebelmann B et al (2007) Primary glomerulonephritis with isolated C3 deposits: a new entity which shares common genetic risk factors with haemolytic uraemic syndrome. J Med Genet 44:193–199PubMedCrossRef Servais A, Fremeaux-Bacchi V, Lequintrec M, Salomon R, Blouin J, Knebelmann B et al (2007) Primary glomerulonephritis with isolated C3 deposits: a new entity which shares common genetic risk factors with haemolytic uraemic syndrome. J Med Genet 44:193–199PubMedCrossRef
65.
Zurück zum Zitat Servais A, Noel LH, Roumenina LT, Le Quintrec M, Ngo S, Dragon-Durey MA et al (2012) Acquired and genetic complement abnormalities play a critical role in dense deposit disease and other C3 glomerulopathies. Kidney Int 82:454–464PubMedCrossRef Servais A, Noel LH, Roumenina LT, Le Quintrec M, Ngo S, Dragon-Durey MA et al (2012) Acquired and genetic complement abnormalities play a critical role in dense deposit disease and other C3 glomerulopathies. Kidney Int 82:454–464PubMedCrossRef
66.
Zurück zum Zitat Iatropoulos P, Noris M, Mele C, Piras R, Valoti E, Bresin E et al (2016) Complement gene variants determine the risk of immunoglobulin-associated MPGN and C3 glomerulopathy and predict long-term renal outcome. Mol Immunol 71:131–142PubMedCrossRef Iatropoulos P, Noris M, Mele C, Piras R, Valoti E, Bresin E et al (2016) Complement gene variants determine the risk of immunoglobulin-associated MPGN and C3 glomerulopathy and predict long-term renal outcome. Mol Immunol 71:131–142PubMedCrossRef
67.
Zurück zum Zitat Marinozzi MC, Roumenina LT, Chauvet S, Hertig A, Bertrand D, Olagne J et al (2017) Anti-factor B and anti-C3b autoantibodies in C3 glomerulopathy and Ig-associated membranoproliferative GN. J Am Soc Nephrol: JASN 28:1603–1613PubMedCrossRef Marinozzi MC, Roumenina LT, Chauvet S, Hertig A, Bertrand D, Olagne J et al (2017) Anti-factor B and anti-C3b autoantibodies in C3 glomerulopathy and Ig-associated membranoproliferative GN. J Am Soc Nephrol: JASN 28:1603–1613PubMedCrossRef
69.
Zurück zum Zitat Larsen CP, Messias NC, Walker PD, Fidler ME, Cornell LD, Hernandez LH et al (2015) Membranoproliferative glomerulonephritis with masked monotypic immunoglobulin deposits. Kidney Int 88:867–873PubMedPubMedCentralCrossRef Larsen CP, Messias NC, Walker PD, Fidler ME, Cornell LD, Hernandez LH et al (2015) Membranoproliferative glomerulonephritis with masked monotypic immunoglobulin deposits. Kidney Int 88:867–873PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Chauvet S, Fremeaux-Bacchi V, Petitprez F, Karras A, Daniel L, Burtey S et al (2017) Treatment of B-cell disorder improves renal outcome of patients with monoclonal gammopathy-associated C3 glomerulopathy. Blood 129:1437–1447PubMedCrossRef Chauvet S, Fremeaux-Bacchi V, Petitprez F, Karras A, Daniel L, Burtey S et al (2017) Treatment of B-cell disorder improves renal outcome of patients with monoclonal gammopathy-associated C3 glomerulopathy. Blood 129:1437–1447PubMedCrossRef
71.
Zurück zum Zitat Thompson RA, Winterborn MH (1981) Hypocomplementaemia due to a genetic deficiency of beta 1H globulin. Clin Exp Immunol 46:110–119PubMedPubMedCentral Thompson RA, Winterborn MH (1981) Hypocomplementaemia due to a genetic deficiency of beta 1H globulin. Clin Exp Immunol 46:110–119PubMedPubMedCentral
72.
Zurück zum Zitat Pickering MC, Cook HT, Warren J, Bygrave AE, Moss J, Walport MJ et al (2002) Uncontrolled C3 activation causes membranoproliferative glomerulonephritis in mice deficient in complement factor H. Nat Genet 31:424–428PubMedCrossRef Pickering MC, Cook HT, Warren J, Bygrave AE, Moss J, Walport MJ et al (2002) Uncontrolled C3 activation causes membranoproliferative glomerulonephritis in mice deficient in complement factor H. Nat Genet 31:424–428PubMedCrossRef
73.
Zurück zum Zitat Dragon-Durey MA, Fremeaux-Bacchi V, Loirat C, Blouin J, Niaudet P, Deschenes G et al (2004) Heterozygous and homozygous factor h deficiencies associated with hemolytic uremic syndrome or membranoproliferative glomerulonephritis: report and genetic analysis of 16 cases. J Am Soc Nephrol 15:787–795PubMedCrossRef Dragon-Durey MA, Fremeaux-Bacchi V, Loirat C, Blouin J, Niaudet P, Deschenes G et al (2004) Heterozygous and homozygous factor h deficiencies associated with hemolytic uremic syndrome or membranoproliferative glomerulonephritis: report and genetic analysis of 16 cases. J Am Soc Nephrol 15:787–795PubMedCrossRef
74.
Zurück zum Zitat Alfandary H, Davidovits M (2015) Novel factor H mutation associated with familial membranoproliferative glomerulonephritis type I. Pediatr Nephrol 30:2129–2134PubMedCrossRef Alfandary H, Davidovits M (2015) Novel factor H mutation associated with familial membranoproliferative glomerulonephritis type I. Pediatr Nephrol 30:2129–2134PubMedCrossRef
75.
Zurück zum Zitat Vaziri-Sani F, Holmberg L, Sjoholm AG, Kristoffersson AC, Manea M, Fremeaux-Bacchi V et al (2006) Phenotypic expression of factor H mutations in patients with atypical hemolytic uremic syndrome. Kidney Int 69:981–988PubMedCrossRef Vaziri-Sani F, Holmberg L, Sjoholm AG, Kristoffersson AC, Manea M, Fremeaux-Bacchi V et al (2006) Phenotypic expression of factor H mutations in patients with atypical hemolytic uremic syndrome. Kidney Int 69:981–988PubMedCrossRef
76.
Zurück zum Zitat Boyer O, Noel LH, Balzamo E, Guest G, Biebuyck N, Charbit M et al (2008) Complement factor H deficiency and posttransplantation glomerulonephritis with isolated C3 deposits. Am J Kidney Dis: Off J Natl Kidney Found 51:671–677CrossRef Boyer O, Noel LH, Balzamo E, Guest G, Biebuyck N, Charbit M et al (2008) Complement factor H deficiency and posttransplantation glomerulonephritis with isolated C3 deposits. Am J Kidney Dis: Off J Natl Kidney Found 51:671–677CrossRef
77.
Zurück zum Zitat Gnappi E, Allinovi M, Vaglio A, Bresin E, Sorosina A, Pilato FP et al (2012) Membrano-proliferative glomerulonephritis, atypical hemolytic uremic syndrome, and a new complement factor H mutation: report of a case. Pediatr Nephrol 27:1995–1999PubMedCrossRef Gnappi E, Allinovi M, Vaglio A, Bresin E, Sorosina A, Pilato FP et al (2012) Membrano-proliferative glomerulonephritis, atypical hemolytic uremic syndrome, and a new complement factor H mutation: report of a case. Pediatr Nephrol 27:1995–1999PubMedCrossRef
78.
