Inflammation and its role in age-related macular degeneration

Anu Kauppinen, Jussi J Paterno, Janusz Blasiak, Antero Salminen, Kai Kaarniranta, Anu Kauppinen, Jussi J Paterno, Janusz Blasiak, Antero Salminen, Kai Kaarniranta

Abstract

Inflammation is a cellular response to factors that challenge the homeostasis of cells and tissues. Cell-associated and soluble pattern-recognition receptors, e.g. Toll-like receptors, inflammasome receptors, and complement components initiate complex cellular cascades by recognizing or sensing different pathogen and damage-associated molecular patterns, respectively. Cytokines and chemokines represent alarm messages for leukocytes and once activated, these cells travel long distances to targeted inflamed tissues. Although it is a crucial survival mechanism, prolonged inflammation is detrimental and participates in numerous chronic age-related diseases. This article will review the onset of inflammation and link its functions to the pathogenesis of age-related macular degeneration (AMD), which is the leading cause of severe vision loss in aged individuals in the developed countries. In this progressive disease, degeneration of the retinal pigment epithelium (RPE) results in the death of photoreceptors, leading to a loss of central vision. The RPE is prone to oxidative stress, a factor that together with deteriorating functionality, e.g. decreased intracellular recycling and degradation due to attenuated heterophagy/autophagy, induces inflammation. In the early phases, accumulation of intracellular lipofuscin in the RPE and extracellular drusen between RPE cells and Bruch's membrane can be clinically detected. Subsequently, in dry (atrophic) AMD there is geographic atrophy with discrete areas of RPE loss whereas in the wet (exudative) form there is neovascularization penetrating from the choroid to retinal layers. Elevations in levels of local and systemic biomarkers indicate that chronic inflammation is involved in the pathogenesis of both disease forms.

Keywords: Aging; Eye; Immune system; NLRP3; Retina; Signaling.

Figures

Fig. 1
Fig. 1
a A schematic transverse section through the human eyeball. The macula is located in the posterior pole of the eye. In the center of the macula, a shallow depression in the retina (the fovea) marks the area with the highest visual acuity. Light enters the eye and bends to the sensory retina in the fovea by passing through the transparent media including cornea, lens, and the vitreous body. The sensory retina converts light into nerve impulses, processes the information, and sends it along the visual pathway to the visual cortex. b A normal human retina. A colored photograph of the fundus from the left eye of a healthy subject. The macula is located in the center of the retina. c A cross-section of the normal macula. An OCT scan through the fovea of the healthy left eye reveals the normal organization of the retinal layers. Normal anatomy of the fovea is important for accurate central vision. Modern OCT is an important in vivo tool for ophthalmologists since it allows them to monitor different pathologies non-invasively in this important but tiny and cell-dense location. d A fundus photograph from the left eye of an individual with dry AMD. This demonstrates the presence of numerous yellow deposits, known as drusen, in the central macula. e A cross-section of the macula from an individual with dry AMD. The OCT scan through the fovea of the left eye shows three drusen under the RPE layer. This eye would be expected to suffer from image distortion, as central drusen are prone to reshape the normal foveal pit. Large drusen are associated with decreased visual acuity and disruption of energy homeostasis in the retina. f A fundus photograph from the right eye of an individual with wet AMD. Significant macular edema and exudates together with foveal hemorrhage occur but only small sparse drusen are present centrally. g A cross-section from the macula in the right eye of an individual with wet AMD. An OCT scan through the location of the fovea shows the formation of intraretinal fluid cysts in the fovea. Edema causes the foveal pit to disappear. The local retinal swelling in wet AMD is due to the leaky, abnormal vessels sprouting from the underlying choroid. Intraretinal edema disrupts the normal retinal layer organization and leads to a retinal dysfunction. The OCT scan reveals also a potential hemorrhage and fibrotic lesion development in the fovea. This is another typical finding in wet AMD, likely to result in a permanent central visual field loss, if left untreated. AMD age-related macular degeneration, OCT optical coherence tomography, RPE retinal pigment epithelium
Fig. 2
Fig. 2
Major aspects of TLR signaling. Ligand recognition by LRR domains triggers the dimerisation of TLR proteins. MyD88 and TRIF are the principal adaptor proteins interacting with intracellular TIR domains and mediating the activation of transcription factors, such as NF-κB and IRFs for the production of pro-inflammatory cytokines and type I interferons. IRFs interferon regulatory factors, LRR leucine-rich repeat, MyD88 myeloid differentiation-primary response gene 88, NF-κB nuclear factor kappa B, TIR Toll/IL-1 receptor domain, TRIF TIR-domain-containing adaptor inducing IFN-β
Fig. 3
Fig. 3
Pro-inflammatory inflammasomes. Four NLRs, two DNA sensors, and an RLR are currently the most well-known inflammasomes that promote inflammation by resulting in the release of inflammatory mediators. Receptors lacking the CARD domain are dependent on the adaptor protein ASC for their interaction with pro-caspase 1, which becomes activated by autocleavage into 20 and 10 kDa subunits by the complex assembly
Fig. 4
Fig. 4
Overview of the NLRP3 inflammasome. Ligand recognition through LRR domains results in a conformational change and oligomerization of NLRP3 receptor proteins (a). Thereafter, the adaptor protein ASC binds NLRP3 by PYD–PYD interactions (b). Binding of pro-caspase-1 to ASC through CASP–CASP interactions promotes autocleavage and thereby activation of the caspase-1 enzyme (c). Finally, caspase-1 cleaves the pro-inflammatory cytokines IL-1β and IL-18 into their mature and secreted forms (d)
Fig. 5
Fig. 5
Initiation of the inflammatory response. Recognition of PAMPs and DAMPs by PRRs triggers intracellular signaling resulting in the production of pro-inflammatory cytokines and chemokines. The released mediators contribute to the activation of endothelium, e.g. elevated expression of adhesion molecules and increased vascular permeabilization. Circulating leukocytes interact with adhesion molecules expressed by endothelium, slow down their speed and start rolling along the endothelial layer. The chemokine gradient which originates from the inflamed tissue becomes sensed by leukocytes that start expressing integrins to permit their tighter binding to endothelial cells. Finally, leukocytes leave the circulation to seek out the inflamed tissue where monocytes differentiate into macrophages and dendritic cells according to the local conditions

References

    1. Thomas CJ, Schroder K. Pattern recognition receptor function in neutrophils. Trends Immunol. 2013;34:317–328. doi: 10.1016/j.it.2013.02.008.
    1. Lambrecht BN, Hammad H. The airway epithelium in asthma. Nat Med. 2012;18:684–692. doi: 10.1038/nm.2737.
    1. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–254. doi: 10.1111/j.1749-6632.2000.tb06651.x.
    1. Salvioli S, Capri M, Valensin S, Tieri P, Monti D, Ottaviani E, Franceschi C. Inflamm-aging, cytokines and aging: state of the art, new hypotheses on the role of mitochondria and new perspectives from systems biology. Curr Pharm Des. 2006;12:3161–3171. doi: 10.2174/138161206777947470.
    1. Jarrett SG, Boulton ME. Consequences of oxidative stress in age-related macular degeneration. Mol Aspects Med. 2012;33:399–417. doi: 10.1016/j.mam.2012.03.009.
    1. Ambati J, Atkinson JP, Gelfand BD. Immunology of age-related macular degeneration. Nat Rev Immunol. 2013;13:438–451. doi: 10.1038/nri3459.
    1. Viiri J, Amadio M, Marchesi N, Hyttinen JM, Kivinen N, Sironen R, Rilla K, Akhtar S, Provenzani A, D’Agostino VG, Govoni S, Pascale A, Agostini H, Petrovski G, Salminen A, Kaarniranta K. Autophagy activation clears ELAVL1/HuR-mediated accumulation of SQSTM1/p62 during proteasomal inhibition in human retinal pigment epithelial cells. PLoS One. 2013;8:e69563. doi: 10.1371/journal.pone.0069563.
    1. Mitter SK, Song C, Qi X, Mao H, Rao H, Akin D, Lewin A, Grant M, Dunn W, Jr, Ding J, Bowes Rickman C, Boulton M. Dysregulated autophagy in the RPE is associated with increased susceptibility to oxidative stress and AMD. Autophagy. 2014;10:1989–2005. doi: 10.4161/auto.36184.
    1. Piippo N, Korkmaz A, Hytti M, Kinnunen K, Salminen A, Atalay M, Kaarniranta K, Kauppinen A. Decline in cellular clearance systems induces inflammasome signaling in human ARPE-19 cells. Biochim Biophys Acta. 2014;1843:3038–3046. doi: 10.1016/j.bbamcr.2014.09.015.
    1. Ferrington DA, Sinha D, Kaarniranta K. Defects in retinal pigment epithelial cell proteolysis and the pathology associated with age-related macular degeneration. Prog Retin Eye Res. 2015
    1. Nowak JZ. Age-related macular degeneration (AMD): pathogenesis and therapy. Pharmacol Rep. 2006;58:353–363.
    1. Stefanini FR, Badaro E, Falabella P, Koss M, Farah ME, Maia M. Anti-VEGF for the management of diabetic macular edema. J Immunol Res. 2014;2014:632307. doi: 10.1155/2014/632307.
    1. Friedman DS, O’Colmain BJ, Munoz B, Tomany SC, McCarty C, de Jong PT, Nemesure B, Mitchell P, Kempen J, Eye Diseases Prevalence Research Group Prevalence of age-related macular degeneration in the US. Arch Ophthalmol. 2004;122:564–572. doi: 10.1001/archopht.122.7.1019.
    1. Prokofyeva E, Zrenner E. Epidemiology of major eye diseases leading to blindness in Europe: a literature review. Ophthalmic Res. 2012;47:171–188. doi: 10.1159/000329603.
    1. Pascolini D, Mariotti SP. Global estimates of visual impairment: 2010. Br J Ophthalmol. 2012;96:614–618. doi: 10.1136/bjophthalmol-2011-300539.
    1. De Falco S. Antiangiogenesis therapy: an update after the first decade. Korean J Intern Med. 2014;29:1–11. doi: 10.3904/kjim.2014.29.1.1.
    1. CATT Research Group. Martin DF, Maguire MG, Ying GS, Grunwald JE, Fine SL, Jaffe GJ. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. N Engl J Med. 2011;364:1897–1908. doi: 10.1056/NEJMoa1102673.
    1. Kaarniranta K, Kauppinen A, Blasiak J, Salminen A. Autophagy regulating kinases as potential therapeutic targets for age-related macular degeneration (AMD) Future Med Chem. 2012;4:2153–2161. doi: 10.4155/fmc.12.169.
    1. Chew EY, Clemons TE, Agron E, Sperduto RD, Sangiovanni JP, Kurinij N, Davis MD, Age-Related Eye Disease Study Research Group Long-term effects of vitamins C and E, beta-carotene, and zinc on age-related macular degeneration: AREDS Report No. 35. Ophthalmology. 2013;120(1604):1611.e4.
