Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Immunology of age-related macular degeneration

Key Points

  • Age-related macular degeneration (AMD) is the leading cause of blindness in elderly individuals in the developed world. It features a progressive deterioration of the central retina. Numerous, sometimes contradictory, pro-inflammatory signals and pathways have been implicated in disease initiation and progression.

  • In the healthy retina, ocular immune-based surveillance has an essential role in maintaining visual homeostasis.

  • As the retina ages and deteriorates, diverse pro-inflammatory agonists induce improper local immune activation. Diverse immune pathways including complement, the inflammasome, Toll-like receptor activation, adaptive immunity and others are implicated in driving retinal damage in AMD.

  • Extraocular immune cell recruitment mediates pathological angiogenesis in neovascular AMD, which is a prevalent condition that is defined by the growth of unwanted, abnormal blood vessels in the normally avascular macular area.

  • Numerous completed and ongoing immune-based clinical trials for AMD have been unanimously unsuccessful. This may be the result of our collective lack of understanding and appreciation of the 'friend–foe' relationship between retinal degeneration and immunity.

Abstract

Age-related macular degeneration (AMD) is a leading cause of blindness in aged individuals. Recent advances have highlighted the essential role of immune processes in the development, progression and treatment of AMD. In this Review we discuss recent discoveries related to the immunological aspects of AMD pathogenesis. We outline the diverse immune cell types, inflammatory activators and pathways that are involved. Finally, we discuss the future of inflammation-directed therapeutics to treat AMD in the growing aged population.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Retinal anatomy in health and disease.
Figure 2: An integrated model of AMD pathogenesis.

Similar content being viewed by others

References

  1. The Global Economic Cost of Visual Impairment. AMD Alliance International [online], http://www.icoph.org/resources/146/The-Global-Economic-Cost-of-Visual-Impairment.html (2010).

  2. Haines, J. L. et al. Complement factor H variant increases the risk of age-related macular degeneration. Science 308, 419–421 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Edwards, A. O. et al. Complement factor H polymorphism and age-related macular degeneration. Science 308, 421–424 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Klein, R. J. et al. Complement factor H polymorphism in age-related macular degeneration. Science 308, 385–389 (2005). References 2–4 are landmark studies showing increased statistical risk of AMD in individuals who have a single CFH polymorphism; these studies were the first of their kind for a complex human disease.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hageman, G. S. et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc. Natl Acad. Sci. USA 102, 7227–7232 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Tuo, J., Grob, S., Zhang, K. & Chan, C. C. Genetics of immunological and inflammatory components in age-related macular degeneration. Ocul. Immunol. Inflamm. 20, 27–36 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Richards, A., Kavanagh, D. & Atkinson, J. P. Inherited complement regulatory protein deficiency predisposes to human disease in acute injury and chronic inflammatory statesthe examples of vascular damage in atypical hemolytic uremic syndrome and debris accumulation in age-related macular degeneration. Adv. Immunol. 96, 141–177 (2007).

    Article  CAS  PubMed  Google Scholar 

  8. Khandhadia, S., Cipriani, V., Yates, J. R. & Lotery, A. J. Age-related macular degeneration and the complement system. Immunobiology 217, 127–146 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Anderson, D. H. et al. The pivotal role of the complement system in aging and age-related macular degeneration: hypothesis re-visited. Prog. Retin. Eye Res. 29, 95–112 (2010). References 6–9 provide a comprehensive review and analysis of the role of the complement system in AMD.

    Article  CAS  PubMed  Google Scholar 

  10. Saint-Geniez, M. et al. Endogenous VEGF is required for visual function: evidence for a survival role on müller cells and photoreceptors. PLoS ONE 3, e3554 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bill, A., Sperber, G. & Ujiie, K. Physiology of the choroidal vascular bed. Int. Ophthalmol. 6, 101–107 (1983).

    Article  CAS  PubMed  Google Scholar 

  12. Hageman, G. S. et al. An integrated hypothesis that considers drusen as biomarkers of immune-mediated processes at the RPE — Bruch's membrane interface in aging and age-related macular degeneration. Prog. Retin. Eye Res. 20, 705–732 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Friedman, D. S. et al. Prevalence of age-related macular degeneration in the United States. Arch. Ophthalmol. 122, 564–572 (2004).

