ArticlePDF Available

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

Authors:

Abstract and Figures

The regulators of complement activation cluster at chromosome 1q32 contains the complement factor H (CFH) and five complement factor H-related (CFHR) genes. This area of the genome arose from several large genomic duplications, and these low-copy repeats can cause genome instability in this region. Genomic disorders affecting these genes have been described in atypical hemolytic uremic syndrome, arising commonly through nonallelic homologous recombination. We describe a novel CFH/CFHR3 hybrid gene secondary to a de novo 6.3-kb deletion that arose through microhomology-mediated end joining rather than nonallelic homologous recombination. We confirmed a transcript from this hybrid gene and showed a secreted protein product that lacks the recognition domain of factor H and exhibits impaired cell surface complement regulation. The fact that the formation of this hybrid gene arose as a de novo event suggests that this cluster is a dynamic area of the genome in which additional genomic disorders may arise.
Identification of a novel FH/FHR3 hybrid protein in patient serum. (A) The protein product of wild-type FH and the predicted FH/FHR3 hybrid protein consisting of CCP1-17 of FH and CCP1-5 of FHR3. The CCPs originating from FHR3 are highlighted in gray. The patient is heterozygous for a common polymorphism in CCP7 of FH (Y402H). Binding epitopes for the mAbs are shown: Ox24-CCP5, MBI7-CCP7 amino acid 402 histidine variant, MBI6-CCP7 amino acid 402 tyrosine variant, and L20/3-CCP20. (B) Serum Western blot using OX24 showing two additional bands in the patient in addition to FH. The upper band is consistent with the predicted size of the FH/FHR3 hybrid. (C) Serum Western blot using L20/3. When using this FH C terminus-specific antibody, no additional bands are seen, consistent with the hybrid protein lacking CCP18-20. (D) Serum Western blot using MBI7. This shows that the patient has a normal allele producing FH with the histidine at amino acid 402. (E) Serum Western blot using MBI6. The patient has three additional bands and no wild-type FH band. This is consistent with the FH/FHR3 hybrid protein carrying the tyrosine amino acid at position 402. The two additional upper bands represent differentially glycosylated hybrid protein, consistent with that seen in the native FHR3. There is a faint degradation product only detected with the MBI6 antibody. This is consistent with the breakdown product being generated only from the hybrid protein and suggests a less stable protein product. The controls were unaffected, unrelated, genotyped samples. In B and C, a sample from an individual heterozygous at Y402H was used. In D and E, samples from individuals homozygous for Y402 (Y/Y), homozygous for H402 (H/H), or heterozygous (Y/H) were used.
… 
Content may be subject to copyright.
BRIEF COMMUNICATION www.jasn.org
ADe Novo Deletion in the Regulators of Complement
Activation Cluster Producing a Hybrid Complement
Factor H/Complement Factor HRelated 3 Gene in
Atypical Hemolytic Uremic Syndrome
Rachel C. Challis,* Geisilaine S.R. Araujo,* Edwin K.S. Wong,* Holly E. Anderson,* Atif Awan,
Anthony M. Dorman,
Mary Waldron,
Valerie Wilson,* Vicky Brocklebank,* Lisa Strain,*
B. Paul Morgan,
§
Claire L. Harris,
§
Kevin J. Marchbank,
|
Timothy H.J. Goodship,* and
David Kavanagh*
*Institutes of Genetic Medicine and
|
Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom;
Department of Nephrology, Our Ladys Childrens Hospital, Crumlin, Dublin;
Department of Renal Pathology,
Beaumont Hospital, Royal College of Surgeons in Ireland, Dublin, Ireland; and
§
Institute of Infection and Immunity,
Cardiff University School of Medicine, Cardiff, United Kingdom
ABSTRACT
The regulators of complement activation cluster at chromosome 1q32 contains the
complement factor H (CFH)andve complement factor Hrelated (CFHR)genes.
This area of the genome arose from several large genomic duplications, and these
low-copy repeats can cause genome instability in this region. Genomic disorders
affecting these genes have been described in atypical hemolytic uremic syndrome,
arising commonly through nonallelic homologous recombination. We describe a
novel CFH/CFHR3 hybrid gene secondary to a de novo 6.3-kb deletion that arose
through microhomologymediated end joining rather than nonallelic homologous
recombination. We conrmed a transcript from this hybrid gene and showed a se-
creted protein product that lacks the recognitiondomainoffactorHandexhibits
impaired cell surface complement regulation. The fact that the formation of this
hybrid gene arose as a de novo event suggests that this cluster is a dynamic area of
the genome in which additional genomic disorders may arise.
J Am Soc Nephrol 27: cccccc, 2015. doi: 10.1681/ASN.2015010100
The complement factor H (CFH)and
complement factor Hrelated (CFHR1
CFHR5) genes reside in a 360-kb region
in the regulators of complement activa-
tion (RCA) cluster on chromosome
1q32 (Figure 1A).
1
This is an area of
the genome that arose from several large
genomic duplications,
2,3
and these low-
copy repeats can cause genome instabil-
ity in this region.
Mutations in CFH are the most com-
mon genetic predispositions to the
thrombotic microangiopathy: atypical he-
molytic uremic syndrome (aHUS).
4
The
majority of mutations in CFH occur at the
C-terminal end responsible for host poly-
anion binding.
57
Several of these CFH
mutations (e.g., S1191L and V1197A)
have been shown to be gene conversion
events between CFH and CFHR1.
8
Non-
allelic homologous recombination involv-
ing CFH and CFHR1 can result in CFH/
CFHR1 hybrid genes (Supplemental Fig-
ure 1).
9,10
A single family with a hybrid
CFH/CFHR3 gene (factor H [FH] protein
complement control protein modules
[CCPs] 119 and factor Hrelated 3
[FHR3] CCP1CCP5) has been reported
to arise through microhomology
mediated end joining (MMEJ)
11
(Supple-
mental Figure 1). These genetic variants
have all been shown to impair C3b/host
polyanion binding on host cells, thus im-
pairing local complement regulation.
5,8,11
Recently, a reverse CFHR1/CFH
hybrid gene arising through nonallelic
homologous recombination has been
described in aHUS. This encodes an
FHR1, where the C-terminal CCPs are
replaced by the C-terminal CCPs of
FH.
12,13
Unlike previously reported hy-
brid proteins, this does not impair FH
cell surface binding but instead, acts as a
competitive inhibitor of FH.
13
Received January 27, 2015. Accepted September
7, 2015.
R.C.C., G.S.R.A., and E.K.S.W. contributed equally
to this work.
Published online ahead of print. Publication date
available at www.jasn.org.
Correspondence: Dr.DavidKavanagh,Instituteof
Genetic Medicine, International Centre for Life,
Central Parkway, Newcastle upon Tyne, NE1 3BZ,
UK. Email: david.kavanagh@ncl.ac.uk
Copyright © 2015 by the American Society of
Nephrology
J Am Soc Nephrol 27: cccccc, 2015 ISSN : 1046-6673/2706-ccc 1
Figure 1. A 6.3-kb deletion in the RCA cluster results in a novel CFH/CFHR3 hybrid gene transcript. (A) Genomic organization of the RCA
cluster region containing the CFH and CFHR genes. (B) MLPA of CFH,CFHR3,CFHR1,CFHR2,andCFHR5 showing the copy number
ratio. The dotted lines represent ratios considered within the normal range. (C) Identication of breakpoint. Genomic DNA was amplied
2Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
In this study, we report a novel CFH/
CFHR3 hybrid gene arising through
MMEJ that impairs cell surface comple-
ment regulation.
An 8-month-old boy presented with a
diarrheal illness; on admission, creati-
nine was 52 mmol/L, and urea was
11.1 mmol/L. A blood lm showed mi-
croangiopathic hemolytic anemia, and
lactate dehydrogenase was 5747 units/L.
Stool culture was positive for Escherichia
coli O157:H7, and a diagnosis of Shiga
toxinproducing E. coli (STEC) hemo-
lyticuremicsyndrome(HUS)was
made. He did not require RRT and did
not receive plasma exchange. He was
discharged 2 weeks later with a creati-
nine of 107 mmol/L. He was readmitted
2 weeks later with an upper respira-
tory tract infection. Creatinine on read-
mission was 141 mmol/L, urea was
20.6 mmol/L, lactate dehydrogenase
was 2398 units/L, and platelets were
128310
9
/L, with evidence of microan-
giopathic hemolytic anemia on blood
lm. This relapse suggested a diagnosis
of aHUS rather than STEC HUS. Serum
complement levels were within the nor-
mal range: C3, 1.05 g/L (0.681.80 g/L);
C4, 0.30 g/L (0.180.60 g/L); and FH,
0.51 g/L (0.350.59 g/L). He com-
menced plasma exchange and required
three sessions of hemoltration. A renal
biopsy conrmed a thrombotic micro-
angiopathy (Figure 2). Creatinine re-
turned to a baseline of approximately
100 mmol/L. He was treated with weekly
plasma exchange for 3.5 years before
starting Eculizumab, on which he has
been maintained for 3 years. Creatinine
is currently 200 mmol/L, and there have
been no additional episodes of aHUS
while on treatment with Eculizumab
(Supplemental Figure 2).
