Molecular Vision 2014; 20:1253-1257 <http://www.molvis.org/molvis/v20/1253>
Received 13 February 2014 | Accepted 11 September 2014 | Published 13 September 2014

Complement factor I and age-related macular degeneration

Philip Alexander,1 Jane Gibson,2 Angela J. Cree,1 Sarah Ennis,3 Andrew J. Lotery1

The first two authors contributed equally to this study.

1Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK; 2Centre for Biological Sciences, Faculty of Natural and Environmental Studies University of Southampton, Southampton, UK; 3Human Genetics and Genomic Medicine, Faculty of Medicine, University of Southampton, Southampton, UK

Correspondence to: Andrew Lotery, Clinical & Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK; Phone: +44 (0) 23 8120 5049; FAX: +44 (0) 709 212 5081; a.j.lotery@soton.ac.uk

Abstract

Purpose: The complement system has been implicated in the pathogenesis of age-related macular degeneration (AMD). Complement factor I (CFI) is a serum protease that inhibits all complement pathways. A previous multicenter study identified a single missense CFI mutation (p.Gly119Arg) in 20/3,567 (0.56%) of AMD cases versus 1/3,937 (0.025%) of controls, thus suggesting that this mutation confers a high risk of AMD. A second CFI mutation, p.Gly188Ala, was identified in one patient with AMD.

Methods: We screened 521 unrelated AMD cases and 627 controls for the p.Gly119Arg and p.Gly188Ala variants. All participants were Caucasian and >55 years, and recruited through Southampton Eye Unit or research clinics in Guernsey. All participants underwent dilated fundal examination by an experienced retinal specialist. SNP assays were performed using KASP™ biochemistry.

Results: The p.Gly119Arg mutation was identified in 7/521 AMD cases compared to 1/627 age-matched controls (odds ratio [OR] = 8.47, confidence interval [CI] = 1.04–69.00, p = 0.027). There was a varied phenotype among the seven cases with the mutation, which was present in 4/254 (1.6%) cases with active or end-stage wet AMD and 3/267 dry AMD cases (1.1%). The p.Gly188Ala substitution was identified in 1/521 cases and 1/627 controls.

Conclusions: Our results identified a much higher frequency of heterozygosity for p.Gly119Arg in both cases and controls than in previous studies. Of note is that our sub-cohort from Guernsey had a particularly high frequency of p.Gly119Arg heterozygosity in affected individuals (4%) compared to our sub-cohort from the mainland (0.71%). Although these data support the conclusions of van de Ven et al. that the p.Gly119Arg substitution confers a high risk of AMD, our data suggest that this missense mutation is not as rare or as highly penetrant as previously reported. There was no difference in frequency for a second CFI variant, p.Gly188Ala, between the cases and the controls.

Introduction

Age-related macular degeneration (AMD) is the leading cause of blindness in the developed world and is characterized by progressive degeneration of the RPE and secondary photoreceptor loss, resulting in visual loss. AMD is a complex multifactorial disease with a strong genetic component, and it is estimated that more than 50% of the heritability can be accounted for by the genetic loci that have already been identified [1-3], consisting of numerous genes in the complement pathway and a region of chromosome 10.

Complement factor I (CFI) is a serum protease that regulates the classical alternative complement pathways, by cleaving C4b and inactivating C3 [4]. Wang et al. have implicated CFI dysregulation in AMD, potentially resulting in chronic low-grade inflammation, and have shown that amyloid beta, a component of drusen, binds to CFI and interferes with its ability to cleave C3b [5].

Fagerness et al. identified two SNPs (rs10033900 and rs13117504) close to the CFI gene (gene ID 3426, OMIM 217030) with a combined haplotype that was strongly associated with AMD, but the span of the linkage disequilibrium included the last two exons of CFI and all four exons of phospholipase A(2) group 12A (PLA2G12A; gene ID 81579, OMIM 611652) [6]. This latter gene is hypothesized to modulate T helper cell function, and could not be excluded as the source of association with AMD. Kondo et al. demonstrated the association of rs10033900 with neovascular AMD in a Japanese cohort [7], and Qian et al. recently replicated this association in the Han Chinese population [8]. Notably, Cipriani et al. found no association between rs10033900 and AMD in a study of two separate cohorts in the United Kingdom (UK) [9].

