ArticlePDF Available

Systemic Complement Activation in Age-Related Macular Degeneration

PLOS
PLOS ONE
Authors:
  • Oxford Eye Hospital

Abstract and Figures

Dysregulation of the alternative pathway (AP) of complement cascade has been implicated in the pathogenesis of age-related macular degeneration (AMD), the leading cause of blindness in the elderly. To further test the hypothesis that defective control of complement activation underlies AMD, parameters of complement activation in blood plasma were determined together with disease-associated genetic markers in AMD patients. Plasma concentrations of activation products C3d, Ba, C3a, C5a, SC5b-9, substrate proteins C3, C4, factor B and regulators factor H and factor D were quantified in patients (n = 112) and controls (n = 67). Subjects were analyzed for single nucleotide polymorphisms in factor H (CFH), factor B-C2 (BF-C2) and complement C3 (C3) genes which were previously found to be associated with AMD. All activation products, especially markers of chronic complement activation Ba and C3d (p<0.001), were significantly elevated in AMD patients compared to controls. Similar alterations were observed in factor D, but not in C3, C4 or factor H. Logistic regression analysis revealed better discriminative accuracy of a model that is based only on complement activation markers Ba, C3d and factor D compared to a model based on genetic markers of the complement system within our study population. In both the controls' and AMD patients' group, the protein markers of complement activation were correlated with CFH haplotypes. This study is the first to show systemic complement activation in AMD patients. This suggests that AMD is a systemic disease with local disease manifestation at the ageing macula. Furthermore, the data provide evidence for an association of systemic activation of the alternative complement pathway with genetic variants of CFH that were previously linked to AMD susceptibility.
Content may be subject to copyright.
Systemic Complement Activation in Age-Related Macular
Degeneration
Hendrik P. N. Scholl
1.
, Peter Charbel Issa
1.
, Maja Walier
2
, Stefanie Janzer
1
, Beatrix Pollok-Kopp
3
,
Florian Bo
¨
rncke
3
, Lars G. Fritsche
4
, Ngaihang V. Chong
5
, Rolf Fimmers
2
, Thomas Wienker
2
, Frank G.
Holz
1
, Bernhard H. F. Weber
4
, Martin Oppermann
3
*
1 Department of Ophthalmology, University of Bonn, Bonn, Germany, 2 Institute of Medical Biometry, Informatics and Epidemiology, University of Bonn, Bonn, Germany,
3 Department of Cellular and Molecular Immunology, University of Go
¨
ttingen, Go
¨
ttingen, Germany, 4 Institute of Human Genetics, University of Regensburg, Regensburg,
Germany, 5 Oxford Eye Hospital, University of Oxford, Oxford, United Kingdom
Abstract
Dysregulation of the alternative pathway (AP) of complement cascade has been implicated in the pathogenesis of age-
related macular degeneration (AMD), the leading cause of blindness in the elderly. To further test the hypothesis that
defective control of complement activation underlies AMD, parameters of complement activation in blood plasma were
determined together with disease-associated genetic markers in AMD patients. Plasma concentrations of activation
products C3d, Ba, C3a, C5a, SC5b-9, substrate proteins C3, C4, factor B and regulators factor H and factor D were quantified
in patients (n = 112) and controls (n = 67). Subjects were analyzed for single nucleotide polymorphisms in factor H (CFH),
factor B-C2 (BF-C2) and complement C3 (C3) genes which were previously found to be associated with AMD. All activation
products, especially markers of chronic complement activation Ba and C3d (p,0.001), were significantly elevated in AMD
patients compared to controls. Similar alterations were observed in factor D, but not in C3, C4 or factor H. Logistic
regression analysis revealed better discriminative accuracy of a model that is based only on complement activation markers
Ba, C3d and factor D compared to a model based on genetic markers of the complement system within our study
population. In both the controls’ and AMD patients’ group, the protein markers of complement activation were correlated
with CFH haplotypes. This study is the first to show systemic complement activation in AMD patients. This suggests that
AMD is a systemic disease with local disease manifestation at the ageing macula. Furthermore, the data provide evidence
for an association of systemic activation of the alternative complement pathway with genetic variants of CFH that were
previously linked to AMD susceptibility.
Citation: Scholl HPN, Issa PC, Walier M, Janzer S, Pollok-Kopp B, et al. (2008) Systemic Complement Activation in Age-Related Macular Degeneration. PLoS
ONE 3(7): e2593. doi:10.1371/journal.pone.0002593
Editor: Amanda Ewart Toland, Ohio State University Medical Center, United States of America
Received March 12, 2008; Accepted May 31, 2008; Publi shed July 2, 2008
Copyright: ß 2008 Scholl et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: Pro Retina Foundation Pro-Re/Seed/Issa.1; European Commission (EU FP6), Integrated Project ‘‘EVIGENORET’’ (LSHG-CT-2005-512036); German
Research Foundation (DFG) Heisenberg Fellowship SCHO 734/2-1, HO 1926/1-3. The funders had no role in study design, data collection and analysis, decision to
publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: mopperm@gwdg.de
. These authors contributed equally to this work.
Introduction
Age-related macular degeneration (AMD) is the leading cause of
blindness in all populations of European origin [1]. The
pathogenesis of AMD is not well understood with both genetic
and environmental factors known to influence susceptibility to this
disease [2]. A hallmark of early disease are drusen, lipoproteinac-
eous deposits which accumulate in the space between the retinal
pigment epithelium (RPE) and Bruch’s membrane. Late AMD is
broadly classified into two clinical forms, a dry form with
geographic atrophy (GA), characterized by loss of RPE and outer
neurosensory retinal cells, and a wet form with choroidal
neovascularization (CNV). An estimated 1.75 million US Amer-
icans suffer from late AMD and another 7.3 million have signs of
early AMD putting them at substantial risk for vision loss from this
devastating disease [1].
Studies on the molecular composition of drusen have implicated
inflammation, and particularly local activation of the alternative
pathway (AP) of the complement cascade in the retina, in the
pathogenesis of AMD [3]. Furthermore, strong evidence for a role of
complement in this disease derives from an independent line of
research which showed that variants in the complement factor H
(CFH) gene are significantly associated with an increased risk for
AMD in Caucasian populations [4–7]. These genetic studies were
recently extended by the observation that polymorphisms in other
complement genes, notably those coding for factor B-complement
component C2 (BF-C2) and complement C3 (C3), are also associated
with AMD [8–11]. Multiple haplotypes in the CFH and BF genes
appear to modulate the AMD disease risk and both disease-
predisposing and protective gene variants were identified [7,8,12].
Activation of the AP of complement on cellular surfaces results
from the failure to downregulate the spontaneous low-level
activation of C3. Factor H, the major inhibitor of the AP of
complement activation in the fluid-phase, binds to host cells and
inhibits complement activation by its ability to interfere with the
formation and activity of the alternative C3 convertase, C3bBb. It
PLoS ONE | www.plosone.org 1 July 2008 | Volume 3 | Issue 7 | e2593
accelerates the decay of this convertase and acts as a cofactor for
the factor I-mediated proteolytic inactivation of C3b into iC3b
and C3dg [13]. In the absence of factor H, C3b binds factor B,
allowing its cleavage by the serine protease factor D to yield the
fragments Ba and Bb. Eventually, this results in the formation of
the alternative C5 convertase and assembly of terminal comple-
ment components into the C5b-9 membrane-attack complex
(Fig. 1). Factor H is among the most abundant complement
proteins in serum, synthesized predominantly in the liver, but to a
lesser extent also locally in the eye by RPE cells [14]. Within the
superfamily of functionally and structurally related cofactors for
Factor I-mediated C3b degradation (factor H, CR1, CR2, MCP)
and for the acceleration of the decay of the C3 convertases (factor
H, CR1, DAF), factor H is the main regulator which acts as a
soluble protein. This possibly explains systemic consequences
which could result from polymorphic variation of this protein.