Zurück zum Zitat Zhang Y, Meyer NC, Wang K, Nishimura C, Frees K, Jones M et al (2012) Causes of alternative pathway dysregulation in dense deposit disease. Clin J Am Soc Nephrol: CJASN 7:265–274PubMedPubMedCentralCrossRef Zhang Y, Meyer NC, Wang K, Nishimura C, Frees K, Jones M et al (2012) Causes of alternative pathway dysregulation in dense deposit disease. Clin J Am Soc Nephrol: CJASN 7:265–274PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Sethi S, Fervenza FC, Zhang Y, Zand L, Vrana JA, Nasr SH et al (2012) C3 glomerulonephritis: clinicopathological findings, complement abnormalities, glomerular proteomic profile, treatment, and follow-up. Kidney Int 82:465–473PubMedPubMedCentralCrossRef Sethi S, Fervenza FC, Zhang Y, Zand L, Vrana JA, Nasr SH et al (2012) C3 glomerulonephritis: clinicopathological findings, complement abnormalities, glomerular proteomic profile, treatment, and follow-up. Kidney Int 82:465–473PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat Licht C, Heinen S, Jozsi M, Loschmann I, Saunders RE, Perkins SJ et al (2006) Deletion of Lys224 in regulatory domain 4 of factor H reveals a novel pathomechanism for dense deposit disease (MPGN II). Kidney Int 70:42–50PubMedCrossRef Licht C, Heinen S, Jozsi M, Loschmann I, Saunders RE, Perkins SJ et al (2006) Deletion of Lys224 in regulatory domain 4 of factor H reveals a novel pathomechanism for dense deposit disease (MPGN II). Kidney Int 70:42–50PubMedCrossRef
81.
Zurück zum Zitat Yu Y, Triebwasser MP, Wong EK, Schramm EC, Thomas B, Reynolds R et al (2014) Whole-exome sequencing identifies rare, functional CFH variants in families with macular degeneration. Hum Mol Genet 23:5283–5293PubMedPubMedCentralCrossRef Yu Y, Triebwasser MP, Wong EK, Schramm EC, Thomas B, Reynolds R et al (2014) Whole-exome sequencing identifies rare, functional CFH variants in families with macular degeneration. Hum Mol Genet 23:5283–5293PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Sethi S, Fervenza FC, Zhang Y, Nasr SH, Leung N, Vrana J et al (2011) Proliferative glomerulonephritis secondary to dysfunction of the alternative pathway of complement. Clin J Am Soc Nephrol: CJASN 6:1009–1017PubMedPubMedCentralCrossRef Sethi S, Fervenza FC, Zhang Y, Nasr SH, Leung N, Vrana J et al (2011) Proliferative glomerulonephritis secondary to dysfunction of the alternative pathway of complement. Clin J Am Soc Nephrol: CJASN 6:1009–1017PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Wong EK, Anderson HE, Herbert AP, Challis RC, Brown P, Reis GS et al (2014) Characterization of a factor H mutation that perturbs the alternative pathway of complement in a family with membranoproliferative GN. J Am Soc Nephrol: JASN 25:2425–2433PubMedPubMedCentralCrossRef Wong EK, Anderson HE, Herbert AP, Challis RC, Brown P, Reis GS et al (2014) Characterization of a factor H mutation that perturbs the alternative pathway of complement in a family with membranoproliferative GN. J Am Soc Nephrol: JASN 25:2425–2433PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Martinez-Barricarte R, Heurich M, Valdes-Canedo F, Vazquez-Martul E, Torreira E, Montes T et al (2010) Human C3 mutation reveals a mechanism of dense deposit disease pathogenesis and provides insights into complement activation and regulation. J Clin Invest 120:3702–3712PubMedPubMedCentralCrossRef Martinez-Barricarte R, Heurich M, Valdes-Canedo F, Vazquez-Martul E, Torreira E, Montes T et al (2010) Human C3 mutation reveals a mechanism of dense deposit disease pathogenesis and provides insights into complement activation and regulation. J Clin Invest 120:3702–3712PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Chauvet S, Roumenina LT, Bruneau S, Marinozzi MC, Rybkine T, Schramm EC et al (2015) A familial C3GN secondary to defective C3 regulation by complement receptor 1 and factor H. J Am Soc Nephrol 27:1665–1677PubMedPubMedCentralCrossRef Chauvet S, Roumenina LT, Bruneau S, Marinozzi MC, Rybkine T, Schramm EC et al (2015) A familial C3GN secondary to defective C3 regulation by complement receptor 1 and factor H. J Am Soc Nephrol 27:1665–1677PubMedPubMedCentralCrossRef
86.
Zurück zum Zitat Janssen van Doorn K, Dirinck E, Verpooten GA, Couttenye MM (2013) Complement factor H mutation associated with membranoproliferative glomerulonephritis with transformation to atypical haemolytic uraemic syndrome. Clin Kidney J 6:216–219PubMedPubMedCentralCrossRef Janssen van Doorn K, Dirinck E, Verpooten GA, Couttenye MM (2013) Complement factor H mutation associated with membranoproliferative glomerulonephritis with transformation to atypical haemolytic uraemic syndrome. Clin Kidney J 6:216–219PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Bu F, Borsa NG, Jones MB, Takanami E, Nishimura C, Hauer JJ et al (2015) High-throughput genetic testing for thrombotic microangiopathies and C3 glomerulopathies. J Am Soc Nephrol: JASN 27:1245–1253PubMedPubMedCentralCrossRef Bu F, Borsa NG, Jones MB, Takanami E, Nishimura C, Hauer JJ et al (2015) High-throughput genetic testing for thrombotic microangiopathies and C3 glomerulopathies. J Am Soc Nephrol: JASN 27:1245–1253PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat Salmon JE, Heuser C, Triebwasser M, Liszewski MK, Kavanagh D, Roumenina L et al (2011) Mutations in complement regulatory proteins predispose to preeclampsia: a genetic analysis of the PROMISSE cohort. PLoS Med 8:e1001013PubMedPubMedCentralCrossRef Salmon JE, Heuser C, Triebwasser M, Liszewski MK, Kavanagh D, Roumenina L et al (2011) Mutations in complement regulatory proteins predispose to preeclampsia: a genetic analysis of the PROMISSE cohort. PLoS Med 8:e1001013PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Bienaime F, Dragon-Durey MA, Regnier CH, Nilsson SC, Kwan WH, Blouin J et al (2010) Mutations in components of complement influence the outcome of factor I-associated atypical hemolytic uremic syndrome. Kidney Int 77:339–349PubMedCrossRef Bienaime F, Dragon-Durey MA, Regnier CH, Nilsson SC, Kwan WH, Blouin J et al (2010) Mutations in components of complement influence the outcome of factor I-associated atypical hemolytic uremic syndrome. Kidney Int 77:339–349PubMedCrossRef
90.