Subsequently, Ennis et al. studied six SNPs in the CFI region [10], including four SNPs previously studied by Fagerness et al. Although four of the six SNPs achieved nominal significance in initial analysis, thus implicating CFI genomic variation with AMD disease susceptibility, no single SNP maintained marginal significance after Bonferroni correction for multiple testing.

Seddon et al. sequenced coding regions of all known AMD loci and identified a significantly increased burden in only one gene, CFI. Rare missense variants in CFI were more common in AMD cases than controls (7.8% versus 2.3%), but despite an in-depth study of 59 variants in the CFI gene, these investigators were unable to identify any individual variants associated with AMD, and suggested that this may be due to low population frequency [11]. However, one of these variants, c.355G4A (p.Gly119Arg), was identified by van de Ven et al. as a rare, highly penetrant missense mutation that confers a high risk of AMD due to altered C3b degradation [12]. These authors identified the p.Gly119Arg substitution in 20 of 3,567 AMD cases, versus only one of 3,937 controls. A second CFI mutation, p. Gly188Ala, was identified in one patient with AMD and three affected family members, but not in 809 unrelated AMD cases.

In this study, we focused on the CFI gene in our cohort of patients with AMD and controls. We screened our cohort for the mutations identified by van de Ven et al. to determine whether these mutations were as rare and highly penetrant in our population as previously described.

Methods

There were 521 unrelated AMD cases (62% female, mean age at recruitment 78.9 years [SD 7.9]) and 627 unaffected controls (56% female, mean age at recruitment 68.0 years [SD 9.8]) in this case-control study. Recruitment and eligibility of this cohort have been described previously [13]. All participants were Caucasian, aged >55 years and systemically well at the time of recruitment. Participants were recruited through the Southampton Eye Unit or from research clinics undertaken in Guernsey. All participants underwent dilated fundal examination by an experienced retinal specialist (AJL). All AMD patients had an AREDS classification of 2 or greater. Of the 521 AMD cases, 254 had active or end-stage wet AMD; the remaining 267 patients had dry AMD. Control subjects were either spouses or partners of AMD patients, or had attended the eye clinics for an unrelated eye condition. SNP assays were performed using KASP™ biochemistry (see Table 1; LGC). Recruitment was approved by Southampton and Southwest Hants local research ethics committee and followed the tenets of the Declaration of Helsinki. All participants provided informed written consent.

Association testing was performed using a Fisher’s exact test, and odds ratios (ORs) and 95% confidence intervals (CIs) were calculated. All statistical analyses were performed using PLINK, the whole genome association analysis toolset [14].

Results

The p.Gly119Arg mutation was identified in 7/521 patients with AMD and 1/627 controls. Of the patients with AMD with this mutation, four patients had evidence of active or end-stage neovascular AMD in at least one eye. The remaining three patients had dry AMD of whom two had central geographic atrophy involving the fovea. The p.Gly188Ala substitution was found in 1/521 patients with AMD and 1/627 controls. The demographic data and clinical phenotypes of the patients and controls positive for either mutation are shown in Table 1.

These data support a significant excess of the allele in CFI encoding p.Gly119Arg in the AMD cases compared to the age-matched controls (OR = 8.47, CI = 1.04–69.00, p = 0.027). Table 2 shows the frequency of this mutation in our cases and controls, and compares our results to those reported by van de Ven et al.

These results demonstrate a much higher frequency of heterozygosity for p.Gly119Arg in the cases and the controls than in the cohort reported by van de Ven et al. Of note is that our sub-cohort from Guernsey has a particularly high frequency of p.Gly119Arg heterozygosity in the affected individuals compared to our sub-cohort from the mainland. The prevalence of p.Gly119Arg heterozygosity in each sub-cohort is compared to van den Ven et al.’s findings in Table 2.