    1. Schmidt-Erfurth U, Kaiser PK, Korobelnik JF, Brown DM, Chong V, Nguyen QD, Ho AC, Ogura Y, Simader C, Jaffe GJ, Slakter JS, Yancopoulos GD, Stahl N, Vitti R, Berliner AJ, Soo Y, Anderesi M, Sowade O, Zeitz O, Norenberg C, Sandbrink R, Heier JS. Intravitreal aflibercept injection for neovascular age-related macular degeneration: 96-week results of the VIEW studies. Ophthalmology. 2014;121:193–201. doi: 10.1016/j.ophtha.2013.08.011.
    1. Damico FM, Gasparin F, Scolari MR, Pedral LS, Takahashi BS. New approaches and potential treatments for dry age-related macular degeneration. Arq Bras Oftalmol. 2012;75:71–76.
    1. Sparrow JR, Hicks D, Hamel CP. The retinal pigment epithelium in health and disease. Curr Mol Med. 2010;10:802–823. doi: 10.2174/156652410793937813.
    1. Strauss O. The retinal pigment epithelium in visual function. Physiol Rev. 2005;85:845–881. doi: 10.1152/physrev.00021.2004.
    1. Williams RA, Brody BL, Thomas RG, Kaplan RM, Brown SI. The psychosocial impact of macular degeneration. Arch Ophthalmol. 1998;116:514–520. doi: 10.1001/archopht.116.4.514.
    1. Kaarniranta K, Sinha D, Blasiak J, Kauppinen A, Vereb Z, Salminen A, Boulton ME, Petrovski G. Autophagy and heterophagy dysregulation leads to retinal pigment epithelium dysfunction and development of age-related macular degeneration. Autophagy. 2013;9:973–984. doi: 10.4161/auto.24546.
    1. Winkler BS, Boulton ME, Gottsch JD, Sternberg P. Oxidative damage and age-related macular degeneration. Mol Vis. 1999;5:32.
    1. Feeney-Burns L, Berman ER, Rothman H. Lipofuscin of human retinal pigment epithelium. Am J Ophthalmol. 1980;90:783–791. doi: 10.1016/S0002-9394(14)75193-1.
    1. Masters SL, De Nardo D. Innate immunity. Curr Opin Immunol. 2014;26:v–vi. doi: 10.1016/j.coi.2013.12.006.
    1. Schmidt AM, Vianna M, Gerlach M, Brett J, Ryan J, Kao J, Esposito C, Hegarty H, Hurley W, Clauss M. Isolation and characterization of two binding proteins for advanced glycosylation end products from bovine lung which are present on the endothelial cell surface. J Biol Chem. 1992;267:14987–14997.
    1. Neeper M, Schmidt AM, Brett J, Yan SD, Wang F, Pan YC, Elliston K, Stern D, Shaw A. Cloning and expression of a cell surface receptor for advanced glycosylation end products of proteins. J Biol Chem. 1992;267:14998–15004.
    1. Lin L. RAGE on the Toll road? Cell Mol Immunol. 2006;3:351–358.
    1. Gonzalez I, Romero J, Rodriguez BL, Perez-Castro R, Rojas A. The immunobiology of the receptor of advanced glycation end-products: trends and challenges. Immunobiology. 2013;218:790–797. doi: 10.1016/j.imbio.2012.09.005.
    1. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–384. doi: 10.1038/ni.1863.
    1. Kumar H, Kawai T, Akira S. Pathogen recognition by the innate immune system. Int Rev Immunol. 2011;30:16–34. doi: 10.3109/08830185.2010.529976.
    1. Jin B, Sun T, Yu XH, Yang YX, Yeo AET. The effects of TLR activation on T-cell development and differentiation. Clin Dev Immunol. 2012;2012:836485. doi: 10.1155/2012/836485.
    1. Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014;5:461.
    1. Schroder K, Zhou R, Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327:296–300. doi: 10.1126/science.1184003.
    1. Kaarniranta K, Salminen A, Haapasalo A, Soininen H, Hiltunen M. Age-related macular degeneration (AMD): Alzheimer’s disease in the eye? J Alzheimers Dis. 2011;24:615–631.
    1. Geijtenbeek TB, Gringhuis SI. Signalling through C-type lectin receptors: shaping immune responses. Nat Rev Immunol. 2009;9:465–479. doi: 10.1038/nri2569.
    1. Chen GY. Role of Nlrp6 and Nlrp12 in the maintenance of intestinal homeostasis. Eur J Immunol. 2014;44:321–327. doi: 10.1002/eji.201344135.
    1. Ting JP, Lovering RC, Alnemri ES, Bertin J, Boss JM, Davis BK, Flavell RA, Girardin SE, Godzik A, Harton JA, Hoffman HM, Hugot JP, Inohara N, Mackenzie A, Maltais LJ, Nunez G, Ogura Y, Otten LA, Philpott D, Reed JC, Reith W, Schreiber S, Steimle V, Ward PA. The NLR gene family: a standard nomenclature. Immunity. 2008;28:285–287. doi: 10.1016/j.immuni.2008.02.005.
    1. Hsu LC, Ali SR, McGillivray S, Tseng PH, Mariathasan S, Humke EW, Eckmann L, Powell JJ, Nizet V, Dixit VM, Karin M. A NOD2-NALP1 complex mediates caspase-1-dependent IL-1beta secretion in response to Bacillus anthracis infection and muramyl dipeptide. Proc Natl Acad Sci USA. 2008;105:7803–7808. doi: 10.1073/pnas.0802726105.
    1. Davis BK, Wen H, Ting JP. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol. 2011;29:707–735. doi: 10.1146/annurev-immunol-031210-101405.
    1. Katagiri N, Shobuike T, Chang B, Kukita A, Miyamoto H. The human apoptosis inhibitor NAIP induces pyroptosis in macrophages infected with Legionella pneumophila. Microbes Infect. 2012;14:1123–1132. doi: 10.1016/j.micinf.2012.03.006.
    1. Khare S, Dorfleutner A, Bryan NB, Yun C, Radian AD, de Almeida L, Rojanasakul Y, Stehlik C. An NLRP7-containing inflammasome mediates recognition of microbial lipopeptides in human macrophages. Immunity. 2012;36:464–476. doi: 10.1016/j.immuni.2012.02.001.
    1. Vladimer GI, Weng D, Paquette SW, Vanaja SK, Rathinam VA, Aune MH, Conlon JE, Burbage JJ, Proulx MK, Liu Q, Reed G, Mecsas JC, Iwakura Y, Bertin J, Goguen JD, Fitzgerald KA, Lien E. The NLRP12 inflammasome recognizes Yersinia pestis. Immunity. 2012;37:96–107. doi: 10.1016/j.immuni.2012.07.006.
    1. Minkiewicz J, de Rivero Vaccari JP, Keane RW. Human astrocytes express a novel NLRP2 inflammasome. Glia. 2013;61:1113–1121. doi: 10.1002/glia.22499.
    1. Allen IC. Non-inflammasome forming NLRs in inflammation and tumorigenesis. Front Immunol. 2014;5:169. doi: 10.3389/fimmu.2014.00169.
    1. Lupfer C, Kanneganti TD. Unsolved mysteries in NLR biology. Front Immunol. 2013;4:285. doi: 10.3389/fimmu.2013.00285.
    1. O’Connor W, Jr, Harton JA, Zhu X, Linhoff MW, Ting JP. Cutting edge: CIAS1/cryopyrin/PYPAF1/NALP3/CATERPILLER 1.1 is an inducible inflammatory mediator with NF-kappa B suppressive properties. J Immunol. 2003;171:6329–6333. doi: 10.4049/jimmunol.171.12.6329.
    1. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241. doi: 10.1038/nature04516.
    1. Martinon F, Gaide O, Petrilli V, Mayor A, Tschopp J. NALP inflammasomes: a central role in innate immunity. Semin Immunopathol. 2007;29:213–229. doi: 10.1007/s00281-007-0079-y.
    1. Petrilli V, Dostert C, Muruve DA, Tschopp J. The inflammasome: a danger sensing complex triggering innate immunity. Curr Opin Immunol. 2007;19:615–622. doi: 10.1016/j.coi.2007.09.002.
    1. Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, Latz E, Fitzgerald KA. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009;458:514–518. doi: 10.1038/nature07725.
    1. Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009;458:509–513. doi: 10.1038/nature07710.
    1. Burckstummer T, Baumann C, Bluml S, Dixit E, Durnberger G, Jahn H, Planyavsky M, Bilban M, Colinge J, Bennett KL, Superti-Furga G. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol. 2009;10:266–272. doi: 10.1038/ni.1702.
    1. Roberts TL, Idris A, Dunn JA, Kelly GM, Burnton CM, Hodgson S, Hardy LL, Garceau V, Sweet MJ, Ross IL, Hume DA, Stacey KJ. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science. 2009;323:1057–1060. doi: 10.1126/science.1169841.
    1. Takaoka A, Wang Z, Choi MK, Yanai H, Negishi H, Ban T, Lu Y, Miyagishi M, Kodama T, Honda K, Ohba Y, Taniguchi T. DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response. Nature. 2007;448:501–505. doi: 10.1038/nature06013.
    1. Unterholzner L, Keating SE, Baran M, Horan KA, Jensen SB, Sharma S, Sirois CM, Jin T, Latz E, Xiao TS, Fitzgerald KA, Paludan SR, Bowie AG. IFI16 is an innate immune sensor for intracellular DNA. Nat Immunol. 2010;11:997–1004. doi: 10.1038/ni.1932.
    1. Kerur N, Veettil MV, Sharma-Walia N, Bottero V, Sadagopan S, Otageri P, Chandran B. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe. 2011;9:363–375. doi: 10.1016/j.chom.2011.04.008.
    1. Ansari MA, Singh VV, Dutta S, Veettil MV, Dutta D, Chikoti L, Lu J, Everly D, Chandran B. Constitutive interferon-inducible protein 16-inflammasome activation during Epstein-Barr virus latency I, II, and III in B and epithelial cells. J Virol. 2013;87:8606–8623. doi: 10.1128/JVI.00805-13.
    1. Liao JC, Lam R, Brazda V, Duan S, Ravichandran M, Ma J, Xiao T, Tempel W, Zuo X, Wang YX, Chirgadze NY, Arrowsmith CH. Interferon-inducible protein 16: insight into the interaction with tumor suppressor p53. Structure. 2011;19:418–429. doi: 10.1016/j.str.2010.12.015.
    1. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, Taira K, Akira S, Fujita T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat Immunol. 2004;5:730–737. doi: 10.1038/ni1087.