    Article  PubMed  Google Scholar 

  14. Rosenfeld, P. J. et al. Ranibizumab for neovascular age-related macular degeneration. N. Engl. J. Med. 355, 1419–1431 (2006).

    Article  CAS  Google Scholar 

  15. Brown, D. M. et al. Ranibizumab versus verteporfin for neovascular age-related macular degeneration. N. Engl. J. Med. 355, 1432–1444 (2006). References 14 and 15 report Phase III clinical trials showing the efficacy of VEGFA-targeted antibody therapy for neovascular AMD, which is the standard of care for this disease.

    Article  CAS  PubMed  Google Scholar 

  16. Bird, A. C. Therapeutic targets in age-related macular disease. J. Clin. Invest. 120, 3033–3041 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sunness, J. S. et al. Enlargement of atrophy and visual acuity loss in the geographic atrophy form of age-related macular degeneration. Ophthalmology 106, 1768–1779 (1999).

    Article  CAS  PubMed  Google Scholar 

  18. Martin, D. F. et al. Ranibizumab and bevacizumab for treatment of neovascular age-related macular degeneration: two-year results. Ophthalmology 119, 1388–1398 (2012).

    Article  PubMed  Google Scholar 

  19. Marneros, A. G. et al. Vascular endothelial growth factor expression in the retinal pigment epithelium is essential for choriocapillaris development and visual function. Am. J. Pathol. 167, 1451–1459 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sepp, T. et al. Complement factor H variant Y402H is a major risk determinant for geographic atrophy and choroidal neovascularization in smokers and nonsmokers. Invest. Ophthalmol. Visual Sci. 47, 536–540 (2006).

    Article  Google Scholar 

  21. Sofat, R. et al. Complement factor H genetic variant and age-related macular degeneration: effect size, modifiers and relationship to disease subtype. Int. J. Epidemiol. 41, 250–262 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Droz, I. et al. Genotype-phenotype correlation of age-related macular degeneration: influence of complement factor H polymorphism. Br. J. Ophthalmol. 92, 513–517 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Magnusson, K. P. et al. CFH Y402H confers similar risk of soft drusen and both forms of advanced AMD. PLoS Med. 3, e5 (2006).

    Article  CAS  PubMed  Google Scholar 

  24. Cameron, D. J. et al. HTRA1 variant confers similar risks to geographic atrophy and neovascular age-related macular degeneration. Cell Cycle 6, 1122–1125 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Sobrin, L. et al. Heritability and genome-wide association study to assess genetic differences between advanced age-related macular degeneration subtypes. Ophthalmology 119, 1874–1875 (2012).

    Article  PubMed  Google Scholar 

  26. Young, R. W. Pathophysiology of age-related macular degeneration. Surv. Ophthalmol. 31, 291–306 (1987).

    Article  CAS  PubMed  Google Scholar 

  27. Ambati, J., Ambati, B. K., Yoo, S. H., Ianchulev, S. & Adamis, A. P. Age-related macular degeneration: etiology, pathogenesis, and therapeutic strategies. Surv. Ophthalmol. 48, 257–293 (2003).

    Article  PubMed  Google Scholar 

  28. Sugita, S. Role of ocular pigment epithelial cells in immune privilege. Arch. Immunol. Ther. Exp. (Warsz.) 57, 263–268 (2009).

    Article  Google Scholar 

  29. Streilein, J. W. Immunological non-responsiveness and acquisition of tolerance in relation to immune privilege in the eye. Eye (Lond.) 9, 236–240 (1995).

    Article  Google Scholar 

  30. Morohoshi, K., Goodwin, A. M., Ohbayashi, M. & Ono, S. J. Autoimmunity in retinal degeneration: autoimmune retinopathy and age-related macular degeneration. J. Autoimmun. 33, 247–254 (2009).

    Article  CAS  PubMed  Google Scholar 

  31. Xu, H., Chen, M., Manivannan, A., Lois, N. & Forrester, J. V. Age-dependent accumulation of lipofuscin in perivascular and subretinal microglia in experimental mice. Aging Cell 7, 58–68 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Ma, W., Zhao, L., Fontainhas, A. M., Fariss, R. N. & Wong, W. T. Microglia in the mouse retina alter the structure and function of retinal pigmented epithelial cells: a potential cellular interaction relevant to AMD. PLoS ONE 4, e7945 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chen, M., Forrester, J. V. & Xu, H. Dysregulation in retinal para-inflammation and age-related retinal degeneration in CCL2 or CCR2 deficient mice. PLoS ONE 6, e22818 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tuo, J. et al. Murine ccl2/cx3cr1 deficiency results in retinal lesions mimicking human age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 48, 3827–3836 (2007).