Discussion

Our results identified a much higher frequency of heterozygosity for p.Gly119Arg in the cases and the controls than in the cohort reported by van de Ven et al. [12]. Of note is that our sub-cohort from Guernsey has a particularly high frequency of p.Gly119Arg heterozygosity in affected individuals compared to our sub-cohort from the mainland. Although all known relatives, detected by clinical records and/or standard (PLINK) software [14], were excluded, within this isolated island population there are probably levels of population substructure that cannot be detected by these processes; this is the most likely explanation for the higher incidence of the p.Gly119Arg mutation in this subset. Despite an extensive literature review, no epidemiological or genetic data are available that would enable estimation of the prevalence of related individuals in Guernsey compared to the mainland UK population.

We acknowledge that our cohort is a relatively small sample size, and it is therefore difficult to infer the frequency of rare mutations. However, our findings suggest a similar frequency of the p.Gly119Arg mutation in dry AMD and neovascular AMD. A recent clinical trial of intravitreal lampalizumab, a complement factor D inhibitor, identified an undisclosed CFI sequence variation in 55% of patients with advanced dry AMD and suggested that this variation was prognostic of treatment response [15]. This indicates the direct clinical significance of genetic variants in CFI in patients with AMD. The prevalence of CFI mutations found in our study was much lower (1.3%), even when only patients with dry AMD are considered (1.1%).

These data confirm the findings of van de Ven et al. that the p.Gly119Arg substitution confers a high risk of AMD. However, our data suggest that this missense mutation is not as rare or as highly penetrant as previously reported [12]. There was no difference in prevalence for a second CFI variant, p.Gly188Ala, between the cases and the controls. With the advent of novel treatments for complement inhibition, these results may have implications for stratifying patients for AMD therapies based on genotype.

Acknowledgments

Andrew Lotery (a.j.lotery@soton.ac.uk) and Sarah Ennis (s.ennis@soton.ac.uk) contributed equally to the work presented here. We are grateful to the Brian Mercer Charitable Trust, Macular Society, TFC Frost Charitable Trust, and the Gift of Sight Appeal for providing funding for this study. We are grateful to Helen Griffiths for her technical expertise and Marie Nelson and the Wellcome Trust Clinical Research Facility in Southampton for patient recruitment, and the patients themselves for their participation in the study.

References

  1. Yu Y, Bhangale TR, Fagerness J, Ripke S, Thorleifsson G, Tan PL, Souied EH, Richardson AJ, Merriam JE, Buitendijk GH, Reynolds R, Raychaudhuri S, Chin KA, Sobrin L, Evangelou E, Lee PH, Lee AY, Leveziel N, Zack DJ, Campochiaro B, Campochiaro P, Smith RT, Barile GR, Guymer RH, Hogg R, Chakravarthy U, Robman LD, Gustafsson O, Sigurdsson H, Ortmann W, Behrens TW, Stefansson K, Uitterlinden AG, van Duijn CM, Vingerling JR, Klaver CC, Allikmets R, Brantley MA, , Jr Baird PN, Katsanis N, Thorsteinsdottir U, Ioannidis JP, Daly MJ, Graham RR, Seddon JM. Common variants near FRK/COL10A1 and VEGFA are associated with advanced age-related macular degeneration. Hum Mol Genet. 2011; 20:3699-709. [PMID: 21665990]
  2. Chen W, Stambolian D, Edwards AO, Branham KE, Othman M, Jakobsdottir J, Tosakulwong N, Pericak-Vance MA, Campochiaro PA, Klein ML, Tan PL, Conley YP, Kanda A, Kopplin L, Li Y, Augustaitis KJ, Karoukis AJ, Scott WK, Agarwal A, Kovach JL, Schwartz SG, Postel EA, Brooks M, Baratz KH, Brown WL, Brucker AJ, Orlin A, Brown G, Ho A, Regillo C, Donoso L, Tian L, Kaderli B, Hadley D, Hagstrom SA, Peachey NS, Klein R, Klein BE, Gotoh N, Yamashiro K, Ferris Iii F, Fagerness JA, Reynolds R, Farrer LA, Kim IK, Miller JW, Corton M, Carracedo A, Sanchez-Salorio M, Pugh EW, Doheny KF, Brion M, Deangelis MM, Weeks DE, Zack DJ, Chew EY, Heckenlively JR, Yoshimura N, Iyengar SK, Francis PJ, Katsanis N, Seddon JM, Haines JL, Gorin MB, Abecasis GR, Swaroop A. Genetic variants near TIMP3 and high-density lipoprotein-associated loci influence susceptibility to age-related macular degeneration. Proc Natl Acad Sci USA. 2010; 107:7401-6. [PMID: 20385819]
  3. Fritsche LG, Chen W, Schu M, Yaspan BL, Yu Y, Thorleifsson G, Zack DJ, Arakawa S, Cipriani V, Ripke S, Igo RP, , Jr Buitendijk GH, Sim X, Weeks DE, Guymer RH, Merriam JE, Francis PJ, Hannum G, Agarwal A, Armbrecht AM, Audo I, Aung T, Barile GR, Benchaboune M, Bird AC, Bishop PN, Branham KE, Brooks M, Brucker AJ, Cade WH, Cain MS, Campochiaro PA, Chan CC, Cheng CY, Chew EY, Chin KA, Chowers I, Clayton DG, Cojocaru R, Conley YP, Cornes BK, Daly MJ, Dhillon B, Edwards AO, Evangelou E, Fagerness J, Ferreyra HA, Friedman JS, Geirsdottir A, George RJ, Gieger C, Gupta N, Hagstrom SA, Harding SP, Haritoglou C, Heckenlively JR, Holz FG, Hughes G, Ioannidis JP, Ishibashi T, Joseph P, Jun G, Kamatani Y, Katsanis N. C NK, Khan JC, Kim IK, Kiyohara Y, Klein BE, Klein R, Kovach JL, Kozak I, Lee CJ, Lee KE, Lichtner P, Lotery AJ, Meitinger T, Mitchell P, Mohand-Said S, Moore AT, Morgan DJ, Morrison MA, Myers CE, Naj AC, Nakamura Y, Okada Y, Orlin A, Ortube MC, Othman MI, Pappas C, Park KH, Pauer GJ, Peachey NS, Poch O, Priya RR, Reynolds R, Richardson AJ, Ripp R, Rudolph G, Ryu E, Sahel JA, Schaumberg DA, Scholl HP, Schwartz SG, Scott WK, Shahid H, Sigurdsson H, Silvestri G, Sivakumaran TA, Smith RT, Sobrin L, Souied EH, Stambolian DE, Stefansson H, Sturgill-Short GM, Takahashi A, Tosakulwong N, Truitt BJ, Tsironi EE, Uitterlinden AG, van Duijn CM, Vijaya L, Vingerling JR, Vithana EN, Webster AR, Wichmann HE, Winkler TW, Wong TY, Wright AF, Zelenika D, Zhang M, Zhao L, Zhang K, Klein ML, Hageman GS, Lathrop GM, Stefansson K, Allikmets R, Baird PN, Gorin MB, Wang JJ, Klaver CC, Seddon JM, Pericak-Vance MA, Iyengar SK, Yates JR, Swaroop A, Weber BH, Kubo M, Deangelis MM, Leveillard T, Thorsteinsdottir U, Haines JL, Farrer LA, Heid IM, Abecasis GR. Seven new loci associated with age-related macular degeneration. Nat Genet. 2013; 45:433-9. [PMID: 23455636]
  4. Khandhadia S, Cipriani V, Yates JR, Lotery AJ. Age-related macular degeneration and the complement system. Immunobiology. 2012; 217:127-46. [PMID: 21868123]
  5. Wang J, Ohno-Matsui K, Yoshida T, Kojima A, Shimada N, Nakahama K, Safranova O, Iwata N, Saido TC, Mochizuki M, Morita I. Altered function of factor I caused by amyloid beta: implication for pathogenesis of age-related macular degeneration. Baltimore, MD: 1950. Drusen. J Immunol. 2008; 181:712-20. [PMID: 18566438]
  6. Fagerness JA, Maller JB, Neale BM, Reynolds RC, Daly MJ, Seddon JM. Variation near complement factor I is associated with risk of advanced AMD. European journal of human genetics Eur J Hum Genet. 2009; 17:100-4. [PMID: 18685559]
  7. Kondo N, Bessho H, Honda S, Negi A. Additional evidence to support the role of a common variant near the complement factor I gene in susceptibility to age-related macular degeneration. European journal of human genetics Eur J Hum Genet. 2010; 18:634-5. [PMID: 20087399]
  8. Qian D, Kan M, Weng X, Huang Y, Zhou C, Yu G, Wang T, Zhou D, Zhang Z, Zhang D, Tang W, Liu Y. Common variant rs10033900 near the complement factor I gene is associated with age-related macular degeneration risk in Han Chinese population. European journal of human genetics Eur J Hum Genet. 2014; 37 [PMID: 24642830]
  9. Cipriani V, Matharu BK, Khan JC, Shahid H, Hayward C, Wright AF, Armbrecht AM, Dhillon B, Harding SP, Bishop PN, Bunce C, Clayton DG, Moore AT, Yates JR. No evidence of association between complement factor I genetic variant rs10033900 and age-related macular degeneration. European journal of human genetics Eur J Hum Genet. 2012; 20:1-2. [PMID: 21989362]
  10. Ennis S, Gibson J, Cree AJ, Collins A, Lotery AJ. Support for the involvement of complement factor I in age-related macular degeneration. European journal of human genetics Eur J Hum Genet. 2010; 18:15-6. [PMID: 19603066]
  11. Seddon JM, Yu Y, Miller EC, Reynolds R, Tan PL, Gowrisankar S, Goldstein JI, Triebwasser M, Anderson HE, Zerbib J, Kavanagh D, Souied E, Katsanis N, Daly MJ, Atkinson JP, Raychaudhuri S. Rare variants in CFI, C3 and C9 are associated with high risk of advanced age-related macular degeneration. Nat Genet. 2013; 45:1366-70. [PMID: 24036952]
  12. van de Ven JP, Nilsson SC, Tan PL, Buitendijk GH, Ristau T, Mohlin FC, Nabuurs SB, Schoenmaker-Koller FE, Smailhodzic D, Campochiaro PA, Zack DJ, Duvvari MR, Bakker B, Paun CC, Boon CJ, Uitterlinden AG, Liakopoulos S, Klevering BJ, Fauser S, Daha MR, Katsanis N, Klaver CC, Blom AM, Hoyng CB, den Hollander AI. A functional variant in the CFI gene confers a high risk of age-related macular degeneration. Nat Genet. 2013; 45:813-7. [PMID: 23685748]
  13. Ennis S, Jomary C, Mullins R, Cree A, Chen X, Macleod A, Jones S, Collins A, Stone E, Lotery A. Association between the SERPING1 gene and age-related macular degeneration: a two-stage case-control study. Lancet. 2008; 372:1828-34. [PMID: 18842294]
  14. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, Maller J, Sklar P, de Bakker PI, Daly MJ, Sham PC. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007; 81:559-75. [PMID: 17701901]
  15. Regillo CD. Lampalizumab (Anti-factor D) in patients with Geographic Atrophy: The MAHALO Phase II Results. American Academy of Ophthalmology (AAO) Annual Meeting; 2013; New Orleans, LA, USA.