    1. Tarallo V, Hirano Y, Gelfand BD, Dridi S, Kerur N, Kim Y, Cho WG, Kaneko H, Fowler BJ, Bogdanovich S, Albuquerque RJ, Hauswirth WW, Chiodo VA, Kugel JF, Goodrich JA, Ponicsan SL, Chaudhuri G, Murphy MP, Dunaief JL, Ambati BK, Ogura Y, Yoo JW, Lee DK, Provost P, Hinton DR, Nunez G, Baffi JZ, Kleinman ME, Ambati J. DICER1 loss and Alu RNA induce age-related macular degeneration via the NLRP3 inflammasome and MyD88. Cell. 2012;149:847–859. doi: 10.1016/j.cell.2012.03.036.
    1. Doyle SL, Campbell M, Ozaki E, Salomon RG, Mori A, Kenna PF, Farrar GJ, Kiang AS, Humphries MM, Lavelle EC, O’Neill LA, Hollyfield JG, Humphries P. NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nat Med. 2012;18:791–798. doi: 10.1038/nm.2717.
    1. Kauppinen A, Niskanen H, Suuronen T, Kinnunen K, Salminen A, Kaarniranta K. Oxidative stress activates NLRP3 inflammasomes in ARPE-19 cells-implications for age-related macular degeneration (AMD) Immunol Lett. 2012;147:29–33. doi: 10.1016/j.imlet.2012.05.005.
    1. Anderson OA, Finkelstein A, Shima DT. A2E induces IL-1β production in retinal pigment epithelial cells via the NLRP3 inflammasome. PLoS One. 2013;8:e67263. doi: 10.1371/journal.pone.0067263.
    1. Liu RT, Gao J, Cao S, Sandhu N, Cui JZ, Chou CL, Fang E, Matsubara JA. Inflammatory mediators induced by amyloid-beta in the retina and RPE in vivo: implications for inflammasome activation in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2013;54:2225–2237. doi: 10.1167/iovs.12-10849.
    1. Zhao T, Gao J, Van J, To E, Wang A, Cao S, Cui JZ, Guo JP, Lee M, McGeer PL, Matsubara JA. Age-related increases in amyloid beta and membrane attack complex: evidence of inflammasome activation in the rodent eye. J Neuroinflammation. 2015;12:121. doi: 10.1186/s12974-015-0337-1.
    1. Triantafilou K, Hughes TR, Triantafilou M, Morgan BP. The complement membrane attack complex triggers intracellular Ca2+ fluxes leading to NLRP3 inflammasome activation. J Cell Sci. 2013;126:2903–2913. doi: 10.1242/jcs.124388.
    1. Brandstetter C, Mohr LK, Latz E, Holz FG, Krohne TU. Light induces NLRP3 inflammasome activation in retinal pigment epithelial cells via lipofuscin-mediated photooxidative damage. J Mol Med (Berl) 2015;93:905–916. doi: 10.1007/s00109-015-1275-1.
    1. Gelfand BD, Wright CB, Kim Y, Yasuma T, Yasuma R, Li S, Fowler BJ, Bastos-Carvalho A, Kerur N, Uittenbogaard A, Han YS, Lou D, Kleinman ME, McDonald WH, Nunez G, Georgel P, Dunaief JL, Ambati J. Iron toxicity in the retina requires Alu RNA and the NLRP3 inflammasome. Cell Rep. 2015;11:1686–1693. doi: 10.1016/j.celrep.2015.05.023.
    1. Blasiak J, Szaflik J, Szaflik JP. Implications of altered iron homeostasis for age-related macular degeneration. Front Biosci (Landmark Ed) 2011;16:1551–1559. doi: 10.2741/3804.
    1. Shi G, Chen S, Wandu WS, Ogbeifun O, Nugent LF, Maminishkis A, Hinshaw SJ, Rodriguez IR, Gery I. Inflammasomes induced by 7-ketocholesterol and other stimuli in RPE and in bone marrow-derived cells differ markedly in their production of IL-1beta and IL-18. Invest Ophthalmol Vis Sci. 2015;56:1658–1664. doi: 10.1167/iovs.14-14557.
    1. Marneros AG. NLRP3 inflammasome blockade inhibits VEGF-A-induced age-related macular degeneration. Cell Rep. 2013;4:945–958. doi: 10.1016/j.celrep.2013.08.002.
    1. Tseng WA, Thein T, Kinnunen K, Lashkari K, Gregory MS, D’Amore PA, Ksander BR. NLRP3 inflammasome activation in retinal pigment epithelial cells by lysosomal destabilization: implications for age-related macular degeneration. Invest Ophthalmol Vis Sci. 2013;54:110–120. doi: 10.1167/iovs.12-10655.
    1. Kerur N, Hirano Y, Tarallo V, Fowler BJ, Bastos-Carvalho A, Yasuma T, Yasuma R, Kim Y, Hinton DR, Kirschning CJ, Gelfand BD, Ambati J. TLR-independent and P2X7-dependent signaling mediate Alu RNA-induced NLRP3 inflammasome activation in geographic atrophy. Invest Ophthalmol Vis Sci. 2013;54:7395–7401. doi: 10.1167/iovs.13-12500.
    1. Hu SJ, Calippe B, Lavalette S, Roubeix C, Montassar F, Housset M, Levy O, Delarasse C, Paques M, Sahel JA, Sennlaub F, Guillonneau X. Upregulation of P2RX7 in Cx3cr1-deficient mononuclear phagocytes leads to increased interleukin-1beta secretion and photoreceptor neurodegeneration. J Neurosci. 2015;35:6987–6996. doi: 10.1523/JNEUROSCI.3955-14.2015.
    1. Indaram M, Ma W, Zhao L, Fariss RN, Rodriguez IR, Wong WT. 7-Ketocholesterol increases retinal microglial migration, activation, and angiogenicity: a potential pathogenic mechanism underlying age-related macular degeneration. Sci Rep. 2015;5:9144. doi: 10.1038/srep09144.
    1. Kim B, Lee S, Suvas S, Rouse BT. Application of plasmid DNA encoding IL-18 diminishes development of herpetic stromal keratitis by antiangiogenic effects. J Immunol. 2005;175:509–516. doi: 10.4049/jimmunol.175.1.509.
    1. Qiao H, Sonoda KH, Ikeda Y, Yoshimura T, Hijioka K, Jo YJ, Sassa Y, Tsutsumi-Miyahara C, Hata Y, Akira S, Ishibashi T. Interleukin-18 regulates pathological intraocular neovascularization. J Leukoc Biol. 2007;81:1012–1021. doi: 10.1189/jlb.0506342.
    1. Shen J, Choy DF, Yoshida T, Iwase T, Hafiz G, Xie B, Hackett SF, Arron JR, Campochiaro PA. Interleukin-18 has antipermeablity and antiangiogenic activities in the eye; reciprocal suppression with VEGF. J Cell Physiol. 2014;229:974–983. doi: 10.1002/jcp.24575.
    1. Doyle SL, Ozaki E, Brennan K, Humphries MM, Mulfaul K, Keaney J, Kenna PF, Maminishkis A, Kiang AS, Saunders SP, Hams E, Lavelle EC, Gardiner C, Fallon PG, Adamson P, Humphries P, Campbell M. IL-18 attenuates experimental choroidal neovascularization as a potential therapy for wet age-related macular degeneration. Sci Transl Med. 2014;6:230ra44. doi: 10.1126/scitranslmed.3007616.
    1. Ijima R, Kaneko H, Ye F, Nagasaka Y, Takayama K, Kataoka K, Kachi S, Iwase T, Terasaki H. Interleukin-18 induces retinal pigment epithelium degeneration in mice. Invest Ophthalmol Vis Sci. 2014;55:6673–6678. doi: 10.1167/iovs.14-15367.
    1. Kim Y, Tarallo V, Kerur N, Yasuma T, Gelfand BD, Bastos-Carvalho A, Hirano Y, Yasuma R, Mizutani T, Fowler BJ, Li S, Kaneko H, Bogdanovich S, Ambati BK, Hinton DR, Hauswirth WW, Hakem R, Wright C, Ambati J. DICER1/Alu RNA dysmetabolism induces Caspase-8-mediated cell death in age-related macular degeneration. Proc Natl Acad Sci USA. 2014;111:16082–16087. doi: 10.1073/pnas.1403814111.
    1. Hirano Y, Yasuma T, Mizutani T, Fowler BJ, Tarallo V, Yasuma R, Kim Y, Bastos-Carvalho A, Kerur N, Gelfand BD, Bogdanovich S, He S, Zhang X, Nozaki M, Ijima R, Kaneko H, Ogura Y, Terasaki H, Nagai H, Haro I, Nunez G, Ambati BK, Hinton DR, Ambati J. IL-18 is not therapeutic for neovascular age-related macular degeneration. Nat Med. 2014;20:1372–1375. doi: 10.1038/nm.3671.
    1. Doyle SL, Adamson P, Lopez FJ, Humphries P, Campbell M. Reply to IL-18 is not therapeutic for neovascular age-related macular degeneration. Nat Med. 2014;20:1376–1377. doi: 10.1038/nm.3741.
    1. Doyle SL, Adamson P, Lopez FJ, Humphries P, Campbell M. Interleukin-18 bioactivity and dose: data interpretation at a crossroads. Invest Ophthalmol Vis Sci. 2014;55:8349–8350. doi: 10.1167/iovs.14-15786.
    1. Kaneko H. Author response: interleukin-18 bioactivity and dose: data interpretation at a crossroads. Invest Ophthalmol Vis Sci. 2014;55:8350–8352. doi: 10.1167/iovs.14-15878.
    1. Liu RT, Wang A, To E, Gao J, Cao S, Cui JZ, Matsubara JA. Vinpocetine inhibits amyloid-beta induced activation of NF-kappaB, NLRP3 inflammasome and cytokine production in retinal pigment epithelial cells. Exp Eye Res. 2014;127:49–58. doi: 10.1016/j.exer.2014.07.003.
    1. Ildefonso CJ, Jaime H, Biswal MR, Boye SE, Li Q, Hauswirth WW, Lewin AS. Gene therapy with the caspase activation and recruitment domain reduces the ocular inflammatory response. Mol Ther. 2015;23:875–884. doi: 10.1038/mt.2015.30.
    1. Dunkelberger JR, Song WC. Complement and its role in innate and adaptive immune responses. Cell Res. 2010;20:34–50. doi: 10.1038/cr.2009.139.
    1. Hawlisch H, Kohl J. Complement and Toll-like receptors: key regulators of adaptive immune responses. Mol Immunol. 2006;43:13–21. doi: 10.1016/j.molimm.2005.06.028.
    1. Nilsson G, Johnell M, Hammer CH, Tiffany HL, Nilsson K, Metcalfe DD, Siegbahn A, Murphy PM. C3a and C5a are chemotaxins for human mast cells and act through distinct receptors via a pertussis toxin-sensitive signal transduction pathway. J Immunol. 1996;157:1693–1698.
    1. DiScipio RG, Schraufstatter IU. The role of the complement anaphylatoxins in the recruitment of eosinophils. Int Immunopharmacol. 2007;7:1909–1923. doi: 10.1016/j.intimp.2007.07.006.