    Article  PubMed  Google Scholar 

  35. Xu, H., Chen, M. & Forrester, J. V. Para-inflammation in the aging retina. Prog. Retin. Eye Res. 28, 348–368 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Sakurai, E., Anand, A., Ambati, B. K., van Rooijen, N. & Ambati, J. Macrophage depletion inhibits experimental choroidal neovascularization. Invest. Ophthalmol. Vis. Sci. 44, 3578–3585 (2003).

    Article  PubMed  Google Scholar 

  37. Espinosa-Heidmann, D. G. et al. Macrophage depletion diminishes lesion size and severity in experimental choroidal neovascularization. Invest. Ophthalmol. Vis. Sci. 44, 3586–3592 (2003).

    Article  PubMed  Google Scholar 

  38. Shen, W. Y., Yu, M. J., Barry, C. J., Constable, I. J. & Rakoczy, P. E. Expression of cell adhesion molecules and vascular endothelial growth factor in experimental choroidal neovascularisation in the rat. Br. J. Ophthalmol. 82, 1063–1071 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ambati, J. et al. An animal model of age-related macular degeneration in senescent Ccl-2- or Ccr-2-deficient mice. Nature Med. 9, 1390–1397 (2003). CCL2- and CCR2-deficient mice were the first animal model shown to reproduce numerous features of early, intermediate and neovascular AMD. This study also directly implicates macrophage migration in retinal degeneration.

    Article  CAS  PubMed  Google Scholar 

  40. Combadiere, C. et al. CX3CR1-dependent subretinal microglia cell accumulation is associated with cardinal features of age-related macular degeneration. J. Clin. Invest. 117, 2920–2928 (2007). This study advanced the epidemiological finding that the Thr280Met CX 3 CR1 polymorphism confers AMD risk by identifying dysfunctional microglial migration in patients carrying the 280Met mutation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Ross, R. J. et al. Immunological protein expression profile in Ccl2/Cx3cr1 deficient mice with lesions similar to age-related macular degeneration. Exp. Res. 86, 675–683 (2008).

    CAS  Google Scholar 

  42. Luhmann, U. F. et al. The drusenlike phenotype in aging Ccl2-knockout mice is caused by an accelerated accumulation of swollen autofluorescent subretinal macrophages. Invest. Ophthalmol. Vis. Sci. 50, 5934–5943 (2009).

    Article  PubMed  Google Scholar 

  43. Bruban, J. et al. CCR2/CCL2-mediated inflammation protects photoreceptor cells from amyloid-β-induced apoptosis. Neurobiol. Dis. 42, 55–72 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Raoul, W. et al. Lipid-bloated subretinal microglial cells are at the origin of drusen appearance in CX3CR1-deficient mice. Ophthalmic Res. 40, 115–119 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Tuo, J. et al. The involvement of sequence variation and expression of CX3CR1 in the pathogenesis of age-related macular degeneration. FASEB J. 18, 1297–1299 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Chen, H., Liu, B., Lukas, T. J. & Neufeld, A. H. The aged retinal pigment epithelium/choroid: a potential substratum for the pathogenesis of age-related macular degeneration. PLoS ONE 3, e2339 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Seidler, S., Zimmermann, H. W., Bartneck, M., Trautwein, C. & Tacke, F. Age-dependent alterations of monocyte subsets and monocyte-related chemokine pathways in healthy adults. BMC Immunol. 11, 30 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Liu, J. et al. Relationship between complement membrane attack complex, chemokine (C-C motif) ligand 2 (CCL2) and vascular endothelial growth factor in mouse model of laser-induced choroidal neovascularization. J. Biol. Chem. 286, 20991–21001 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Xie, P. et al. Suppression and regression of choroidal neovascularization in mice by a novel CCR2 antagonist, INCB3344. PLoS ONE 6, e28933 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Raychaudhuri, S. et al. A rare penetrant mutation in CFH confers high risk of age-related macular degeneration. Nature Genet. 43, 1232–1236 (2011). The initial clear-cut description of a rare variant in CFH with high penetrance, in which the functional studies demonstrate a defect as a result of this variant. Presumably, the first of many rare variants in regulators and activators of the alternative complement pathway that predispose to AMD.

    Article  CAS  PubMed  Google Scholar 

  51. Anderson, D. H., Mullins, R. F., Hageman, G. S. & Johnson, L. V. A role for local inflammation in the formation of drusen in the aging eye. Am. J. Ophthalmol. 134, 411–431 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Baudouin, C. et al. Immunohistological study of subretinal membranes in age-related macular degeneration. Jpn. J. Ophthalmol. 36, 443–451 (1992). The histological examination in this study was the first to implicate complement disturbances in AMD, preceding GWASs by 13 years.

    CAS  PubMed  Google Scholar 

  53. Clark, S. J. et al. Impaired binding of the age-related macular degeneration-associated complement factor H 402H allotype to Bruch's membrane in human retina. J. Biol. Chem. 285, 30192–30202 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Sjöberg, A. P. et al. The factor H variant associated with age-related macular degeneration (His-384) and the non-disease-associated form bind differentially to C-reactive protein, fibromodulin, DNA, and necrotic cells. J. Biol. Chem. 282, 10894–10900 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Laine, M. et al. Y402H polymorphism of complement factor H affects binding affinity to C-reactive protein. J. Immunol. 178, 3831–3836 (2007).

    Article  CAS  PubMed  Google Scholar 

  56. Johnson, P. T. et al. Individuals homozygous for the age-related macular degeneration risk-conferring variant of complement factor H have elevated levels of CRP in the choroid. Proc. Natl Acad. Sci. USA 103, 17456–17461 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Hollyfield, J. G. et al. Oxidative damage-induced inflammation initiates age-related macular degeneration. Nature Med. 14, 194–198 (2008).

    Article  CAS  PubMed  Google Scholar 

  58. Doyle, S. L. et al. NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nature Med. 18, 791–798 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Tarallo, V. et al. DICER1 loss and Alu RNA induce age-related macular degeneration via the NLRP3 inflammasome and MyD88. Cell 149, 847–859 (2012). The first study to show that inflammasome activation in RPE is a crucial driver of geographic atrophy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kaneko, H. et al. DICER1 deficit induces Alu RNA toxicity in age-related macular degeneration. Nature 471, 325–330 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Tseng, W. A. et al. NLRP3 inflammasome activation in retinal pigment epithelial cells by lysosomal destabilization: implications for age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 54, 110–120 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kauppinen, A. et al. Oxidative stress activates NLRP3 inflammasomes in ARPE-19 cells-Implications for age-related macular degeneration (AMD). Immunol. Lett. 147, 29–33 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Liu, R. T. et al. Inflammatory mediators induced by amyloid-β in the retina and RPE in vivo: implications for inflammasome activation in age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 54, 2225–2237 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Kleinman, M. E. et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature 452, 591–597 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Kleinman, M. E. et al. Short-interfering RNAs induce retinal degeneration via TLR3 and IRF3. Mol. Ther. 20, 101–108 (2012).

    Article  CAS  PubMed  Google Scholar 

  66. Lavalette, S. et al. Interleukin-1β inhibition prevents choroidal neovascularization and does not exacerbate photoreceptor degeneration. Am. J. Pathol. 178, 2416–2423 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kumar, M. V., Nagineni, C. N., Chin, M. S., Hooks, J. J. & Detrick, B. Innate immunity in the retina: toll-like receptor (TLR) signaling in human retinal pigment epithelial cells. J. Neuroimmunol. 153, 7–15 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Yang, Z. et al. Toll-like receptor 3 and geographic atrophy in age-related macular degeneration. N. Engl. J. Med. 359, 1456–1463 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zareparsi, S. et al. Toll-like receptor 4 variant D299G is associated with susceptibility to age-related macular degeneration. Hum. Mol. Genet. 14, 1449–1455 (2005).

    Article  CAS  PubMed  Google Scholar 

  70. Cho, Y. et al. Toll-like receptor polymorphisms and age-related macular degeneration: replication in three case-control samples. Invest. Ophthalmol. Vis. Sci. 50, 5614–5618 (2009).

    Article  PubMed  Google Scholar 

  71. Shiose, S. et al. Toll-like receptor 3 is required for development of retinopathy caused by impaired all-trans-retinal clearance in mice. J. