ArticlePDF Available

The disease-protective complement factor H allotypic variant Ile62 shows increased binding affinity for C3b and enhanced cofactor activity

Authors:

Abstract and Figures

Mutations and polymorphisms in the gene encoding factor H (CFH) have been associated with atypical haemolytic uraemic syndrome, dense deposit disease and age-related macular degeneration. The disease-predisposing CFH variants show a differential association with pathology that has been very useful to unravel critical events in the pathogenesis of one or other disease. In contrast, the factor H (fH)-Ile(62) polymorphism confers strong protection to all three diseases. Using ELISA-based methods and surface plasmon resonance analyses, we show here that the protective fH-Ile(62) variant binds more efficiently to C3b than fH-Val(62) and competes better with factor B in proconvertase formation. Functional analyses demonstrate an increased cofactor activity for fH-Ile(62) in the factor I-mediated cleavage of fluid phase and surface-bound C3b; however, the two fH variants show no differences in decay accelerating activity. From these data, we conclude that the protective effect of the fH-Ile(62) variant is due to its better capacity to bind C3b, inhibit proconvertase formation and catalyze inactivation of fluid-phase and surface-bound C3b. This demonstration of the functional consequences of the fH-Ile(62) polymorphism provides relevant insights into the complement regulatory activities of fH that will be useful in disease prediction and future development of effective therapeutics for disorders caused by complement dysregulation.
Content may be subject to copyright.
The disease-protective complement factor H allotypic variant
Ile62 shows increased binding affinity for C3b and enhanced
cofactor activity
Agustín Tortajada1, Tamara Montes1, Ruben Martinez-Barricarte1, B. Paul Morgan2, Claire
L. Harris2,*, and Santiago Rodríguez de Córdoba1,*
1)Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Centro
de Investigación Biomédica en Enfermedades Raras and Instituto Reina Sofía de Investigaciones
Nefrológicas, Ramiro de Maeztu 9, 28040 Madrid, Spain.
2)Department of Medical Biochemistry and Immunology, School of Medicine, Cardiff University,
Cardiff, CF14 4XN, UK.
Summary
Mutations and polymorphisms in the gene encoding factor H (
CFH
) have been associated with
atypical haemolytic uraemic syndrome, dense deposit disease and age-related macular
degeneration. The disease-predisposing
CFH
variants show a differential association with
pathology that has been very useful to unravel critical events in the pathogenesis of one or other
disease. In contrast, the fH-Ile62 polymorphism confers strong protection to all three diseases.
Using ELISA-based methods and surface plasmon resonance analyses we show here that the
protective fH-Ile62 variant binds more efficiently to C3b than fH-Val62 and competes better with
factor B in proconvertase formation. Functional analyses demonstrate an increased cofactor
activity for fH-Ile62 in the factor I-mediated cleavage of fluid phase and surface-bound C3b;
however, the two fH variants show no differences in decay accelerating activity. From these data
we conclude that the protective effect of the fH-Ile62 variant is due to its better capacity to bind
C3b, inhibit proconvertase formation and catalyse inactivation of fluid-phase and surface-bound
C3b. This demonstration of the functional consequences of the fH-Ile62 polymorphism provides
relevant insights into the complement regulatory activities of fH that will be useful in disease
prediction and future development of effective therapeutics for disorders caused by complement
dysregulation.
Introduction
Complement is a major component of innate immunity with crucial roles in microbial
killing, apoptotic cell clearance and immune complex handling. Activation of complement
Corresponding authors: Dr. Santiago Rodríguez de Córdoba, Complement Genetics and Molecular Pathology Unit, Depart. of Cellular
and Molecular Physiopathology, Centro de Investigaciones Biologicas, Ramiro de Maeztu 9, Madrid 28040, Spain. Tel: (+34) 91
7373112 x4432 Fax: (+34) 91 5360432 srdecordoba@cib.csic.es Dr. Claire L Harris, Complement Biology Group, Depart. of Medical
Biochemistry and Immunology, School of Medicine, Cardiff University, Henry Wellcome Building, Heath Park, Cardiff, CF14 4XN,
UK. Tel: (+44) 29 20687012 Fax: (+44) 29 20687079 HarrisCL@cardiff.ac.uk.
*)These two authors contributed equally to this work.
Author’s contributions CLH, BPM and SRdeC designed research, analysed the data and wrote the paper. CLH and SRdeC contribute
equally to this work. AT, TM and RMB prepared the proteins. AT and CLH performed the binding and functional assays.
Publisher's Disclaimer: This is a pre-copy-editing, author-produced PDF of an article accepted for publication in Human Molecular
genetics following peer review. The definitive publisher-authenticated version [Hum. Mol. Genet. (2009) 18 (18): 3452-3461. doi:
10.1093/hmg/ddp289 First published online: June 23, 2009] is available at http://hmg.oxfordjournals.org/content/18/18/3452.long
Conflicts of interest Authors declare no conflict of interest
Europe PMC Funders Group
Author Manuscript
Hum Mol Genet. Author manuscript; available in PMC 2012 February 05.
Published in final edited form as:
Hum Mol Genet
. 2009 September 15; 18(18): 3452–3461. doi:10.1093/hmg/ddp289.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
by foreign surfaces (alternative pathway; AP), antibody (classical pathway; CP) or mannan
(lectin pathway; LP), causes target opsonisation, leukocyte recruitment, and cell lysis. The
critical steps in complement activation are the formation of unstable protease complexes,
named C3-convertases (AP, C3bBb; CP/LP, C4b2a) and the cleavage of C3 to generate C3b.
Convertase-generated C3b can form more AP C3-convertase, providing exponential
amplification to the initial activation. Binding of C3b to the C3-convertases generates the
C5-convertases with the capacity to bind and cleave C5, initiating formation of the lytic
membrane attack complex (MAC).
Nascent C3b binds indiscriminately to pathogens and adjacent host cells. To prevent damage
to self and to avoid wasteful consumption of components, complement is under the control
of multiple regulatory proteins that limit complement activation by inactivating C3b or C4b,
dissociating the multimolecular C3/C5 convertases or inhibiting MAC formation. In health,
activation of C3 in the blood is kept at a low level and deposition of C3b and further
activation of complement is limited to the surface of pathogens (1).
Factor H (fH) is a relatively abundant plasma protein that is essential to maintain
complement homeostasis and to restrict the action of complement to activating surfaces. fH
binds to C3b, accelerates the decay of the alternative pathway C3-convertase (C3bBb) and
acts as a cofactor for the fI-mediated proteolytic inactivation of C3b (2-4). fH regulates
complement both in fluid phase and on cellular surfaces (5-7). The factor H molecule is a
single polypeptide chain glycoprotein of 155 kDa composed of 20 repetitive units of ~60
amino acids (8), named short consensus repeats (SCR), arranged end-to-end like ‘beads on a
string’. fH presents different interaction sites for C3b and polyanions which delineate
distinct functional domains at the N- and C-termini. The C3b binding site in SCR1-4 is the
only site essential for the C3-convertase decay accelerating and fI cofactor activities of fH.
Similarly, the C3b/polyanion-binding site located within SCR19-20 is the most important
site for preventing alternative pathway activation through binding to host cell membranes
(9).
Several reports in the last few years have established that membranoproliferative
glomerulonephritis type II or dense deposit disease (MPGN2/DDD) (10-13), atypical
haemolytic uraemic syndrome (aHUS) (14-17) and age-related macular degeneration
(AMD) (18-21), are each associated with mutations or polymorphisms in the
CFH
gene. The
available data support the hypothesis that AP dysregulation is a unifying pathogenetic
feature of these diverse conditions. They also illustrate a remarkable genotype-phenotype
correlation in which distinct genetic variations at
CFH
specifically predispose to aHUS,
AMD or MPGN2. In addition to these
CFH
variants conferring increased risk to disease, one
common extended haplotype in the
CFH
gene has been described associated with lower risk
to aHUS, AMD and MPGN2/DDD (18, 22). This
CFH
haplotype carries the Ile62 variant
within the SCR1 domain in the N-terminal region that is essential for fH regulatory
activities. It is, therefore, possible that the substitution of Val for Ile at position 62 may
increase the fH regulatory activity and thus confer lower risk to AMD, MPGN2/DDD and
aHUS by reducing AP activation.
To test this hypothesis we have purified the two fH variants from the plasma of fH-Val62
and fH-Ile62 homozygote donors and performed a series of binding and functional analyses.
Our data show that the fH-Ile62 variant exhibits increased binding to C3b compared to fH-
Val62, and is also a more efficient cofactor for fI in the proteolytic inactivation of C3b.
Together these data provide an explanation for why fH-Ile62 protects from diseases
associated with AP dysregulation.
Tortajada et al. Page 2
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Results
Interaction of fH-Ile62 and fH-Val62 with surface-bound C3b
Purified C3b was immobilized on microtiter plates and serial dilutions of fH-Ile62 or fH-
Val62 variants, ‘polished’ free from potential aggregates by gel filtration, were allowed to
interact with C3b for two hours at 37°C. Factor H bound to C3b was detected using an
antifH mAb (35H9) that recognises equally both variants as described in Materials and
Methods. Binding of the protective fH-Ile62 variant to surface-bound C3b was significantly
higher than that of the fH-Val62 variant (P<0.0001) (Figure 1a). These data suggest that the
Val62Ile polymorphism influences the interaction between fH and C3b. To confirm these
findings in a different assay, we performed SPR studies using chips coated with identical
amounts of fH-Ile62 or fH-Val62 variants and flowed increasing concentrations of C3b.