    1. Daffern PJ, Pfeifer PH, Ember JA, Hugli TE. C3a is a chemotaxin for human eosinophils but not for neutrophils. I. C3a stimulation of neutrophils is secondary to eosinophil activation. J Exp Med. 1995;181:2119–2127. doi: 10.1084/jem.181.6.2119.
    1. Kildsgaard J, Hollmann TJ, Matthews KW, Bian K, Murad F, Wetsel RA. Cutting edge: targeted disruption of the C3a receptor gene demonstrates a novel protective anti-inflammatory role for C3a in endotoxin-shock. J Immunol. 2000;165:5406–5409. doi: 10.4049/jimmunol.165.10.5406.
    1. Asgari E, Le Friec G, Yamamoto H, Perucha E, Sacks SS, Kohl J, Cook HT, Kemper C. C3a modulates IL-1beta secretion in human monocytes by regulating ATP efflux and subsequent NLRP3 inflammasome activation. Blood. 2013;122:3473–3481. doi: 10.1182/blood-2013-05-502229.
    1. Laudisi F, Spreafico R, Evrard M, Hughes TR, Mandriani B, Kandasamy M, Morgan BP, Sivasankar B, Mortellaro A. Cutting edge: the NLRP3 inflammasome links complement-mediated inflammation and IL-1beta release. J Immunol. 2013;191:1006–1010. doi: 10.4049/jimmunol.1300489.
    1. An LL, Mehta P, Xu L, Turman S, Reimer T, Naiman B, Connor J, Sanjuan M, Kolbeck R, Fung M. Complement C5a potentiates uric acid crystal-induced IL-1beta production. Eur J Immunol. 2014;44:3669–3679. doi: 10.1002/eji.201444560.
    1. Samstad EO, Niyonzima N, Nymo S, Aune MH, Ryan L, Bakke SS, Lappegard KT, Brekke OL, Lambris JD, Damas JK, Latz E, Mollnes TE, Espevik T. Cholesterol crystals induce complement-dependent inflammasome activation and cytokine release. J Immunol. 2014;192:2837–2845. doi: 10.4049/jimmunol.1302484.
    1. Cumpelik A, Ankli B, Zecher D, Schifferli JA. Neutrophil microvesicles resolve gout by inhibiting C5a-mediated priming of the inflammasome. Ann Rheum Dis. 2015
    1. Agrawal A, Singh PP, Bottazzi B, Garlanda C, Mantovani A. Pattern recognition by pentraxins. Adv Exp Med Biol. 2009;653:98–116. doi: 10.1007/978-1-4419-0901-5_7.
    1. Deban L, Bottazzi B, Garlanda C, de la Torre YM, Mantovani A. Pentraxins: multifunctional proteins at the interface of innate immunity and inflammation. Biofactors. 2009;35:138–145. doi: 10.1002/biof.21.
    1. Mousavi M, Armstrong RA. Genetic risk factors and age-related macular degeneration (AMD) J Optom. 2013;6:176–184. doi: 10.1016/j.optom.2013.07.002.
    1. Edwards AO, Ritter R, 3rd, Abel KJ, Manning A, Panhuysen C, Farrer LA. Complement factor H polymorphism and age-related macular degeneration. Science. 2005;308:421–424. doi: 10.1126/science.1110189.
    1. Haines JL, Hauser MA, Schmidt S, Scott WK, Olson LM, Gallins P, Spencer KL, Kwan SY, Noureddine M, Gilbert JR, Schnetz-Boutaud N, Agarwal A, Postel EA, Pericak-Vance MA. Complement factor H variant increases the risk of age-related macular degeneration. Science. 2005;308:419–421. doi: 10.1126/science.1110359.
    1. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, Haynes C, Henning AK, SanGiovanni JP, Mane SM, Mayne ST, Bracken MB, Ferris FL, Ott J, Barnstable C, Hoh J. Complement factor H polymorphism in age-related macular degeneration. Science. 2005;308:385–389. doi: 10.1126/science.1109557.
    1. Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, Hardisty LI, Hageman JL, Stockman HA, Borchardt JD, Gehrs KM, Smith RJ, Silvestri G, Russell SR, Klaver CC, Barbazetto I, Chang S, Yannuzzi LA, Barile GR, Merriam JC, Smith RT, Olsh AK, Bergeron J, Zernant J, Merriam JE, Gold B, Dean M, Allikmets R. 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. doi: 10.1073/pnas.0501536102.
    1. Horie-Inoue K, Inoue S. Genomic aspects of age-related macular degeneration. Biochem Biophys Res Commun. 2014;452:263–275. doi: 10.1016/j.bbrc.2014.08.013.
    1. Okemefuna AI, Nan R, Miller A, Gor J, Perkins SJ. Complement factor H binds at two independent sites to C-reactive protein in acute phase concentrations. J Biol Chem. 2010;285:1053–1065. doi: 10.1074/jbc.M109.044529.
    1. Laine M, Jarva H, Seitsonen S, Haapasalo K, Lehtinen MJ, Lindeman N, Anderson DH, Johnson PT, Jarvela I, Jokiranta TS, Hageman GS, Immonen I, Meri S. Y402H polymorphism of complement factor H affects binding affinity to C-reactive protein. J Immunol. 2007;178:3831–3836. doi: 10.4049/jimmunol.178.6.3831.
    1. Deban L, Jarva H, Lehtinen MJ, Bottazzi B, Bastone A, Doni A, Jokiranta TS, Mantovani A, Meri S. Binding of the long pentraxin PTX3 to factor H: interacting domains and function in the regulation of complement activation. J Immunol. 2008;181:8433–8440. doi: 10.4049/jimmunol.181.12.8433.
    1. Kaemmerer E, Schutt F, Krohne TU, Holz FG, Kopitz J. Effects of lipid peroxidation-related protein modifications on RPE lysosomal functions and POS phagocytosis. Invest Ophthalmol Vis Sci. 2007;48:1342–1347. doi: 10.1167/iovs.06-0549.
    1. Weismann D, Hartvigsen K, Lauer N, Bennett KL, Scholl HP, Charbel Issa P, Cano M, Brandstatter H, Tsimikas S, Skerka C, Superti-Furga G, Handa JT, Zipfel PF, Witztum JL, Binder CJ. Complement factor H binds malondialdehyde epitopes and protects from oxidative stress. Nature. 2011;478:76–81. doi: 10.1038/nature10449.
    1. Chen M, Forrester JV, Xu H. Synthesis of complement factor H by retinal pigment epithelial cells is down-regulated by oxidized photoreceptor outer segments. Exp Eye Res. 2007;84:635–645. doi: 10.1016/j.exer.2006.11.015.
    1. Wu Z, Lauer TW, Sick A, Hackett SF, Campochiaro PA. Oxidative stress modulates complement factor H expression in retinal pigmented epithelial cells by acetylation of FOXO3. J Biol Chem. 2007;282:22414–22425. doi: 10.1074/jbc.M702321200.
    1. Chen M, Muckersie E, Robertson M, Forrester JV, Xu H. Up-regulation of complement factor B in retinal pigment epithelial cells is accompanied by complement activation in the aged retina. Exp Eye Res. 2008;87:543–550. doi: 10.1016/j.exer.2008.09.005.
    1. Kaarniranta K, Salminen A. Age-related macular degeneration: activation of innate immunity system via pattern recognition receptors. J Mol Med (Berl) 2009;87:117–123. doi: 10.1007/s00109-008-0418-z.
    1. Thurman JM, Renner B, Kunchithapautham K, Ferreira VP, Pangburn MK, Ablonczy Z, Tomlinson S, Holers VM, Rohrer B. Oxidative stress renders retinal pigment epithelial cells susceptible to complement-mediated injury. J Biol Chem. 2009;284:16939–16947. doi: 10.1074/jbc.M808166200.
    1. Kunchithapautham K, Atkinson C, Rohrer B. Smoke exposure causes endoplasmic reticulum stress and lipid accumulation in retinal pigment epithelium through oxidative stress and complement activation. J Biol Chem. 2014;289:14534–14546. doi: 10.1074/jbc.M114.564674.
    1. Aredo B, Li T, Chen X, Zhang K, Wang CX, Gou D, Zhao B, He Y, Ufret-Vincenty RL. A chimeric Cfh transgene leads to increased retinal oxidative stress, inflammation, and accumulation of activated subretinal microglia in mice. Invest Ophthalmol Vis Sci. 2015;56:3427–3440. doi: 10.1167/iovs.14-16089.
    1. Luo C, Zhao J, Madden A, Chen M, Xu H. Complement expression in retinal pigment epithelial cells is modulated by activated macrophages. Exp Eye Res. 2013;112:93–101. doi: 10.1016/j.exer.2013.04.016.
    1. Yates JR, Sepp T, Matharu BK, Khan JC, Thurlby DA, Shahid H, Clayton DG, Hayward C, Morgan J, Wright AF, Armbrecht AM, Dhillon B, Deary IJ, Redmond E, Bird AC, Moore AT, Genetic Factors in AMD Study Group Complement C3 variant and the risk of age-related macular degeneration. N Engl J Med. 2007;357:553–561. doi: 10.1056/NEJMoa072618.
    1. Maller JB, Fagerness JA, Reynolds RC, Neale BM, Daly MJ, Seddon JM. Variation in complement factor 3 is associated with risk of age-related macular degeneration. Nat Genet. 2007;39:1200–1201. doi: 10.1038/ng2131.
    1. Seitsonen SP, Onkamo P, Peng G, Xiong M, Tommila PV, Ranta PH, Holopainen JM, Moilanen JA, Palosaari T, Kaarniranta K, Meri S, Immonen IR, Jarvela IE. Multifactor effects and evidence of potential interaction between complement factor H Y402H and LOC387715 A69S in age-related macular degeneration. PLoS One. 2008;3:e3833. doi: 10.1371/journal.pone.0003833.
    1. 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;45:1366–1370. doi: 10.1038/ng.2741.
    1. 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;45:813–817. doi: 10.1038/ng.2640.
    1. Zhan X, Larson DE, Wang C, Koboldt DC, Sergeev YV, Fulton RS, Fulton LL, Fronick CC, Branham KE, Bragg-Gresham J, Jun G, Hu Y, Kang HM, Liu D, Othman M, Brooks M, Ratnapriya R, Boleda A, Grassmann F, von Strachwitz C, Olson LM, Buitendijk GH, Hofman A, van Duijn CM, Cipriani V, Moore AT, Shahid H, Jiang Y, Conley YP, Morgan DJ, Kim IK, Johnson MP, Cantsilieris S, Richardson AJ, Guymer RH, Luo H, Ouyang H, Licht C, Pluthero FG, Zhang MM, Zhang K, Baird PN, Blangero J, Klein ML, Farrer LA, DeAngelis MM, Weeks DE, Gorin MB, Yates JR, Klaver CC, Pericak-Vance MA, Haines JL, Weber BH, Wilson RK, Heckenlively JR, Chew EY, Stambolian D, Mardis ER, Swaroop A, Abecasis GR. Identification of a rare coding variant in complement 3 associated with age-related macular degeneration. Nat Genet. 2013;45:1375–1379. doi: 10.1038/ng.2758.