Sanger sequencing of aHUS-associated
genes (CFH,CFI,CFB,CD46,C3,THBD,
and DGKE) did not reveal any muta-
tions, although heterozygosity was found
for the common Y402H polymorphism
(rs1061170) in CFH. Multiplex ligation
dependent probe amplication (MLPA)
of CFH and the CFHRs revealed a dele-
tion in the CFH gene (Figure 1B). To de-
ne the extent of the deletion and the
breakpoint, Sanger sequencing of geno-
mic DNA was under taken. This showed a
6.3-kb deletion extending from CFH in-
tron 20 to the CFH 39intergenic region
incorporating exons 2123 of CFH (Fig-
ure 1C). Directly anking the breakpoint
was a 7-bp region of microhomology
(Figure 1D). The deletion was not de-
tected in either parent, suggesting that
this was a de novo event.
We hypothesized that the loss of CFH
exons 2123 and the 39UTR of CFH
would lead to aberrant splicing. In silico
analysis showed that the next acceptor
splice site following the CFH exon 20
donor site was 59of CFHR3 exon 2,
thus potentially producing a transcript
for a hybrid CFH/CFHR3 gene (Supple-
mental Figure 3). mRNA for this puta-
tive hybrid CFH/CFHR3genewas
conrmed by amplifying patient and
control cDNA with CFHand CFHR3
specic primers. This showed a product
in the patient and not in healthy controls
(Figure 1E).
To conrm that this hybrid transcript
led to the synthesis and secretion of a
hybrid protein, Western blotting was
performed using a series of epitope
dened antiFH mAbs (Figure 3A). An
initial blot probed with OX24, an FH
CCP5specic antibody, showed, in ad-
dition to FH, two additional bands only
in the patient (Figure 3B). The species at
approximately 160 kD was consistent
with the predicted intact hybrid
FH/FHR3. A species at approximately
120kDwashypothesizedtobea
degradation product. No abnormal bands
were detected in either parent (Supple-
mental Figure 4A). An FH C terminus
specic antibody (L20/3) did not reveal
additional aberrant bands, indicating that
these protein species lacked FH CCP20
(Figure 3C). Genotyping showed the pa-
tient to be heterozygous for the Y402H
polymorphism in FH. This allowed the
use of mAbs specic for histidine or
tyrosine in CCP7 of FH.
14
The normal
alleles product reacted to the histidine-
specic mAb (MBI7) and gave a single
band of approximately 150 kD, consis-
tent with FH. The hybrid allele reacted
to the tyrosine-specicmAb(MBI6),
showingadoubletatapproximately
160 kD (Figure 3D). FHR3 is known to
be alternately glycosylated,
1517
and we
hypothesize that the doublet is caused
by an alternately glycosylated hybrid pro-
tein. Additionally, it can be seen that the
presumed degradation product at 120 kD
is only seen using the tyrosine-specic
mAb, consistent with this only arising
from the hybrid protein.
To conrm these ndings, FH species
were puried from serum using afnity
chromatography with an OX24 mAb.
These were separated by a 6% SDS-PAGE
before trypsin digestion. Mass spec-
trometrywasthencarriedoutonthese
three puried bands (Figure 4A). Pep-
tide species identied from the approx-
imately 160-kD band conrmed that
this was a hybrid FH/FHR3 protein.
Protein fragments from the 150-kD
band were consistent with FH. Frag-
ments from CCP5, CCP6, CCP8,
CCP9, and CCP14 of FH were seen in
the band at approximately 120 kD. Cov-
erage at CCP7 was insufcient to deter-
mine whether there was a tyrosine or
histidine at position 402. The Western
blot analysis and mass spectrometry
data together show that breakdown
using a forward primer specicforCFH exon 20 (1f) and a reverse primer in the CFH 39region intergenic region (1r) and sequenced. The
deletion of exons 2123 of CFH is shown (shaded box), and the breakpoint is highlighted. (D) The CFH and CFHR3 sequence anking the
breakpoint. The 7-b p area of microhomology is shown (b old and boxed). (E) Conrmation of hyb rid CFH/CFHR3 mRNA. A message for th e
hybrid CFH/CFHR3 gene was conrmed by amplif ying patient and control cDNA with cross CFH and CFHR3 gene primers (bla ck arrows 2f
and 2r in C). The agarose gel shows amplied cDNA. The patient lane shows an amplied product consistent with a hybrid CFH/CFHR3
gene, which is not present in either control cDNA.
J Am Soc Nephrol 27: cccccc, 2015 Hybrid CFH/CFHR3 Gene and aHUS 3
www.jasn.org BRIEF COMMUNICATION
products from only the hybrid protein
are seen in serum.
To examine the functional signi-
cance, the FH/FHR3 hybrid protein
was puried from serum using afnity
chromatography with an MBI6 mAb
followed by gel ltration (Supplemental
Figure 4B). The FH/FHR3 hybrid pro-
tein displayed both impaired cell surface
decay acceleration (Figure 4B) and co-
factor activity (Figure 4C, Supplemental
Figure 5).
Thus, in this study, we show a de-
letion in the RCA cluster resulting in a
novel de novo CFH/CFHR3 hybrid gene.
Microhomology in the sequence ank-
ing the breakpoint suggests that the
deletion has occurred through MMEJ.
Genomic disorders affecting CFH and
the CFHRs as a result of nonallelic ho-
mologous recombination are found in
approximately4.5%ofpatientswith
aHUS. In contrast, only one patient
with a genomic disorder secondary to
MMEJ has been described.
11
We have shown that the product of
this gene, a 22 CCP domain protein, is
secreted, albeit with degradation frag-
ments present in the serum, suggesting
impaired stability of the protein. Func-
tional analysis of the puried FH/FHR3
protein showed impaired cell surface
complement regulation.
This FH/FHR3 hybrid protein is sim-
ilar to that d escribed by Francis et al.,
11
in
that in both, the C-terminal end of FH is
replaced by all ve CCPs of FHR3 (Sup-
plemental Figure 1). Although both hy-
brids lack CCP20 of FH responsible for
cell surface protection, the hybrid
reported here also lacks CCP18 and
CCP19.
The functional role of FHR3 is un-
clear, with various regulatory activities
being suggested.
16,18
Unsurprisingly,
given its high homology with CCP7 of
FH, FHR3 binds to heparin.
16
FHR3
also binds to C3b and C3d through
CCP4 and CCP5.
16,19
Competition be-
tween FHR3 and FH for surface-bound
C3b has been described.
18
The hybrid
described by Francis et al.
11
showed
normal uidphase complement
Figure 2. A renal biopsy from the patient demonstrating haemolytic uraemic syndrome. Renal
biopsyof thepatientshowing thrombi(arrows)oncapillaryloops. (A) hematoxylinandeosin.(B)
Masson trichrome. A developing membranoproliferative pattern of injury can be seen in (C)
characterized by capillary loop double contours (arrows). Silver counterstained H&E, 3400.
4Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
regulatory activity but a profoundly re-
duced cell surface complement regula-
tory activity.
ThefactthatsuchanFH/FHR3hy-
brid should lack cell surface regulatory
activity is not surprising. Both of these
hybrids lack the CCP20 domain of FH
responsible for cell surface localization.
6
Additionally, structural analysis has
revealed a specichairpinstructure
suggesting a model whereby cell surface
bound C3b is engaged by both CCP14
and CCP19 and CCP20 of FH concur-
rently.
20
The elongation of FH by the ad-
dition of extra CCPs would prevent such
an orientation.
Although the initial presentation of
disease was seen in association with
STEC, the rapid relapse suggested
aHUS rather than STEC HUS, and this
was conrmed by the nding of the CFH/
CFHR3 hybrid gene. In individuals car-
rying mutations in complement genes,
additional triggers (e.g.,pregnancy
21
and infection
22
) are required for disease
to manifest.
4,22
In this case, STEC served
to unmask the genetic predisposi-
tion to disease, such as in two previously
reported patients with STEC-triggered
aHUS.