Subtle differences in plasma concentrations or functional
activities of negative (factors H and I) or positive (factor D)
complement regulatory proteins, as well as differences in the
substrates factor B and C3, could have a significant impact on the
magnitude of local complement activation in response to a given
stimulus. Consequently, low-level activation of the AP of
complement may result in the local release of pro-inflammatory
and angiogenic mediators as well as tissue damage in the retina
which eventually may lead to manifest disease. Based on the
hypothesis that defective control of complement activation leads to
the release of complement cleavage products which are detectable
in the circulation, we performed a comprehensive investigation of
AP of complement protein plasma concentrations in a cohort of
AMD patients and controls. The findings were correlated with
polymorphisms in the CFH, BF-C2, and C3 genes.
Results
The study population included 112 AMD patients and 67 control
subjects of similar age, gender and smoking habits which showed no
signs of macular disease (Table 1). Plasma concentrations of
complement proteins in the study population are shown in Table 2.
All complement activation products, and most prominently markers
of chronic complement activation C3d and Ba (p,0.001), were
significantly elevated in AMD patients as compared to controls. The
small C3a and C5a anaphylatoxins, which are rapidly eliminated
from blood plasma, and SC5b-9, which is generated downstream of
C3 and factor B activation, were also detectable at higher levels,
although these differences were less pronounced. Complement
activation cannot be attributed to different plasma concentrations of
factor H, nor of C3 or C4, which were found to be very similar in
both study groups. Factor D plasma levels were significantly
(p,0.001) higher in the patients’ group.
An analysis of phenotypic subgroups revealed that only the C3d
concentration in plasma was differently distributed (ANOVA,
uncorrected p,0.001), the CNV subgroup exhibiting lowest C3d
levels compared to patients with geographic atrophy and patients
with early AMD. The degree to which this finding can be
generalized is limited due to the small number of patients which
were studied within the different phenotypic subgroups.
To determine whether AP of complement activation in AMD
patients is related to complement gene polymorphisms previously
associated with the disease, all probands were genotyped for six
SNPs in the CFH gene, five in BF-C2, and one polymorphism in
C3 (Fig. 1 and Table S1). In accordance with previous reports [9–
12], four markers in CFH (I62V, Y402H, A473A, IVS 15), two
markers in BF-C2 (IVS 10, R32Q) and R102G in C3 were
Figure 1. The Alternative Pathway of Complement: Polymorphic Variants and Complement Proteins under Study. Complement gene
SNPs (boxed with dotted lines) and protein plasma concentrations (boxed with solid lines) were determined in all AMD patients and controls. C3, C4
and factor B are substrates (open rectangles), factor H and factor D are regulators (open ellipses), Ba, C3d and SC5b-9 are markers of chronic
activation (filled rectangles), and C3a and C5a are markers of acute activation (filled ellipses) of the alternative complement pathway.
doi:10.1371/journal.pone.0002593.g001
Complement Activation in AMD
PLoS ONE | www.plosone.org 2 July 2008 | Volume 3 | Issue 7 | e2593
significantly associated with AMD. Haplotype analysis of the SNPs
in CFH revealed one risk haplotype, GCGGGC, and two
protective haplotypes, ATGAAC and GTGAAC (Table S2). For
the definition of the CFH risk haplotype, the Y402H polymor-
phism was sufficient. The risk haplotype was observed in 57% of
cases but only 36% of controls, whereas the protective haplotypes
were observed in 12% and 8% of cases and in 23% and 18% of
controls, respectively. The distribution of haplotypes was different
between cases and controls for CFH (p,0.001), but not for BF-C2
(p = 0.14).
Logistic regression analysis yielded significance for six variables
including three protein markers and three genetic markers. A
subsequent stepwise logistic regression analysis including exclu-
sively genetic markers revealed the three genetic markers A473A
at CFH, IVS 10 at BF-C2, and R102G at C3 to be the best genetic
predictors of risk for AMD with a high accuracy for discrimination
and an area under the curve (AUC) value of 0.726 (Fig. 2). A
stepwise logistic regression analysis including only protein markers
resulted in a model with the three markers Ba, C3d and factor D.
The corresponding ROC curve showed an even better discrim-
inatory accuracy (AUC = 0.816; p = 0.05). When both genetic and
protein markers were included into the stepwise regression
procedure, the resulting model revealed only a marginally better
fit (AUC = 0.850).
To detect a direct link between genetic variants at CFH and
protein markers of AP of complement activation, plasma levels
were compared between carriers of the CFH risk haplotype and
carriers of the protective haplotypes, both within the controls’ and
the AMD patients’ group (Table 3). Within both subject groups
(with the exception of C5a in the patients’ group), carriers of the
CFH risk haplotype showed consistently higher complement
activation parameters than those carrying the protective haplo-
types. A correlation between genetic variants in BF-C2 or C3 and
complement protein levels was not observed.
Discussion
A low-key but persistent inflammatory process which involves
activation of the complement system has been proposed to underlie
the sight-threatening manifestations in AMD. In support of this
hypothesis, various inflammatory mediators including complement
proteins, their activation products and regulators have been
identified in retinal deposits of AMD patients. In particular,
complement components C3 and C5, the membrane attack complex
C5b-9 and factor H, the main regulator of alternative pathway
activation, are constituents of drusen in AMD patients [16]. Further
evidence for a role of complement in this disease derives from two
recent studies which showed that laser-induced CNV, an accelerated
Table 1. Clinical and Demographic Characteristics of the
Study Population.
Variable Controls AMD patients
(N = 67) (N = 112)
Disease status - no. (%) *
Early AMD (Drusen) - 9 (8%)
Choroidal neovascularization - 78 (70%)
Geographic atrophy - 25 (22%)
Gender - no. (%) **
Male 37 (55%) 41 (37%)
Female 30 (45%) 71 (63%)
Mean age - years (SD; rang e)*** 70.1 (6.0; 60–86) 75.6 (6.6; 59–94)
Smoking history no. (%)
Never smoked 35 (52%) 59 (53%)
Ex-smoker 26 (39%) 43 (38%)
Current smoker 6 (9%) 10 (9%)
*
AMD patients were categorized into mutually exclusive groups: All subjects
classified as ‘‘early AMD’’ (n = 9) had extensive intermediate and/or large
drusen in at least one eye and all qualified as category III of the Age-Related
Eye Disease Study group. Patients with GA in both eyes or GA in one eye in the
absence of CNV in the fell ow eye were classified as ‘‘GA’’ (n = 25). Nineteen of
these had subfoveal GA in at least one eye; the remaining six patients
exclusively showed extrafoveal GA. If a CNV due to AMD was present in at least
one eye, patients were classified as ‘‘CNV’’ (n = 78). Subjects with changes such
as many hard drusen and/or pigmentary alterations in both eyes were
excluded as these changes may refer to normal aging processes not necessarily
linked to AMD.
** Difference between cases and controls, p = 0.02;
***
p,0.001.
doi:10.1371/journal.pone.0002593.t001
Table 2. Plasma Concentrations of Complement Proteins in AMD Patients and Controls.
Complement protein * Units
Controls AMD patients
p**
Median 5
th
,95
th
Ptcl. Median 5
th
,95
th
Ptcl.
C3 [mg/ml] 1.18 0.85–1.48 1.12 0.89–1.53 0.85
C4 [mg/ml] 0.24 0.15–0.34 0.23 0.15–0.38 1.0
Factor B [mg/ml] 642 378–1354 803 497–1489 0.02
Factor H [mg/ml] 515 365–711 546 396–758 0.21
Factor D [mg/ml] 0.95 0.50–1.65 1.26 0.69–2.30 ,0.001
C3a [ng/ml] 14.3 10.6–21.2 15.5 11.2–24.1 0.03
C5a [ng/ml] 1.67 0.66–2.32 1.85 0.78–2.66 0.04
Ba [mg/ml] 1.09 0.60–1.71 1.33 0.90–2.09 ,0.001
C3d [mg/ml] 46.9 32.2–68.5 55.2 35.7–94.1 ,0.001
SC5b-9 [units] 159 90–710 188 107–777 0.01
*
Complement proteins in this table are arranged in groups of substrates (C3, C4, factor B), regulators (factor H, factor D), markers of acute (C3a, C5a) and chronic
activation (Ba, C3d, SC5b-9) of the alternative pathway of complement (Fig. 1).