    1. Alexander P, Gibson J, Cree AJ, Ennis S, Lotery AJ. Complement factor I and age-related macular degeneration. Mol Vis. 2014;20:1253–1257.
    1. Gibson J, Cree A, Collins A, Lotery A, Ennis S. Determination of a gene and environment risk model for age-related macular degeneration. Br J Ophthalmol. 2010;94:1382–1387. doi: 10.1136/bjo.2010.182568.
    1. Tanaka K, Nakayama T, Mori R, Sato N, Kawamura A, Yuzawa M. Associations of complement factor B and complement component 2 genotypes with subtypes of polypoidal choroidal vasculopathy. BMC Ophthalmol. 2014;14:83. doi: 10.1186/1471-2415-14-83.
    1. Schnabolk G, Coughlin B, Joseph K, Kunchithapautham K, Bandyopadhyay M, O’Quinn EC, Nowling T, Rohrer B. Local production of the alternative pathway component factor B is sufficient to promote laser-induced choroidal neovascularization. Invest Ophthalmol Vis Sci. 2015;56:1850–1863. doi: 10.1167/iovs.14-15910.
    1. Gold B, Merriam JE, Zernant J, Hancox LS, Taiber AJ, Gehrs K, Cramer K, Neel J, Bergeron J, Barile GR, Smith RT, AMD Genetics Clinical Study Group. Hageman GS, Dean M, Allikmets R. Variation in factor B (BF) and complement component 2 (C2) genes is associated with age-related macular degeneration. Nat Genet. 2006;38:458–462. doi: 10.1038/ng1750.
    1. Spencer KL, Hauser MA, Olson LM, Schmidt S, Scott WK, Gallins P, Agarwal A, Postel EA, Pericak-Vance MA, Haines JL. Protective effect of complement factor B and complement component 2 variants in age-related macular degeneration. Hum Mol Genet. 2007;16:1986–1992. doi: 10.1093/hmg/ddm146.
    1. Thakkinstian A, McEvoy M, Chakravarthy U, Chakrabarti S, McKay GJ, Ryu E, Silvestri G, Kaur I, Francis P, Iwata T, Akahori M, Arning A, Edwards AO, Seddon JM, Attia J. The association between complement component 2/complement factor B polymorphisms and age-related macular degeneration: a HuGE review and meta-analysis. Am J Epidemiol. 2012;176:361–372. doi: 10.1093/aje/kws031.
    1. Wang X, Zhang Y, Zhang MN. Complement factor B polymorphism (rs641153) and susceptibility to age-related macular degeneration: evidence from published studies. Int J Ophthalmol. 2013;6:861–867.
    1. Nozaki M, Raisler BJ, Sakurai E, Sarma JV, Barnum SR, Lambris JD, Chen Y, Zhang K, Ambati BK, Baffi JZ, Ambati J. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc Natl Acad Sci USA. 2006;103:2328–2333. doi: 10.1073/pnas.0408835103.
    1. Bora NS, Kaliappan S, Jha P, Xu Q, Sivasankar B, Harris CL, Morgan BP, Bora PS. CD59, a complement regulatory protein, controls choroidal neovascularization in a mouse model of wet-type age-related macular degeneration. J Immunol. 2007;178:1783–1790. doi: 10.4049/jimmunol.178.3.1783.
    1. Cashman SM, Ramo K, Kumar-Singh R. A non membrane-targeted human soluble CD59 attenuates choroidal neovascularization in a model of age related macular degeneration. PLoS One. 2011;6:e19078. doi: 10.1371/journal.pone.0019078.
    1. Joseph K, Kulik L, Coughlin B, Kunchithapautham K, Bandyopadhyay M, Thiel S, Thielens NM, Holers VM, Rohrer B. Oxidative stress sensitizes retinal pigmented epithelial (RPE) cells to complement-mediated injury in a natural antibody-, lectin pathway-, and phospholipid epitope-dependent manner. J Biol Chem. 2013;288:12753–12765. doi: 10.1074/jbc.M112.421891.
    1. Fernandez-Godino R, Garland DL, Pierce EA. A local complement response by RPE causes early-stage macular degeneration. Hum Mol Genet. 2015;24:5555–5559. doi: 10.1093/hmg/ddv287.
    1. Wang J, Ohno-Matsui K, Yoshida T, Shimada N, Ichinose S, Sato T, Mochizuki M, Morita I. Amyloid-beta up-regulates complement factor B in retinal pigment epithelial cells through cytokines released from recruited macrophages/microglia: another mechanism of complement activation in age-related macular degeneration. J Cell Physiol. 2009;220:119–128. doi: 10.1002/jcp.21742.
    1. Wang L, Kondo N, Cano M, Ebrahimi K, Yoshida T, Barnett BP, Biswal S, Handa JT. Nrf2 signaling modulates cigarette smoke-induced complement activation in retinal pigmented epithelial cells. Free Radic Biol Med. 2014;70:155–166. doi: 10.1016/j.freeradbiomed.2014.01.015.
    1. Liu B, Wei L, Meyerle C, Tuo J, Sen HN, Li Z, Chakrabarty S, Agron E, Chan CC, Klein ML, Chew E, Ferris F, Nussenblatt RB. Complement component C5a promotes expression of IL-22 and IL-17 from human T cells and its implication in age-related macular degeneration. J Transl Med. 2011;9:1–12. doi: 10.1186/1479-5876-9-1.
    1. Wei L, Liu B, Tuo J, Shen D, Chen P, Li Z, Liu X, Ni J, Dagur P, Sen HN, Jawad S, Ling D, Park S, Chakrabarty S, Meyerle C, Agron E, Ferris FL, 3rd, Chew EY, McCoy JP, Blum E, Francis PJ, Klein ML, Guymer RH, Baird PN, Chan CC, Nussenblatt RB. Hypomethylation of the IL17RC promoter associates with age-related macular degeneration. Cell Rep. 2012;2:1151–1158. doi: 10.1016/j.celrep.2012.10.013.
    1. Ardeljan D, Wang Y, Park S, Shen D, Chu XK, Yu CR, Abu-Asab M, Tuo J, Eberhart CG, Olsen TW, Mullins RF, White G, Wadsworth S, Scaria A, Chan CC. Interleukin-17 retinotoxicity is prevented by gene transfer of a soluble interleukin-17 receptor acting as a cytokine blocker: implications for age-related macular degeneration. PLoS One. 2014;9:e95900. doi: 10.1371/journal.pone.0095900.
    1. Chen Y, Zhong M, Liang L, Gu F, Peng H. Interleukin-17 induces angiogenesis in human choroidal endothelial cells in vitro. Invest Ophthalmol Vis Sci. 2014;55:6968–6975. doi: 10.1167/iovs.14-15029.
    1. Zhang S, Liu Y, Lu S, Cai X. Genetic variants of interleukin 17A are functionally associated with increased risk of age-related macular degeneration. Inflammation. 2015;38:658–663. doi: 10.1007/s10753-014-9973-3.
    1. Zhang M, Xu G, Liu W, Ni Y, Zhou W. Role of fractalkine/CX3CR1 interaction in light-induced photoreceptor degeneration through regulating retinal microglial activation and migration. PLoS One. 2012;7:e35446. doi: 10.1371/journal.pone.0035446.
    1. Faure S, Meyer L, Costagliola D, Vaneensberghe C, Genin E, Autran B, Delfraissy JF, McDermott DH, Murphy PM, Debre P, Theodorou I, Combadiere C. Rapid progression to AIDS in HIV+ individuals with a structural variant of the chemokine receptor CX3CR1. Science. 2000;287:2274–2277. doi: 10.1126/science.287.5461.2274.
    1. Tuo J, Bojanowski CM, Zhou M, Shen D, Ross RJ, Rosenberg KI, Cameron DJ, Yin C, Kowalak JA, Zhuang Z, Zhang K, Chan CC. Murine ccl2/cx3cr1 deficiency results in retinal lesions mimicking human age-related macular degeneration. Invest Ophthalmol Vis Sci. 2007;48:3827–3836. doi: 10.1167/iovs.07-0051.
    1. Ross RJ, Zhou M, Shen D, Fariss RN, Ding X, Bojanowski CM, Tuo J, Chan CC. Immunological protein expression profile in Ccl2/Cx3cr1 deficient mice with lesions similar to age-related macular degeneration. Exp Eye Res. 2008;86:675–683. doi: 10.1016/j.exer.2008.01.014.
    1. Chen M, Hombrebueno JR, Luo C, Penalva R, Zhao J, Colhoun L, Pandi SP, Forrester JV, Xu H. Age- and light-dependent development of localised retinal atrophy in CCL2(−/−)CX3CR1(GFP/GFP) mice. PLoS One. 2013;8:e61381. doi: 10.1371/journal.pone.0061381.
    1. Sennlaub F, Auvynet C, Calippe B, Lavalette S, Poupel L, Hu SJ, Dominguez E, Camelo S, Levy O, Guyon E, Saederup N, Charo IF, Rooijen NV, Nandrot E, Bourges JL, Behar-Cohen F, Sahel JA, Guillonneau X, Raoul W, Combadiere C. CCR2(+) monocytes infiltrate atrophic lesions in age-related macular disease and mediate photoreceptor degeneration in experimental subretinal inflammation in Cx3cr1 deficient mice. EMBO Mol Med. 2013;5:1775–1793. doi: 10.1002/emmm.201302692.
    1. Liang KJ, Lee JE, Wang YD, Ma W, Fontainhas AM, Fariss RN, Wong WT. Regulation of dynamic behavior of retinal microglia by CX3CR1 signaling. Invest Ophthalmol Vis Sci. 2009;50:4444–4451. doi: 10.1167/iovs.08-3357.
    1. Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454:428–435. doi: 10.1038/nature07201.
    1. Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Prog Retin Eye Res. 2009;28:348–368. doi: 10.1016/j.preteyeres.2009.06.001.
    1. McDermott DH, Fong AM, Yang Q, Sechler JM, Cupples LA, Merrell MN, Wilson PW, D’Agostino RB, O’Donnell CJ, Patel DD, Murphy PM. Chemokine receptor mutant CX3CR1-M280 has impaired adhesive function and correlates with protection from cardiovascular disease in humans. J Clin Invest. 2003;111:1241–1250. doi: 10.1172/JCI16790.
    1. Daoudi M, Lavergne E, Garin A, Tarantino N, Debre P, Pincet F, Combadiere C, Deterre P. Enhanced adhesive capacities of the naturally occurring Ile249-Met280 variant of the chemokine receptor CX3CR1. J Biol Chem. 2004;279:19649–19657. doi: 10.1074/jbc.M313457200.
    1. Chan CC, Tuo J, Bojanowski CM, Csaky KG, Green WR. Detection of CX3CR1 single nucleotide polymorphism and expression on archived eyes with age-related macular degeneration. Histol Histopathol. 2005;20:857–863.
    1. Yang X, Hu J, Zhang J, Guan H. Polymorphisms in CFH, HTRA1 and CX3CR1 confer risk to exudative age-related macular degeneration in Han Chinese. Br J Ophthalmol. 2010;94:1211–1214. doi: 10.1136/bjo.2009.165811.
    1. Vessey KA, Greferath U, Jobling AI, Phipps JA, Ho T, Waugh M, Fletcher EL. Ccl2/Cx3cr1 knockout mice have inner retinal dysfunction but are not an accelerated model of AMD. Invest Ophthalmol Vis Sci. 2012;53:7833–7846. doi: 10.1167/iovs.12-10650.
    1. Luhmann UF, Carvalho LS, Robbie SJ, Cowing JA, Duran Y, Munro PM, Bainbridge JW, Ali RR. Ccl2, Cx3cr1 and Ccl2/Cx3cr1 chemokine deficiencies are not sufficient to cause age-related retinal degeneration. Exp Eye Res. 2013;107:80–87. doi: 10.1016/j.exer.2012.11.015.
    1. Schaumberg DA, Rose L, DeAngelis MM, Semba RD, Hageman GS, Chasman DI. Prospective study of common variants in CX3CR1 and risk of macular degeneration: pooled analysis from 5 long-term studies. JAMA Ophthalmol. 2014;132:84–95. doi: 10.1001/jamaophthalmol.2013.5506.
    1. Luhmann UF, Lange CA, Robbie S, Munro PM, Cowing JA, Armer HE, Luong V, Carvalho LS, MacLaren RE, Fitzke FW, Bainbridge JW, Ali RR. Differential modulation of retinal degeneration by Ccl2 and Cx3cr1 chemokine signalling. PLoS One. 2012;7:e35551. doi: 10.1371/journal.pone.0035551.
    1. Mattapallil MJ, Wawrousek EF, Chan CC, Zhao H, Roychoudhury J, Ferguson TA, Caspi RR. The Rd8 mutation of the Crb1 gene is present in vendor lines of C57BL/6N mice and embryonic stem cells, and confounds ocular induced mutant phenotypes. Invest Ophthalmol Vis Sci. 2012;53:2921–2927. doi: 10.1167/iovs.12-9662.
    1. Ramkumar HL, Tuo J, de Shen F, Zhang J, Cao X, Chew EY, Chan CC. Nutrient supplementation with n3 polyunsaturated fatty acids, lutein, and zeaxanthin decrease A2E accumulation and VEGF expression in the retinas of Ccl2/Cx3cr1-deficient mice on Crb1rd8 background. J Nutr. 2013;143:1129–1135. doi: 10.3945/jn.112.169649.
    1. Zhang J, Tuo J, Cao X, Shen D, Li W, Chan CC. Early degeneration of photoreceptor synapse in Ccl2/Cx3cr1-deficient mice on Crb1(rd8) background. Synapse. 2013;67:515–531. doi: 10.1002/syn.21674.
    1. Luhmann UF, Robbie S, Munro PM, Barker SE, Duran Y, Luong V, Fitzke FW, Bainbridge JW, Ali RR, MacLaren RE. The drusenlike phenotype in aging Ccl2-knockout mice is caused by an accelerated accumulation of swollen autofluorescent subretinal macrophages. Invest Ophthalmol Vis Sci. 2009;50:5934–5943. doi: 10.1167/iovs.09-3462.
    1. Chen M, Forrester JV, Xu H. Dysregulation in retinal para-inflammation and age-related retinal degeneration in CCL2 or CCR2 deficient mice. PLoS One. 2011;6:e22818. doi: 10.1371/journal.pone.0022818.
    1. Despriet DD, Bergen AA, Merriam JE, Zernant J, Barile GR, Smith RT, Barbazetto IA, van Soest S, Bakker A, de Jong PT, Allikmets R, Klaver CC. Comprehensive analysis of the candidate genes CCL2, CCR2, and TLR4 in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2008;49:364–371. doi: 10.1167/iovs.07-0656.
    1. Lentsch AB, Ward PA. Regulation of inflammatory vascular damage. J Pathol. 2000;190:343–348. doi: 10.1002/(SICI)1096-9896(200002)190:3<343::AID-PATH522>;2-M.
    1. Blann AD. Endothelial cell activation, injury, damage and dysfunction: separate entities or mutual terms? Blood Coagul Fibrinolysis. 2000;11:623–630. doi: 10.1097/00001721-200010000-00006.
    1. Sprague AH, Khalil RA. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol. 2009;78:539–552. doi: 10.1016/j.bcp.2009.04.029.
    1. Weber C, Fraemohs L, Dejana E. The role of junctional adhesion molecules in vascular inflammation. Nat Rev Immunol. 2007;7:467–477. doi: 10.1038/nri2096.
    1. Rather LJ. Disturbance of function (functio laesa): the legendary fifth cardinal sign of inflammation, added by Galen to the four cardinal signs of Celsus. Bull N Y Acad Med. 1971;47:303–322.
    1. Mitchinson MJ. Fluor: another cardinal sign of inflammation. Lancet. 1989;2:1520. doi: 10.1016/S0140-6736(89)92957-7.
    1. Baggiolini M, Loetscher P, Moser B. Interleukin-8 and the chemokine family. Int J Immunopharmacol. 1995;17:103–108. doi: 10.1016/0192-0561(94)00088-6.
    1. Kobayashi Y. The role of chemokines in neutrophil biology. Front Biosci. 2008;13:2400–2407. doi: 10.2741/2853.
    1. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535. doi: 10.1126/science.1092385.
    1. Mayadas TN, Cullere X, Lowell CA. The multifaceted functions of neutrophils. Annu Rev Pathol. 2014;9:181–218. doi: 10.1146/annurev-pathol-020712-164023.
    1. Cui BB, Tan CY, Schorn C, Tang HH, Liu Y, Zhao Y. Neutrophil extracellular traps in sterile inflammation: the story after dying? Autoimmunity. 2012;45:593–596. doi: 10.3109/08916934.2012.719952.
    1. Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011;11:519–531. doi: 10.1038/nri3024.
    1. Ohki Y, Heissig B, Sato Y, Akiyama H, Zhu Z, Hicklin DJ, Shimada K, Ogawa H, Daida H, Hattori K, Ohsaka A. Granulocyte colony-stimulating factor promotes neovascularization by releasing vascular endothelial growth factor from neutrophils. FASEB J. 2005;19:2005–2007.
    1. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol. 2011;11:762–774. doi: 10.1038/nri3070.
    1. Swirski FK, Robbins CS. Neutrophils usher monocytes into sites of inflammation. Circ Res. 2013;112:744–745. doi: 10.1161/CIRCRESAHA.113.300867.
    1. Shi Y, Zheng W, Rock KL. Cell injury releases endogenous adjuvants that stimulate cytotoxic T cell responses. Proc Natl Acad Sci USA. 2000;97:14590–14595. doi: 10.1073/pnas.260497597.
    1. Shi Y, Rock KL. Cell death releases endogenous adjuvants that selectively enhance immune surveillance of particulate antigens. Eur J Immunol. 2002;32:155–162. doi: 10.1002/1521-4141(200201)32:1<155::AID-IMMU155>;2-P.
    1. Pawelec G, Goldeck D, Derhovanessian E. Inflammation, ageing and chronic disease. Curr Opin Immunol. 2014;29C:23–28. doi: 10.1016/j.coi.2014.03.007.
    1. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448.
    1. Mosser DM. The many faces of macrophage activation. J Leukoc Biol. 2003;73:209–212. doi: 10.1189/jlb.0602325.
    1. Fernandez-Velasco M, Gonzalez-Ramos S, Bosca L. Involvement of monocytes/macrophages as key factors in the development and progression of cardiovascular diseases. Biochem J. 2014;458:187–193. doi: 10.1042/BJ20131501.
    1. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32:593–604. doi: 10.1016/j.immuni.2010.05.007.
    1. Zhao H, Roychoudhury J, Doggett TA, Apte RS, Ferguson TA. Age-dependent changes in FasL (CD95L) modulate macrophage function in a model of age-related macular degeneration. Invest Ophthalmol Vis Sci. 2013;54:5321–5331. doi: 10.1167/iovs.13-12122.
    1. Edwards JP, Zhang X, Frauwirth KA, Mosser DM. Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol. 2006;80:1298–1307. doi: 10.1189/jlb.0406249.
    1. Vallejo AN. CD28 extinction in human T cells: altered functions and the program of T-cell senescence. Immunol Rev. 2005;205:158–169. doi: 10.1111/j.0105-2896.2005.00256.x.
    1. Frasca D, Blomberg BB. Aging affects human B cell responses. J Clin Immunol. 2011;31:430–435. doi: 10.1007/s10875-010-9501-7.
    1. Aspinall R, Lapenna A, B-Lynch C, Lang PO. Cellular signalling pathways in immune aging and regeneration. Biochem Soc Trans. 2014;42:651–656. doi: 10.1042/BST20140021.
    1. Lemster BH, Michel JJ, Montag DT, Paat JJ, Studenski SA, Newman AB, Vallejo AN. Induction of CD56 and TCR-independent activation of T cells with aging. J Immunol. 2008;180:1979–1990. doi: 10.4049/jimmunol.180.3.1979.
    1. Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol. 2013;13:875–887. doi: 10.1038/nri3547.
    1. Aprahamian T, Takemura Y, Goukassian D, Walsh K. Ageing is associated with diminished apoptotic cell clearance in vivo. Clin Exp Immunol. 2008;152:448–455. doi: 10.1111/j.1365-2249.2008.03658.x.
    1. Bird AC, Bressler NM, Bressler SB, Chisholm IH, Coscas G, Davis MD, de Jong PT, Klaver CC, Klein BE, Klein R, The International ARM Epidemiological Study Group An international classification and grading system for age-related maculopathy and age-related macular degeneration. Surv Ophthalmol. 1995;39:367–374. doi: 10.1016/S0039-6257(05)80092-X.
    1. Sallo FB, Peto T, Leung I, Xing W, Bunce C, Bird AC. The International Classification system and the progression of age-related macular degeneration. Curr Eye Res. 2009;34:238–240. doi: 10.1080/02713680802714058.
    1. Fraccaro P, Nicolo M, Bonetto M, Giacomini M, Weller P, Traverso CE, Prosperi M, O Sullivan D. Combining macula clinical signs and patient characteristics for age-related macular degeneration diagnosis: a machine learning approach. BMC Ophthalmol. 2015;15:10. doi: 10.1186/1471-2415-15-10.
    1. Mullins RF, Russell SR, Anderson DH, Hageman GS. Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease. FASEB J. 2000;14:835–846.
    1. Ohno-Matsui K. Parallel findings in age-related macular degeneration and Alzheimer’s disease. Prog Retin Eye Res. 2011;30:217–238. doi: 10.1016/j.preteyeres.2011.02.004.
    1. Burns RP, Feeney-Burns L. Clinico-morphologic correlations of drusen of Bruch’s membrane. Trans Am Ophthalmol Soc. 1980;78:206–225.
    1. Ishibashi T, Patterson R, Ohnishi Y, Inomata H, Ryan SJ. Formation of drusen in the human eye. Am J Ophthalmol. 1986;101:342–353. doi: 10.1016/0002-9394(86)90830-5.
    1. Hageman GS, Mullins RF, Russell SR, Johnson LV, Anderson DH. Vitronectin is a constituent of ocular drusen and the vitronectin gene is expressed in human retinal pigmented epithelial cells. FASEB J. 1999;13:477–484.
    1. Ozaki S, Johnson LV, Mullins RF, Hageman GS, Anderson DH. The human retina and retinal pigment epithelium are abundant sources of vitronectin mRNA. Biochem Biophys Res Commun. 1999;258:524–529. doi: 10.1006/bbrc.1999.0672.
    1. Anderson DH, Mullins RF, Hageman GS, Johnson LV. A role for local inflammation in the formation of drusen in the aging eye. Am J Ophthalmol. 2002;134:411–431. doi: 10.1016/S0002-9394(02)01624-0.
    1. Crabb JW, Miyagi M, Gu X, Shadrach K, West KA, Sakaguchi H, Kamei M, Hasan A, Yan L, Rayborn ME, Salomon RG, Hollyfield JG. Drusen proteome analysis: an approach to the etiology of age-related macular degeneration. Proc Natl Acad Sci USA. 2002;99:14682–14687. doi: 10.1073/pnas.222551899.
    1. Sakaguchi H, Miyagi M, Shadrach KG, Rayborn ME, Crabb JW, Hollyfield JG. Clusterin is present in drusen in age-related macular degeneration. Exp Eye Res. 2002;74:547–549. doi: 10.1006/exer.2002.1186.
    1. Anderson DH, Talaga KC, Rivest AJ, Barron E, Hageman GS, Johnson LV (2004) Characterization of beta amyloid assemblies in drusen: the deposits associated with aging and age-related macular degeneration. Exp Eye Res 78:243–256. pii:S0014483503003440
    1. An E, Lu X, Flippin J, Devaney JM, Halligan B, Hoffman EP, Strunnikova N, Csaky K, Hathout Y. Secreted proteome profiling in human RPE cell cultures derived from donors with age related macular degeneration and age matched healthy donors. J Proteome Res. 2006;5:2599–2610. doi: 10.1021/pr060121j.
    1. Johnson LV, Forest DL, Banna CD, Radeke CM, Maloney MA, Hu J, Spencer CN, Walker AM, Tsie MS, Bok D, Radeke MJ, Anderson DH. Cell culture model that mimics drusen formation and triggers complement activation associated with age-related macular degeneration. Proc Natl Acad Sci USA. 2011;108:18277–18282. doi: 10.1073/pnas.1109703108.
    1. Ebrahimi KB, Handa JT. Lipids, lipoproteins, and age-related macular degeneration. J Lipids. 2011;2011:802059. doi: 10.1155/2011/802059.
    1. Wang AL, Lukas TJ, Yuan M, Du N, Tso MO, Neufeld AH. Autophagy and exosomes in the aged retinal pigment epithelium: possible relevance to drusen formation and age-related macular degeneration. PLoS One. 2009;4:e4160. doi: 10.1371/journal.pone.0004160.
    1. Krohne TU, Holz FG, Kopitz J. Apical-to-basolateral transcytosis of photoreceptor outer segments induced by lipid peroxidation products in human retinal pigment epithelial cells. Invest Ophthalmol Vis Sci. 2010;51:553–560. doi: 10.1167/iovs.09-3755.
    1. Hageman GS, Luthert PJ, Victor Chong NH, Johnson LV, Anderson DH, Mullins RF. 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. 2001;20:705–732. doi: 10.1016/S1350-9462(01)00010-6.
    1. Johnson LV, Ozaki S, Staples MK, Erickson PA, Anderson DH. A potential role for immune complex pathogenesis in drusen formation. Exp Eye Res. 2000;70:441–449. doi: 10.1006/exer.1999.0798.
    1. Johnson LV, Leitner WP, Staples MK, Anderson DH. Complement activation and inflammatory processes in Drusen formation and age related macular degeneration. Exp Eye Res. 2001;73:887–896. doi: 10.1006/exer.2001.1094.
    1. Johnson LV, Leitner WP, Rivest AJ, Staples MK, Radeke MJ, Anderson DH. The Alzheimer’s A beta -peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration. Proc Natl Acad Sci USA. 2002;99:11830–11835. doi: 10.1073/pnas.192203399.
    1. Anderson DH, Radeke MJ, Gallo NB, Chapin EA, Johnson PT, Curletti CR, Hancox LS, Hu J, Ebright JN, Malek G, Hauser MA, Rickman CB, Bok D, Hageman GS, Johnson LV. The pivotal role of the complement system in aging and age-related macular degeneration: hypothesis re-visited. Prog Retin Eye Res. 2010;29:95–112. doi: 10.1016/j.preteyeres.2009.11.003.
    1. Gehrs KM, Jackson JR, Brown EN, Allikmets R, Hageman GS. Complement, age-related macular degeneration and a vision of the future. Arch Ophthalmol. 2010;128:349–358. doi: 10.1001/archophthalmol.2010.18.
    1. Liu C, Cao L, Yang S, Xu L, Liu P, Wang F, Xu D. Subretinal injection of amyloid-beta peptide accelerates RPE cell senescence and retinal degeneration. Int J Mol Med. 2015;35:169–176.
    1. Chen L, Yang P, Kijlstra A. Distribution, markers, and functions of retinal microglia. Ocul Immunol Inflamm. 2002;10:27–39. doi: 10.1076/ocii.10.1.27.10328.
    1. Xu H, Chen M, Manivannan A, Lois N, Forrester JV. Age-dependent accumulation of lipofuscin in perivascular and subretinal microglia in experimental mice. Aging Cell. 2008;7:58–68. doi: 10.1111/j.1474-9726.2007.00351.x.
    1. Griffith TS, Brunner T, Fletcher SM, Green DR, Ferguson TA. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science. 1995;270:1189–1192. doi: 10.1126/science.270.5239.1189.
    1. Griffith TS, Yu X, Herndon JM, Green DR, Ferguson TA. CD95-induced apoptosis of lymphocytes in an immune privileged site induces immunological tolerance. Immunity. 1996;5:7–16. doi: 10.1016/S1074-7613(00)80305-2.
    1. Elzey BD, Griffith TS, Herndon JM, Barreiro R, Tschopp J, Ferguson TA. Regulation of Fas ligand-induced apoptosis by TNF. J Immunol. 2001;167:3049–3056. doi: 10.4049/jimmunol.167.6.3049.
    1. Ferguson TA, Green DR. Fas-ligand and immune privilege: the eyes have it. Cell Death Differ. 2001;8:771–772. doi: 10.1038/sj.cdd.4400891.
    1. Roychoudhury J, Herndon JM, Yin J, Apte RS, Ferguson TA. Targeting immune privilege to prevent pathogenic neovascularization. Invest Ophthalmol Vis Sci. 2010;51:3560–3566. doi: 10.1167/iovs.09-3890.
    1. Ardeljan D, Chan CC. Aging is not a disease: distinguishing age-related macular degeneration from aging. Prog Retin Eye Res. 2013;37:68–69. doi: 10.1016/j.preteyeres.2013.07.003.
    1. Nita M, Grzybowski A, Ascaso FJ, Huerva V. Age-related macular degeneration in the aspect of chronic low-grade inflammation (pathophysiological parainflammation) Mediators Inflamm. 2014;2014:930671. doi: 10.1155/2014/930671.
    1. Wu WK, Llewellyn OP, Bates DO, Nicholson LB, Dick AD. IL-10 regulation of macrophage VEGF production is dependent on macrophage polarisation and hypoxia. Immunobiology. 2010;215:796–803. doi: 10.1016/j.imbio.2010.05.025.
    1. Faber C, Singh A, Kruger Falk M, Juel HB, Sorensen TL, Nissen MH. Age-related macular degeneration is associated with increased proportion of CD56(+) T cells in peripheral blood. Ophthalmology. 2013;120:2310–2316. doi: 10.1016/j.ophtha.2013.04.014.
    1. Koreck A, Suranyi A, Szony BJ, Farkas A, Bata-Csorgo Z, Kemeny L, Dobozy A. CD3+CD56+NK T cells are significantly decreased in the peripheral blood of patients with psoriasis. Clin Exp Immunol. 2002;127:176–182. doi: 10.1046/j.1365-2249.2002.01721.x.
    1. Ahn JK, Chung H, Lee DS, Yu YS, Yu HG. CD8brightCD56+ T cells are cytotoxic effectors in patients with active Behcet’s uveitis. J Immunol. 2005;175:6133–6142. doi: 10.4049/jimmunol.175.9.6133.
    1. Michel JJ, Turesson C, Lemster B, Atkins SR, Iclozan C, Bongartz T, Wasko MC, Matteson EL, Vallejo AN. CD56-expressing T cells that have features of senescence are expanded in rheumatoid arthritis. Arthritis Rheum. 2007;56:43–57. doi: 10.1002/art.22310.
    1. Li WX, Pan HF, Hu JL, Wang CZ, Zhang N, Li J, Li XP, Xu JH, Ye DQ. Assay of T- and NK-cell subsets and the expression of NKG2A and NKG2D in patients with new-onset systemic lupus erythematosus. Clin Rheumatol. 2010;29:315–323. doi: 10.1007/s10067-009-1322-9.
    1. Penfold PL, Provis JM, Furby JH, Gatenby PA, Billson FA. Autoantibodies to retinal astrocytes associated with age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol. 1990;228:270–274. doi: 10.1007/BF00920033.
    1. Gu X, Meer SG, Miyagi M, Rayborn ME, Hollyfield JG, Crabb JW, Salomon RG. Carboxyethylpyrrole protein adducts and autoantibodies, biomarkers for age-related macular degeneration. J Biol Chem. 2003;278:42027–42035. doi: 10.1074/jbc.M305460200.
    1. Patel N, Ohbayashi M, Nugent AK, Ramchand K, Toda M, Chau KY, Bunce C, Webster A, Bird AC, Ono SJ, Chong V. Circulating anti-retinal antibodies as immune markers in age-related macular degeneration. Immunology. 2005;115:422–430. doi: 10.1111/j.1365-2567.2005.02173.x.
    1. Dhillon B, Wright AF, Tufail A, Pappworth I, Hayward C, Moore I, Strain L, Kavanagh D, Barlow PN, Herbert AP, Schmidt CQ, Armbrecht AM, Laude A, Deary IJ, Staniforth SJ, Holmes LV, Goodship TH, Marchbank KJ. Complement factor H autoantibodies and age-related macular degeneration. Invest Ophthalmol Vis Sci. 2010;51:5858–5863. doi: 10.1167/iovs.09-5124.
    1. Morohoshi K, Ohbayashi M, Patel N, Chong V, Bird AC, Ono SJ. Identification of anti-retinal antibodies in patients with age-related macular degeneration. Exp Mol Pathol. 2012;93:193–199. doi: 10.1016/j.yexmp.2012.03.007.
    1. Camelo S. Potential sources and roles of adaptive immunity in age-related macular degeneration: Shall we rename AMD into autoimmune macular disease? Autoimmune Dis. 2014;2014:532487.
    1. Bergstrom I, Backteman K, Lundberg A, Ernerudh J, Jonasson L. Persistent accumulation of interferon-gamma-producing CD8+CD56+ T cells in blood from patients with coronary artery disease. Atherosclerosis. 2012;224:515–520. doi: 10.1016/j.atherosclerosis.2012.07.033.
    1. Klein R, Cruickshanks KJ, Myers CE, Sivakumaran TA, Iyengar SK, Meuer SM, Schubert CR, Gangnon RE, Klein BE. The relationship of atherosclerosis to the 10-year cumulative incidence of age-related macular degeneration: the Beaver Dam studies. Ophthalmology. 2013;120:1012–1019. doi: 10.1016/j.ophtha.2012.11.003.
    1. Guymer RH, Tao LW, Goh JK, Liew D, Ischenko O, Robman LD, Aung K, Cipriani T, Cain M, Richardson AJ, Baird PN, Langham R. Identification of urinary biomarkers for age-related macular degeneration. Invest Ophthalmol Vis Sci. 2011;52:4639–4644. doi: 10.1167/iovs.10-7120.
    1. Reynolds R, Hartnett ME, Atkinson JP, Giclas PC, Rosner B, Seddon JM. Plasma complement components and activation fragments: associations with age-related macular degeneration genotypes and phenotypes. Invest Ophthalmol Vis Sci. 2009;50:5818–5827. doi: 10.1167/iovs.09-3928.
    1. Hecker LA, Edwards AO, Ryu E, Tosakulwong N, Baratz KH, Brown WL, Charbel Issa P, Scholl HP, Pollok-Kopp B, Schmid-Kubista KE, Bailey KR, Oppermann M. Genetic control of the alternative pathway of complement in humans and age-related macular degeneration. Hum Mol Genet. 2010;19:209–215. doi: 10.1093/hmg/ddp472.
    1. Stanton CM, Yates JR, den Hollander AI, Seddon JM, Swaroop A, Stambolian D, Fauser S, Hoyng C, Yu Y, Atsuhiro K, Branham K, Othman M, Chen W, Kortvely E, Chalmers K, Hayward C, Moore AT, Dhillon B, Ueffing M, Wright AF. Complement factor D in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2011;52:8828–8834. doi: 10.1167/iovs.11-7933.
    1. Silva AS, Teixeira AG, Bavia L, Lin F, Velletri R, Belfort R, Jr, Isaac L. Plasma levels of complement proteins from the alternative pathway in patients with age-related macular degeneration are independent of Complement Factor H Tyr(4)(0)(2)His polymorphism. Mol Vis. 2012;18:2288–2299.
    1. Haas P, Aggermann T, Nagl M, Steindl-Kuscher K, Krugluger W, Binder S. Implication of CD21, CD35, and CD55 in the pathogenesis of age-related macular degeneration. Am J Ophthalmol. 2011;152(396–399):e1.
    1. Singh A, Faber C, Falk M, Nissen MH, Hviid TV, Sorensen TL. Altered expression of CD46 and CD59 on leukocytes in neovascular age-related macular degeneration. Am J Ophthalmol. 2012;154(193–199):e2.
    1. Cipriani V, Matharu BK, Khan JC, Shahid H, Stanton CM, Hayward C, Wright AF, Bunce C, Clayton DG, Moore AT, Yates JR. Genetic variation in complement regulators and susceptibility to age-related macular degeneration. Immunobiology. 2012;217:158–161. doi: 10.1016/j.imbio.2011.09.002.
    1. Cao S, Ko A, Partanen M, Pakzad-Vaezi K, Merkur AB, Albiani DA, Kirker AW, Wang A, Cui JZ, Forooghian F, Matsubara JA. Relationship between systemic cytokines and complement factor H Y402H polymorphism in patients with dry age-related macular degeneration. Am J Ophthalmol. 2013;156:1176–1183. doi: 10.1016/j.ajo.2013.08.003.
    1. An E, Gordish-Dressman H, Hathout Y. Effect of TNF-alpha on human ARPE-19-secreted proteins. Mol Vis. 2008;14:2292–2303.
    1. Leung KW, Barnstable CJ, Tombran-Tink J. Bacterial endotoxin activates retinal pigment epithelial cells and induces their degeneration through IL-6 and IL-8 autocrine signaling. Mol Immunol. 2009;46:1374–1386. doi: 10.1016/j.molimm.2008.12.001.
    1. Lee IT, Liu SW, Chi PL, Lin CC, Hsiao LD, Yang CM. TNF-alpha mediates PKCdelta/JNK1/2/c-Jun-dependent monocyte adhesion via ICAM-1 induction in human retinal pigment epithelial cells. PLoS One. 2015;10:e0117911. doi: 10.1371/journal.pone.0117911.
    1. Oh H, Takagi H, Takagi C, Suzuma K, Otani A, Ishida K, Matsumura M, Ogura Y, Honda Y. The potential angiogenic role of macrophages in the formation of choroidal neovascular membranes. Invest Ophthalmol Vis Sci. 1999;40:1891–1898.
    1. Cousins SW, Espinosa-Heidmann DG, Csaky KG. Monocyte activation in patients with age-related macular degeneration: a biomarker of risk for choroidal neovascularization? Arch Ophthalmol. 2004;122:1013–1018. doi: 10.1001/archopht.122.7.1013.
    1. Liu B, Faia L, Hu M, Nussenblatt RB. Pro-angiogenic effect of IFNgamma is dependent on the PI3K/mTOR/translational pathway in human retinal pigmented epithelial cells. Mol Vis. 2010;16:184–193.
    1. Juel HB, Faber C, Udsen MS, Folkersen L, Nissen MH. Chemokine expression in retinal pigment epithelial ARPE-19 cells in response to coculture with activated T cells. Invest Ophthalmol Vis Sci. 2012;53:8472–8480. doi: 10.1167/iovs.12-9963.
    1. Faber C, Jehs T, Juel HB, Singh A, Falk MK, Sorensen TL, Nissen MH. Early and exudative age-related macular degeneration is associated with increased plasma levels of soluble TNF receptor II. Acta Ophthalmol. 2015;93:242–247. doi: 10.1111/aos.12581.
    1. Chau KY, Sivaprasad S, Patel N, Donaldson TA, Luthert PJ, Chong NV. Plasma levels of matrix metalloproteinase-2 and -9 (MMP-2 and MMP-9) in age-related macular degeneration. Eye (Lond) 2008;22:855–859. doi: 10.1038/sj.eye.6702722.
    1. Seddon JM, George S, Rosner B, Rifai N. Progression of age-related macular degeneration: prospective assessment of C-reactive protein, interleukin 6, and other cardiovascular biomarkers. Arch Ophthalmol. 2005;123:774–782. doi: 10.1001/archopht.123.6.774.
    1. Mooijaart SP, Koeijvoets KM, Sijbrands EJ, Daha MR, Westendorp RG. Complement Factor H polymorphism Y402H associates with inflammation, visual acuity, and cardiovascular mortality in the elderly population at large. Exp Gerontol. 2007;42:1116–1122. doi: 10.1016/j.exger.2007.08.001.
    1. Klein R, Myers CE, Cruickshanks KJ, Gangnon RE, Danforth LG, Sivakumaran TA, Iyengar SK, Tsai MY, Klein BE. Markers of inflammation, oxidative stress, and endothelial dysfunction and the 20-year cumulative incidence of early age-related macular degeneration: the Beaver Dam Eye Study. JAMA Ophthalmol. 2014;132:446–455. doi: 10.1001/jamaophthalmol.2013.7671.
    1. Mo FM, Proia AD, Johnson WH, Cyr D, Lashkari K. Interferon gamma-inducible protein-10 (IP-10) and eotaxin as biomarkers in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2010;51:4226–4236. doi: 10.1167/iovs.09-3910.
    1. Hong T, Tan AG, Mitchell P, Wang JJ. A review and meta-analysis of the association between C-reactive protein and age-related macular degeneration. Surv Ophthalmol. 2011;56:184–194. doi: 10.1016/j.survophthal.2010.08.007.
    1. Mitta VP, Christen WG, Glynn RJ, Semba RD, Ridker PM, Rimm EB, Hankinson SE, Schaumberg DA. C-reactive protein and the incidence of macular degeneration: pooled analysis of 5 cohorts. JAMA Ophthalmol. 2013;131:507–513. doi: 10.1001/jamaophthalmol.2013.2303.
    1. Yip JL, Khawaja AP, Chan MP, Broadway DC, Peto T, Tufail A, Luben R, Hayat S, Bhaniani A, Wareham NJ, Khaw KT, Foster PJ. Cross sectional and longitudinal associations between cardiovascular risk factors and age related macular degeneration in the EPIC-Norfolk eye study. PLoS One. 2015;10:e0132565. doi: 10.1371/journal.pone.0132565.
    1. Smith W, Mitchell P, Leeder SR, Wang JJ. Plasma fibrinogen levels, other cardiovascular risk factors, and age-related maculopathy: the Blue Mountains Eye Study. Arch Ophthalmol. 1998;116:583–587. doi: 10.1001/archopht.116.5.583.
    1. Schaumberg DA, Christen WG, Buring JE, Glynn RJ, Rifai N, Ridker PM. High-sensitivity C-reactive protein, other markers of inflammation, and the incidence of macular degeneration in women. Arch Ophthalmol. 2007;125:300–305. doi: 10.1001/archopht.125.3.300.
    1. Jiang S, Moriarty-Craige SE, Li C, Lynn MJ, Cai J, Jones DP, Sternberg P. Associations of plasma-soluble fas ligand with aging and age-related macular degeneration. Invest Ophthalmol Vis Sci. 2008;49:1345–1349. doi: 10.1167/iovs.07-0308.
    1. Sharma NK, Prabhakar S, Gupta A, Singh R, Gupta PK, Gupta PK, Anand A. New biomarker for neovascular age-related macular degeneration: eotaxin-2. DNA Cell Biol. 2012;31:1618–1627. doi: 10.1089/dna.2012.1786.
    1. Juel HB, Faber C, Munthe-Fog L, Bastrup-Birk S, Reese-Petersen AL, Falk MK, Singh A, Sorensen TL, Garred P, Nissen MH. Systemic and ocular long Pentraxin 3 in patients with age-related macular degeneration. PLoS One. 2015;10:e0132800. doi: 10.1371/journal.pone.0132800.

Source: PubMed

3
Abonner