23
In summary, we describe a deletion
in the RCA cluster arising through
MMEJ that results in a novel CFH/
CFHR3 hybrid gene. The fact that this
arose as a de novo event suggests that
this is a dynamic area of the genome
where we should expect additional ge-
nomic disorders.
CONCISE METHODS
The study was approved by Newcastle and
North Tyneside 1 Research Ethics Com-
mittee, and informed consent was obtained
in accordance with the Declaration of
Helsinki.
Complement Assays
C3 and C4 levels were measured by rate
nephelometry (Beckman Coulter Array 360).
FH levels were measured by radial immuno-
diffusion (Binding Site). Screening for FH
autoantibodies was undertaken using ELISA
as described previously.
24
Genetic Analyses and MLPA
Mutation screening of CFH,
25
CFI,
26
CFB,
27
MCP,
28
C3,
29
and DGKE
30
was un-
dertaken using Sanger sequencing as
Figure 3. Identication of a novel FH/FHR3 hybrid protein in patient serum. (A) The protein
product of wild-type FH and the predicted FH/FHR3 hybrid protein consisting of CCP117 of FH
and CCP15 of FHR3. The CCPs originating from FHR3 are highlighted in gray. The patient is
heterozygous for a common polymorphism in CCP7 of FH (Y402H). Binding epitopes for the mAbs
are shown: Ox24-CCP5, MBI7-CCP7 amino acid 402 histidine variant, MBI6-CCP7 amino acid 402
tyrosine variant, and L20/3-CCP20. (B) Serum Western blot using OX24 showing two additional
bands in the patient in addition to FH. The upper band is consistent with the predicted size of the
FH/FHR3 hybrid. (C) Serum Western blot using L20/3. When using this FH C terminusspecic
antibody, no additional bands are seen, consistent with the hybrid protein lacking CCP1820. (D)
Serum Western blot using MBI7. This shows that the patient has a normal allele producing FH with
the histidine at amino acid 402. (E) Serum Western blot using MBI6. The patient has three addi-
tional bands and no wildtype FH band. This is consistent with the FH/FHR3 hybrid protein carrying
the tyrosine amino acid at position 402. The two additional upper bands represent differentially
glycosylated hybrid protein, consistent with that seen in the native FHR3. There is a faint degra-
dation product only detected with the MBI6 antibody. This is consistent with the breakdown
product being generated only from the hybrid protein and suggests a less stable protein product.
The controls were unaffected, unrelated, genotyped samples. In B and C, a sample from an in-
dividual heterozygous at Y402H was used. In D and E, samples from individuals homozygous for
Y402 (Y/Y), homozygous for H402 (H/H), or heterozygous (Y/H) were used.
J Am Soc Nephrol 27: cccccc, 2015 Hybrid CFH/CFHR3 Gene and aHUS 5
www.jasn.org BRIEF COMMUNICATION
previously described. Screening for geno-
mic disorders affecting CFH,CFHR1,
CFHR2,CFHR3,andCFHR5 was under-
taken using MLPA in both the affected
individual and 500 normal controls as pre-
viously described.
11
Aproprietarykitfrom
MRC Holland (SALSA MLPA Kit P236-A1
ARMD; www.mlpa.com) was supplemented
by specicinhouse CFH probes (Supple-
mental Material). GeneMarker software
(Version 1.90) was used to calculate dosage
quotients.
Figure 4. Impaired cell surface co-factor and decay acceleration activity of hybrid FH/FHR3 hybrid protein. (A) Mass spectrometry of
puried proteins. The FH, FH/FHR3 hybrid, and degradation product were puried using afnity chromatography with an OX24
column. These FH species were separated by 6% SDS-PAGE, stained using Coomassie, cut from the gel (left panel), and submitted for
trypsin digest and mass spectrometry. Peptides sequences identied using mass spectrometry are indicated with asterisks. The
peptides detected in the hybrid degradation product by mass spectrometry are annotated on a fulllength FH/FHR3 hybrid protein.
CCPs that cannot be directly inferred from mass spectrometry data are outlined with a dashed line. A molecular mass of approximately
120 kD is consistent with an approximately 17 CCP protein. (B) Decay acceleration assays on sheep erythrocytes. The puried FH/
FHR3 hybrid from the patient showed impaired cell surface complement regulation compared with wild type (WT) FH puried from
control. Alternative pathway convertase (C3bBb) was formed on sheep erythrocytes. Cells were incubated for 15 minutes with di-
lutions of puried FH/FHR3 hybrid and WT FH before triggering lysis with NHSDBDH. Maximal lysis occurs in buffer-only (0 mM FH)
conditions. Addition of WT FH caused decay of the C3 convertase and decay of convertase resulting in inhibition of lysis. The FH/
FHR3 hybrid was up to 2-fold less efcient at inhibiting lysis. (C) C3b cofactor activity on sheep erythrocytes. WT FH and puried FH/
FHR3 hybrid were tested for the abilitytoactasacofactorforfactorIcatalyzed inactivation of C3b deposited on the surfaces of sheep
erythrocytes. The C3 convertase (C3bBb) was formed on residual C3b, and lysis was triggered by adding NHSDBDH. Maximal lysis
occurs in the presence of buffer only (0 mM FH). The addition of WT FH and factor I produces iC3b, which decreases convertase
formation and subsequent lysis, and this is shown as increasing amounts of inhibition of lysis (expressed as percentage of maximal
lysis) after incubation with factor I and WT FH (black circles) or FH/FHR3 hybrid (white circles). The FH/FHR3 hybrid can be seen to be
markedly less active than WT.
6Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
Genomic Breakpoint Analyses
To identify the breakpoint of the deletion that
resulted in the CFH/CFHR3 hybrid gene, genomic
DNAwas amplied using a forward primer specic
for CFH in exon 20 and a reverse primer in the
CFH 39region (Figure 1C). The sequence of the
forward primer was GTAACTGTTATCAGTT-
GATTTGC, and the reverse was ACGGATTG-
CATGTATAAGTG. The product was then
sequenced using direct uorescent sequencing.
Conrmation of CFH/CFHR3 RNA
Product
mRNA was extracted from peripheral blood lym-
phocytes of the patient, and cDNA was prepared.
This was amplied using a forward primer in CFH
exon 20 (TGGATGGAGCCAGTAATGTAA-
CATGCAT) and a reverse primer in CFHR3 exon
2 (GAAATAGACCTCCATGTTTAATGTCTG)
(Figure 1C). The PCR product was detected in an
ethidium bromidestained 1.5% agarose gel.
Western Blotting
Detection of abnormal protein products in
serum arising as a consequence of the deletion
was undertaken by Western blotting. Sera were
diluted 1:100, and under nonreducing condi-
tions, they were electrophoresed on 6% SDS-
PAGE gel and transferred onto nitrocellulose.
mAbsof knownspecicity to FH(OX24,CCP5,
L20/3,CCP20, MBI6, CCP7402Y, MBI7 CCP7,
and 402H) were used with sheep antimouse Ig
HRP (Jackson Immuno Research) at dilutions
of 1:2000 (primary antibody) and 1:4000 (sec-
ondary antibody) (Supplemental Material).
After washes in 13TBST, blots were developed
using Pierce ECL Western Blotting Substrate
(Thermo Scientic).
Purication of FH Species
FH species (wild-type FH from normal allele,
FH/FHR3 hybrid, and FH/FHR3 breakdown
product all with CCP5 of FH) were puried
from serum with afnity chromatography
using immobilized mAb to FH (OX24) on a
1-ml HiTrap NHS HP Column (GE Health-
care). After washing with PBS, the bound
proteins were then eluted using 0.1 M glycine
(pH 2.7). The eluted material was pooled and
concentratedfor analysis bymass spectrometry.
The FH/FHR3 hybr id protein was puried
from patient serum with afnity chromatog-
raphy using immobilized mAb to FH CCP7
402Y (MBI6)
31
on a 1-ml HiTrap NHS HP
Column (GE Healthcare). After washing with
PBS, the bound proteins were eluted using
PBS/diethylamine (50 mM). Subsequently,
the FH/FHR3 protein was puried from its
degradation product and FHL1 using a Su-
perdex 200 Size Exclusion Column. We also
puried wildtype FH protein from a healthy
control. FH or FH/FHR3 hybrid protein pu-
ried using the MBI6 column was used for
cofactor and decay acceleration assays.
Mass Spectrometry
A 6% SDS-PAGE was run, a nd the three b ands
identied by Coomassi e staining were excised
from the gel as indic ated in Figure 4A. Trypsin
digest and mass spectrometry were then un-
dertaken (Supplemental Material).
Cell Surface Decay Acceleration
Assays
Decay acceleration on sheep erythrocytes was
performed as previously described
32
and in
Supplemental Material. Briey, FH/FHR3
hybrid from the patient and FH from con-
trols were puried by immunoafnity chro-
matography as above and gel ltered into
PBS. Alternative pathway convertase
(C3bBb) was formed on sheep erythrocytes.
Cells were incubated for 15 minutes with
1.2450 nM patient FH/FHR3 hybrid or con-
trol FH. The molar concentration of the pu-
ried patient FH/FHR3 was estimated using
the extinction coefcient (272,170 M cm
21
),
whereas the extinction coefcient of FH
(246,800 M cm
21
) was used for the control.
Lysis was initiated with 4% normal human
serum depleted of factor B and FH
(NHSDBDH). Maximal lysis was achieved
by adding NHSDBDHtonoFHwells(buffer
only). To determine the amount of lysis, cells
were pelleted by centrifugation, and hemo-
globin release was measured at 420 nm
(A
420
). Percentage of inhibition of lysis in
the presence of increasing concentrations
was dened as (A
420
[buffer only] 2
A
420
[FH])/A
420
[buffer only] 3100%.
Cell Surface Cofactor Assays
Cofactor activity on sheep erythrocytes was
performed as previously described.
32
Briey
washed C3bcoated sheep erythrocyte cells
were resuspended in AP buffer and incubated
with an equal volume of a range of concen-
trations of FH/FHR3 hybrid and wild-type
FH and 2.5 mg/ml factor I (CompTech) for
8 minutes at 25°C. After three washes in AP
buffer, AP convertase was formed on the re-
maining C3b. Lysis was initiated with 4%
NHSDBDH. Again, cells were pelleted, and
hemoglobin release was measured at 420
nm. Percentage of inhibition from lysis was
calculated by the formula (A
420
[buffer only]
2A
420
[FH])/A
420
[buffer only] 3100%.
ACKNOWLEDGMENTS
We thank Achim Treumann for technical help
with mass spectrometry.
The research leading to these results has re-
ceived funding from European Unions Seventh
Framework Programme FP7/2007-2013 Grant
305608 (EURenOmics). Funding for this
study was provided by United Kingdom
Medical Research Council Grant G0701325.
G.S.R.A. is funded by Conselho Nacional de
Desenvolvimento Cientíco e Tecnológico.
E.K.S.W. is a Medical Research Council
Clinical Training Fellow. D.K. is a Wellcome
Trust Intermediate Clinical Fellow.
DISCLOSURES
A.A. has received fees from Alexion Pharmaceu-
ticals for lectures. C.L.H. is also employed by
GlaxoSmithKline and has shares in this company.
Newcastle University has received fees from Alexion
Pharmaceuticals for lectures and consultancy un-
dertaken by T.H.J.G. and D.K.
REFERENCES
1. Díaz-Guillén MA, Rodríguez de Córdoba S,
Heine-Suñer D: A radiation hybrid map of
complement factor H and factor H-related
genes. Immunogenetics 49: 549552, 1999
2. Male DA, Ormsby RJ, Ranganathan S,
Giannakis E, Gordon DL: Complement factor
H: Sequence analysis of 221 kb of human ge-
nomic DNAcontaining the entire fH, fHR-1 and
fHR-3 genes. Mol Immunol 37: 4152, 2000
3. Zipfel PF, Jokiranta TS , Hellwage J, Koistinen
V, Meri S: The factor H protein family. Im-
munopharmacology 42: 5360, 1999
4. Kavanagh D, Goodship TH, Richards A:
Atypical hemolytic uremic syndrome. Semin
Nephrol 33: 508530, 2013
5. Ferreira VP, Herbert AP, Co rtés C, McKee KA,
Blaum BS, Esswein ST, Uhrín D, Barlow PN,
Pangburn MK, Kavanagh D: The binding of
factor H to a complex of physiological poly-
anions and C3b on cells is impaired in atypi-
cal hemolytic uremic syndrome. JImmunol
182: 70097018, 2009
J Am Soc Nephrol 27: cccccc, 2015 Hybrid CFH/CFHR3 Gene and aHUS 7
www.jasn.org BRIEF COMMUNICATION
6. Schmidt CQ, Herbert AP, Kavanagh D,
Gandy C, Fenton CJ, Blaum BS, Lyon M,
Uhrín D, Barlow PN: A new map of glycos-
aminoglycan and C3b binding sites on factor
H. JImmunol181: 26102619, 2008
7. Blaum BS, Hannan JP, Herbert AP, Kavanagh
D, Uhrín D, Stehle T: Structural basis for sialic
acid-mediated self-recognition by comple-
ment factor H. Nat ChemBiol 11: 7782, 2015
8. Heinen S, Sanchez-Corral P, Jackson MS,
Strain L, Goodship JA, Kemp EJ, Skerka C,
Jokiranta TS, Meyers K, Wagner E, Robitaille
P, Esparza-Gordil lo J, Rodriguez de Cordoba
S, Zipfel PF, Goodship TH: 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: 292293, 2006
9. Venables JP, Strain L, Routledge D, Bourn D,
Powell HM, Warwicker P, Diaz-Torres ML,
Sampson A, Mead P, Webb M, Pirson Y,
Jackson MS, Hughes A, W ood KM, Goodship
JA, Goodship TH: Atypical haemolytic urae-
mic syndrome associated with a hybrid
complement gene. PLoS Med 3: e431, 2006
10. Maga TK, Meyer NC, Belsha C, Nishimura
CJ, Zhang Y, Smith RJ: A novel del etion in the
RCA gene cluster causes atypical hemolytic
uremic syndrome. Nephrol Dial Transplant
26: 739741, 2011
11. Francis NJ, McNicholas B, Awan A, Waldron
M, Reddan D, Sadlier D, Kavanagh D, Strain
L, Marchbank KJ, Harris CL, Goodship TH: A
novel hybrid CFH/CFHR3 gene generated
by a microhomology-mediated deletion in
familial atypical hemolytic uremic syndrome.
Blood 119: 591601, 2012
12. Eyler SJ, Meyer NC, Zhang Y, Xiao X, Nester
CM, Smith RJ: A novel hybrid CFHR1/CFH
gene causes atypical hemolytic uremic syn-
drome. Pediatr Nephrol 28: 22212225, 2013
13. Valoti E, Alberti M, Tortajada A, Garcia-
Fernandez J, Gastoldi S, Besso L, Bresin E,
Remuzzi G, Rodriguez de Cordoba S, Noris
M: A novel atypical hemolytic uremic syn-
drome-associated hybrid CFHR1/CFH gene
encoding a fusion protein that antagonizes
factor H-dependen t complement regulation.
JAmSocNephrol26: 209219, 2015
14. Hakobyan S, Tortajada A, Harris CL, de
Córdoba SR, Morgan BP: Variant-specic
quantication of factor H in plasma identies
null alleles associated with atypical hemolytic
uremic syndrome. Kidney Int 78: 782788, 2010
15. Skerka C, Kühn S, Günther K, Lingelbach K,
Zipfel PF: A novel short consensus repeat-
containing molecule is related to human
complement factor H. JBiolChem268:
29042908, 1993
16. Hellwage J, Jokiranta TS, Koistinen V,
Vaarala O, Meri S, Zipfel PF: Functional
properties of complement factor H-related
proteins FHR-3 and FHR-4: Binding to the
C3d region of C3b and diff erential regulation
by heparin. FEBS Lett 462: 345352, 1999
17. Skerka C, Chen Q, Fremeaux-Bacchi V,
Roumenina LT: Comp lement factor H related
proteins (CFHRs). Mol Immunol 56: 170180,
2013
18. Fritsche LG, Lauer N, Hartmann A, Stippa S,
Keilhauer CN, Oppermann M, Pandey MK,
Köhl J, Zipfel PF, Weber BH, Skerka C: An
imbalance of human complement regulatory
proteins CFHR1, CFHR3 and factor H inu-
ences risk for age-related macular de-
generation (AMD). Hum Mol Genet 19:
46944704, 2010
19. Hellwage J, Jokiranta TS, Friese MA, Wolk TU,
Kampen E, Zipfel PF, Meri S: Complement
C3b/C3d and cell surface polyanions are rec-
ognized by overlapping binding sites on the
most carboxyl-terminal domainof complement
factor H. J Immunol 169: 69356944, 2002
20. Morgan HP, Schmidt CQ, Guariento M,
Blaum BS, Gillespie D, Herbe rt AP, Kavanagh
D, Mertens HD, Svergun DI, Johansson CM,
Uhrín D, Barlow PN, Hannan JP: Structural
basis for engagement by complement factor
H of C3b on a self surface. Nat Struct Mol Biol
18: 463470, 2011
21. Fakhouri F, Roumenina L, Provot F, Sallée M,
Caillard S, Couzi L, Essig M, Ribes D, Dragon-
Durey MA, Bridoux F, R ondeau E, Frémeaux-
Bacchi V: Pregnancy-associated hemolytic
uremic syndrome revisited in the era of
complement gene mutations. JAmSoc
Nephrol 21: 859867, 2010
22. Caprioli J, Noris M, Brioschi S, Pianetti G,
Castelletti F, Bettinaglio P, Mele C, Bresin E,
Cassis L, Gamba S, Porrati F, Bucchioni S,
Monteferrante G, Fang CJ, Liszewski MK,
Kavanagh D, Atkinson JP, Remuzzi G; In-
ternational Reg istry of Recurrent and Familial
HUS/TTP: Genetics of HUS: The impact of
MCP, CFH, and IF mutations on clinical pre-
sentation, response to treatment, and out-
come. Blood 108: 12671279, 2006
23. Alberti M, Valoti E, Piras R, Bresin E,
Galbusera M, Tripodo C, Thaiss F, Remuzzi
G, Noris M: Two patients with history of
STEC-HUS, posttransplant recurrence and
complement gene mutations. Am J Trans-
plant 13: 22012206, 2013
24. Moore I, Strain L, Pappworth I, Kavanagh D,
Barlow PN, Herbert AP, Schmidt CQ,
Staniforth SJ, Holmes LV, Ward R, Morgan L,
Goodship TH, Marchbank KJ: 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: 379387, 2010
25. RichardsA, Buddles MR, Donne RL, KaplanBS,
Kirk E, Venning MC, Tielemans CL, Goodship
JA, Goodship TH: Factor H mutations in
hemolytic uremic syndrome cluster in exons
18-20, a domain important for host cell rec-
ognition.Am J Hum Genet 68: 485490, 2001
26. KavanaghD, Kemp EJ, Mayland E, Winney RJ,
Dufeld JS, Warwick G, Richards A, Ward R,
Goodship JA, Goodship TH: Mutations in
complement factor I predispose to develop-
ment of atypical hemolytic uremic syndrome.
J Am Soc Nephrol 16: 21502155, 2005
27. Kavanagh D, Kemp EJ, Richards A, Burgess
RM, Mayland E, Goodship JA, Goodship TH:
Does complement factor B have a role in the
pathogenesis of atypical HUS? Mol Immunol
43: 856859, 2006
28. Richards A, Kemp EJ, Liszewski MK,
Goodship JA, Lampe AK, Decorte R,
Müslümano
glu MH, Kavukcu S, Filler G,
Pirson Y, Wen LS, Atkinso n JP, Goodship TH:
Mutations in human complement regulator,
membrane cofactor protein (CD46), pre-
dispose to developm ent of familial hemolytic
uremic syndrome. Proc Natl Acad Sci U S A
100: 1296612971, 2003
29. Frémeaux-Bacchi V, Miller EC, Liszewski MK,
Strain L, Blouin J, Brown AL, Moghal N,
Kaplan BS, Weiss RA, Lhotta K, Kapur G,
Mattoo T, Nivet H, Wong W, Gie S, Hurault
de Ligny B, Fischbac h M, Gupta R, Hauhart R,
Meunier V, Loirat C, Dragon-Durey MA,
Fridman WH, Janssen BJ, Goodship TH,
Atkinson JP: Mutations in complement C3
predispose to development of atypical he-
molytic uremic syndrome. Blood 112: 4948
4952, 2008
30. Lemaire M, Frémeaux-Bacchi V, Schaefer F,
Choi M, Tang WH, Le Quintrec M, Fakhouri
F,TaqueS,NobiliF,MartinezF,JiW,
Overton JD, Mane SM, Nürnberg G,
Altmüller J, Thiele H, Morin D, Deschenes G,
Baudouin V, Llanas B, Collard L, Majid MA,
Simkova E, Nürnberg P, Rioux-Leclerc N,
Moeckel GW, Gubler MC, Hwa J, Loirat C,
Lifton RP: Recessive mutations in DGKE
cause atypical hemolytic-uremic syndrome.
Nat Genet 45: 531536, 2013
31. Hakobyan S, Harris CL, Tortajada A,
Goicochea de Jorge E, García-Layana A,
Fernández-Robredo P, Rodríguez de
Córdoba S, Morgan BP: Measurement of
factor H variants in plasma using variant-
specic monoclonal antibodies: Application
to assessing risk of age-related macular de-
generation. Invest Ophthalmol Vis Sci 49:
19831990, 2008
32. Wong EK, Anderson HE, Herbert AP, Challis
RC, Brown P, Reis GS, Tellez JO, Strain L,
Fluck N, Humphrey A, Macleod A, Richards
A, Ahlert D, Santibanez-Koref M, Barlow PN,
Marchbank KJ, Harris CL, Goodship TH,
Kavanagh D: Characterization of a factor H
mutation that perturbs the alternative path-
way of complement in a family with mem-
branoproliferati ve GN. JAmSocNephrol25:
24252433, 2014
This article contains supplemental material online
at http://jasn.asnjournals.org/lookup/suppl/doi:10.
1681/ASN.2015010100/-/DCSupplemental.
8Journal of the American Society of Nephrology J Am Soc Nephrol 27: cccccc,2015
BRIEF COMMUNICATION www.jasn.org
... To date, six types of hybrid genes were reported in the CFH/ CFHR gene cluster (Fig. 3) [8][9][10][11][12][13]. In four of these, C-terminaldeficient CFH proteins were fused to the C-terminus of the CFHR protein ("CFH-CFHR protein") [8][9][10][11], similar to our cases. ...
... To date, six types of hybrid genes were reported in the CFH/ CFHR gene cluster (Fig. 3) [8][9][10][11][12][13]. In four of these, C-terminaldeficient CFH proteins were fused to the C-terminus of the CFHR protein ("CFH-CFHR protein") [8][9][10][11], similar to our cases. Because the impaired function of the CFH C-terminus, i.e., the SCR 19-20 surface binding domains [16], results in reduced recognition of and binding to host cell surfaces, the CFH-CFHR hybrid proteins devoid of this domain can cause over-activation of the complement system on cell surfaces. ...
... The family members of patient-2 denied consent and could not be analyzed in this study. It is possible that his is a sporadic case caused by a de novo genomic rearrangement [9,11]. ...
Article
Full-text available
Atypical hemolytic uremic syndrome (aHUS) is a rare complement-mediated disease that manifests as the triad of thrombotic microangiopathy. We identified two aHUS patients with neither anti-complement factor H (CFH) antibodies nor causative variants of seven aHUS-related genes (CFH, CFI, CFB, C3, MCP, THBD, and DGKE); however, their plasma showed increased levels of hemolysis by hemolytic assay, which strongly suggests CFH-related abnormalities. Using a copy number variation (CNV) analysis of the CFH/CFHR gene cluster, we identified CFH-CFHR1 hybrid genes in these patients. We verified the absence of aHUS-related abnormal CNVs of the CFH gene in control genomes of 2036 individuals in the general population, which suggests that pathogenicity is related to these hybrid genes. Our study emphasizes that, for patients suspected of having aHUS, it is important to perform an integrated analysis based on a clinical examination, functional analysis, and detailed genetic investigation.
... cluster spanning over approximately 360 kb. 3 Because of successive large genomic duplication events during human evolution, CFH and its five related genes (CFHR1 to CFHR5) show high sequence and structural homologies and lie in a head-to-tail arrangement within the regulators of the complement activation gene cluster on chromosome 1q32. 4e6 These genomic structural variants are most probably caused by nonallelic homologous recombination 7,8 and interlocus gene conversion 2 or a mechanism called microhomology-mediated end joining, reported by Challis et al 9 and Francis et al, 10 respectively. The circulating plasma protein CFH is primarily produced in the liver and is composed of individual folding domains called short consensus repeats. ...
... Genomic aberrations or structural variants affecting the complement factor H (CFH ) gene cluster were reported in approximately 4.5% of patients, in a cohort of 154 individuals with aHUS analyzed by multiplex ligationdependent probe amplification (MLPA), 7 and are often caused by nonallelic homologous recombination events. 7,9,10 Incomplete penetrance is frequently observed in families with autosomal dominant aHUS, where unaffected family members carry the causative aberration as well. 30,31 These variants either decrease the activity of the complementregulating proteins or increase the activity of complement activator proteins, leading to a decreased threshold for inappropriate pathologic complement activation. ...
... The current routine molecular diagnostics for aHUS patients consist of next-generation sequencing (NGS) or Sanger sequencing, augmented by deletion and duplication testing using MLPA. 7,9,32 CFH/CFHR hybrid gene identification can be performed using either chromosomal microarray 7,33 or PCR-based approaches, the latter of which may also allow for breakpoint detection. 7,9,33 CFH autoantibodies are associated with the frequent homozygous CFHR3/CFHR1 deletions and are often tested with enzyme-linked immunosorbent assay. ...
Article
Full-text available
Complement factor H (CFH) and its related proteins have an essential role in regulating the alternative pathway of the complement system. Mutations and structural variants (SVs) of the CFH gene cluster, consisting of CFH and its five related genes (CFHR1-5), have been reported in renal pathologies as well as in complex immune diseases like age-related macular degeneration and systemic lupus erythematosus. SV analysis of this cluster is challenging due to its high degree of sequence homology. Following first-line NGS gene panel sequencing, we applied Genomic Vision’s Molecular Combing Technology, to detect and visualize SVs within the CFH gene cluster and resolve its structural haplotypes completely. This approach was tested in three patients with atypical hemolytic uremic syndrome (aHUS) and known SVs, and 18 patients with aHUS or complement factor 3 glomerulopathy with unknown CFH gene cluster haplotypes. Three SVs, a CFH/CFHR1 hybrid gene in two patients and a rare heterozygous CFHR4/CFHR1 deletion in trans with the common CFHR3/CFHR1 deletion in a third patient were newly identified. For the latter, the breakpoints were determined using a targeted enrichment approach for long DNA fragments (Samplix Xdrop) in combination with Oxford Nanopore sequencing. Molecular combing in addition to NGS was able to improve the molecular genetic yield in this pilot study. This (cost-)effective approach warrants validation in larger cohorts with CFH/CFHR-associated disease.
... Immune-complex-MPGN and C3 glomerulopathy are rare, with estimated incidence of 1-4 cases per million population 3,4 . Acquired and genetic abnormalities associated with fluid phase dysregulation of the alternative pathway of complement have been identified in immunecomplex MPGN and C3 glomerulopathy [5][6][7][8][9][10][11][12][13][14][15][16] . ...
... Genetic screening of all exons and flanking regions of C3 5 , CFB 6 , CFH 7 , CFI 8 , CD46 9 and DGKE 10 was performed and rare genetic variants and common polymorphisms were identified following targeted next generation sequencing and confirmatory Sanger sequencing. Rare genetic variants were defined as minor allele frequency <0.01 in the exome variant server database (evs.gs.washington.edu). ...
Article
BACKGROUND AND OBJECTIVES: Membranoproliferative GN and C3 glomerulopathy are rare and overlapping disorders associated with dysregulation of the alternative complement pathway. Specific etiologic data for pediatric membranoproliferative GN/C3 glomerulopathy are lacking, and outcome data are based on retrospective studies without etiologic data. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: A total of 80 prevalent pediatric patients with membranoproliferative GN/C3 glomerulopathy underwent detailed phenotyping and long-term follow-up within the National Registry of Rare Kidney Diseases (RaDaR). Risk factors for kidney survival were determined using a Cox proportional hazards model. Kidney and transplant graft survival was determined using the Kaplan-Meier method. RESULTS: Central histology review determined 39 patients with C3 glomerulopathy, 31 with immune-complex membranoproliferative GN, and ten with immune-complex GN. Patients were aged 2-15 (median, 9; interquartile range, 7-11) years. Median complement C3 and C4 levels were 0.31 g/L and 0.14 g/L, respectively; acquired (anticomplement autoantibodies) or genetic alternative pathway abnormalities were detected in 46% and 9% of patients, respectively, across all groups, including those with immune-complex GN. Median follow-up was 5.18 (interquartile range, 2.13-8.08) years. Eleven patients (14%) progressed to kidney failure, with nine transplants performed in eight patients, two of which failed due to recurrent disease. Presence of >50% crescents on the initial biopsy specimen was the sole variable associated with kidney failure in multivariable analysis (hazard ratio, 6.2; 95% confidence interval, 1.05 to 36.6; P<0.05). Three distinct C3 glomerulopathy prognostic groups were identified according to presenting eGFR and >50% crescents on the initial biopsy specimen. CONCLUSIONS: Crescentic disease was a key risk factor associated with kidney failure in a national cohort of pediatric patients with membranoproliferative GN/C3 glomerulopathy and immune-complex GN. Presenting eGFR and crescentic disease help define prognostic groups in pediatric C3 glomerulopathy. Acquired abnormalities of the alternative pathway were commonly identified but not a risk factor for kidney failure.
... Immune-complex-MPGN and C3 glomerulopathy are rare, with estimated incidence of 1-4 cases per million population 3,4 . Acquired and genetic abnormalities associated with fluid phase dysregulation of the alternative pathway of complement have been identified in immunecomplex MPGN and C3 glomerulopathy [5][6][7][8][9][10][11][12][13][14][15][16] . ...
... Genetic screening of all exons and flanking regions of C3 5 , CFB 6 , CFH 7 , CFI 8 , CD46 9 and DGKE 10 was performed and rare genetic variants and common polymorphisms were identified following targeted next generation sequencing and confirmatory Sanger sequencing. Rare genetic variants were defined as minor allele frequency <0.01 in the exome variant server database (evs.gs.washington.edu). ...
Article
Background and objectives : Membranoproliferative Glomerulonephritis (MPGN) and C3 Glomerulopathy are rare and overlapping disorders associated with dysregulation of the alternative complement pathway. Specific aetiological data for paediatric MPGN/C3 glomerulopathy are lacking, and outcome data are based upon retrospective studies without aetiological data. Design, setting, participants, and measurements : Eighty prevalent pediatric patients with MPGN/C3 glomerulopathy underwent detailed phenotyping and long-term follow-up within the National Registry of Rare Kidney Diseases (RaDaR). Risk factors for kidney survival were determined using COX proportional hazards model. Kidney and transplant graft survival was determined using Kaplan-Meier method. Results : Central histology review determined 39 C3 glomerulopathy, 31 immune-complex MPGN and 10 immune-complex glomerulonephritis (GN) cases. Patients were aged 2-15 (median 9 (IQR 7-11) years. Median complement C3 and C4 levels were 0.31g/L and 0.14g/L respectively; acquired (anti-complement autoantibodies) or genetic alternative pathway abnormalities were detected in 46% and 9% patients respectively, across all groups including immune-complex GN. Median follow-up was 5.18 (IQR 2.13-8.08) years. Eleven patients (14%) progressed to kidney failure with 9 transplants performed in 8 patients, 2 of which failed due to recurrent disease. Presence of >50% crescents on initial biopsy was the sole variable associated with kidney failure in multivariable analysis (Hazard Ratio 6.2, p = 0.045; 95% CI 1.05 to 36.6). Three distinct C3 glomerulopathy prognostic groups were identified according to presenting eGFR and >50% crescents on initial biopsy. Conclusions : Crescentic disease was a key risk factor associated with kidney failure in a national cohort of pediatric MPGN/C3 glomerulopathy and immune-complex GN. Presenting eGFR and crescentic disease help define prognostic groups in pediatric C3 glomerulopathy. Acquired abnormalities of the alternative pathway were commonly identified but not a risk factor for kidney failure.
... All individuals referred to the NRCTC with suspected CaHUS were considered for study ( Figure 1). Renal transplant associated CaHUS cases were excluded 9 Genetic analysis for known aHUS associated genes and autoantibody analysis was as previously described [10][11][12][13][14][15][16][17][18][19][20] . Where whole exome sequencing was undertaken on a research basis as described 21 , the analysis was approved by the Northern and Yorkshire Multicentre ...