**
Wilcoxon rank-sum test; corrected for multiple testing by Bonferroni-Holm procedure.
doi:10.1371/journal.pone.0002593.t002
Complement Activation in AMD
PLoS ONE | www.plosone.org 3 July 2008 | Volume 3 | Issue 7 | e2593
model of neovascular AMD in mice, requires intact C3 and cellular
C3aR/C5aR receptors [17,18]. Thus, uncontrolled activation of the
AP of complement within the central retina may play a major role in
the pathophysiology of AMD through its ability to promote local
tissue damage and angiogenesis [17].
In the present study we show that several parameters which
reflect systemic complement activation are significantly elevated in
the circulation of AMD patients as compared to controls. We
exercised great caution to avoid in vitro activation of complement.
As illustrated in a recent study on complement activation in AMD
patients, erroneously elevated concentrations of complement
fragments are detected, when heparin- rather than EDTA-
anticoagulated blood was used for analysis [19]. Although we
concur with the overall conclusions from this particular study, the
presented data are largely artifactual and result from ex vivo
complement activation in the heparinized blood tubes [40,41]. As
previous studies pointed to a role of complement activation in
AMD we focused our analysis on the quantification of the major
breakdown products of C3 and factor B, i.e. C3a, C3d, and Ba.
Among these, Ba and C3d are particularly well suited as markers
of chronic AP of complement activation at low levels, since their
respective half-lives in-vivo are in the order of several hours rather
than minutes [20]. Since complement turnover in vivo is further
determined by biosynthetic rates of the precursor proteins as well
as by concentrations of complement control proteins, we
determined plasma levels of the corresponding precursor proteins
C3 and factor B, and the main positive and negative regulators of
the AP of complement, factors D and H. C5a and SC5b-9 were
quantified as markers of terminal pathway of complement
activation which are generated downstream of C3 and factor B.
The simultaneous quantification of the main cleavage products,
substrates and of control proteins of the AP of complement is a
unique feature of this study and allowed to precisely document the
state of complement activation in all patients.
Our notion of enhanced systemic complement activation in
AMD is mainly based on the finding that in patients all
complement activation products determined in this study were
elevated as compared to controls. This difference was most
strikingly observed with regard to Ba and C3d, two sensitive
markers of chronic AP of complement activation in-vivo. In
contrast, the complement proteins C3, C4 and factor H did not
significantly differ between the two groups. We also observed
elevated levels of factors B and D in AMD patients and this could
possibly be due to an acute phase response-mediated upregulation
of factor B or by polymorphic variation in the FD gene which may
affect factor D plasma levels. Upregulation of these two positive
regulators may further contribute to enhanced AP of complement
activation, however, subtle changes in factors B and D alone
cannot explain the enhanced turnover of complement substrates
[21]. While Ba and C3d concentrations in AMD patients were
only modestly elevated by a factor of 1.2 to 1.3 compared to
controls, C3d levels in the plasma of patients with rheumatoid
arthritis are also increased to a similar degree [22]. Since local C3d
concentrations in synovial fluids from these patients are much
higher than in plasma [23], a similar gradient between sites of local
complement activation and blood plasma may also exist in AMD.
Our results further demonstrate that a combination of
complement activation markers can be used to most reliably
discriminate AMD patients from controls in our study population.
The discriminatory ability of these complement proteins
(AUC = 0.816) appears superior or at least similar to the
discriminatory ability of genetic markers of complement genes
Figure 2. Receiver Operating Characteristic (ROC) Curves for
the Discriminative Capability of Genetic and Protein markers
of the Complement System. Receiver operating characteristic curves
for genetic markers (dotted line; A473A of CFH, IVS 10 of BF-C2 and
R102G of C3) and complement protein markers (solid line; Ba, C3d, and
factor D) are shown. AUC = Area under ROC curve.
doi:10.1371/journal.pone.0002593.g002
Table 3. Plasma Concentrations (mean6S.E.M.) of Complement Proteins in Carriers of Risk-Conferring and Protective Haplotypes
of the CFH Gene.
Complement
protein Units
Controls AMD patients
All Risk haplotype
Protective
haplotypes All Risk haplotype
Protective
haplotypes
(n = 67) (n = 17) (n = 23) (n = 112) (n = 67) (n = 16)
Ba [mg/ml] 1.1160.04 1.2160.25 1.0460.08 1.4160.04 1.4660.05 1.2760.11
C3d [mg/ml] 48.161.40 50.963.71 45.462.34 59.161.80 60.662.11 52.764.88
C3a [ng/ml] 14.760.38 15.360.70 14.060.62 16.460.37 16.660.53 15.360.89
C5a [ng/ml] 1.5560.06 1.6460.12 1.4660.11 1.7760.06 1.7260.07 1.8660.16
Factor D [mg/ml] 0.9860.04 1.0460.05 0.8560.07 1.3560.05 1.3860.06 1.2660.12
doi:10.1371/journal.pone.0002593.t003
Complement Activation in AMD
PLoS ONE | www.plosone.org 4 July 2008 | Volume 3 | Issue 7 | e2593
for the prediction of AMD as determined in this and other
similarly designed studies [24]. Because the protein markers are
closer to the presumed mechanisms of AMD pathogenesis, this
finding appears plausible. Prospective studies will be required to
determine whether plasma concentrations of complement proteins
could be useful as markers of AMD at less advanced stages, either
alone or in combination with genetic markers. Since this study was
limited to genes and proteins of the alternative pathway of
complement, a general conclusion on the superiority of protein
markers compared to genetic markers is not possible. Such a
conclusion may be derived from an even more comprehensive
study including additional genetic and protein markers. Since the
LOC387715/HTRA1/ARMS2 locus has been shown to be of
similar significance as CFH [25], polymorphisms within ARMS2
would have to be considered. Recently, the discriminative
accuracy of testing polymorphisms in CFH and BF-C2 together
with ARMS2 for the prediction of AMD was found to be 80% [24],
which is similar to the score we found for testing protein markers
of the alternative pathway of complement.
Our study has linked elevated plasma concentrations of AP of
complement activation products in AMD patients to polymorphic
variations in the CFH gene which codes for the main regulator of the
AP of complement activation in the fluid phase and on cell surfaces
[13]. We observed within both study groups that individuals who
carry the AMD-associated CFH risk haplotype had higher plasma
concentrations of complement activation products, and conversely
that protective CFH haplotypes were associated with lower levels of
activation products (p = 0.05, MANOVA).
In particular, the association between complement activation
and the CFH risk haplotype which includes Y402H, a non-
synonymous SNP in CFH which leads to a tyrosine to histidine
exchange at position 402, is biologically plausible. Several recent
studies have shown that the His402 variant binds less well to
heparin, C reactive protein and RPE cell surfaces [14,26,27].
While the consequences of defective factor H function are most
likely not restricted to the eye, but result in inappropriate
complement control at other cell surfaces throughout the body,
the retina appears to be especially sensitive towards the effects of
complement activation.
Our data suggest that AMD indeed is a systemic disease with
local disease manifestation at the ageing macula. Such local
manifestation of systemic pathophysiology is not without prece-
dence. An example are mutations in the PRPF3 gene causing a
tissue-specific phenotype of autosomal dominant retinitis pigmen-
tosa although PRPF3 is an element of the ubiquitously expressed
RNA splicing machinery [28]. In addition, the macula is more
easily damaged than the retinal periphery, e.g. because it exhibits
decreased thickness and integrity of the elastic layer of Bruch’s
membrane [29]. The hypothesis of AMD being a systemic disease
certainly raises the possibility of other organ manifestations that
have so far not been detected. In support of this hypothesis,
patients with membranoproliferative glomerulonephritis type II
(MPGN II) and systemic complement activation develop retinal
deposits at early ages which resemble drusen in AMD patients
[30]. Since MPGN II patients benefit from substitution therapy
with intact complement control proteins, local or systemic
administration of AP of complement inhibitors may be considered
as a future therapeutic option in AMD.