Zurück zum Zitat Kavanagh D, Yu Y, Schramm EC, Triebwasser M, Wagner EK, Raychaudhuri S et al (2015) Rare genetic variants in the CFI gene are associated with advanced age-related macular degeneration and commonly result in reduced serum factor I levels. Hum Mol Genet 24:3861–3870PubMedPubMedCentral Kavanagh D, Yu Y, Schramm EC, Triebwasser M, Wagner EK, Raychaudhuri S et al (2015) Rare genetic variants in the CFI gene are associated with advanced age-related macular degeneration and commonly result in reduced serum factor I levels. Hum Mol Genet 24:3861–3870PubMedPubMedCentral
91.
Zurück zum Zitat Kavanagh D, Richards A, Noris M, Hauhart R, Liszewski MK, Karpman D et al (2008) Characterization of mutations in complement factor I (CFI) associated with hemolytic uremic syndrome. Mol Immunol 45:95–105PubMedCrossRef Kavanagh D, Richards A, Noris M, Hauhart R, Liszewski MK, Karpman D et al (2008) Characterization of mutations in complement factor I (CFI) associated with hemolytic uremic syndrome. Mol Immunol 45:95–105PubMedCrossRef
92.
Zurück zum Zitat Nilsson SC, Kalchishkova N, Trouw LA, Fremeaux-Bacchi V, Villoutreix BO, Blom AM (2010) Mutations in complement factor I as found in atypical hemolytic uremic syndrome lead to either altered secretion or altered function of factor I. Eur J Immunol 40:172–185PubMedCrossRef Nilsson SC, Kalchishkova N, Trouw LA, Fremeaux-Bacchi V, Villoutreix BO, Blom AM (2010) Mutations in complement factor I as found in atypical hemolytic uremic syndrome lead to either altered secretion or altered function of factor I. Eur J Immunol 40:172–185PubMedCrossRef
93.
Zurück zum Zitat Gleeson PJ, Wilson V, Cox TE, Sharma SD, Smith-Jackson K, Strain L et al (2016) Chromosomal rearrangement—a rare cause of complement factor I associated atypical haemolytic uraemic syndrome. Immunobiology 221:1124–1130PubMedCrossRef Gleeson PJ, Wilson V, Cox TE, Sharma SD, Smith-Jackson K, Strain L et al (2016) Chromosomal rearrangement—a rare cause of complement factor I associated atypical haemolytic uraemic syndrome. Immunobiology 221:1124–1130PubMedCrossRef
94.
Zurück zum Zitat Nilsson SC, Karpman D, Vaziri-Sani F, Kristoffersson AC, Salomon R, Provot F et al (2007) A mutation in factor I that is associated with atypical hemolytic uremic syndrome does not affect the function of factor I in complement regulation. Mol Immunol 44:1835–1844PubMedCrossRef Nilsson SC, Karpman D, Vaziri-Sani F, Kristoffersson AC, Salomon R, Provot F et al (2007) A mutation in factor I that is associated with atypical hemolytic uremic syndrome does not affect the function of factor I in complement regulation. Mol Immunol 44:1835–1844PubMedCrossRef
95.
Zurück zum Zitat Imamura H, Konomoto T, Tanaka E, Hisano S, Yoshida Y, Fujimura Y et al (2015) Familial C3 glomerulonephritis associated with mutations in the gene for complement factor B. Nephrol Dial Transplant 30:862–864PubMedCrossRef Imamura H, Konomoto T, Tanaka E, Hisano S, Yoshida Y, Fujimura Y et al (2015) Familial C3 glomerulonephritis associated with mutations in the gene for complement factor B. Nephrol Dial Transplant 30:862–864PubMedCrossRef
96.
Zurück zum Zitat Kavanagh D, Anderson HE (2012) Interpretation of genetic variants of uncertain significance in atypical hemolytic uremic syndrome. Kidney Int 81:11–13PubMedCrossRef Kavanagh D, Anderson HE (2012) Interpretation of genetic variants of uncertain significance in atypical hemolytic uremic syndrome. Kidney Int 81:11–13PubMedCrossRef
97.
Zurück zum Zitat Goicoechea de Jorge E, Caesar JJ, Malik TH, Patel M, Colledge M, Johnson S et al (2013) Dimerization of complement factor H-related proteins modulates complement activation in vivo. Proc Natl Acad Sci U S A 110:4685–4690PubMedPubMedCentralCrossRef Goicoechea de Jorge E, Caesar JJ, Malik TH, Patel M, Colledge M, Johnson S et al (2013) Dimerization of complement factor H-related proteins modulates complement activation in vivo. Proc Natl Acad Sci U S A 110:4685–4690PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Gale DP, de Jorge EG, Cook HT, Martinez-Barricarte R, Hadjisavvas A, McLean AG et al (2010) Identification of a mutation in complement factor H-related protein 5 in patients of Cypriot origin with glomerulonephritis. Lancet 376:794–801PubMedPubMedCentralCrossRef Gale DP, de Jorge EG, Cook HT, Martinez-Barricarte R, Hadjisavvas A, McLean AG et al (2010) Identification of a mutation in complement factor H-related protein 5 in patients of Cypriot origin with glomerulonephritis. Lancet 376:794–801PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Chen Q, Wiesener M, Eberhardt HU, Hartmann A, Uzonyi B, Kirschfink M et al (2014) Complement factor H-related hybrid protein deregulates complement in dense deposit disease. J Clin Invest 124:145–155PubMedCrossRef Chen Q, Wiesener M, Eberhardt HU, Hartmann A, Uzonyi B, Kirschfink M et al (2014) Complement factor H-related hybrid protein deregulates complement in dense deposit disease. J Clin Invest 124:145–155PubMedCrossRef
100.
Zurück zum Zitat Chen Q, Manzke M, Hartmann A, Buttner M, Amann K, Pauly D et al (2015) Complement factor H-related 5-hybrid proteins anchor properdin and activate complement at self-surfaces. J Am Soc Nephrol: JASN 27:1413–1425PubMedPubMedCentralCrossRef Chen Q, Manzke M, Hartmann A, Buttner M, Amann K, Pauly D et al (2015) Complement factor H-related 5-hybrid proteins anchor properdin and activate complement at self-surfaces. J Am Soc Nephrol: JASN 27:1413–1425PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Zhang Y, Xiao X, Garcia-Fernandez J, de Cordoba SR, Zipfel P, Meyer N et al (2013) A novel fusion gene CHFR5-CFHR2 causes C3 glomerulonephritis. Mol Immunol 56:297–297CrossRef Zhang Y, Xiao X, Garcia-Fernandez J, de Cordoba SR, Zipfel P, Meyer N et al (2013) A novel fusion gene CHFR5-CFHR2 causes C3 glomerulonephritis. Mol Immunol 56:297–297CrossRef
102.
Zurück zum Zitat Tortajada A, Yebenes H, Abarrategui-Garrido C, Anter J, Garcia-Fernandez JM, Martinez-Barricarte R et al (2013) C3 glomerulopathy-associated CFHR1 mutation alters FHR oligomerization and complement regulation. J Clin Invest 123:2434–2446PubMedPubMedCentralCrossRef Tortajada A, Yebenes H, Abarrategui-Garrido C, Anter J, Garcia-Fernandez JM, Martinez-Barricarte R et al (2013) C3 glomerulopathy-associated CFHR1 mutation alters FHR oligomerization and complement regulation. J Clin Invest 123:2434–2446PubMedPubMedCentralCrossRef
103.