These SPR assays replicated and extended the findings from ELISA experiments, showing
that fH-Ile62 binds C3b with a higher affinity than fH-Val62 (Figure 2a). Steady state
analysis under defined buffer conditions gave a KD of 1.04μM for fH-Ile62 and 1.33μM for
fH-Val62 (Figure 2b).
Cofactor activity for fI-mediated proteolysis of fluid phase C3b
In order to study the fI cofactor activity of the fH-Ile62 and fH-Val62 variants we first
performed a fluid phase cofactor activity assay. Identical amounts of purified fH-Ile62 and
fH-Val62 variants were added to purified C3b in the presence of fI and incubated for 2.5, 5,
7.5 and 10 minutes at 37°C. Under the conditions of these experiments 100% of C3b
cleavage was reached after 20 minutes of incubation. Controls for 0% cleavage were
obtained in the absence of fI. The ratio between α′chain / βchain of C3b, determined by
densitometry, was used to determine the percentage of C3b cleavage. Figure 3a illustrates
one experiment representative of several, showing that the fH-Ile62 variant is more efficient
as a cofactor for fI in the cleavage of C3b in the fluid phase. Figure 3b shows a significant
difference (P=0.0012) in the % C3b cleavage catalysed by identical amounts of purified fH-
Ile62 and fH-Val62 variants at different incubation times. Double regression plotting and
statistical analysis of the slopes for the linearized curves reveal significant differences
between the cofactor activities of the fH-Ile62 and fH-Val62 variants. Figure 3c shows the
densitometry analysis for the differences in cofactor activities between the fH-Ile62 and fH-
Val62 variants at 6 minutes incubation time in an independent set of assays. From these
experiments it was calculated that fH-Ile62 is approximately 20% more active than fH-Val62
as a cofactor for the fI-mediated cleavage of fluid phase C3b.
Cofactor activity of fI-mediated inactivation of surface-bound C3b
To determine whether the fH-Ile62 variant is also more active than fH-Val62 as cofactor for
the fI-mediated inactivation of surface-bound C3b we used a haemolytic assay. C3b
deposited onto sheep erythrocytes was subjected to degradation by fI in the presence of
increasing amounts of purified fH-Ile62 or fH-Val62. For each fH concentration, the residual
surface-bound C3b was determined by measuring sheep erythrocyte lysis after lytic pathway
reconstitution (see Materials and Methods).
Three different experiments, each in triplicate, were performed with identical results (Figure
4). Calculated EC50 were 22.6nM and 14nM for fH-Ile62 and fH-Val62, respectively. These
experiments consistently show that fH-Ile62 is significantly more active than fH-Val62 as a
cofactor for the fI-mediated proteolysis of surface bound C3b (P=0.0025; two-tailed
unpaired T test). From these experiments it was estimated that the dose of fH-Val62 needed
to achieve 50% fI-mediated inactivation of C3b is 1.6-1.8 fold that required when fH-Ile62 is
used.
Tortajada et al. Page 3
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Decay accelerating activity of the alternative pathway C3-convertase
To measure AP convertase decay accelerating activity of the fH-Ile62 and fH-Val62 variants,
sheep erythrocytes were coated with AP convertase (C3bBb) and incubated with increasing
amounts of purified fH-Ile62 or fH-Val62 in the absence of fI. Residual AP convertase on the
sheep erythrocytes was determined by measuring erythrocyte lysis after lytic pathway
reconstitution (see Materials and Methods). In three independent experiments, these
hemolytic assays showed that fH-Ile62 and fH-Val62 have equivalent decay accelerating
activity (Figure 5a). Independent confirmation of this finding was sought using Biacore
(Figure 5b). AP C3 convertase was assembled on a C3b-coated chip and allowed to decay
naturally for 160 seconds; fH-Ile62 or fH-Val62 at a concentration of 73nM were then flowed
over the chip. Binding of fH and accelerated convertase decay occurred simultaneously.
Following dissociation of fH from the surface, remaining convertase was measured, this was
identical for each fH variant. Note the increased binding of fH-Ile62 to the surface in
agreement with Figure 2a.
Competition between fH and fB for binding to C3b
From the experiments presented above it is clear that the differences in binding affinity for
C3b of the fH-Ile62 and fH-Val62 variants affect their capacity to function as cofactor for fI
in the proteolysis of C3b. To explore whether these differences in affinity also influence the
ability of fH to prevent formation of the C3 proconvertase by competing with fB for binding
to C3b competition assays were performed on Biacore. We first showed, in keeping with
previous reports, that fH does not accelerate decay of the pre-formed proconvertase C3bB
(Figure 6a). When fB together with increasing amounts of fH was flowed over a C3b
surface, competition between fB and fH for binding to C3b was apparent from the fH-
dependent decrease in the formation of proconvertase measured following dissociation of fH
(Figure 6b). Next, fB was flowed over C3b and binding competed using identical amounts
of the fH-Ile62 and fH-Val62 variants. As expected, fH-Ile62, shown to bind better to C3b,
was a more efficient competitor and caused a small but consistent decreased formation of
the proconvertase (Figure 6c). These data illustrate that the increased C3b-binding affinity of
the fH-Ile62 variant makes it not only a better cofactor for the fI-dependent inactivation of
C3b, but also a more efficient inhibitor of the formation of the C3 proconvertase.
Combined effects of the fH Val62Ile and fB Arg32Gln polymorphisms in the formation of
the AP C3 convertase
Previously, we have characterized the common fB polymorphism, fB-Arg32/fB-Gln32/fB-
Trp32, and found that the AMD-protective allele fB-Gln32 had decreased affinity for C3b
compared with the fB-Arg32 and fBTrp32 alleles. SPR comparison revealed markedly
different proenzyme formation activities; fB-Arg32 bound C3b with 4-fold higher affinity
than fB-Gln32, and formation of activated convertase was enhanced (29). Here we tested
combinations of these two variants of fB with the two variants of fH characterised above in
order to explore the consequences of different combinations of variant components and
regulators. In haemolytic assays, we found that the combinations complemented each other
as predicted from their individual activities (Figure 7). The fH-Ile62-fB-Gln32 combination
was the least lytic and the fH-Val62-fB-Arg32 combination the most lytic (Figure 7).
Calculated EC50s were 4.3nM and 3.5nM (for the fH-Ile62-fB-Gln32 and fH-Val62-fB-Gln32
combinations, respectively) and 3nM and 2.1nM (for the fH-Ile62-fB-Arg32 and fH-Val62-
fB-Arg32 combinations, respectively). Differences in the EC50 were statistically significant
between the combinations fH-Val62-fB-Arg32 and fH-Ile62-fB-Gln32 (P<0.001); fH-Val62-
fB-Arg32 and fH-Ile62-fB-Arg32 (P=0.004); and fH-Val62-fB-Gln32 and fH-Ile62-fB-Gln32
(P=0.034). P values were calculated using a two-tailed unpaired T test. No significant
differences were observed between the combinations fH-Val62-fB-Gln32 and fH-Ile62-fB-
Arg32.
Tortajada et al. Page 4
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Discussion
Factor H (fH) plays a key role in regulating the alternative pathway by acting as a cofactor
for fI-mediated cleavage of C3b to iC3b, by accelerating the dissociation of the alternative
pathway C3 convertases and by competing with factor B for binding to C3b in
proconvertase formation (9). All these activities are mediated by the interaction between fH
and C3b. Functional studies using truncated molecules have demonstrated that fH possesses
binding sites for C3b located at the N-terminus (SCR1-4), the C-terminus (SCR19-20) and
in the middle of the molecule (SCR7) (30, 31). The C3b-binding sites at the C-terminal and
N-terminal ends are well characterized, whereas that in SCR7 is a very weak binding site of
unknown function. The C3b-binding site in SCR19-20 shows the highest affinity for C3b
and plays a critical role in recognition of foreign surfaces by fH. At the other end of the
molecule, the C3b-binding site in SCR1-4 is essential for the regulatory activities of fH as it
carries the fI-mediated cofactor and decay-accelerating activities of fH. Deletion
mutagenesis studies have demonstrated that the N-terminal four SCRs are necessary and
sufficient for these activities of fH, suggesting that multiple interactions occur between C3b
and the N-terminal region of fH (32, 33).
Here we report that the Val62Ile substitution in SCR1 of fH increases its affinity for C3b; as
a consequence, when compared to fH-Val62, fH-Ile62 competes more efficiently with fB for
C3b binding in proconvertase formation and acquires enhanced cofactor activity for the
factor-I mediated cleavage of C3b proteolysis; however, its decay accelerating activity is not
altered. These findings show that fH-Ile62 is a better AP convertase inhibitor and provide an
explanation for the association of the fH-Ile62 variant with protection in three distinct
disorders linked by AP dysregulation. The fact that the Val62Ile substitution affects binding
to C3b but not decay accelerating activity suggests that different regions in fH may be
involved in binding C3b/cofactor activity and in decay accelerating activity.
SCR1 is necessary for both cofactor and decay accelerating activities (32, 33). Our findings
imply that the C3b-binding site in SCR1 is not directly involved in decay accelerating
activity and that SCR1 may contain distinct, although perhaps overlapping, sites for cofactor
and decay accelerating activities. This scenario dictates that the interactions of fH with C3b
and with C3bBb are structurally distinct. Previously, we showed that the aHUS-associated
fB mutation, K323E, located remote from the C3b-fB interaction site, makes the C3bBb
convertase resistant to decay by decay accelerating factor (DAF) and fH (24, 34). The
mutation apparently affects a complement regulator binding site in the von Willebrand
factor type A (vWA) domain of fB (24). We have also previously showed that DAF-SCR2
interacts with Bb, whereas DAF-SCR4 interacts with C3b in the C3bBb complex (27). From
comparison with DAF it is likely that decay accelerating activity of fH also requires binding
to both Bb and C3b. We suggest that there are two distinct binding sites in SCR1, one
including the Val62Ile fH polymorphism that is necessary for cofactor activity, and a second
that binds fB at, or close to, K323 in fB that is essential for decay accelerating activity. We
also postulate that fH has a C3b binding site in SCR3/SCR4 that contributes to both cofactor
and decay accelerating activities.
Overwhelming evidence has associated MPGN2/DDD, aHUS and AMD with mutations or
polymorphisms in the
CFH
gene and provided conclusive data that AP dysregulation is a
unifying pathogenetic feature of these diverse conditions (35). However, only MPGN2/DDD
and AMD have pathological similarities. Indeed, occasionally, they occur in the same
patient (36). The hallmark of AMD is drusen, a complex, complement-containing material
that accumulates beneath the retinal pigmented epithelium; in MPGN2/DDD, accumulation
of a drusen-like C3 and electron-dense material occurs along the glomerular basement
membrane (GBM). In contrast to these ‘debris-associated’ conditions, aHUS is characterized
Tortajada et al. Page 5
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
by renal endothelial cell injury and thrombosis (thrombotic microangiopathy), resulting in
haemolytic anaemia, thrombocytopenia and renal failure. Consistent with these differences,
distinct functional alterations in fH associate with pathogenesis in these disorders. Mutations
or polymorphisms altering the C3b/polyanions-binding site located at the C–terminal region
of fH are strongly associated with aHUS because they impair the capacity of fH to protect
host cells but have no effect on fluid-phase fH activities. On the other hand, mutations that
disrupt the capacity of fH to inhibit complement activation in plasma result in massive
activation of C3 that causes MPGN2/DDD. This clear genotype-phenotype correlation
contrasts with the association of the fH Val62Ile polymorphism, associated with lower risk
for the three diseases (18, 22).
To understand why the fH-Ile62 variant confers protection from aHUS, MPGN2/DDD and
AMD, we purified to homogeneity both fH-Val62 and fH-Ile62 variants and compared in a
series of functional assays for potential effects on proenzyme formation and cofactor and
decay accelerating activities in fluid phase and on cell surfaces. Using four different
experimental approaches, we showed that fH-Ile62 binds better to C3b, competes better with
fB to reduce proenzyme formation, and performs more efficiently as a cofactor of fI in the
proteolysis of fluid phase and surface-bound C3b. These enhanced activities explain the
protective role of fH-Ile62 both in diseases associated with fluid phase complement
dysregulation, like MPGN2/DDD, and membrane-restricted dysregulation as is the case in
aHUS.
One important conclusion from this report is that the protective effect of the fH-Ile62 variant
is subtle, with alterations in activities of between 20% and 50% depending on the assay
used. This is consistent with the recent observation (37) that the Val62Ile polymorphism
causes a very minor perturbation in the structure of SCR1, this contrasts with the larger
structural disturbance caused by an aHUS-associated mutation (Arg53His) which has
detrimental consequences on the functional activities of fH. Nevertheless, the very nature of
the complement system will amplify these small effects. Further, as we show here by
combining known functional variants in fB with fH-Ile62 and fH-Val62, particular
combinations of variants in components and regulators will result in very different AP
characteristics, markedly affecting formation and regulation of the AP C3 convertase in
plasma and on cell surfaces. Identification of individuals carrying ‘high risk’ or ‘low risk’
combinations (‘complotypes’) of the polymorphic complement component and regulator
variants will be of great importance for prediction of disease risk and may also help in
diagnosis and choice of treatment for diseases involving complement dysregulation.
Materials and Methods
Purification of complement components and activation fragments
Normal healthy volunteers were screened for mutations/polymorphisms in the
CFH
gene by
automatic DNA sequencing of PCR amplified fragments. Genomic DNA was prepared from
peripheral blood cells according to standard procedures (23). Each exon of the
CFH
gene
was amplified from genomic DNA by using specific primers derived from the 5 and 3
intronic sequences as described (14). Automatic sequencing was performed in an ABI 3730
sequencer using a dye terminator cycle sequencing kit (Applied Biosystems, Foster City,
CA).
Factor H was purified from individuals homozygous for either the fH-Ile62 and fH-Val62
variants who were identical at all other amino acid residues. Fresh EDTA plasma (100 ml)
was precipitated with 7% polyethylene glycol 8000 overnight at 4°C. The precipitate was re-
dissolved in PBS, dialysed extensively against 20 mM Tris-HCl (pH 7.4), 50 mM NaCl, 5
mM EDTA and applied to a heparin-Sepharose column (Heparin 6B Fast Flow, Amersham)
Tortajada et al. Page 6
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
equilibrated in the same buffer. The proteins bound to the column were eluted with a
100-200 mM NaCl gradient in 20mM Tris-HCl, pH 7.4, 5mM EDTA. Fractions containing
fH were identified by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-
PAGE), pooled, dialysed against 20 mM Tris-HCl; pH 7.6, 20 mM NaCl and 10 mM EDTA
and applied to a DEAE-Sephacel column. Bound proteins were eluted with a 20-300 mM
NaCl gradient. Fractions containing fH were identified by SDS-PAGE, pooled and further
purified by gel filtration on a Superose™ 6 10/300 column (GE Healthcare). The fH peak
fractions were pooled and stored frozen at −70°C. The fH used in haemolysis assays and
Biacore studies was purified by affinity chromatography using immobilised anti-fH (35H9;
in house). Protein was eluted with 0.1M Glycine/HCl pH 2,5 and gel filtered into assay
buffer using a Superdex 200 10/300 column (GE Healthcare) immediately prior to analysis.
The purity of the final preparations was confirmed by SDS-PAGE. Preparations of fH-Ile62
and fH-Val62 were obtained without any detectable contaminants or aggregates (Figure 1b).
C3 and Factor B were purified by affinity chromatography and gel filtration as described
previously (24). Concentration of proteins was assessed using absorbance at A280,
molarities were calculated using an extinction coefficient for fH of 1.95 (25), for fB of 1.43
and for C3 of 0.98 (coefficients were obtained by using Protean Software, DNAStar). C3b
was generated by limited digestion with trypsin or convertase as previously described (24,
26) and re-purified by ion exchange and/or gel filtration as described above (GE
Healthcare). C3b was obtained without any detectable contaminants or aggregates. Factor I,
factor D and properdin were purchased from Comptech (Tyler, TX).
ELISA C3b–binding Assay
The binding of fH variants to surface-bound C3b was determined by ELISA. In a 96-well
polystyrene microtiter plate, C3b (5 μg/ml) in coupling buffer (0.1 M NaHCO3 pH 9.5) was
coated overnight at 4°C. The plate was blocked with washing buffer (20 mM Tris, 150 mM
NaCl and 0.1% Tween 20) with 1% Bovine Serum Albumin for 1 hour at room temperature
(RT). After washing, serial dilutions of fH variants (10μg/ml) in blocking buffer containing
150 mM NaCl, 5mM EDTA, were added and incubated with surface-bound C3b for 2 hours
at 37°C. After washing, the plate was incubated with anti-fH monoclonal antibody (mAb)
35H9 (in house) in blocking buffer, for 1 hour at RT, and then with a secondary antibody
coupled with horseradish peroxidase (DAKO). Colour reaction was developed with o-
phenylene-diamine (DAKO) and absorbance measured at 492nm. fH preparations used in
the ligand assay were quantified in duplicate in the same ELISA plate using immobilised
polyclonal anti-fH antibody to capture fH and the same anti-fH mAb, 35H9, and secondary
antibodies to measure the amount of protein. Concentrations of fH were calculated from
curves obtained using purified standard samples.
Biosensor Analysis
Kinetic analyses (Figure 2) were carried out on a Biacore T100, all other analyses were
carried out using a Biacore 3000 (GE Healthcare). To measure affinity, fH was amine
coupled to a CM5 (carboxymethylated dextran) chip as instructed by the manufacturer
(NHS/EDC coupling kit). Number of RUs loaded for both variants were 1004RU (fH Ile62)
and 1003RU (fH Val62). C3b was flowed across the surface at different concentrations and
bound protein was allowed to decay naturally, the buffer was 10mM Hepes pH7.4, 100mM
NaCl, 0.005% Surfactant P20. Data were collected at 25°C at a flow rate of 30μl/minute and
were double-referenced (data from reference cell and blank inject were subtracted) to
control for bulk refractive index changes. To calculate Kd values (Figure 2) we repeated this
experiment on three different surfaces: twice with C3b flowing, and once with hydrolysed
C3 flowing. Pooling the data from different runs is difficult. However, the ratio of the
derived Kd values was the same for each run as follows: fH-Ile62 was 0.77, 0.78 or 0.8 fold
Tortajada et al. Page 7
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
lower than the fH-Val62 form. We flowed C3b over the surface (rather than fH over C3b) in
order to minimise the avidity effects seen when flowing fH over the surface. In order to
obtain the best quality data, the C3b was gel-filtered prior to use to remove any aggregates
and then used in the experiment without further concentration. The C3b needed to be at a
very high concentration pre-filtration in order to achieve 1mg/ml post-filtration, this was the
maximum concentration that we could use without precipitating the protein pre-filtration.
Although we did not achieve saturation in these experiments, in each case the concentration
of C3b used exceeded the Kd value (2.2-fold for fH-Ile62 and 1.7-fold for fH-Val62).
In the following experiments the buffer was 10mM Hepes pH 7.4, 150mM NaCl, 1mM
Mg2+. To test the decay activity of fH (Figure 5), fB at 100μg/ml (1.1μM) and fD (2μg/
mL), were flowed across the C3b surface to form the AP C3 convertase as previously
described (27). The fH variants were subsequently flowed across the C3b surface at 11.3μg/
ml (73nM) and decay was monitored. To examine competition between fH and fB for
binding to C3b (Figure 6), both proteins were mixed at the indicated concentrations and
flowed at 30μl/min across the C3b surface in the absence of fD. To determine whether fH
accelerated decay of the proenzyme, fH was flowed over the surface subsequent to the fB
injection rather than being premixed.
Cofactor activity for fI-mediated proteolysis of fluid phase C3b
The fluid-phase cofactor activity of factor H was determined in a C3b proteolysis assay
using purified proteins. In brief, C3b, fH and fI were mixed in 10mM Hepes pH 7.5, 150mM
NaCl, 0.02% Tween 20 at final concentrations of 50 μg/ml (263nM), 4 μg/ml (25.8nM) and
10 μg/ml (114nM), respectively. Mixtures were incubated at 37°C in a water bath and 20μl
aliquots were collected at 2.5, 5, 7.5 and 10 minutes. The reaction was stopped by the
addition of 3μl of SDS sample buffer (2% SDS, 62.5mM Tris, 10% Glycerol, 0.75%
Bromophenol Blue). Samples were analyzed in 10% SDS-PAGE under reducing conditions.
Gels were stained with Coomassie brilliant blue R-250 (Bio Rad) and proteolysis of C3b
determined by measuring the cleavage of the α’-chain using a GS-800 calibrated
densitometer (BioRAD) and the MultiGauge software package (FUJIFILM). The C3b β-
chain was used as an internal control to normalize the % of cleavage between samples.
Percentage of cleavage was determined by the ratio between α′chain / βchain of C3b and
setting as 0% the amount of α’-chain at time 0.
Factor H-dependent haemolysis assays
NHS was sequentially depleted of fB and fH (NHSΔBΔH) by flowing over immobilised
anti-Bb (JC1 mAb; in house) and immobilised anti-fH (35H9; in house) affinity columns in
complement fixation diluent (CFD; Oxoid), undiluted depleted serum was pooled and used
in haemolysis assays as described below. Antibody-coated sheep erythrocytes (EA) were
prepared by incubating sheep E (2% v/v) with Amboceptor (1/1000 dilution; Behring
Diagnostics) in complement fixation diluent (CFD; Oxoid) for 30 minutes at 37°C, EA were
washed and resuspended at 2% (v/v) in CFD. To deposit C3b on the E surface (E-C3b),
equal volumes of EA and NHSΔBΔH (8% v/v) were incubated at 37°C for 10 minutes, the
C5 inhibitor (OmCI; 6μg/ml; (28) was added to block the terminal pathway).
To test fH dependent decay accelerating activity, washed E-C3b cells were resuspended to
2% (v/v) in AP buffer (5 mM sodium barbitone pH 7.4, 150 mM NaCl, 7 mM MgCl2, 10
mM EGTA) and AP convertase was formed on the cell surface by incubating with fB 42μg/
ml (0.46μM) and fD (0.4μg/ml) at 37°C for 15 minutes. 1/25 volume of PBS/0.25M EDTA
was added to prevent further enzyme formation and cells (50μl) were mixed and incubated
with 50μl of fH (serial dilution from 15.4μg/ml (99nM)) in PBS/10mM EDTA for 12
minutes. Lysis was developed by adding 50μl NHSΔBΔH (4%, v/v) in PBS/EDTA and
Tortajada et al. Page 8
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
incubating at 37°C for 20 minutes. To calculate lysis, cells were pelleted by centrifugation,
and hemoglobin release was measured by absorbance at 415 nm. Control incubations
included 0%lysis (buffer only) and 100%lysis (0.1% Nonidet-P40). Percentage lysis
100*(A415 test sample-A415 0% control)/(A415 100% control-A415 0% control).
To test fH cofactor activity, washed EA-C3b cells were resuspended to 2% in AP buffer and
incubated with an equal volume of different concentrations of fH as indicated and constant
fI (2.5μg/mL) for 7 minutes at 22°C. After three washes in AP buffer, 50μl cells (2%) were
mixed with 50μl of 70μg/ml fB (0.75μM; fB32R or fB32Q) and fD (0.4μg/ml) and incubated
for 10 minutes at 22°C to form convertase on residual C3b (EA-C3bBb). Lysis was
developed by adding 50μl NHSΔBΔH (4%, v/v) in PBS/EDTA and incubating at 37°C for
20 minutes. Percentage lysis was calculated as described above. To assess the effect on lysis
by combining different polymorphic variants of fB and fH, the above two assays were
combined and modified as follows. EA-C3b cells were incubated with 80ng/ml (0.5nM) fH-
Ile62 or fH-Val62 variant and 2.5μg/ml fI for 7 minutes at 22°C. Washed cells were
incubated as described above with different concentrations of fBArg32 or fBGln32, fD and
properdin (1μg/ml) and lysis was developed using NHSΔBΔH.
Acknowledgments
This work was supported by MRC Project Grant Ref 84908 (to CLH and BPM), Ministerio de Ciencia e Innovación
Ref SAF 2005-00913 (to SRdeC) the CIBER de Enfermedades Raras and Fundación Renal Iñigo Alvarez de
Toledo (to SRdeC). We thank the blood donors for their invaluable contribution to the project.
Abbreviations
CFH gene encoding factor H
AP alternative pathway
CP classical pathway
LP lectin pathway
MAC membrane attack complex
fH Factor H
SCR short consensus repeats
MPGN2/DDD membranoproliferative glomerulonephritis type II or dense deposit
disease
aHUS atypical haemolytic uraemic syndrome
AMD age-related macular degeneration
SPR surface plasmon resonance
fI factor I
fB factor B
NHS normal human serum
CFD complement fixation diluent
EA antibody-coated sheep erythrocytes
DAF decay accelerating factor
vWA von Willebrand factor type A
Tortajada et al. Page 9
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
References
1. Law, S.; Reid, K. Complement. 2 ed. IRL Press; Oxford, UK: 1995.
2. Pangburn MK, Schreiber RD, Müller-Eberhard HJ. Human complement C3b inactivator: isolation,
characterization, and demonstration of an absolute requirement for the serum protein beta1H for
cleavage of C3b and C4b in solution. J. Exp. Med. 1977; 146:257–70. [PubMed: 301546]
3. Weiler JM, Daha MR, Austen KF, Fearon DT. Control of the amplification convertase of
complement by the plasma protein beta1H. Proc. Natl. Acad. Sci. USA. 1976; 73:3268–3272.
[PubMed: 1067618]
4. Whaley K, Ruddy S. Modulation of the alternative complement pathway by beta1H globulin. J. Exp.
Med. 1976; 144:1147–1163. [PubMed: 62817]
5. Fearon DT. Regulation by membrane sialic acid of beta1H-dependent decay-dissociation of
amplification C3 convertase of the alternative complement pathway. Proc. Natl. Acad. Sci. USA.
1978; 75:1971–1975. [PubMed: 273923]
6. Kazatchkine MD, Fearon DT, Austen KF. Human alternative complement pathway: Membrane-
associated sialic acid regulates the competition between B and beta1H for cell-bound C3b. J.
Immunol. 1979; 122:75–81. [PubMed: 762425]
7. Pangburn MK, Schreiber RD, Muller Eberhard HJ. C3b deposition during activation of the
alternative complement pathway and the effect of deposition on the activating surface. J. Immunol.
1983; 131:1930–1935. [PubMed: 6225800]
8. Ripoche J, Day AJ, Harris TJR, Sim RB. The complete amino acid sequence of human complement
factor H. Biochem. J. 1988; 249:593–602. [PubMed: 2963625]
9. Rodriguez de Cordoba S, Esparza-Gordillo J, Goicoechea de Jorge E, Lopez-Trascasa M, Sanchez-
Corral P. The human complement factor H: functional roles, genetic variations and disease
associations. Mol. Immunol. 2004; 41:355–67. [PubMed: 15163532]
10. Dragon-Durey M-A, Fremeaux-Bacchi V, Loirat C, Blouin J, Niaudet P, Deschenes G, Coppo P,
Herman Fridman W, Weiss L. 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. 2004; 15:787–795. [PubMed: 14978182]
11. Levy M, Halbwachs-Mecarelli L, Gubler MC. H deficiency in two brothers with atypical dense
intramembranous deposit disease. Kidney Int. 1986; 30:949–956. [PubMed: 2950269]
12. Licht C, Heinen S, Józsi M, Löschmann I, Saunders RE, Perkins SJ, Waldherr R, Skerka C,
Kirschfink M, Hoppe B, et al. Deletion of Lys224 in regulatory domain 4 of Factor H reveals a
novel pathomechanism for dense deposit disease (MPGN II). Kidney Int. 2006; 70:42–50.
[PubMed: 16612335]
13. López-Larrea C, Dieguez MA, Enguix A, Dominguez O, Marin B, Gómez E. A familial deficiency
of complement factor H. Biochem. Soc. T. 1987; 15:648–649.
14. Perez-Caballero D, Gonzalez-Rubio C, Gallardo M. Esther, Vera M, Lopez-Trascasa M, Rodriguez
de Cordoba S, Sanchez-Corral P. Clustering of missense mutations in the C-terminal region of
factor H in atypical hemolytic uremic syndrome. Am. J. Hum. Genet. 2001; 68:478–484.
[PubMed: 11170895]
15. Caprioli J, Castelletti F, Bucchioni S, Bettinaglio P, Bresin E, Pianetti G, Gamba S, Brioschi S,
Daina E, Remuzzi G, et al. Complement factor H mutations and gene polymorphisms in
haemolytic uraemic syndrome: The C-257T, the A2089G and the G2881T polymorphisms are
strongly associated with the disease. Hum. Mol. Genet. 2003; 12:3385–3395. [PubMed:
14583443]
16. Richards A, Buddles MR, Donne RL, Kaplan BS, Kirk E, Venning MC, Tielemans CL, Goodship
JA, Goodship THJ. Factor H mutations in hemolytic uremic syndrome cluster in exons 18-20, a
domain important for host cell recognition. Am. J. Hum. Genet. 2001; 68:485–490. [PubMed:
11170896]
17. Warwicker P, Goodship THJ, Donne RL, Pirson Y, Nicholls A, Ward RM, Turnpenny P, Goodship
JA. Genetic studies into inherited and sporadic hemolytic uremic syndrome. Kidney Int. 1998;
53:836–844. [PubMed: 9551389]
Tortajada et al. Page 10
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
18. Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, Hardisty LI, Hageman JL,
Stockman HA, Borchardt JD, Gehrs KM, et al. A common haplotype in the complement
regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration.
Proc. Natl. Acad. Sci. USA. 2005; 102:7227–7232. [PubMed: 15870199]
19. Edwards AO, Ritter R, Abel KJ, Manning A, Panhuysen C, Farrer LA. Complement Factor H
Polymorphism and Age-Related Macular Degeneration. Science. 2005; 308:421–424. [PubMed:
15761121]
20. Haines JL, Hauser MA, Schmidt S, Scott WK, Olson LM, Gallins P, Spencer KL, Kwan SY,
Noureddine M, Gilbert JR, et al. Complement Factor H Variant Increases the Risk of Age-Related
Macular Degeneration. Science. 2005; 308:419–421. [PubMed: 15761120]
21. Klein RJ, Zeiss C, Chew EY, Tsai J-Y, Sackler RS, Haynes C, Henning AK, SanGiovanni JP,
Mane SM, Mayne ST, et al. Complement Factor H Polymorphism in Age-Related Macular
Degeneration. Science. 2005; 308:385–389. [PubMed: 15761122]
22. Pickering MC, Goicoechea de Jorge E, Martinez-Barricarte R, Recalde S, Garcia-Layana A, Rose
KL, Moss J, Walport MJ, Cook HT, Rodriguez de Cordoba S, et al. Spontaneous hemolytic uremic
syndrome triggered by complement factor H lacking surface recognition domains. J. Exp. Med.
2007; 204:1249–1256. [PubMed: 17517971]
23. Miller SA, Dykes DD, Polesky HF. A simple salting out procedure for extracting DNA from
human nucleated cells. Nucleic Acids Res. 1988; 16:1215. [PubMed: 3344216]
24. Goicoechea De Jorge E, Harris CL, Esparza-Gordillo J, Carreras L, Arranz E. Aller, Garrido C.
Abarrategui, Lopez-Trascasa M, Sanchez-Corral P, Morgan BP, Rodriguez De Cordoba S. Gain-
of-function mutations in complement factor B are associated with atypical hemolytic uremic
syndrome. Proc. Natl. Acad. Sci. USA. 2007; 104:240–245. [PubMed: 17182750]
25. Hakobyan S, Harris CL, Tortajada A, Goicochea de Jorge E, Garcia-Layana A, Fernandez-
Robredo P, Rodriguez de Cordoba S, Morgan BP. Measurement of factor H variants in plasma
using variant-specific monoclonal antibodies: application to assessing risk of age-related macular
degeneration. Invest. Ophthalmol. Vis. Sci. 2008; 49:1983–1990. [PubMed: 18436830]
26. Sanchez-Corral P, Anton LC, Alcolea JM, Marques G, Sanchez A, Vivanco F. Separation of active
and inactive forms of the third component of human complement C3, by fast protein liquid
chromatography (FPLC). J. Inmunol. 1989; 122:105–113.
27. Harris CL, Abbott RJM, Smith RA, Morgan BP, Lea SM. Molecular dissection of interactions
between components of the alternative pathway of complement and decay accelerating factor
(CD55). J. Biol. Chem. 2005; 280:2569–2578. [PubMed: 15536079]
28. Hepburn NJ, Williams AS, Nunn MA, Chamberlain-Banoub JC, Hamer J, Morgan BP, Harris CL.
In Vivo Characterization and Therapeutic Efficacy of a C5-specific Inhibitor from the Soft Tick
Ornithodoros moubata. J. Biol. Chem. 2007; 282:8292–8299. [PubMed: 17215252]
29. Montes T, Tortajada A, Morgan BP, Rodriguez de Cordoba S, Harris CL. Functional basis of
protection against age-related macular degeneration conferred by a common polymorphism in
complement factor B. Proc. Natl. Acad. Sci. USA. 2009; 106:4366–4371. [PubMed: 19255449]
30. Sharma AK, Pangburn MK. Identification of three physically and functionally distinct binding sites
for C3b in human complement factor H by deletion mutagenesis. Proc. Natl. Acad. Sci. USA.
1996; 93:10996–11001. [PubMed: 8855297]
31. Schmidt CQ, Herbert AP, Kavanagh D, Gandy C, Fenton CJ, Blaum BS, Lyon M, Uhrin D,
Barlow PN. A new map of glycosaminoglycan and C3b binding sites on factor H. J. Inmunol.
2008; 181:2610–2619.
32. Kuhn S, Skerka C, Zipfel PF. Mapping of the complement regulatory domains in the human factor
H-like protein I and in factor H. J. Immunol. 1995; 155:5663–5670. [PubMed: 7499851]
33. Gordon DL, Kaufman RM, Blackmore TK, Kwong J, Lublin DM. Identification of complement
regulatory domains in human factor H. J. Immunol. 1995; 155:348–356. [PubMed: 7541419]
34. Torreira E, Tortajada A, Montes T, Rodriguez de Cordoba S, Llorca O. 3D structure of the C3bB
complex provides insights into the activation and regulation of the complement alternative
pathway convertase. Proc. Natl. Acad. Sci. USA. 2009; 106:882–887. [PubMed: 19136636]
Tortajada et al. Page 11
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
35. Rodriguez de Cordoba S, Goicoechea de Jorge E. Translational Mini-Review Series on
Complement Factor H: Genetics and disease associations of human complement factor H. Clin.
Exp. Immunol. 2008; 151:1–13. [PubMed: 18081690]
36. Montes T, Goicoechea de Jorge E, Ramos R, Goma M, Pujol O, Sanchez-Corral P, Rodriguez de
Cordoba S. Genetic deficiency of complement factor H in a patient with age-related macular
degeneration and membranoproliferative glomerulonephritis. Mol. Immunol. 2008; 45:2897–2904.
[PubMed: 18336910]
37. Hocking HG, Herbert AP, Kavanagh D, Soares DC, Ferreira VP, Pangburn MK, Uhrin D, Barlow
PN. Structure of the N-terminal Region of Complement Factor H and Conformational Implications
of Disease-linked Sequence Variations. J. Biol. Chem. 2008; 283:9475–9487. [PubMed:
18252712]
Tortajada et al. Page 12
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 1. ELISA of fH-Ile62 and fH-Val62 binding to C3b
(a) Interaction between serial dilutions of purified fH-Ile62 (open circles) or fH-Val62 (filled
circles) with C3b deposited in 96-well plates is expressed as Abs492. Means ± S.D. of three
independent experiments are shown. Inset panel shows the double reciprocal plot of the fH-
Ile62 (open circles) and fH-Val62 (filled circles) C3b-binding curves. Multiple linear
regression analysis revealed significant differences between Val62 and Ile62 binding to C3b
(P<0.0001).
(b) SDS-PAGE illustrating the fH-Ile62 and fH-Val62 purified from the plasma of
homozygote carriers as described in Materials and Methods and then gel filtered to remove
aggregates.
Tortajada et al. Page 13
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 2. SPR analysis of fH-Ile62 and fH-Val62 binding to C3b
(a) Identical amounts of fH were immobilised onto a CM5 chip (fH-Ile62 1004RU
immobilised; fH-Val62 1003RU immobilised). C3b (2.2μM-8.6nM; 1/2 serial dilution) was
flowed across the fH-Ile62 or fH-Val62 surfaces in 10mm Hepes pH 7.4, 100mM NaCl,
0.005% surfactant P20. Data from a reference cell was subtracted to control for any bulk
changes in refractive index. Sensorgrams resulting from fH-Ile62 are solid lines and fH-
Val62 are dotted lines; identical concentrations are illustrated for the two variants.
(b) Steady state analysis of the data in these buffer conditions indicate the affinities for C3b
are: KD fH-Ile62: 1.03μM, KD fH-Val62: 1.33μM. The standard errors (SE) in the fits are
0.14μM for fH-Val62 and 0.12μM for fH-Ile62.
Tortajada et al. Page 14
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 3. Cofactor activity of fH-Ile62 and fH-Val62 variants in the proteolysis of fluid phase C3b
(a) SDS-PAGE of C3b proteolysis. C3b, fH and fI were incubated for the times indicated,
the reaction was stopped by the addition of SDS sample buffer. Samples were analyzed by
SDS-PAGE under reducing conditions and gels were Coomassie-stained.
(b) Densitometric analysis of C3b proteolysis. Fluid phase cofactor activity was measured be
examining C3b cleavage at 2.5, 5, 7.5 and 10 minutes reaction for both fH-Ile62 (open
circles) and fH-Val62 (filled circles) variants. Percentage of cofactor activity was determined
by the ratio of cleaved α′chain:βchain, normalized to 0% proteolysis of control samples.
Inset panel shows the double reciprocal plot of the fH-Ile62 (open circles) and fH-Val62
(filled circles) of the cofactor activity curves. Multiple linear regression analysis revealed
significant differences between the slopes for fH-Val62 and fH-Ile62 cofactor activities
(P=0.0012).
(c) Densitometric analysis of C3b proteolysis from an independent set of assays at 6 minutes
incubation time. Difference in percentage of cofactor activity between fH-Val62 and fH-Ile62
was significant (P<0.001).
Tortajada et al. Page 15
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 4. Cofactor activity of fH-Ile62 and fH-Val62 variants in the proteolysis of surface-bound
C3b
The ability of the fH variants to mediate fI-catalysed inactivation of surface-bound C3b was
assessed using a haemolysis assay. C3b was deposited on the surface of sheep E using the
classical pathway as described in Methods. E-C3b were incubated in AP buffer with
different concentrations of fH-Ile62 (open circles) or fH-Val62 (filled circles) and constant fI
for 7 minutes at 22°C. Cells were washed and AP convertase was formed using purified fB
and fD. Lysis was developed in EDTA-containing buffer using serum depleted of fB and fH.
Percent lysis was calculated for each concentration of fH. The log10 of fB concentration
(final concentration in the incubation) was plotted on the
x
axis, and percentage lysis on the
y
axis. Data points represent mean ±SD of 3 determinations. The curves were fitted by using
nonlinear regression analysis to calculate the EC50. There are significant differences
(P=0.0025) between the EC50 values corresponding to the fH-Ile62 (14nM) and fH-Val62
(22.6nM) variants.
Tortajada et al. Page 16
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 5. Decay accelerating activity of fH-Ile62 and fH-Val62 variants on surface-bound AP
convertase
The ability of the fH variants to accelerate decay of the AP convertase, C3bBb, was assessed
using haemolysis assays with convertase coated sheep E as target (a), and in real time using
SPR (b). (a) C3b was deposited on the surface of sheep E using the classical pathway as
described in Methods. AP convertase was formed on the cell surface using purified fB and
fD, convertase formation was stopped after 15 minutes using EDTA. E-C3bBb were
incubated in EDTA with different concentrations of fH-Ile62 (open circles) or fH-Val62
(filled circles) for 12 minutes to allow decay of the convertase and lysis was developed
using serum depleted of fB and fH. Percent lysis was calculated for each concentration of
fH. (b) AP convertase was formed on the surface of a C3b-coated Biacore chip by flowing
fB and fD over the surface. Convertase decayed naturally for 160s prior to injection of either
fH variant (73nM). Change in RU (y-axis) during the fH injection represents the combined
effect of fH binding to the surface and to C3bBb, and loss of Bb from the convertase due to
fH-mediated accelerated decay. Despite enhanced binding of fH-Ile62 to the surface (grey
line), an identical amount of Bb was decayed from the surface as measured following
complete dissociation of fH from the chip surface.
Tortajada et al. Page 17
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 6. Competition between fH and fB for binding to C3b
(a) Proconvertase was formed on the surface of a C3b-coated Biacore chip by flowing fB,
this was allowed to decay naturally for a short time before injection of fH as indicated. As
expected, fH did not accelerate decay of the proenzyme. The binding profile of fH on the
C3b surface only (no fB injected) is illustrated in grey for comparison. (b) In order to
demonstrate competition between fB and fH for binding to the C3b-coated surface, fB
(662nM) was flowed across the C3b-coated surface alone (black line), or was premixed with
26 or 66nM fH (dotted grey and solid grey lines respectively) before injection. Note that the
change in RU (y-axis) represents the sum of both fB and fH binding to the surface.
Decreased proconvertase formation is evident with increasing fH. (c) In order to analyse
differential effects of fH-Ile62 and fH-Val62 on proconvertase formation, 132nM of either
variant was premixed with fB (338nM) and injected over the surface. Comparison of
binding curves (following dissociation of fH from the surface) with fB binding in the
absence of any fH demonstrates that both fH variants prevent proconvertase formation and
that the fH-Ile62 variant is more effective.
Tortajada et al. Page 18
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
Figure 7. Hemolytic activity of different fH and fB variant combinations
To test the combined effect of the fH-Ile62, fH-Val62 and the fBArg32, fBGln32 variants,
C3b was deposited on the surface of sheep E using the classical pathway as described in
Methods. E-C3b were incubated in AP buffer with 1nM of fH-Ile62 or fH-Val62 (final
concentration) and constant fI for 7 minutes at 22°C. Cells were washed and AP convertase
was formed using different concentrations of purified fBArg32 or fBGln32, and constant fD
and properdin. Lysis was developed in EDTA-containing buffer using serum depleted of fB
and fH. Percent lysis was calculated for each concentration of fH.
The log10 of fB concentration (final concentration in the incubation) was plotted on the
x
axis, and percentage lysis on the
y
axis. Data points represent mean ±SD of 3
determinations. The curves were fitted by using nonlinear regression analysis to calculate
the EC50. Two-tailed unpaired T test showed significant differences in the EC50 between
the combinations fH-Val62-fB-Arg32 (filled circles) and fH-Ile62-fB-Gln32 (open triangles)
(P<0.001); fH-Val62-fB-Arg32 (filled circles) and fH-Ile62-fB-Arg32 (open circles)
(P=0.004); and fH-Val62-fB-Gln32 (filled triangles) and fH-Ile62-fB-Gln32 (open triangles)
(P=0.034).
Tortajada et al. Page 19
Hum Mol Genet
. Author manuscript; available in PMC 2012 February 05.
Europe PMC Funders Author Manuscripts Europe PMC Funders Author Manuscripts
... The first form of genetic protection is conferred by a common polymorphism, tagged by rs800292, that encodes a valine to isoleucine substitution at amino acid position 62 of FH/FHL-1. The I62 allotype confers increased complement co-factor and C3 convertase decay acceleration activities that result in reduced AP activation 35,36 . The second form of AMD protection arises from a deletion of both the CFHR3 and CFHR1 genes (CFHR3/1 deletion), two genes that are expressed by liver, but not ocular tissue 27,30,37 . ...
Article
Full-text available
Dysregulation of the alternative pathway (AP) of the complement system is a significant contributor to age-related macular degeneration (AMD), a primary cause of irreversible vision loss worldwide. Here, we assess the contribution of the liver-produced complement factor H-related 4 protein (FHR-4) to AMD initiation and course of progression. We show that FHR-4 variation in plasma and at the primary location of AMD-associated pathology, the retinal pigment epithelium/Bruch’s membrane/choroid interface, is entirely explained by three independent quantitative trait loci (QTL). Using two distinct cohorts composed of a combined 14,965 controls and 20,741 cases, we ascertain that independent QTLs for FHR-4 are distinct from variants causally associated with AMD, and that FHR-4 variation is not independently associated with disease. Additionally, FHR-4 does not appear to influence AMD progression course among patients with disease driven predominantly by AP dysregulation. Modulation of FHR-4 is therefore unlikely to be an effective therapeutic strategy for AMD.
... CFH/ FHL-1 is a key blocking protein for C3 and CFH genetic variant Y402H, as well as CFH knockdown via miRNA, have been associated with increased risk for AD [9]. CFH V62I was reported to be a bene t gene in AMD (OR = 0.81) [9] and its protective role was due to that CFH V62I possessed increased binding a nity for C3b and enhanced the cofactor activity for C3b inactivation [30]. ...
Preprint
Full-text available
Background Studies demonstrated that the complement system was involved in the pathogenesis of Alzheimer’s disease (AD). A genetic screening study in a Chinese cohort identified that two single nucleotide polymorphisms (SNPs) of the complement regulator Factor H (CFH) rs1061170 (Y402H) and rs800292 (V62I) were robustly associated with AD. FH-like protein 1 (FHL-1) is a short alternative splicing derived from CFH gene. Due to its smaller size and more diffuseness, FHL-1 may provide greater protection via blocking complement 3 (C3). This study aims to investigate the role of FHL-1 and its two mutants V62I and Y402H via lentiviral overexpression of FHL-1 wild type (FHL-1WT), FHL-1V62I, and FHL-1Y402H in 5×FAD mice. Methods We produced lentivirus of control, FHL-1 wild type (FHL-1WT), FHL-1V62I, and FHL-1Y402H and injected into the hippocampus 5×FAD mice. We employed immunostaining and behavior test to investigate the role of these vectors in AD model. Results The result showed that overexpression of FHL-1WT and FHL-1V62I but not FHL-1Y402H ameliorated cognitive impairment in 5×FAD mice. In the other hand, overexpression of FHL-1WT, FHL-1V62I, and FHL-1Y402H did not differently affected the plaque load and astrocytic status. The V62I mutation lightly increased the diffuseness index of amyloid plaque and reduced the number of plaque-associated microglia. Notably, overexpression of FHL-1Y402H prominently promoted synapse phagocytosis by microglia in 5×FAD mice compared to FHL-1WT and FHL-1V62I. Conclusions this indicated that microglia-mediated synapse phagocytosis via complement system may be a key contributor to the cognitive impairment in 5×FAD mice independent on amyloid plaque. Our study provides a clue that blocking microglia-mediated synapse phagocytosis would be an effective therapeutic approach and strategy for AD.
... This SNP has been reported by Abrera-Abeleda et al. [16], Paun et al. [26] and Pickering et al. [27] to be protective against complement-mediated MPGN, age-related macular degeneration, and atypical haemolytic uremic syndrome respectively. Tortajada et al. [28] have found that the protective effect of the CFH allele A variant is owing to its better ability to bind C3b, compete with factor B in proconvertase formation, and enhance the inactivation of fluid-phase and surface-bound C3b. This confirms that there is no contradiction between our results as this polymorphism is a nonsynonymous missense polymorphism that affects the function of CFH, not its serum level. ...
Article
Full-text available
Objectives: It is not known why only some hepatitis C virus (HCV) infected patients develop glomerulonephritis (GN). Therefore, we investigated the role of soluble complement regulators in the development of HCV associated GN. Methods: Patients with HCV associated GN who were admitted to our nephrology unit between July 2016 and July 2018 were recruited to the study (group 1). Two other age and sex matched groups were studied as control groups: patients with HCV without GN (group 2) and healthy HCV negative volunteers (group 3). There were 26 participants in each of the three groups at the end of the recruitment period. An assay of serum fluid-phase complement regulators was performed using enzyme linked immunosorbent assay technique. Three complement single nucleotide polymorphisms (SNPs) were analyzed using real time polymerase chain reaction (Taqman; thermo fisher scientific): rs2230199 and rs1047286 for complement 3 (C3) and rs800292 for complement factor H (CFH). Results: Serum levels of complement 4 binding protein (C4BP) were significantly lower in group 1 (median 70 ng/ml) than in groups 2 (median 88.8 ng/ml) and 3 (median 82.8 ng/ml) with p value of 0.007. The minor allele (allele A) of rs800292 for CFH was significantly higher in group 2 and group 3 (G 54% and A 46%) than in group 1 (G 73% and A 27%), p = 0.04. Conclusions: Low C4BP levels are associated with GN in HCV infected patients. In addition, rs800292 SNP in CFH protects against GN in patients with HCV.
Article
Full-text available
Most drugs that target the complement system are designed to inhibit the complement pathway at either the proximal or terminal levels. The use of a natural complement regulator such as factor H (FH) could provide a superior treatment option by restoring the balance of an overactive complement system while preserving its normal physiological functions. Until now, the systemic treatment of complement-associated disorders with FH has been deemed unfeasible, primarily due to high production costs, risks related to FH purified from donors’ blood, and the challenging expression of recombinant FH in different host systems. We recently demonstrated that a moss-based expression system can produce high yields of properly folded, fully functional, recombinant FH. However, the half-life of the initial variant (CPV-101) was relatively short. Here we show that the same polypeptide with modified glycosylation (CPV-104) achieves a pharmacokinetic profile comparable to that of native FH derived from human serum. The treatment of FH-deficient mice with CPV-104 significantly improved important efficacy parameters such as the normalization of serum C3 levels and the rapid degradation of C3 deposits in the kidney compared to treatment with CPV-101. Furthermore, CPV-104 showed comparable functionality to serum-derived FH in vitro, as well as similar performance in ex vivo assays involving samples from patients with atypical hemolytic uremic syndrome, C3 glomerulopathy and paroxysomal nocturnal hematuria. CPV-104 – the human FH analog expressed in moss – will therefore allow the treatment of complement-associated human diseases by rebalancing instead of inhibiting the complement cascade.
Article
Full-text available
Introduction: As the global pandemic continues, new complications of COVID-19 in pediatric population have turned up, one of them being hemolytic uremic syndrome (HUS), a complement-mediated thrombotic microangiopathy (CM-TMA) characterized by triad of thrombocytopenia, microangiopathic hemolytic anemia and acute kidney injury (AKI). With both multisystem inflammatory syndrome in children (MIS-C) and HUS sharing complement dysregulation as one of the key factors, the aim of this case report is to highlight differences between these two conditions and also emphasize the importance of complement blockade as a treatment modality. Case report: We describe a 21-month-old toddler who initially presented with fever and confirmed COVID-19. His condition quickly deteriorated and he developed oliguria, accompanied with diarrhea, vomiting and oral intake intolerance. HUS was suspected, supported with compelling laboratory findings, including decreased platelets count and C3 levels, elevated LDH, urea, serum creatinine and sC5b-9 and presence of schistocytes in peripheral blood, negative fecal Shiga toxin and normal ADAMTS13 metalloprotease activity. The patient was given C5 complement blocker Ravulizumab and started to display rapid improvement. Conclusion: Although reports of HUS in the setting of COVID-19 continue to pour in, the questions of exact mechanism and similarities to MIS-C remain. Our case for the first time accentuates the use of complement blockade as a valuable treatment option in this scenario. We sincerely believe that reporting on HUS as a complication of COVID-19 in children will give rise to improved diagnosis and treatment, as well as better understanding of both of these intricating diseases.
Article
Full-text available
Age-related macular degeneration (AMD) is linked to 2 main disparate genetic pathways: a chromosome 10 risk locus and the alternative pathway (AP) of complement. Rare genetic variants in complement factor H (CFH; FH) and factor I (CFI; FI) are associated with AMD. FH acts as a soluble cofactor to facilitate FI’s cleavage and inactivation of the central molecule of the AP, C3b. For personalised treatment, sensitive assays are required to define the functional significance of individual AP genetic variants. Generation of recombinant FI for functional analysis has thus far been constrained by incomplete processing resulting in a preparation of active and inactive protein. Using an internal ribosomal entry site (IRES)-Furin-CFI expression vector, fully processed FI was generated with activity equivalent to serum purified FI. By generating FI with an inactivated serine protease domain (S525A FI), a real-time surface plasmon resonance assay of C3b:FH:FI complex formation for characterising variants in CFH and CFI was developed and correlated well with standard assays. Using these methods, we further demonstrate that patient-associated rare genetic variants lacking enzymatic activity (e.g. CFI I340T) may competitively inhibit the wild-type FI protein. The dominant negative effect identified in inactive factor I variants could impact on the pharmacological replacement of FI currently being investigated for the treatment of dry AMD.
Article
Full-text available
The factor H (FH) protein family is emerging as a complex network of proteins controlling the fate of the complement alternative pathway (AP) and dictating susceptibility to a wide range of diseases including infectious, inflammatory, autoimmune, and degenerative diseases and cancer. Composed, in man, of seven highly related proteins, FH, factor H‐like 1, and 5 factor H‐related proteins, some of the FH family proteins are devoted to down‐regulating the AP, while others exert an opposite function by promoting AP activation. Recent findings have provided insights into the molecular mechanisms defining their biological roles and their pathogenicity, illustrating the relevance that the balance between the regulators and the activators within this protein family has in defining the outcome of complement activation on cell surfaces. In this review we will discuss the emerging roles of the factor H protein family, their impact in the complement cascade, and their involvement in the pathogenesis of complement‐mediated diseases associated with the AP dysregulation.