Materials and Methods
Cases and Controls
112 patients with a clinical diagnosis of AMD and 67 control
subjects were included in the study. Absence of AMD, early and
late AMD was defined according to the criteria of the ARM
Epidemiological Study Group [31]. Exclusion criteria included age
below 55 years; any evidence of retinal disease (in the control
group) with the exception of AMD (in the AMD group)
ascertained by history, clinical examination, digital fundus
photography and grading of fundus images (see below); any
systemic disease known to affect the complement system (e.g.
rheumatoid arthritis) ascertained by a standardized case report
form (CRF) derived from the multicenter FAM-Study [32] and
abnormal renal clearance (ascertained by creatinine and cystatin C
values). In the recruitment of the control subjects particular
attention was paid to match for smoking habits (ascertained by the
CRF), since smoking has been shown to be by far the most
significant environmental risk factor for AMD. Moreover, earlier
studies have suggested an effect of smoking on factor H blood
levels [13]. As a result, the control group was of similar age and
gender, and exhibited identical smoking habits; minor differences
in age and gender of the two groups were regarded irrelevant for
complement protein concentrations [41]. All subjects were of
Caucasian descent and were recruited within the same time period
from the Department of Ophthalmology, University of Bonn
between January and October 2006. Control subjects did not have
any signs of AMD, especially no early changes such as many small
drusen, intermediate or large drusen. Informed consent was
obtained from all subjects. The research protocol was in keeping
with the provisions of the Declaration of Helsinki, and approval
was obtained from the institutional ethics committee. Digital
fundus photographs were obtained from all participants. In
patients with CNV, optical coherence tomography and fluorescein
angiography were performed. Fundus autofluorescence imaging
was performed in patients with GA. All fundus images were
evaluated separately by two independent readers (HPNS and PCI);
digital fundus images were graded according to the classification
system of the International ARM Epidemiological Study Group
[31,33,34]. Clinical characteristics and demographic data of the
study populations are provided in Table 1.
Blood samples
Venous blood was collected from all subjects into tubes
containing dipotassium EDTA at a final concentration of 8 mM.
The plasma was separated from the blood cells by centrifugation
(20 min/10006 g) within 3 hours after venipuncture and frozen in
aliquots at 280uC until analysis. One subject with elevated
complement levels due to mild chronic renal failure was excluded
from the analysis of factor D and Ba since the catabolism of these
two complement proteins is determined by the glomerular
filtration rate [21]. All other subjects had normal creatinine and
cystatin C values; both subject groups were not significantly
different for these variables.
Analysis of comp lement proteins
Assays for the quantitation of complement components factor B,
Ba, C3a, C3d, C5a, SC5b-9, factor D and factor H have been
developed previously [35–37]. All complement activation assays
were based on monoclonal antibodies with specificities for
activation-induced neoepitopes present on the different comple-
ment split products which are absent from the respective native
precursor proteins. In the case of the ‘C3d’-assay, a capture mAb
(I3/15) was used which reacts with a common neoepitope on C3b,
iC3b, and C3dg, in combination with a polyclonal rabbit anti-C3d
as the detecting antibody [35]. Plasma concentrations of C3 and
C4 were determined by rate nephelometry. All patients and
control samples were handled identically and analyzed simulta-
neously in order to ensure stable assay conditions. As reported
Complement Activation in AMD
PLoS ONE | www.plosone.org 5 July 2008 | Volume 3 | Issue 7 | e2593
before [36], the inter-assay coefficient of variation of ELISA
procedures was below 10%. The intra-individual stability of
complement plasma levels was assessed in a subset of AMD
patients (n = 14). From these patients, a second ETDA plasma
sample was obtained 12 months after the first venipuncture.
Within this period of time, Ba, C3d and factor D values varied by
less than 15% compared to the initial values.
Genotyping
Genomic DNA was extracted from peripheral blood leukocytes
following established protocols. Genotyping was done by TaqMan
SNP Genotyping or by direct sequencing of SNPs. TaqMan Pre-
Designed SNP Genotyping Assays (Applied Biosystems, Foster
City, U.S.A.) were performed according to the manufacturer’s
instructions and were analyzed with a 7900HT Fast Real-Time
PCR System (Applied Biosystems). Direct sequencing was
performed with the Big Dye Terminator Cycle Sequencing Kit
Version 1.1 (Applied Biosystems) according to the manufacturer’s
instructions. Reactions were analyzed with an ABI Prism Model
3130xl Sequencer (Applied Biosystems). Individual genotypes that
were ambiguous or missing were reanalyzed resulting in a call-rate
of 100% for all SNPs tested. SNP-IDs for three complement gene
loci CFH, BF-C2, and C3 are indicated in Fig. 1.
Statistical Analysis
Based on data of assay variability of the key proteins of chronic
AP of complement activation (C3d, Ba, SC5b-9) derived from our
laboratory [35,36], the study was designed to detect a difference of
2/3 of a standard deviation between cases and controls with a
power of at least 90%, with a two sided test at a level of a = 0.05/
3. The level had been chosen to account for the fact that three
biochemical markers had to be compared. The recruitment was
planned to be imbalanced with a case control ratio of 2:1 resulting
in 120 cases and 60 controls to be included.
Allele and genotype frequencies were determined. All markers
were in Hardy-Weinberg equilibrium (all p.0.20). All allele
frequencies were within the ranges reported in the Entrez SNP
database (www.ncbi.nih.gov) and in previous publications [4–12].
To test for genetic association, genotype frequencies were
compared between cases and controls by Armitage’s trend test.
A retrospective power analysis based on the empirical values for
allele frequencies and relative risks found in our data was
performed [38]. Results are reported in Table S3. Haplotypes
for the markers in CFH and BF-C2 were estimated using
FAMHAP [39]. The difference in distribution of haplotypes was
tested by likelihood ratio test.
Stepwise logistic regression analysis was used to explore models
to predict the risk for AMD depending on genetic markers and
complement protein markers and to gain more insight into the
relevance of these risk parameters in relation to one another.
Results were visualized by receiver operating characteristic (ROC)
curves for the scores resulting from the logistic regression. ROC
curves were compared using the method proposed by DeLong et
al [15]. Multivariate analysis of variance (MANOVA) was applied
to the joint distribution of complement activation markers to verify
the observation of simultaneously increased values depending on
CFH haplotypes and disease status. Data were handled in SAS
(SAS software package for Windows, version 9.1.; SAS Institute
Inc., Cary, NC, USA; http://www.sas.com).
Supporting Information
Table S1 Genotypes
Found at: doi:10.1371/journal.pone.0002593.s001 (0.09 MB
PDF)
Table S2 Haplotypes
Found at: doi:10.1371/journal.pone.0002593.s002 (0.08 MB
PDF)
Table S3 Retrospective Power Analysis
Found at: doi:10.1371/journal.pone.0002593.s003 (0.17 MB
PDF)
Author Contributions
Concei ved and designed the experiments: MO HS NC TW BW.
Performed the experiments: LF MO HS PI SJ BP FB. Analyzed the data:
RF MW TW. Wrote the paper: LF RF MO HS PI MW SJ BP FB NC TW
FH BW.
References
1. Congdon N, O’Colmain B, Klaver CC, Klein R, Munoz B, et al. (200 4) Causes
and prevalence of visual impairment among adults in the United States. Arch
Ophthalmol 122: 477–485.
2. de Jong PT (2006) Age-related macular degeneration. N Engl J Med 355:
1474–1485.
3. Hageman GS, Luthert PJ, Victor Chong NH, Johnson LV, Anderson DH, et al.
(2001) An integrated hypothesis that considers drusen as biomarkers of immune-
mediated processes at the RPE-Bruch’s membrane interface in aging and age-
related macular degeneration. Prog Retin Eye Res 20: 705–732.
4. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, et al. (2005) Complement
factor H polymorphism in age-related macular degeneration. Science 308:
385–389.
5. Edwards AO, Ritter R III, Abel KJ, Manning A, Panhuysen C, Farrer LA (2005)
Complement factor H polymorphism and age-related macular degeneration.
Science 308: 421–424.
6. Haines JL, Hauser MA, Schmidt S, Scott WK, Olson LM, et al. (2005)
Complement factor H variant increases the risk of age-related macular
degeneration. Science 308: 419–421.
7. Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, et al. (2005)
A common haplotype in the complement regulatory gene factor H (HF1/CFH)
predisposes individuals to age-related macular degeneration. Proc Natl Acad
Sci U S A 102: 7227–7232.
8. Gold B, Merriam JE, Zernant J, Hancox LS, Taiber AJ, et al. (2006) Variation
in factor B (BF) and complement component 2 (C2) genes is associated with age-
related macular degeneration. Nat Genet 38: 458–462.
9. Spencer KL, Hauser MA, Olson LM, Schmidt S, Scott WK, et al. (2007)
Protective effect of complement factor B and complement component 2 variants
in age-related macular degeneration. Hum Mol Genet 16: 1986–1992.
10. Yates JR, Sepp T, Matharu BK, Khan JC, Thurlby DA, et al. (2007)
Complement C3 variant and the risk of age-related macular degeneration.
N Engl J Med 357: 553–561.
11. Maller JB, Fagerness JA, Reynolds RC, Neale BM, Daly MJ, et al. (2007)
Variation in complement factor 3 is associated with risk of age-related macular
degeneration. Nat Genet 39: 1200–1201.
12. Li M, Atmaca-Sonmez P, Othman M, Branham KE, Khanna R, et al. (2006)
CFH haplotypes without the Y402H coding variant show strong association with
susceptibility to age-related macular degeneration. Nat Genet 38: 1049–1054.
13. Rodriguez de CS, Esparza-Gordillo J, Goicoechea de JE, Lopez-Trascasa M,
Sanchez-Corral P (2004) The human complement factor H: functional roles,
genetic variations and disease associations. Mol Immunol 41: 355–367.
14. Skerka C, Lauer N, Weinberger AA, Keilhauer CN, Suhnel J, et al. (2007)
Defective complement control of factor H (Y402H) and FHL-1 in age-related
macular degeneration. Mol Immunol 44: 3398–3406.
15. DeLong ER, DeLong DM, Clarke-Pearson DL (1988) Comparing the areas
under two or more correlated receiver operating characteristic curves: a
nonparametric approach. Biometrics 44: 837–845.
16. Johnson PT, Betts KE, Radeke MJ, Hageman GS, Anderson DH, et al. (2006)
Individual s hom oz ygous for the age-r ela ted macular deg ene rati on r isk-
conferring variant of complement factor H have elevated levels of CRP in the
choroid. Proc Natl Acad Sci U S A 103: 17456–17461.
17. Nozaki M, Raisler BJ, Sakurai E, Sarma JV, Barnum SR, et al. (2006) Drusen
complement components C3a and C5a promote choroidal neovascularization.
Proc Natl Acad Sci U S A 103: 2328–2333.
18. Bora PS, Sohn JH, Cruz JM, Jha P, Nishihori H, et al. (2005) Role of
complement and complement membrane attack complex in laser-induced
choroidal neovascularization. J Immunol 174: 491–497.
Complement Activation in AMD
PLoS ONE | www.plosone.org 6 July 2008 | Volume 3 | Issue 7 | e2593
19. Sivaprasad S, Adewoyin T, Bailey TA, Dandekar SS, Jenkins S, et al. (2007)
Estimation of systemic complement C3 activity in age-related macular
degeneration. Arch Ophthalmol 125: 515–519.
20. Mollnes TE, Jokiranta TS, Truedsson L, Nilsson B, Rodriguez de CS, et al.
(2007) Complement analysis in the 21st century. Mol Immunol 44: 3838–3849.
21. Oppermann M, Kurts C, Zierz R, Quentin E, Weber MH, et al. (1991) Elevated
plasma levels of the immunosuppressive complement fragment Ba in renal
failure. Kidney Int 40: 939–947.
22. Bourke BE, Moss IK, Mumford P, Horsfall A, Maini RN (1982) The
complement fixing ability of putative circulating immune complexes in
rheumatoid arthritis and its relationship to extra-articular disease. Clin Exp
Immunol 48: 726–732.
23. Mollnes TE, Lea T, Mellbye OJ, Pahle J, Grand O, et al. (1986) Complement
activation in rheumatoid arthritis evaluated by C3dg and the terminal
complement complex. Arthritis Rheum 29: 715–721.
24. Despriet DD, Klaver CC, van Duijn CC, Janssens AC (2007) Predictive value of
multiple genetic testing for age-related macular degeneration. Arch Ophthalmol
125: 1270–1271.
25. Rivera A, Fisher SA, Fritsche LG, Keilhauer CN, Lichtner P, et al. (2005)
Hypothetical LOC387715 is a second major susceptibility gene for age-related
macular degeneration, contributing independently of complement factor H to
disease risk. Hum Mol Genet 14: 3227–3236.
26. Clark SJ, Higman VA, Mulloy B, Perkins SJ, Lea SM, et al. (2006) His-384
allotypic variant of factor H associated with age-related macular degeneration
has different heparin bindi ng properties from the non-disease-associated form.
J Biol Chem 2 81: 24713–24720.
27. Laine M, Jarva H, Seitsonen S, Haapasalo K, Lehtinen MJ, et al. (2007) Y402H
Polymorphism of Complement Factor H Affects Binding Affinity to C-Reactive
Protein. J Immunol 178: 3831–3836.
28. Comitato A, Spampanato C, Chakarova C, Sanges D, Bhattacharya SS, et al.
(2007) Mutations in splicing factor PRPF3, causing retinal degeneration, form
detrimental aggregates in photoreceptor cells. Hum Mol Genet 16: 1699–1707.
29. Chong NH, Keonin J, Luthert PJ, Frennesson CI, Weingeist DM, et al. (2005)
Decreased thickness and integrity of the macular elastic layer of Bruch’s
membrane correspond to the distribution of lesions associated with age-related
macular degeneration. Am J Pathol 166: 241–251.
30. Huang SJ, Costa DL, Gross NE, Yannuzzi LA (2003) Peripheral drusen in
membranoproliferative glomerulonephritis type II. Retina 23: 429–431.
31. Bird AC, Bressler NM, Bressler SB, Chisholm IH, Coscas G, et al. (1995) An
international classification and grading system for age-related maculopathy and
age-related mac u lar degeneration. The International ARM Epidemiological
Study Group. Surv Ophthalmol 39: 367–374.
32. Holz FG, Bindewald-Wittich A, Fleckenstein M, Dreyhaupt J, Scholl HPN, et al.
(2007) Progression of geographic atrophy and impact of fundus autofluorescence
patterns in age-related macular degeneration. Am J Ophthalmol 143: 463–472.
33. Scholl HPN, Peto T, Dandekar S, Bunce C, Xing W, et al. (2003) Inter- and
intra-observer variability in grading lesions of age-related maculopathy and
macular degeneration. Graefes Arch Clin Exp Ophthalmol 241: 39–47.
34. Scholl HPN, Dandekar SS, Peto T, Bunce C, Xing W, et al. (2004) What is lost
by digitizing stereoscopic fundus color slides for macular grading in age-related
maculopathy and degeneration? Ophthalmology 111: 125–132.
35. Oppermann M, Baumgarten H, Brandt E, Gottsleben W, Kurts C, et al. (1990)
Quantitation of components of the alternative pathway of complement (APC) by
enzyme-linked immunosorbent assays. J Immunol Methods 133: 181–190.
36. Oppermann M, Schulze M, Go¨tze O (1991) A sensitive enzyme immunoassay
for the quantitation of human C5a/C5a(desArg) anaphylatoxin us ing a
monoclonal antibody with specificity for a neoepitope. Complement Inflamm
8: 13–24.
37. Wu¨rzner R, Schulze M, Happe L, Franzke A, Bieber FA, et al. (1991) Inhibition
of terminal complement complex formation and cell lysis by monoclonal
antibodies. Complement Inflamm 8: 328–340.
38. Purcell S, Cherny SS, Sham PC (2003) Genetic Power Calculator: design of
linkage and association genetic mapping studies of complex traits. Bioinformatics
19: 149–150.