Zurück zum Zitat Malik TH, Lavin PJ, Goicoechea de Jorge E, Vernon KA, Rose KL, Patel MP et al (2012) A hybrid CFHR3-1 gene causes familial C3 glomerulopathy. J Am Soc Nephrol 23:1155–1160PubMedPubMedCentralCrossRef Malik TH, Lavin PJ, Goicoechea de Jorge E, Vernon KA, Rose KL, Patel MP et al (2012) A hybrid CFHR3-1 gene causes familial C3 glomerulopathy. J Am Soc Nephrol 23:1155–1160PubMedPubMedCentralCrossRef
104.
Zurück zum Zitat Togarsimalemath SK, Sethi SK, Duggal R, Quintrec ML, Jha P, Daniel R et al (2017) A novel CFHR1-CFHR5 hybrid leads to a familial dominant C3 glomerulopathy. Kidney Int 92:876–887PubMedCrossRef Togarsimalemath SK, Sethi SK, Duggal R, Quintrec ML, Jha P, Daniel R et al (2017) A novel CFHR1-CFHR5 hybrid leads to a familial dominant C3 glomerulopathy. Kidney Int 92:876–887PubMedCrossRef
105.
Zurück zum Zitat Xiao X, Ghossein C, Tortajada A, Zhang Y, Meyer N, Jones M et al (2016) Familial C3 glomerulonephritis caused by a novel CFHR5-CFHR2 fusion gene. Mol Immunol 77:89–96PubMedCrossRef Xiao X, Ghossein C, Tortajada A, Zhang Y, Meyer N, Jones M et al (2016) Familial C3 glomerulonephritis caused by a novel CFHR5-CFHR2 fusion gene. Mol Immunol 77:89–96PubMedCrossRef
106.
Zurück zum Zitat Besbas N, Gulhan B, Gucer S, Korkmaz E, Ozaltin F (2014) A novel CFHR5 mutation associated with C3 glomerulonephritis in a Turkish girl. J Nephrol 27:457–460PubMedCrossRef Besbas N, Gulhan B, Gucer S, Korkmaz E, Ozaltin F (2014) A novel CFHR5 mutation associated with C3 glomerulonephritis in a Turkish girl. J Nephrol 27:457–460PubMedCrossRef
107.
Zurück zum Zitat Abrera-Abeleda MA, Nishimura C, Smith JL, Sethi S, McRae JL, Murphy BF et al (2006) Variations in the complement regulatory genes factor H (CFH) and factor H related 5 (CFHR5) are associated with membranoproliferative glomerulonephritis type II (dense deposit disease). J Med Genet 43:582–589PubMedCrossRef Abrera-Abeleda MA, Nishimura C, Smith JL, Sethi S, McRae JL, Murphy BF et al (2006) Variations in the complement regulatory genes factor H (CFH) and factor H related 5 (CFHR5) are associated with membranoproliferative glomerulonephritis type II (dense deposit disease). J Med Genet 43:582–589PubMedCrossRef
108.
Zurück zum Zitat Abrera-Abeleda MA, Nishimura C, Frees K, Jones M, Maga T, Katz LM et al (2011) Allelic variants of complement genes associated with dense deposit disease. J Am Soc Nephrol: JASN 22:1551–1559PubMedPubMedCentralCrossRef Abrera-Abeleda MA, Nishimura C, Frees K, Jones M, Maga T, Katz LM et al (2011) Allelic variants of complement genes associated with dense deposit disease. J Am Soc Nephrol: JASN 22:1551–1559PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Daha MR, Fearon DT, Austen KF (1976) C3 nephritic factor (C3NeF): stabilization of fluid phase and cell-bound alternative pathway convertase. J Immunol 116:1–7PubMed Daha MR, Fearon DT, Austen KF (1976) C3 nephritic factor (C3NeF): stabilization of fluid phase and cell-bound alternative pathway convertase. J Immunol 116:1–7PubMed
111.
Zurück zum Zitat Daha MR, Austen KF, Fearon DT (1977) The incorporation of C3 nephritic factor (C3NeF) into a stabilized C3 convertase, C3bBb (C3NeF), and its release after decay of convertase function. J Immunol 119:812–817PubMed Daha MR, Austen KF, Fearon DT (1977) The incorporation of C3 nephritic factor (C3NeF) into a stabilized C3 convertase, C3bBb (C3NeF), and its release after decay of convertase function. J Immunol 119:812–817PubMed
112.
Zurück zum Zitat Sissons JG, West RJ, Fallows J, Williams DG, Boucher BJ, Amos N et al (1976) The complement abnormalities of lipodystrophy. N Engl J Med 294:461–465PubMedCrossRef Sissons JG, West RJ, Fallows J, Williams DG, Boucher BJ, Amos N et al (1976) The complement abnormalities of lipodystrophy. N Engl J Med 294:461–465PubMedCrossRef
113.
Zurück zum Zitat Gewurz AT, Imherr SM, Strauss S, Gewurz H, Mold C (1983) C3 nephritic factor and hypocomplementaemia in a clinically healthy individual. Clin Exp Immunol 54:253–258PubMedPubMedCentral Gewurz AT, Imherr SM, Strauss S, Gewurz H, Mold C (1983) C3 nephritic factor and hypocomplementaemia in a clinically healthy individual. Clin Exp Immunol 54:253–258PubMedPubMedCentral
114.
Zurück zum Zitat Strobel S, Zimmering M, Papp K, Prechl J, Jozsi M (2010) Anti-factor B autoantibody in dense deposit disease. Mol Immunol 47:1476–1483PubMedCrossRef Strobel S, Zimmering M, Papp K, Prechl J, Jozsi M (2010) Anti-factor B autoantibody in dense deposit disease. Mol Immunol 47:1476–1483PubMedCrossRef
115.
Zurück zum Zitat Chen Q, Muller D, Rudolph B, Hartmann A, Kuwertz-Broking E, Wu K et al (2011) Combined C3b and factor B autoantibodies and MPGN type II. N Engl J Med 365:2340–2342PubMedCrossRef Chen Q, Muller D, Rudolph B, Hartmann A, Kuwertz-Broking E, Wu K et al (2011) Combined C3b and factor B autoantibodies and MPGN type II. N Engl J Med 365:2340–2342PubMedCrossRef
116.
Zurück zum Zitat Goodship TH, Pappworth IY, Toth T, Denton M, Houlberg K, McCormick F et al (2012) Factor H autoantibodies in membranoproliferative glomerulonephritis. Mol Immunol 52:200–206PubMedCrossRef Goodship TH, Pappworth IY, Toth T, Denton M, Houlberg K, McCormick F et al (2012) Factor H autoantibodies in membranoproliferative glomerulonephritis. Mol Immunol 52:200–206PubMedCrossRef
117.
Zurück zum Zitat Blanc C, Togarsimalemath SK, Chauvet S, Le Quintrec M, Moulin B, Buchler M et al (2015) Anti-factor H autoantibodies in C3 glomerulopathies and in atypical hemolytic uremic syndrome: one target, two diseases. J Immunol 194:5129–5138PubMedCrossRef Blanc C, Togarsimalemath SK, Chauvet S, Le Quintrec M, Moulin B, Buchler M et al (2015) Anti-factor H autoantibodies in C3 glomerulopathies and in atypical hemolytic uremic syndrome: one target, two diseases. J Immunol 194:5129–5138PubMedCrossRef
118.