Article
Full-text available
Historically the majority of patients with complement mediated atypical haemolytic uraemic syndrome (CaHUS) progressed to end stage kidney disease (ESKD). Single arm trials of eculizumab with short follow-up suggested efficacy. We prove for the first time in a genotyped matched CaHUS cohort that the five-year cumulative estimate of ESKD free survival improved from 39.5% in a control cohort to 85.5% in the eculizumab treated cohort; HR 4.95 (95% CI 2.75-8.90), p=0.000, number needed to treat 2.17 (95% CI 1.81-2.73). Outcome following eculizumab is associated with underlying genotype. Lower serum creatinine, lower platelet count, lower blood pressure and younger age at presentation, as well as shorter time between presentation and first dose of eculizumab were associated with eGFR >60 ml/min at 6 months in multivariate analysis. The rate of meningococcal infection in the treated cohort was 550 times greater than the background rate in the general population. The relapse rate upon eculizumab withdrawal was 1 per 9.5 person years for those with a pathogenic mutation and 1 per 10.8 person for those with a variant of uncertain significance. There were no relapses recorded in 67.3 person years off eculizumab in those with no rare genetic variants. Eculizumab was restarted in 6 individuals with functioning kidneys in whom it had been stopped with no individual progressing to ESKD. We demonstrate that biallelic pathogenic mutations in RNA processing genes, including EXOSC3, encoding an essential part of the RNA exosome, cause eculizumab non-responsive aHUS. Recessive HSD11B2 mutations causing apparent mineralocorticoid excess may also present with a thrombotic microangiopathy.
... 30 Additionally, the area of the genome in which the CFH gene resides arose from several large genomic duplications. These low-copy repeats can cause genome instability in this region which results in the formation of hybrid genes [31][32][33][34][35][36] which require copy number analysis to detect in addition to standard sequencing. ...
Article
Full-text available
Thrombotic microangiopathy (TMA) is characterized by thrombocytopenia, microangiopathic haemolytic anaemia and end organ damage. TMAs have varying underlying pathophysiology and can therefore present with an array of clinical presentations. Renal involvement is common as the kidney is particularly susceptible to the endothelial damage and microvascular occlusion. TMAs require rapid assessment, diagnosis, and commencement of appropriate treatment due to the high morbidity and mortality associated with them. Ground‐breaking research into the pathogenesis of TMAs over the past 20 years has driven the successful development of targeted therapeutics revolutionizing patient outcomes. This review outlines the clinical presentations, pathogenesis, diagnostic tests and treatments for TMAs.
... Screening for chromosomal rearrangements affecting CFH, CFHR1, CFHR2, CFHR3, CFHR4, CFHR5, CFI, and CD46 was undertaken using multiplex ligation-dependent probe amplification, as previously described. 38,39 In familial cases (NCL25, NCL26, NCL27, and NCL29) whole exome sequencing was undertaken as previously described. 24 Sanger sequencing as previously described 24 was used to confirm DGKE mutations identified by whole exome sequencing and for routine screening. ...
Article
Full-text available
Recessive mutations in diacylglycerol kinase epsilon (DGKE) display genetic pleiotropy, with pathological features reported as either thrombotic microangiopathy or membranoproliferative glomerulonephritis (MPGN), and clinical features of atypical hemolytic uremic syndrome (aHUS), nephrotic syndrome or both. Pathophysiological mechanisms and optimal management strategies have not yet been defined. In prospective and retrospective studies of aHUS referred to the United Kingdom National aHUS service and prospective studies of MPGN referred to the National Registry of Rare Kidney Diseases for MPGN we defined the incidence of DGKE aHUS as 0.009/million/year and so-called DGKE MPGN as 0.006/million/year, giving a combined incidence of 0.015/million/year. Here, we describe a cohort of sixteen individuals with DGKE nephropathy. One presented with isolated nephrotic syndrome. Analysis of pathological features reveals that DGKE mutations give an MPGN-like appearance to different extents, with but more often without changes in arterioles or arteries. In 15 patients presenting with aHUS, ten had concurrent substantial proteinuria. Identified triggering events were rare but coexistent developmental disorders were seen in six. Nine with aHUS experienced at least one relapse, although in only one did a relapse of aHUS occur after age five years. Persistent proteinuria was seen in the majority of cases. Only two individuals have reached end stage renal disease, 20 years after the initial presentation, and in one, renal transplantation was successfully undertaken without relapse. Six individuals received eculizumab. Relapses on treatment occurred in one individual. In four individuals eculizumab was withdrawn, with one spontaneously resolving aHUS relapse occurring. Thus we suggest that DGKE-mediated aHUS is eculizumab non-responsive and that in individuals who currently receive eculizumab therapy it can be safely withdrawn. This has important patient safety and economic implications.
... Hybrid proteins composed of FH and FHRs (indicated by double colons between the proteins), namely FH::FHR-1, FHR-1::FH and FH::FHR-3 are associated with aHUS, because these changes either replace FH CCP20, which harbors the surface/sialic acid recognition site in FH (FH::FHR-1 and FH::FHR-3), or remove the regulatory CCPs 1-4 domains (FHR1::FH) [25,[209][210][211][212][213][214][215]. Hybrid FHRs containing domains from two proteins (FHR-3::FHR-1, FHR-1::FHR-5, FHR-2::FHR-5, FHR-5::FHR-2) and FHR-1 and FHR-5 with duplicated dimerization domains (CCPs 1-2) due to intragenic duplications are associated with C3G; the hybrids between FHR-1 and FHR-5 or FHR-2 and FHR-5 also have duplicated dimerization domains [22,89,[216][217][218][219][220][221]. ...
Article
The complement system, while being an essential and very efficient effector component of innate immunity, may cause damage to the host and result in various inflammatory, autoimmune and infectious diseases or cancer, when it is improperly activated or regulated. Factor H is a serum glycoprotein and the main regulator of the activity of the alternative complement pathway. Factor H, together with its splice variant factor H-like protein 1 (FHL-1), inhibits complement activation at the level of the central complement component C3 and beyond. In humans, there are also five factor H-related (FHR) proteins, whose function is poorly characterized. While data indicate complement inhibiting activity for some of the FHRs, there is increasing evidence that FHRs have an opposite role compared with factor H and FHL-1, namely, they enhance complement activation directly and also by competing with the regulators FH and FHL-1. This review summarizes the current stand and recent data on the roles of factor H family proteins in health and disease, with focus on the function of FHR proteins.
Article
Full-text available
Background: Atypical hemolytic uremic syndrome (aHUS) is a rare cause of end-stage kidney disease and associated with poor outcomes after kidney transplantation from early disease recurrence. Prophylactic eculizumab treatment at the time of transplantation is used in selected patients with aHUS. We report a retrospective case note review describing transplant outcomes in patients with aHUS transplanted between 1978 and 2017, including those patients treated with eculizumab. Methods: The National Renal Complement Therapeutics Centre database identified 118 kidney transplants in 86 recipients who had a confirmed diagnosis of aHUS. Thirty-eight kidney transplants were performed in 38 recipients who received prophylactic eculizumab. The cohort not treated with eculizumab comprised 80 transplants in 60 recipients and was refined to produce a comparable cohort of 33 transplants in 32 medium and high-risk recipients implanted since 2002. Complement pathway genetic screening was performed. Graft survival was censored for graft function at last follow-up or patient death. Graft survival without eculizumab treatment is described by complement defect status and by Kidney Disease: Improving Global Outcomes risk stratification. Results: Prophylactic eculizumab treatment improved renal allograft survival (P = 0.006) in medium and high-risk recipients with 1-y survival of 97% versus 64% in untreated patients. Our data supports the risk stratification advised by Kidney Disease: Improving Global Outcomes. Conclusions: Prophylactic eculizumab treatment dramatically improves graft survival making transplantation a viable therapeutic option in aHUS.
Article
Significance Statement The human CFHR–Factor H gene cluster encodes the five FHR proteins that are emerging complement and immune modulators and the two complement regulators Factor H and FHL1. Genetic and chromosomal alterations in this cluster are associated with the human kidney diseases atypical hemolytic uremic syndrome and C3 glomerulopathy. Various genetic alterations result in the expression of mutant and altered FHR proteins, or FHR::Factor H and Factor H::FHR hybrid proteins. The modified FHR proteins together with an altered FHR and Factor H plasma repertoire, which often modify complement action in the fluid phase and cause morphologic alteration in the glomerulus, provide important views on FHR protein function in the kidney. Sequence and copy number variations in the human CFHR–Factor H gene cluster comprising the complement genes CFHR1 , CFHR2 , CFHR3 , CFHR4 , CFHR5 , and Factor H are linked to the human kidney diseases atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy. Distinct genetic and chromosomal alterations, deletions, or duplications generate hybrid or mutant CFHR genes, as well as hybrid CFHR–Factor H genes, and alter the FHR and Factor H plasma repertoire. A clear association between the genetic modifications and the pathologic outcome is emerging: CFHR1 , CFHR3 , and Factor H gene alterations combined with intact CFHR2 , CFHR4 , and CFHR5 genes are reported in atypical hemolytic uremic syndrome. But alterations in each of the five CFHR genes in the context of an intact Factor H gene are described in C3 glomerulopathy. These genetic modifications influence complement function and the interplay of the five FHR proteins with each other and with Factor H. Understanding how mutant or hybrid FHR proteins, Factor H::FHR hybrid proteins, and altered Factor H, FHR plasma profiles cause pathology is of high interest for diagnosis and therapy.
Article
Full-text available
Genomic aberrations affecting the genes encoding factor H (FH) and the five FH-related proteins (FHRs) have been described in patients with atypical hemolytic uremic syndrome (aHUS), a rare condition characterized by microangiopathic hemolytic anemia, thrombocytopenia, and ARF. These genomic rearrangements occur through nonallelic homologous recombinations caused by the presence of repeated homologous sequences in CFH and CFHR1-R5 genes. In this study, we found heterozygous genomic rearrangements among CFH and CFHR genes in 4.5% of patients with aHUS. CFH/CFHR rearrangements were associated with poor clinical prognosis and high risk of post-transplant recurrence. Five patients carried known CFH/CFHR1 genes, but we found a duplication leading to a novel CFHR1/CFH hybrid gene in a family with two affected subjects. The resulting fusion protein contains the first four short consensus repeats of FHR1 and the terminal short consensus repeat 20 of FH. In an FH-dependent hemolysis assay, we showed that the hybrid protein causes sheep erythrocyte lysis. Functional analysis of the FHR1 fraction purified from serum of heterozygous carriers of the CFHR1/CFH hybrid gene indicated that the FHR1/FH hybrid protein acts as a competitive antagonist of FH. Furthermore, sera from carriers of the hybrid CFHR1/CFH gene induced more C5b-9 deposition on endothelial cells than control serum. These results suggest that this novel genomic hybrid mediates disease pathogenesis through dysregulation of complement at the endothelial cell surface. We recommend that genetic screening of aHUS includes analysis of CFH and CFHR rearrangements, particularly before a kidney transplant.
Article
Full-text available
Complement C3 activation is a characteristic finding in membranoproliferative GN (MPGN). This activation can be caused by immune complex deposition or an acquired or inherited defect in complement regulation. Deficiency of complement factor H has long been associated with MPGN. More recently, heterozygous genetic variants have been reported in sporadic cases of MPGN, although their functional significance has not been assessed. We describe a family with MPGN and acquired partial lipodystrophy. Although C3 nephritic factor was shown in family members with acquired partial lipodystrophy, it did not segregate with the renal phenotype. Genetic analysis revealed a novel heterozygous mutation in complement factor H (R83S) in addition to known risk polymorphisms carried by individuals with MPGN. Patients with MPGN had normal levels of factor H, and structural analysis of the mutant revealed only subtle alterations. However, functional analysis revealed profoundly reduced C3b binding, cofactor activity, and decay accelerating activity leading to loss of regulation of the alternative pathway. In summary, this family showed a confluence of common and rare functionally significant genetic risk factors causing disease. Data from our analysis of these factors highlight the role of the alternative pathway of complement in MPGN.
Article
Full-text available
Hemolytic uremic syndrome (HUS) is a triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure. The atypical form of HUS is a disease characterized by complement overactivation. Inherited defects in complement genes and acquired autoantibodies against complement regulatory proteins have been described. Incomplete penetrance of mutations in all predisposing genes is reported, suggesting that a precipitating event or trigger is required to unmask the complement regulatory deficiency. The underlying genetic defect predicts the prognosis both in native kidneys and after renal transplantation. The successful trials of the complement inhibitor eculizumab in the treatment of atypical HUS will revolutionize disease management.
Article
Full-text available
Factor H related proteins comprise a group of five plasma proteins: CFHR1, CFHR2, CFHR3, CFHR4 and CFHR5, and each member of this group binds to the central complement component C3b. Mutations, genetic deletions, duplications or rearrangements in the individual CFHR genes are associated with a number of diseases including atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathies (C3 glomerulonephritis (C3GN), dense deposit disease (DDD) and CFHR5 nephropathy), IgA nephropathy, age related macular degeneration (AMD) and systemic lupus erythematosus (SLE). Although complement regulatory functions were attributed to most of the members of the CFHR protein family, the precise role of each CFHR protein in complement activation and the exact contribution to disease pathology is still unclear. Recent publications show that CFHR proteins form homo- as well as heterodimers. Genetic abnormalities within the CFHR gene locus can result in hybrid proteins with affected dimerization or recognition domains which cause defective functions. Here we summarize the recent data about CFHR genes and proteins in order to better understand the role of CFHR proteins in complement activation and in complement associated diseases.
Article
Full-text available
Pathologic thrombosis is a major cause of mortality. Hemolytic-uremic syndrome (HUS) features episodes of small-vessel thrombosis resulting in microangiopathic hemolytic anemia, thrombocytopenia and renal failure. Atypical HUS (aHUS) can result from genetic or autoimmune factors that lead to pathologic complement cascade activation. Using exome sequencing, we identified recessive mutations in DGKE (encoding diacylglycerol kinase ɛ) that co-segregated with aHUS in nine unrelated kindreds, defining a distinctive Mendelian disease. Affected individuals present with aHUS before age 1 year, have persistent hypertension, hematuria and proteinuria (sometimes in the nephrotic range), and develop chronic kidney disease with age. DGKE is found in endothelium, platelets and podocytes. Arachidonic acid-containing diacylglycerols (DAG) activate protein kinase C (PKC), which promotes thrombosis, and DGKE normally inactivates DAG signaling. We infer that loss of DGKE function results in a prothrombotic state. These findings identify a new mechanism of pathologic thrombosis and kidney failure and have immediate implications for treating individuals with aHUS.
Article
The serum protein complement factor H (FH) ensures downregulation of the complement alternative pathway, a branch of innate immunity, upon interaction with specific glycans on host cell surfaces. Using ligand-based NMR, we screened a comprehensive set of sialylated glycans for binding to FH and solved the crystal structure of a ternary complex formed by the two C-terminal domains of FH, a sialylated trisaccharide and the complement C3b thioester-containing domain. Key residues in the sialic acid binding site are conserved from mice to men, and residues linked to atypical hemolytic uremic syndrome cluster within this binding site, suggesting a possible role for sialic acid as a host marker also in other mammals and a critical role in human renal complement homeostasis. Unexpectedly, the FH sialic acid binding site is structurally homologous to the binding sites of two evolutionarily unrelated proteins. The crystal structure also advances our understanding of bacterial immune evasion strategies.
Article
Mutations in complement factor H (CFH) are associated with complement dysregulation and the development of an aggressive form of atypical hemolytic uremic syndrome (aHUS) that progresses to end-stage renal disease (ESRD) and in most patients has a high rate of recurrence following transplantation. Sequence analysis of CFH and its downstream complement factor H-related genes (CFHR1-5) reveals several macrohomologous blocks caused by large genomic duplications. This high degree of sequence identity renders this area susceptible to nonallelic homologous recombination (NAHR) events, resulting in large-scale deletions, duplications, and the generation of hybrid CFH genes. Here, we report the finding of a novel CFHR1/CFH hybrid gene created by a de novo NAHR event in a 14-year-old girl with aHUS. The resulting fusion protein contains the first three short consensus repeats (SCRs) of CFHR1 and the terminal two SCRs of CFH. This finding demonstrates a novel pathogenic mechanism for the development of aHUS. Additionally, since standard Sanger sequencing is unable to detect such rearrangements, all aHUS patients should receive comprehensive genetic screening that includes analysis of copy number variation in order to identify patients with poor clinical prognoses.
Article
Hemolytic uremic syndrome (HUS) is a disease of microangiopathic hemolytic anemia, thrombocytopenia and acute renal failure. About 90% of cases are secondary to infections by Escherichia coli strains producing Shiga-like toxins (STEC-HUS), while 10% are associated with mutations in genes encoding proteins of complement system (aHUS). We describe two patients with a clinical history of STEC-HUS, who developed end-stage renal disease (ESRD) soon after disease onset. They received a kidney transplant but lost the graft for HUS recurrence, a complication more commonly observed in aHUS. Before planning a second renal transplantation, the two patients underwent genetic screening for aHUS-associated mutations that revealed the presence of a heterozygous CFI mutation in patient #1 and a heterozygous MCP mutation in patient #2, and also in her mother who donated the kidney. This finding argues that the two cases originally diagnosed as STEC-HUS had indeed aHUS triggered by STEC infection on a genetic background of impaired complement regulation. Complement gene sequencing should be performed before kidney transplantation in patients who developed ESRD following STEC-HUS since they may be undiagnosed cases of aHUS, at risk of posttransplant recurrence. Furthermore, genetic analysis of donors is mandatory before living-related transplantation to exclude carriers of HUS-predisposing mutations.