Article
Full-text available
Glomerulonephritis (GN) is a complex disease with intricate underlying pathogenic mechanisms. The possible role of underlying complement dysregulation is not fully elucidated in some GN subsets, especially in the setting of autoimmunity or infection. In the current study, diagnosed cases of lupus nephritis (LN) and post-infectious GN (PIGN) were recruited for molecular genetic analysis and targeted next-generation DNA sequencing was performed for two main complement regulating genes: in the fluid phase; CFH, and on tissue surfaces; MCP. Three heterozygous pathogenic variants in CFH (Q172*, W701*, and W1096*) and one likely pathogenic heterozygous variant in MCP (C223R) have been identified in four of the studied LN cases. Additionally, among the several detected variants of uncertain significance, one novel variant (CFH:F614S) was identified in 74% of the studied LN cases and in 65% of the studied PIGN cases. This variant was detected for the first time in the Egyptian population. These findings suggest that subtle mutations may be present in complement regulating genes in patients with immune-complex mediated category of GN that may add to the disease pathogenesis. These findings also call for further studies to delineate the impact of these gene variants on the protein function, the disease course, and outcome. Frontiers in Immunology
Article
Full-text available
The complement regulatory protein decay accelerating factor (DAF; CD55), inhibits the alternative complement pathway by accelerating decay of the convertase enzymes formed by C3b and factor B. We show, using surface plasmon resonance, that in the absence of Mg(2+), DAF binds C3b, factor B, and the Bb subunit with low affinity (K(D), 14 +/- 0.1, 44 +/- 10, and 20 +/- 7 microm, respectively). In the presence of Mg(2+), DAF bound Bb or the von Willebrand factor type A subunit of Bb with higher affinities (K(D), 1.3 +/- 0.5 and 2.2 +/- 0.1 microm, respectively). Interaction with the proenzyme C3bB was investigated by flowing factor B across a C3b-coated surface in the absence of factor D. The dissociation rate was dependent on the time of incubation, suggesting that a time-dependent conformational transition stabilized the C3b-factor B interaction. Activation by factor D (forming C3bBb) increased the complex half-life; however, the enzyme became susceptible to rapid decay by DAF, unlike the proenzyme, which was unaffected. A convertase assembled with cobra venom factor and Bb was decayed by DAF, albeit far less efficiently than C3bBb. DAF did not bind cobra venom factor, implying that Bb decay is accelerated, at least in part, through DAF binding of this subunit. It is likely that DAF binds the complex with higher affinity/avidity, promoting a conformational change in either or both subunits accelerating decay. Such analysis of component and regulator interactions will inform our understanding of inhibitory mechanisms and the ways in which regulatory proteins cooperate to control the complement cascade.
Article
Full-text available
Mutations and polymorphisms in complement genes have been linked with numerous rare and prevalent disorders, implicating dysregulation of complement in pathogenesis. The 3 common alleles of factor B (fB) encode Arg (fB(32R)), Gln (fB(32Q)), or Trp (fB(32W)) at position 32 in the Ba domain. The fB(32Q) allele is protective for age-related macular degeneration, the commonest cause of blindness in developed countries. Factor B variants were purified from plasma of homozygous individuals and were tested in hemolysis assays. The protective variant fB(32Q) had decreased activity compared with fB(32R). Biacore comparison revealed markedly different proenzyme formation; fB(32R) bound C3b with 4-fold higher affinity, and formation of activated convertase was enhanced. Binding and functional differences were confirmed with recombinant fB(32R) and fB(32Q); an intermediate affinity was revealed for fB(32W). To confirm contribution of Ba to binding, affinity of Ba for C3b was determined. Ba-fB(32R) had 3-fold higher affinity compared with Ba-fB(32Q). We demonstrate that the disease-protective effect of fB(32Q) is consequent on decreased potential to form convertase and amplify complement activation. Knowledge of the functional consequences of polymorphisms in complement activators and regulators will aid disease prediction and inform targeting of diagnostics and therapeutics.
Article
Full-text available
Generation of the alternative pathway C3-convertase, the central amplification enzyme of the complement cascade, initiates by the binding of factor B (fB) to C3b to form the proconvertase, C3bB. C3bB is subsequently cleaved by factor D (fD) at a single site in fB, producing Ba and Bb fragments. Ba dissociates from the complex, while Bb remains bound to C3b, forming the active alternative pathway convertase, C3bBb. Using single-particle electron microscopy we have determined the 3-dimensional structures of the C3bB and the C3bBb complexes at approximately 27A resolution. The C3bB structure shows that fB undergoes a dramatic conformational change upon binding to C3b. However, the C3b-bound fB structure was easily interpreted after independently fitting the atomic structures of the isolated Bb and Ba fragments. Interestingly, the divalent cation-binding site in the von Willebrand type A domain in Bb faces the C345C domain of C3b, whereas the serine-protease domain of Bb points outwards. The structure also shows that the Ba fragment interacts with C3b separately from Bb at the level of the alpha'NT and CUB domains. Within this conformation, the long and flexible linker between Bb and Ba is likely exposed and accessible for cleavage by fD to form the active convertase, C3bBb. The architecture of the C3bB and C3bBb complexes reveals that C3b could promote cleavage and activation of fB by actively displacing the Ba domain from the von Willebrand type A domain in free fB. These structures provide a structural basis to understand fundamental aspects of the activation and regulation of the alternative pathway C3-convertase.
Article
We investigate the contribution of the Iberian bat fauna to the cryptic diversity in Europe using mitochondrial (cytb and ND1) and nuclear (RAG2) DNA sequences. For each of the 28 bat species known for Iberia, samples covering a wide geographic range within Spain were compared to samples from the rest of Europe. In this general screening, almost 20% of the Iberian species showed important mitochondrial discontinuities (K2P distance values > 5%) either within the Iberian or between Iberian and other European samples. Within Eptesicus serotinus and Myotis nattereri, levels of genetic divergence between lineages exceeded 16%, indicating that these taxa represent a complex of several biological species. Other well-differentiated lineages (K2P distances between 5–10%) appeared within Hypsugo savii, Pipistrellus kuhlii and Plecotus auritus, suggesting the existence of further cryptic diversity. Most unsuspected lineages seem restricted to Iberia, although two have crossed the Pyrenees to reach, at leas...
Article
The complement regulatory enzyme, C3b inactivator (C3bINA), has been purified from human serum by affinity chromatography on an anti-C3bINA Sepharose column. Subsequent chromatography on DEAE-cellulose and removal of IgG with anti-IgG Sepharose resulted in a product which was found to be homogeneous by polyacrylamide gel electrophoresis at pH 8.9 and by sodium dodecyl sulfate polyacrylamide gel electrophoresis. The molecule is composed of two disulfide bonded polypeptide chains with mol wt of 50,000 and 38,000 daltons. Human CobINA was found to be a glycoprotein containing at least 10.7% carbohydrate and to have a normal serum concentration of 34 +/- 7 mug/ml (mean +/- 1 SD). Highly purified C3bINA cleaved neither free C3b nor free C4b if trace amounts of contaminating beta1H were removed from these proteins with anti- beta1H Sepharose. However, in the presence of highly purified beta1H and C3bINA, both C3bIna, both C3b and C4b were cleaved. Incubation of native C3 or C4 with C3bINA and beta1H had no effect on their cleaved. Incubation of native C3 or C4 with C3bINA and beta1H had no effect on their structure. The action of C3bINA and beta1H on C3b produced two fragments of the alpha1-chain which did not dissociate without reduction of the molecule. These fragments have mol wt of 67,000 and 40,000 daltons. The action of C3bINA and beta1H on C4b resulted in cleavage of the alpha'-chain giving rise to the 150,000-dalton C4c and the 49,000-dalton C4d fragments which dissociated without reduction. To produce from C3b the immunochemically defined C3c and C3d, fragments, the action of an additional serum enzyme appears to be required, the effect of which can be mimicked by trypsin.
Article
Hemolytic uremic syndrome (HUS) in adults carries a high morbidity and mortality, and its cause remains unknown despite many theories. Although familial HUS is rare, it affords a unique opportunity to elucidate underlying mechanisms that may have relevance to acquired HUS. We have undertaken a genetic linkage study based on a candidate gene approach. A common area bounded by the markers D1S212 and D1S306, a distance of 26 cM located at 1q32 segregated with the disease (Z max 3.94). We demonstrate that the gene for factor H lies within the region. Subsequent mutation analysis of the factor H gene has revealed two mutations in patients with HUS. In an individual with the sporadic/relapsing form of the disease we have found a mutation comprising a deletion, subsequent frame shift and premature stop codon leading to half normal levels of serum factor H. In one of the three families there is a point mutation in exon 20 causing an arginine to glycine change, which is likely to alter structure and hence function of the factor H protein. Factor H is a major plasma protein that plays a critical regulatory role in the alternative pathway of complement activation. In light of these findings and previous reports of HUS in patients with factor H deficiency, we postulate that abnormalities of factor H may be involved in the etiology of HUS.