39. Becker T, Cichon S, Jonson E, Knapp M (2005) Multiple testing in the context
of haplotype analysis revisited: application to case-control data. Ann Hum Genet
69: 747–756.
40. Mollnes TE, Garred P, Bergseth G (1998) Effect of time, temperature and
anticoagulants on in vitro complement activation: consequences for collection
and preservation of samples to be examined for complement activation. Clin
Exp Immunol 73: 484–8.
41. Oppermann M, Ho¨pken U, Go¨tze O (1992) Assessment of complement
activation in vivo. Immunopharmacology 24: 119–34.
Complement Activation in AMD
PLoS ONE | www.plosone.org 7 July 2008 | Volume 3 | Issue 7 | e2593
... Notably, CFH, C3, and terminal complement proteins C5-C9 have been identified in drusen deposits in AMD, while C5 was isolated from RPE cells adjacent to drusen [38]. Increased levels of complement proteins C3d and complement factors B and D were observed in the plasma of patients with AMD, suggesting a possible systemic overactivation of the complement cascade in this disease [58]. ...
Article
Full-text available
The complement cascade is a vital system in the human body’s defense against pathogens. During the natural aging process, it has been observed that this system is imperative for ensuring the integrity and homeostasis of the retina. While this system is critical for proper host defense and retinal integrity, it has also been found that dysregulation of this system may lead to certain retinal pathologies, including geographic atrophy and diabetic retinopathy. Targeting components of the complement system for retinal diseases has been an area of interest, and in vivo, ex vivo, and clinical trials have been conducted in this area. Following clinical trials, medications targeting the complement system for retinal disease have also become available. In this manuscript, we discuss the pathophysiology of complement dysfunction in the retina and specific pathologies. We then describe the results of cellular, animal, and clinical studies targeting the complement system for retinal diseases. We then provide an overview of complement inhibitors that have been approved by the Food and Drug Administration (FDA) for geographic atrophy. The complement system in retinal diseases continues to serve as an emerging therapeutic target, and further research in this field will provide additional insights into the mechanisms and considerations for treatment of retinal pathologies.
... CFH is a negative regulator of the complement system and CFH Y402H carriers have slightly increased activity in the complement system with increased levels of complement split-products in plasma [12][13][14] . These splitproducts may interact with T cells and professional antigen-presenting cells via membrane-bound receptors and lead to stronger T cell responses 15,16 . ...
Article
Full-text available
Age-related macular degeneration (AMD) is a common cause of visual loss among the elderly. Genetic variants in the gene encoding complement factor H (CFH) have been identified as an AMD susceptibility gene, however, the mechanistic link is debated. Here, we investigated the link between the CFH Y402H genotype and low-grade inflammation. We recruited 153 healthy individuals, 84 participants with dry stages of AMD, and 148 participants with neovascular AMD. All participants were subjected to detailed retinal examination, and interview regarding comorbidities and lifestyle. Blood samples were analyzed for level of C-Reactive Protein (CRP), white blood cell differential count, and stained with fluorescent antibodies to differentiate CD4⁺ and CD8⁺ T cells. CFH Y402H genotyping was performed using an allele-specific polymerase chain reaction genotyping assay. Splenocytes from young and aged wild type and Cfh null mutant C57BL/6J mice were examined for CD4⁺ and CD8⁺ T cells. Healthy individuals with the CFH Y402H at-risk polymorphism HH had higher levels of CRP and lower proportions of CD4⁺ T cells compared to persons with the YH or YY polymorphism (P = 0.037, Chi-square). Healthy individuals with the HH polymorphism displayed lower proportions of CD4⁺ T cells with ageing (P < 0.01, one-way ANOVA), whereas both young and aged Cfh null mutant mice displayed lower proportions of CD4⁺ T cells (P < 0.001 and P < 0.05; unpaired t test). Participants with dry AMD and the HH polymorphism had similarly lower proportions of CD4⁺ T cells (P = 0.024, one-way ANOVA), but no difference in CRP-levels. In the neovascular stage of AMD, there was no difference in proportion of CD4⁺ cells or CRP levels according to genotype. The risk-associated CFH genotype is associated with an age-related decrease in proportion of CD4⁺ T cells and increased levels of CRP in healthy individuals. This indicates that decreased complement regulation results in extensive changes in innate and adaptive immune compartments that precede development of AMD.
... 78 Complement component 5 (C5) mediates inflammation and was thought to participate in the local chronic inflammatory process in AMD. 79 While research on single nucleotide polymorphisms of C5 did not support an association between C5 and AMD, immunolocalization has provided evidence of the C5 complement activation in AMD. 80,81 The aptamer targeting C5 (avacincaptad pegol) has been used for AMD therapy, 44 and the results have shown that avacincaptad pegol significantly reduces geographic atrophy in AMD. ...
Article
Full-text available
Aptamers are widely applied to diagnosis and therapy because of their targeting. However, the current progress of research into aptamers for the treatment of eye disorders has not been well-documented. The current literature on aptamers was reviewed in this study. Aptamer-related drugs and biochemical sensors have been evaluated for several eye disorders within the past decade; S58 targeting TGF-β receptor II and pegaptanib targeting vascular endothelial growth factor (VEGF) are used to prevent fibrosis after glaucoma filtration surgery. Anti-brain-derived neurotrophic factor aptamer has been used to diagnose glaucoma. The first approved aptamer drug (pegaptanib) has been used to inhibit angiogenesis in age-related macular degeneration (AMD) and diabetic retinopathy (DR), and its efficacy and safety have been demonstrated in clinical trials. Aptamers, including E10030, RBM-007, AS1411, and avacincaptad pegol, targeting other angiogenesis-related biomarkers have also been discovered and subjected to clinical trials. Aptamers, such as C promoter binding factor 1, CD44, and advanced end products in AMD and DR, targeting other signal pathway proteins have also been discovered for therapy, and biochemical sensors for early diagnosis have been developed based on aptamers targeting VEGF, connective tissue growth factor, and lipocalin 1. Aptamers used for early detection and treatment of ocular tumors were derived from other disease biomarkers, such as CD71, nucleolin, and high mobility group A. In this review, the development and application of aptamers in eye disorders in recent years are systematically discussed, which may inspire a new link between aptamers and eye disorders. The aptamer development trajectory also facilitates the discovery of the pathogenesis and therapeutic strategies for various eye disorders.
... This assumption would also explain why there are no differences between AMD patients in the comparison of genetic and smoking-associated risk profiles. In the measurement of complement activity markers in the plasma, such as C3d/C3 ratio, C3a-desarg, or TCC, the different risk allele CFH, ARMS, or CFI carriers display comparable levels of those markers, while healthy donor's plasma displayed increased levels of complement activity (18,19,21,23,(53)(54)(55)(56)(57)(58). Increased markers for complement activity were found when compared to those in control plasma in both patients with dry and wet AMD. ...
Article
Full-text available
Purpose Polymorphisms in complement genes are risk-associated for age-related macular degeneration (AMD). Functional analysis revealed a common deficiency to control the alternative complement pathway by risk-associated gene polymorphisms. Thus, we investigated the levels of terminal complement complex (TCC) in the plasma of wet AMD patients with defined genotypes and the impact of the complement activation of their plasma on second-messenger signaling, gene expression, and cytokine/chemokine secretion in retinal pigment epithelium (RPE) cells. Design Collection of plasma from patients with wet AMD (n = 87: 62% female and 38% male; median age 77 years) and controls (n = 86: 39% female and 61% male; median age 58 years), grouped for risk factor smoking and genetic risk alleles CFH 402HH and ARMS2 rs3750846, determination of TCC levels in the plasma, in vitro analysis on RPE function during exposure to patients’ or control plasma as a complement source. Methods Genotyping, measurement of TCC concentrations, ARPE-19 cell culture, Ca²⁺ imaging, gene expression by qPCR, secretion by multiplex bead analysis of cell culture supernatants. Main outcome measures TCC concentration in plasma, intracellular free Ca²⁺, relative mRNA levels, cytokine secretion. Results TCC levels in the plasma of AMD patients were five times higher than in non-AMD controls but did not differ in plasma from carriers of the two risk alleles. Complement-evoked Ca²⁺ elevations in RPE cells differed between patients and controls with a significant correlation between TCC levels and peak amplitudes. Comparing the Ca²⁺ signals, only between the plasma of smokers and non-smokers, as well as heterozygous (CFH 402YH) and CFH 402HH patients, revealed differences in the late phase. Pre-stimulation with complement patients’ plasma led to sensitization for complement reactions by RPE cells. Gene expression for surface molecules protective against TCC and pro-inflammatory cytokines increased after exposure to patients’ plasma. Patients’ plasma stimulated the secretion of pro-inflammatory cytokines in the RPE. Conclusion TCC levels were higher in AMD patients but did not depend on genetic risk factors. The Ca²⁺ responses to patients’ plasma as second-messenger represent a shift of RPE cells to a pro-inflammatory phenotype and protection against TCC. We conclude a substantial role of high TCC plasma levels in AMD pathology.