Zurück zum Zitat Meri S, Koistinen V, Miettinen A, Tornroth T, Seppala IJ (1992) Activation of the alternative pathway of complement by monoclonal lambda light chains in membranoproliferative glomerulonephritis. J Exp Med 175:939–950PubMedCrossRef Meri S, Koistinen V, Miettinen A, Tornroth T, Seppala IJ (1992) Activation of the alternative pathway of complement by monoclonal lambda light chains in membranoproliferative glomerulonephritis. J Exp Med 175:939–950PubMedCrossRef
119.
Zurück zum Zitat Zand L, Kattah A, Fervenza FC, Smith RJ, Nasr SH, Zhang Y et al (2013) C3 glomerulonephritis associated with monoclonal gammopathy: a case series. Am J Kidney Dis: Off J Natl Kidney Found 62:506–514CrossRef Zand L, Kattah A, Fervenza FC, Smith RJ, Nasr SH, Zhang Y et al (2013) C3 glomerulonephritis associated with monoclonal gammopathy: a case series. Am J Kidney Dis: Off J Natl Kidney Found 62:506–514CrossRef
120.
Zurück zum Zitat Cansick JC, Lennon R, Cummins CL, Howie AJ, McGraw ME, Saleem MA et al (2004) Prognosis, treatment and outcome of childhood mesangiocapillary (membranoproliferative) glomerulonephritis. Nephrol Dial Transplant 19:2769–2777PubMedCrossRef Cansick JC, Lennon R, Cummins CL, Howie AJ, McGraw ME, Saleem MA et al (2004) Prognosis, treatment and outcome of childhood mesangiocapillary (membranoproliferative) glomerulonephritis. Nephrol Dial Transplant 19:2769–2777PubMedCrossRef
121.
Zurück zum Zitat Braun MC, Stablein DM, Hamiwka LA, Bell L, Bartosh SM, Strife CF (2005) Recurrence of membranoproliferative glomerulonephritis type II in renal allografts: the North American Pediatric Renal Transplant Cooperative Study experience. J Am Soc Nephrol: JASN 16:2225–2233PubMedCrossRef Braun MC, Stablein DM, Hamiwka LA, Bell L, Bartosh SM, Strife CF (2005) Recurrence of membranoproliferative glomerulonephritis type II in renal allografts: the North American Pediatric Renal Transplant Cooperative Study experience. J Am Soc Nephrol: JASN 16:2225–2233PubMedCrossRef
122.
Zurück zum Zitat Varade WS, Forristal J, West CD (1990) Patterns of complement activation in idiopathic membranoproliferative glomerulonephritis, types I, II, and III. Am J Kidney Dis: Off J Natl Kidney Found 16:196–206CrossRef Varade WS, Forristal J, West CD (1990) Patterns of complement activation in idiopathic membranoproliferative glomerulonephritis, types I, II, and III. Am J Kidney Dis: Off J Natl Kidney Found 16:196–206CrossRef
123.
Zurück zum Zitat Eisinger AJ, Shortland JR, Moorhead PJ (1972) Renal disease in partial lipodystrophy. Q J Med 41:343–354PubMed Eisinger AJ, Shortland JR, Moorhead PJ (1972) Renal disease in partial lipodystrophy. Q J Med 41:343–354PubMed
124.
Zurück zum Zitat Misra A, Peethambaram A, Garg A (2004) Clinical features and metabolic and autoimmune derangements in acquired partial lipodystrophy: report of 35 cases and review of the literature. Medicine 83:18–34PubMedCrossRef Misra A, Peethambaram A, Garg A (2004) Clinical features and metabolic and autoimmune derangements in acquired partial lipodystrophy: report of 35 cases and review of the literature. Medicine 83:18–34PubMedCrossRef
125.
Zurück zum Zitat Hageman GS, Mullins RF, Russell SR, Johnson LV, Anderson DH (1999) Vitronectin is a constituent of ocular drusen and the vitronectin gene is expressed in human retinal pigmented epithelial cells. FASEB J 13:477–484PubMedCrossRef Hageman GS, Mullins RF, Russell SR, Johnson LV, Anderson DH (1999) Vitronectin is a constituent of ocular drusen and the vitronectin gene is expressed in human retinal pigmented epithelial cells. FASEB J 13:477–484PubMedCrossRef
126.
Zurück zum Zitat Tarshish P, Bernstein J, Tobin JN, Edelmann CM Jr (1992) Treatment of mesangiocapillary glomerulonephritis with alternate-day prednisone—a report of the International Study of Kidney Disease in Children. Pediatr Nephrol 6:123–130PubMedCrossRef Tarshish P, Bernstein J, Tobin JN, Edelmann CM Jr (1992) Treatment of mesangiocapillary glomerulonephritis with alternate-day prednisone—a report of the International Study of Kidney Disease in Children. Pediatr Nephrol 6:123–130PubMedCrossRef
127.
Zurück zum Zitat Faedda R, Satta A, Tanda F, Pirisi M, Bartoli E (1994) Immunosuppressive treatment of membranoproliferative glomerulonephritis. Nephron 67:59–65PubMedCrossRef Faedda R, Satta A, Tanda F, Pirisi M, Bartoli E (1994) Immunosuppressive treatment of membranoproliferative glomerulonephritis. Nephron 67:59–65PubMedCrossRef
128.
Zurück zum Zitat Jones G, Juszczak M, Kingdon E, Harber M, Sweny P, Burns A (2004) Treatment of idiopathic membranoproliferative glomerulonephritis with mycophenolate mofetil and steroids. Nephrol Dial Transplant 19:3160–3164PubMedCrossRef Jones G, Juszczak M, Kingdon E, Harber M, Sweny P, Burns A (2004) Treatment of idiopathic membranoproliferative glomerulonephritis with mycophenolate mofetil and steroids. Nephrol Dial Transplant 19:3160–3164PubMedCrossRef
129.
Zurück zum Zitat Rabasco C, Cavero T, Roman E, Rojas-Rivera J, Olea T, Espinosa M et al (2015) Effectiveness of mycophenolate mofetil in C3 glomerulonephritis. Kidney Int 88:1153–1160PubMedCrossRef Rabasco C, Cavero T, Roman E, Rojas-Rivera J, Olea T, Espinosa M et al (2015) Effectiveness of mycophenolate mofetil in C3 glomerulonephritis. Kidney Int 88:1153–1160PubMedCrossRef
130.
Zurück zum Zitat Zauner I, Bohler J, Braun N, Grupp C, Heering P, Schollmeyer P (1994) Effect of aspirin and dipyridamole on proteinuria in idiopathic membranoproliferative glomerulonephritis: a multicentre prospective clinical trial. Collaborative Glomerulonephritis Therapy Study Group (CGTS). Nephrol Dial Transplant 9:619–622PubMedCrossRef Zauner I, Bohler J, Braun N, Grupp C, Heering P, Schollmeyer P (1994) Effect of aspirin and dipyridamole on proteinuria in idiopathic membranoproliferative glomerulonephritis: a multicentre prospective clinical trial. Collaborative Glomerulonephritis Therapy Study Group (CGTS). Nephrol Dial Transplant 9:619–622PubMedCrossRef
131.
Zurück zum Zitat Donadio JV Jr, Anderson CF, Mitchell JC 3rd, Holley KE, Ilstrup DM, Fuster V et al (1984) Membranoproliferative glomerulonephritis. A prospective clinical trial of platelet-inhibitor therapy. N Engl J Med 310:1421–1426PubMedCrossRef Donadio JV Jr, Anderson CF, Mitchell JC 3rd, Holley KE, Ilstrup DM, Fuster V et al (1984) Membranoproliferative glomerulonephritis. A prospective clinical trial of platelet-inhibitor therapy. N Engl J Med 310:1421–1426PubMedCrossRef
132.
Zurück zum Zitat Donadio JV Jr, Offord KP (1989) Reassessment of treatment results in membranoproliferative glomerulonephritis, with emphasis on life-table analysis. Am J Kidney Dis: Official J Natl Kidney Found 14:445–451CrossRef Donadio JV Jr, Offord KP (1989) Reassessment of treatment results in membranoproliferative glomerulonephritis, with emphasis on life-table analysis. Am J Kidney Dis: Official J Natl Kidney Found 14:445–451CrossRef
133.
Zurück zum Zitat Farooqui M, Alsaad K, Aloudah N, Alhamdan H (2015) Treatment-resistant recurrent membranoproliferative glomerulonephritis in renal allograft responding to rituximab: case report. Transplant Proc 47:823–826PubMedCrossRef Farooqui M, Alsaad K, Aloudah N, Alhamdan H (2015) Treatment-resistant recurrent membranoproliferative glomerulonephritis in renal allograft responding to rituximab: case report. Transplant Proc 47:823–826PubMedCrossRef
134.
Zurück zum Zitat Marques ID, Ramalho J, David DR, Nahas WC, David-Neto E (2014) Rituximab in a B cell-driven regimen for the treatment of recurrent membranoproliferative glomerulonephritis after kidney transplantation. Int Urol Nephrol 46:2053–2054PubMedCrossRef Marques ID, Ramalho J, David DR, Nahas WC, David-Neto E (2014) Rituximab in a B cell-driven regimen for the treatment of recurrent membranoproliferative glomerulonephritis after kidney transplantation. Int Urol Nephrol 46:2053–2054PubMedCrossRef
135.
Zurück zum Zitat Giaime P, Daniel L, Burtey S (2015) Remission of C3 glomerulopathy with rituximab as only immunosuppressive therapy. Clin Nephrol 83:57–60PubMedCrossRef Giaime P, Daniel L, Burtey S (2015) Remission of C3 glomerulopathy with rituximab as only immunosuppressive therapy. Clin Nephrol 83:57–60PubMedCrossRef
137.
Zurück zum Zitat Bomback AS, Smith RJ, Barile GR, Zhang Y, Heher EC, Herlitz L et al (2012) Eculizumab for dense deposit disease and C3 glomerulonephritis. Clin J Am Soc Nephrol: CJASN 7:748–756PubMedPubMedCentralCrossRef Bomback AS, Smith RJ, Barile GR, Zhang Y, Heher EC, Herlitz L et al (2012) Eculizumab for dense deposit disease and C3 glomerulonephritis. Clin J Am Soc Nephrol: CJASN 7:748–756PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat McCaughan JA, O’Rourke DM, Courtney AE (2012) Recurrent dense deposit disease after renal transplantation: an emerging role for complementary therapies. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg 12:1046–1051CrossRef McCaughan JA, O’Rourke DM, Courtney AE (2012) Recurrent dense deposit disease after renal transplantation: an emerging role for complementary therapies. Am J Transplant Off J Am Soc Transplant Am Soc Transplant Surg 12:1046–1051CrossRef
139.
Zurück zum Zitat Rousset-Rouviere C, Cailliez M, Garaix F, Bruno D, Laurent D, Tsimaratos M (2014) Rituximab fails where eculizumab restores renal function in C3nef-related DDD. Pediatr Nephrol 29:1107–1111PubMedCrossRef Rousset-Rouviere C, Cailliez M, Garaix F, Bruno D, Laurent D, Tsimaratos M (2014) Rituximab fails where eculizumab restores renal function in C3nef-related DDD. Pediatr Nephrol 29:1107–1111PubMedCrossRef
140.
Zurück zum Zitat Le Quintrec M, Lionet A, Kandel C, Bourdon F, Gnemmi V, Colombat M et al (2015) Eculizumab for treatment of rapidly progressive C3 glomerulopathy. Am J Kidney Dis: Off J Natl Kidney Found 65:484–489CrossRef Le Quintrec M, Lionet A, Kandel C, Bourdon F, Gnemmi V, Colombat M et al (2015) Eculizumab for treatment of rapidly progressive C3 glomerulopathy. Am J Kidney Dis: Off J Natl Kidney Found 65:484–489CrossRef
141.
Zurück zum Zitat Inman M, Prater G, Fatima H, Wallace E (2015) Eculizumab-induced reversal of dialysis-dependent kidney failure from C3 glomerulonephritis. Clin Kidney J 8:445–448PubMedPubMedCentralCrossRef Inman M, Prater G, Fatima H, Wallace E (2015) Eculizumab-induced reversal of dialysis-dependent kidney failure from C3 glomerulonephritis. Clin Kidney J 8:445–448PubMedPubMedCentralCrossRef
142.
Zurück zum Zitat Habbig S, Mihatsch MJ, Heinen S, Beck B, Emmel M, Skerka C et al (2009) C3 deposition glomerulopathy due to a functional factor H defect. Kidney Int 75:1230–1234PubMedCrossRef Habbig S, Mihatsch MJ, Heinen S, Beck B, Emmel M, Skerka C et al (2009) C3 deposition glomerulopathy due to a functional factor H defect. Kidney Int 75:1230–1234PubMedCrossRef
143.
Zurück zum Zitat Zhang Y, Nester CM, Holanda DG, Marsh HC, Hammond RA, Thomas LJ et al (2013) Soluble CR1 therapy improves complement regulation in C3 glomerulopathy. J Am Soc Nephrol: JASN 24:1820–1829PubMedPubMedCentralCrossRef Zhang Y, Nester CM, Holanda DG, Marsh HC, Hammond RA, Thomas LJ et al (2013) Soluble CR1 therapy improves complement regulation in C3 glomerulopathy. J Am Soc Nephrol: JASN 24:1820–1829PubMedPubMedCentralCrossRef
144.
Zurück zum Zitat Strubing P (1882) Paroxysmale haemoglobinurie. Dtsch Med Wochenschr 8:1–16CrossRef Strubing P (1882) Paroxysmale haemoglobinurie. Dtsch Med Wochenschr 8:1–16CrossRef
145.
Zurück zum Zitat Hill A, DeZern AE, Kinoshita T, Brodsky RA (2017) Paroxysmal nocturnal haemoglobinuria. Nat Rev Dis Primers 3:17028PubMedCrossRef Hill A, DeZern AE, Kinoshita T, Brodsky RA (2017) Paroxysmal nocturnal haemoglobinuria. Nat Rev Dis Primers 3:17028PubMedCrossRef
146.
Zurück zum Zitat Socie G, Schrezenmeier H, Muus P, Lisukov I, Roth A, Kulasekararaj A et al (2016) Changing prognosis in paroxysmal nocturnal haemoglobinuria disease subcategories: an analysis of the International PNH Registry. Intern Med J 46:1044–1053PubMedCrossRef Socie G, Schrezenmeier H, Muus P, Lisukov I, Roth A, Kulasekararaj A et al (2016) Changing prognosis in paroxysmal nocturnal haemoglobinuria disease subcategories: an analysis of the International PNH Registry. Intern Med J 46:1044–1053PubMedCrossRef
147.
Zurück zum Zitat Ware RE, Hall SE, Rosse WF (1991) Paroxysmal nocturnal hemoglobinuria with onset in childhood and adolescence. N Engl J Med 325:991–996PubMedCrossRef Ware RE, Hall SE, Rosse WF (1991) Paroxysmal nocturnal hemoglobinuria with onset in childhood and adolescence. N Engl J Med 325:991–996PubMedCrossRef
148.
Zurück zum Zitat Takeda J, Miyata T, Kawagoe K, Iida Y, Endo Y, Fujita T et al (1993) Deficiency of the GPI anchor caused by a somatic mutation of the PIG-A gene in paroxysmal nocturnal hemoglobinuria. Cell 73:703–711PubMedCrossRef Takeda J, Miyata T, Kawagoe K, Iida Y, Endo Y, Fujita T et al (1993) Deficiency of the GPI anchor caused by a somatic mutation of the PIG-A gene in paroxysmal nocturnal hemoglobinuria. Cell 73:703–711PubMedCrossRef
149.
Zurück zum Zitat Oni SB, Osunkoya BO, Luzzatto L (1970) Paroxysmal nocturnal hemoglobinuria: evidence for monoclonal origin of abnormal red cells. Blood 36:145–152PubMed Oni SB, Osunkoya BO, Luzzatto L (1970) Paroxysmal nocturnal hemoglobinuria: evidence for monoclonal origin of abnormal red cells. Blood 36:145–152PubMed
150.
Zurück zum Zitat Kinoshita T, Medof ME, Silber R, Nussenzweig V (1985) Distribution of decay-accelerating factor in the peripheral blood of normal individuals and patients with paroxysmal nocturnal hemoglobinuria. J Exp Med 162:75–92PubMedCrossRef Kinoshita T, Medof ME, Silber R, Nussenzweig V (1985) Distribution of decay-accelerating factor in the peripheral blood of normal individuals and patients with paroxysmal nocturnal hemoglobinuria. J Exp Med 162:75–92PubMedCrossRef
151.
Zurück zum Zitat Nafa K, Bessler M, Castro-Malaspina H, Jhanwar S, Luzzatto L (1998) The spectrum of somatic mutations in the PIG-A gene in paroxysmal nocturnal hemoglobinuria includes large deletions and small duplications. Blood Cells Mol Dis 24:370–384PubMedCrossRef Nafa K, Bessler M, Castro-Malaspina H, Jhanwar S, Luzzatto L (1998) The spectrum of somatic mutations in the PIG-A gene in paroxysmal nocturnal hemoglobinuria includes large deletions and small duplications. Blood Cells Mol Dis 24:370–384PubMedCrossRef
152.
Zurück zum Zitat Nafa K, Mason PJ, Hillmen P, Luzzatto L, Bessler M (1995) Mutations in the PIG-A gene causing paroxysmal nocturnal hemoglobinuria are mainly of the frameshift type. Blood 86:4650–4655PubMed Nafa K, Mason PJ, Hillmen P, Luzzatto L, Bessler M (1995) Mutations in the PIG-A gene causing paroxysmal nocturnal hemoglobinuria are mainly of the frameshift type. Blood 86:4650–4655PubMed
153.
Zurück zum Zitat Luzzatto L (1999) Paroxysmal murine hemoglobinuria(?): a model for human PNH. Blood 94:2941–2944PubMed Luzzatto L (1999) Paroxysmal murine hemoglobinuria(?): a model for human PNH. Blood 94:2941–2944PubMed
154.
Zurück zum Zitat Rosti V (2002) Murine models of paroxysmal nocturnal hemoglobinuria. Ann N Y Acad Sci 963:290–296PubMedCrossRef Rosti V (2002) Murine models of paroxysmal nocturnal hemoglobinuria. Ann N Y Acad Sci 963:290–296PubMedCrossRef
155.
Zurück zum Zitat Inoue N, Izui-Sarumaru T, Murakami Y, Endo Y, Nishimura J, Kurokawa K et al (2006) Molecular basis of clonal expansion of hematopoiesis in 2 patients with paroxysmal nocturnal hemoglobinuria (PNH). Blood 108:4232–4236PubMedPubMedCentralCrossRef Inoue N, Izui-Sarumaru T, Murakami Y, Endo Y, Nishimura J, Kurokawa K et al (2006) Molecular basis of clonal expansion of hematopoiesis in 2 patients with paroxysmal nocturnal hemoglobinuria (PNH). Blood 108:4232–4236PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Rotoli B, Luzzatto L (1989) Paroxysmal nocturnal hemoglobinuria. Semin Hematol 26:201–207PubMed Rotoli B, Luzzatto L (1989) Paroxysmal nocturnal hemoglobinuria. Semin Hematol 26:201–207PubMed
157.
Zurück zum Zitat Ohishi K, Inoue N, Kinoshita T (2001) PIG-S and PIG-T, essential for GPI anchor attachment to proteins, form a complex with GAA1 and GPI8. EMBO J 20:4088–4098PubMedPubMedCentralCrossRef Ohishi K, Inoue N, Kinoshita T (2001) PIG-S and PIG-T, essential for GPI anchor attachment to proteins, form a complex with GAA1 and GPI8. EMBO J 20:4088–4098PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Krawitz PM, Hochsmann B, Murakami Y, Teubner B, Kruger U, Klopocki E et al (2013) A case of paroxysmal nocturnal hemoglobinuria caused by a germline mutation and a somatic mutation in PIGT. Blood 122:1312–1315PubMedCrossRef Krawitz PM, Hochsmann B, Murakami Y, Teubner B, Kruger U, Klopocki E et al (2013) A case of paroxysmal nocturnal hemoglobinuria caused by a germline mutation and a somatic mutation in PIGT. Blood 122:1312–1315PubMedCrossRef
159.
160.
Zurück zum Zitat Hill A, Kelly RJ, Hillmen P (2013) Thrombosis in paroxysmal nocturnal hemoglobinuria. Blood 121:4985–4996PubMedCrossRef Hill A, Kelly RJ, Hillmen P (2013) Thrombosis in paroxysmal nocturnal hemoglobinuria. Blood 121:4985–4996PubMedCrossRef
161.
Zurück zum Zitat Wong EK, Kavanagh D (2015) Anticomplement C5 therapy with eculizumab for the treatment of paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Transl Res 165:306–320PubMedCrossRef Wong EK, Kavanagh D (2015) Anticomplement C5 therapy with eculizumab for the treatment of paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. Transl Res 165:306–320PubMedCrossRef
162.
Zurück zum Zitat Rother RP, Bell L, Hillmen P, Gladwin MT (2005) The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease. JAMA 293:1653–1662PubMedCrossRef Rother RP, Bell L, Hillmen P, Gladwin MT (2005) The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease. JAMA 293:1653–1662PubMedCrossRef
163.
Zurück zum Zitat Clark DA, Butler SA, Braren V, Hartmann RC, Jenkins DE Jr (1981) The kidneys in paroxysmal nocturnal hemoglobinuria. Blood 57:83–89PubMed Clark DA, Butler SA, Braren V, Hartmann RC, Jenkins DE Jr (1981) The kidneys in paroxysmal nocturnal hemoglobinuria. Blood 57:83–89PubMed
164.
Zurück zum Zitat Nath KA, Vercellotti GM, Grande JP, Miyoshi H, Paya CV, Manivel JC et al (2001) Heme protein-induced chronic renal inflammation: suppressive effect of induced heme oxygenase-1. Kidney Int 59:106–117PubMedCrossRef Nath KA, Vercellotti GM, Grande JP, Miyoshi H, Paya CV, Manivel JC et al (2001) Heme protein-induced chronic renal inflammation: suppressive effect of induced heme oxygenase-1. Kidney Int 59:106–117PubMedCrossRef
165.
Zurück zum Zitat de Latour RP, Mary JY, Salanoubat C, Terriou L, Etienne G, Mohty M et al (2008) Paroxysmal nocturnal hemoglobinuria: natural history of disease subcategories. Blood 112:3099–3106PubMedCrossRef de Latour RP, Mary JY, Salanoubat C, Terriou L, Etienne G, Mohty M et al (2008) Paroxysmal nocturnal hemoglobinuria: natural history of disease subcategories. Blood 112:3099–3106PubMedCrossRef
166.
Zurück zum Zitat Socie G, Mary JY, de Gramont A, Rio B, Leporrier M, Rose C et al (1996) Paroxysmal nocturnal haemoglobinuria: long-term follow-up and prognostic factors. French Society of Haematology. Lancet 348:573–577PubMedCrossRef Socie G, Mary JY, de Gramont A, Rio B, Leporrier M, Rose C et al (1996) Paroxysmal nocturnal haemoglobinuria: long-term follow-up and prognostic factors. French Society of Haematology. Lancet 348:573–577PubMedCrossRef
167.
Zurück zum Zitat Hillmen P, Lewis SM, Bessler M, Luzzatto L, Dacie JV (1995) Natural history of paroxysmal nocturnal hemoglobinuria. N Engl J Med 333:1253–1258PubMedCrossRef Hillmen P, Lewis SM, Bessler M, Luzzatto L, Dacie JV (1995) Natural history of paroxysmal nocturnal hemoglobinuria. N Engl J Med 333:1253–1258PubMedCrossRef
168.
Zurück zum Zitat Kelly RJ, Hill A, Arnold LM, Brooksbank GL, Richards SJ, Cullen M et al (2011) Long-term treatment with eculizumab in paroxysmal nocturnal hemoglobinuria: sustained efficacy and improved survival. Blood 117:6786–6792PubMedCrossRef Kelly RJ, Hill A, Arnold LM, Brooksbank GL, Richards SJ, Cullen M et al (2011) Long-term treatment with eculizumab in paroxysmal nocturnal hemoglobinuria: sustained efficacy and improved survival. Blood 117:6786–6792PubMedCrossRef
169.
Zurück zum Zitat Hillmen P, Young NS, Schubert J, Brodsky RA, Socie G, Muus P et al (2006) The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 355:1233–1243PubMedCrossRef Hillmen P, Young NS, Schubert J, Brodsky RA, Socie G, Muus P et al (2006) The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 355:1233–1243PubMedCrossRef
170.
Zurück zum Zitat Hill A, Hillmen P, Richards SJ, Elebute D, Marsh JC, Chan J et al (2005) Sustained response and long-term safety of eculizumab in paroxysmal nocturnal hemoglobinuria. Blood 106:2559–2565PubMedCrossRef Hill A, Hillmen P, Richards SJ, Elebute D, Marsh JC, Chan J et al (2005) Sustained response and long-term safety of eculizumab in paroxysmal nocturnal hemoglobinuria. Blood 106:2559–2565PubMedCrossRef
171.
Zurück zum Zitat Hillmen P, Muus P, Duhrsen U, Risitano AM, Schubert J, Luzzatto L et al (2007) Effect of the complement inhibitor eculizumab on thromboembolism in patients with paroxysmal nocturnal hemoglobinuria. Blood 110:4123–4128PubMedCrossRef Hillmen P, Muus P, Duhrsen U, Risitano AM, Schubert J, Luzzatto L et al (2007) Effect of the complement inhibitor eculizumab on thromboembolism in patients with paroxysmal nocturnal hemoglobinuria. Blood 110:4123–4128PubMedCrossRef
172.
Zurück zum Zitat Hillmen P, Elebute M, Kelly R, Urbano-Ispizua A, Hill A, Rother RP et al (2010) Long-term effect of the complement inhibitor eculizumab on kidney function in patients with paroxysmal nocturnal hemoglobinuria. Am J Hematol 85:553–559PubMedCrossRef Hillmen P, Elebute M, Kelly R, Urbano-Ispizua A, Hill A, Rother RP et al (2010) Long-term effect of the complement inhibitor eculizumab on kidney function in patients with paroxysmal nocturnal hemoglobinuria. Am J Hematol 85:553–559PubMedCrossRef
173.
Zurück zum Zitat Risitano AM, Notaro R, Marando L, Serio B, Ranaldi D, Seneca E et al (2009) Complement fraction 3 binding on erythrocytes as additional mechanism of disease in paroxysmal nocturnal hemoglobinuria patients treated by eculizumab. Blood 113:4094–4100PubMedCrossRef Risitano AM, Notaro R, Marando L, Serio B, Ranaldi D, Seneca E et al (2009) Complement fraction 3 binding on erythrocytes as additional mechanism of disease in paroxysmal nocturnal hemoglobinuria patients treated by eculizumab. Blood 113:4094–4100PubMedCrossRef
174.
Zurück zum Zitat Rondelli T, Risitano AM, Peffault de Latour R, Sica M, Peruzzi B, Ricci P et al (2014) Polymorphism of the complement receptor 1 gene correlates with the hematologic response to eculizumab in patients with paroxysmal nocturnal hemoglobinuria. Haematologica 99:262–266PubMedPubMedCentralCrossRef Rondelli T, Risitano AM, Peffault de Latour R, Sica M, Peruzzi B, Ricci P et al (2014) Polymorphism of the complement receptor 1 gene correlates with the hematologic response to eculizumab in patients with paroxysmal nocturnal hemoglobinuria. Haematologica 99:262–266PubMedPubMedCentralCrossRef
Metadaten
Titel
Diseases of complement dysregulation—an overview
verfasst von
Edwin K. S. Wong
David Kavanagh
Publikationsdatum
11.01.2018
Verlag
Springer Berlin Heidelberg
Erschienen in
Seminars in Immunopathology / Ausgabe 1/2018
Print ISSN: 1863-2297
Elektronische ISSN: 1863-2300
DOI
https://doi.org/10.1007/s00281-017-0663-8

Weitere Artikel der Ausgabe 1/2018

Seminars in Immunopathology 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.