... There is evidence suggesting that levels of circulating complement activation products may be associated with the severity of AMD, although it is not yet clear whether this is a causal relationship or simply a result of ageing [212][213][214][215][216][217]. Systemic inflammation and endothelial dysfunction have been linked to reduced chorioretinal thickness [218], which may precede the development of AMD. ...
Article
Full-text available
The complement system is crucial for immune surveillance, providing the body’s first line of defence against pathogens. However, an imbalance in its regulators can lead to inappropriate overactivation, resulting in diseases such as age-related macular degeneration (AMD), a leading cause of irreversible blindness globally affecting around 200 million people. Complement activation in AMD is believed to begin in the choriocapillaris, but it also plays a critical role in the subretinal and retinal pigment epithelium (RPE) spaces. Bruch’s membrane (BrM) acts as a barrier between the retina/RPE and choroid, hindering complement protein diffusion. This impediment increases with age and AMD, leading to compartmentalisation of complement activation. In this review, we comprehensively examine the structure and function of BrM, including its age-related changes visible through in vivo imaging, and the consequences of complement dysfunction on AMD pathogenesis. We also explore the potential and limitations of various delivery routes (systemic, intravitreal, subretinal, and suprachoroidal) for safe and effective delivery of conventional and gene therapy-based complement inhibitors to treat AMD. Further research is needed to understand the diffusion of complement proteins across BrM and optimise therapeutic delivery to the retina.
Article
Full-text available
Careful regulation of the complement system is critical for enabling complement proteins to titrate immune defense while also preventing collateral tissue damage from poorly controlled inflammation. In the eye, this balance between complement activity and inhibition is crucial, as a low level of basal complement activity is necessary to support ocular immune privilege, a prerequisite for maintaining vision. Dysregulated complement activation contributes to parainflammation, a low level of inflammation triggered by cellular damage that functions to reestablish homeostasis, or outright inflammation that disrupts the visual axis. Complement dysregulation has been implicated in many ocular diseases, including glaucoma, diabetic retinopathy, and age-related macular degeneration (AMD). In the last two decades, complement activity has been the focus of intense investigation in AMD pathogenesis, leading to the development of novel therapeutics for the treatment of atrophic AMD. This Review outlines recent advances and challenges, highlighting therapeutic approaches that have advanced to clinical trials, as well as providing a general overview of the complement system in the posterior segment of the eye and selected ocular diseases.
Article
Inflammasomes are tightly regulated intracellular multiprotein complexes capable of sensing cytosolic perturbance that can be detrimental for cellular homeostasis. A certain type of inflammasome receptor is activated and an inflammasome multiprotein complex assembled depending on how the activation is triggered in response to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). It is the formation of the inflammasome complex which enables caspase-1 to be activated after dimerization and a subsequent auto-cleavage. Thereafter, active caspase-1 transforms interleukin (IL)-1β and IL-18 into their biologically active forms. Mature IL-1β and IL-18 are secreted out from the cell typically through the pores in the cell membrane formed by gasdermin proteins. The secretion of IL-1β and IL-18 leads to cell membrane permeabilization and inflammatory cell death, a process also called pyroptosis. Typically, inflammasome activation is described as a mechanism of self-defense and a beneficial response to infection and tissue injury. However, prolonged inflammasome activation can be detrimental and cause damage for both the host’s cells and the surrounding tissues. Especially uncontrolled and infection-free inflammasome activation is thought to be present in many chronic inflammatory diseases, e.g., age-related macular degeneration (AMD), Alzheimer’s and Parkinson’s disease, type 2 diabetes, and atherosclerosis. /// As the cornea is located in the anterior part of the eye, it is highly exposed to ultraviolet B (UVB) radiation; one of its properties is to protect the deeper ocular structures from the damaging effects of UVB. Although UVB does not reach the retina in vivo, it serves as an elegant in vitro model to mimic and study retinal pigment epithelium (RPE) and the pathogenesis of AMD, which is the most common severe eye disease among the elderly in Western world. /// The experiments conducted in this thesis evaluated if UVB-induced photochemical damage results in NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in human corneal epithelial (HCE) and human RPE cells. Moreover, it was examined whether mitochondrial damage regulates NLRP3 or absent in melanoma 2 (AIM2) inflammasome activation in human RPE cells. The ability of cis-urocanic acid (cis-UCA) to prevent inflammasome activation was explored in UVB-irradiated HCE and RPE cells. cis-UCA is a naturally occurring chromophore in human epidermis, which has been previously shown to reduce the levels of IL-8 and IL-6 in UVB-irradiated HCE and human conjunctival epithelial cells (HCEC) cells. Sequestome 1 (p62/SQSTM1) recognizes ubiquitinated protein aggregates and damaged mitochondria in the cytoplasm and delivers this kind of cargo for degradation. As a decline in intracellular protein clearance has been demonstrated to contribute to AMD causing also an elevated inflammasome activation in human RPE cells, the role of p62/SQSTM1 on the production of IL-6, IL-8, and monocyte chemoattractant protein 1 (MCP-1) was studied in human RPE cells. /// The results of this thesis revealed that UVB activates NLRP3 inflammasomes in HCE and RPE cells. Typically, one inflammasome complex can regulate the production of IL-1β and IL-18 via the same signaling route. In this thesis, concurrently produced IL-1β and IL-18 were shown to be regulated by separate signaling pathways. The production of IL-1β was dependent on potassium (K+) efflux and this was followed by the activation of the NLRP3 inflammasome, whereas the production of IL-18 was dependent on the generation of reactive oxygen species (ROS) and was determined to be independent of NLRP3 inflammasome activation in the case of the UVB-induced photochemical damage. cis-UCA alleviated UVB-induced DNA damage and the production of IL-18. Additionally, cis-UCA reduced the activation of caspase-1 and the subsequent production of IL-1β. It was also observed that when there were conditions of mitochondrial damage, both AIM2 and NLRP3 inflammasomes were activated in human RPE cells. Lastly, it was found that an excessive amount of p62/SQSTM1 induced the production of IL-8 whereas the depletion of p62/SQSTM1 triggered the production of MCP-1 in human RPE cells. /// This thesis highlights the complex activation mechanisms of inflammasomes in photochemical damage and mitochondrial damage-exposed epithelial cells from human cornea and retina. It is noteworthy that one type of danger signal can activate more than one inflammasome receptor and the production of IL-1β and IL-18 can be regulated by two distinct pathways. Especially the AIM2 inflammasome requires more attention in future studies since it has recently been shown to be involved in the development of AMD. On average, 85–90% of AMD patients suffer from the dry form of the disease for which there is currently no medical treatment. Therefore, basic research to study the pathogenesis of AMD is crucial in order to identify potential targets to stop and prevent the disease progression.
Article
Geographic atrophy (GA) secondary to age-related macular degeneration is a leading cause of visual deterioration in the global elderly population, often causing a decreased quality of life. Perifoveal atrophy can significantly affect reading, low-light vision and driving. Similarly, foveal atrophy can cause defects in central visual acuity. While the complement cascade has been implicated as playing a pivotal role in disease pathogenesis, no approved treatment for GA currently exists. Two concurrent phase III trials, DERBY and OAKS, assessed pegcetacoplan (Apellis Pharmaceuticals, MA, USA), a selective complement component C3 inhibitor, with regard to reduction in GA progression. This article reviews the available clinical trial data relating to the efficacy and safety of pegcetacoplan in the treatment of GA secondary to age-related macular degeneration.
Article
Full-text available
Purpose: To investigate the relationship between complement pathway activities and progression of geographic atrophy (GA) secondary to age-related macular degeneration in samples collected from patients enrolled in the Chroma and Spectri trials. Design: Chroma and Spectri were phase III, double-masked, and sham-controlled, 96-week trials. Participants: Aqueous humor (AH) samples collected at baseline and week 24 visits from 81 patients with bilateral GA across all 3 treatment groups (intravitreal lampalizumab 10 mg every 6 weeks, every 4 weeks, or corresponding sham procedures) were tested, along with patient-matched plasma samples collected at baseline. Methods: Antibody capture assays using the Simoa platform were used to measure the levels of complement factor B, the Bb fragment of complement factor B, intact complement component 3 (C3), processed C3, intact complement C4, and processed C4. Complement factor D levels were measured using enzyme-linked immunosorbent assay. Main outcome measures: Correlations of complement levels and activities (i.e., processed:intact ratio of complement component) in AH and plasma with baseline GA lesion size and growth rate. Results: In baseline AH, there were strong correlations (Spearman's rho ≥ 0.80) between intact complement proteins, between processed complement proteins, and between linked processed and intact complement proteins; weak correlations (rho ≤ 0.24) were found between complement pathway activities. There were no strong correlations between complement protein levels and activities measured in AH and plasma at baseline (rho ≤ 0.37). Baseline complement levels and activities in AH and plasma did not correlate with baseline GA lesion size or change from baseline in GA lesion area at week 48 (i.e., annualized growth rate). There were no strong correlations between changes in complement levels/activities in the AH from baseline to week 24 and annualized GA lesion growth rate. Genotype analysis did not reveal a meaningful correlation between complement-related, age-related macular degeneration risk single-nucleotide polymorphisms and complement levels and activities. Conclusions: Complement levels or activities in AH and plasma did not correlate with GA lesion size or growth rate. This suggests that local complement activation as measured in AH does not appear to be related to GA lesion progression. Financial disclosures: Proprietary or commercial disclosure may be found after the references.
Article
Full-text available
Age-related macular degeneration (AMD) is a leading cause of blindness in the elderly. In its severest form, choroidal neovessels breach the macular Bruch's membrane, an extracellular matrix compartment comprised of elastin and collagen laminae, and grow into the retina. We sought to determine whether structural properties of the elastic lamina (EL) correspond to the region of the macula that is predilected toward degeneration in AMD. Morphometric assessment of the macular and extramacular regions of 121 human donor eyes, with and without AMD, revealed a statistically significant difference in both the integrity (P < 0.0001) and thickness (P < 0.0001) of the EL between the macular and extramacular regions in donors of all ages. The EL was three to six times thinner and two to five times less abundant in the macula than in the periphery. The integrity of the macular EL was significantly lower in donors with early-stage AMD (P = 0.028), active choroidal neovascularization (P = 0.020), and disciform scars (P = 0.003), as compared to unaffected, age-matched controls. EL thickness was significantly lower only in individuals with disciform scars (P = 0.008). The largest gaps in macular EL integrity were significantly larger in all categories of AMD (each P < 0.0001), as compared to controls. EL integrity, thickness, and gap length in donors with geographic atrophy did not differ from those of controls. These structural properties of the macular EL correspond spatially to the distribution of macular lesions associated with AMD and may help to explain why the macula is more susceptible to degenerative events that occur in this disease.
Article
A common detection and classification system is needed for epidemiologic studies of age-related maculopathy (ARM). Such a grading scheme for ARM is described in this paper. ARM is defined as a degenerative disorder in persons ≥50 years of age characterized on grading of color fundus transparencies by the presence of the following abnormalities in the macular area: soft drusen ≥63μm, hyperpigmentation and/or hypopigmentation of the retinal pigment epithelium (RPE), RPE and associated neurosensory detachment, (peri)retinal hemorrhages, geographic atrophy of the RPE, or (peri)retinal fibrous scarring in the absence of other retinal (vascular) disorders. Visual acuity is not used to define the presence of ARM. Early ARM is defined as the presence of drusen and RPE pigmentary abnormalities described above; late ARM is similar to age-related macular degeneration (AMD) and includes dry AMD (geographic atrophy of the RPE in the absence of neovascular AMD) or neovascular AMD (RPE detachment, hemorrhages, and/or scars as described above). Methods to take and grade fundus transparencies are described.
Article
Complement activation was evaluated in plasma and synovial fluid from patients with rheumatoid arthritis. The activation fragment C3dg and the fluid-phase terminal complement complex were used as indicators of initial and terminal activation, respectively. Considerable activation of the whole complement cascade was demonstrated in most synovial fluid samples and in one-third of the plasma samples. No correlation was found between the level of activation products in synovial fluid and the level in plasma. Therefore, both compartments must be examined in order to evaluate local and systemic complement activation in rheumatoid arthritis.
Article
The common variant in the human complement Factor H gene (CFH), with Tyr402His, is linked to age-related macular degeneration (AMD), a prevalent disorder leading to visual impairment and irreversible blindness in elderly patients. Here we show that the risk variant CFH 402His displays reduced binding to C reactive protein (CRP), heparin and retinal pigment epithelial cells. This reduced binding can cause inefficient complement regulation at the cell surface, particularly when CRP is recruited to injured sites and tissue. In addition, we identify the Factor H-like protein 1 (FHL-1), an alternative splice product of the CFH gene as an additional protein that includes the risk residue 402, and thus confers risk for AMD. FHL-1 is expressed in the eye and the FHL-1 402His risk variant shows similar reduced cell binding and likely reduced complement regulatory functions on the cell surface. CFH and FHL-1 may act in concert in the eye and the reduced surface binding may result in inappropriate local complement control, which in turn can lead to inflammation, disturbance of local physiological homeostasis and progression to cell damage. As a consequence, these processes may lead to AMD pathogenesis.
Article
Typescript. Thesis (O.D.)--Pacific University, 1984. Includes bibliographical references (leaves 12-14).
Article
Hemolytic assays that measure the functional integrity of the complement system and the quantitation of individual components by immunochemical techniques have been widely used in the past for the assessment of in vivo complement activation. However, the complement system comprises a large number of interacting serum proteins which are subject to independent synthetic and catabolic processes. The fact that complement proteins are rapidly metabolized under in vivo conditions adds to the complexity of complement analysis. Assays that are based on monoclonal antibodies with specificities for activation-dependent neoepitopes now allow the direct determination of complement fragments in plasma. These methods are superior to the quantitation of native proteins. Several parameters that differentially affect the generation or the catabolism of individual complement activation products still have to be taken into account when elevated plasma levels of complement fragments suggest in vivo complement activation. These factors include the binding to complement fragment receptors, the degradation by serum proteases and renal or hepatic clearance. An accurate estimation of complement activation in vivo requires the simultaneous determination of both the native components and the activation products.
Article
The direct quantitation of C5a/C5a(desArg) in human plasma was achieved by a specific and highly sensitive ELISA which is based on the neoepitope-specific monoclonal antibody C17/5. The error-prone removal of C5 from plasma prior to the assay is therefore not required. With a detection limit of 20 pg C5a/ml plasma, the sensitivity of this assay allowed to define normal ranges (1.94 +/- 1.49 ng C5a/nl, mean +/- SD) of C5a/C5a(desArg) in human blood plasma. This assay will also be applicable to the quantitation of C5a in specimens with low protein content where precipitation-based assays fail to accurately determine C5a. In addition, the mAb C17/5 turned out to efficiently block the binding of C5a/C5a (desArg) to its cellular receptor and therefore provides a valuable tool in the delineation of C5a effects in complex biologic systems in the presence of native C5, such as under in vivo conditions.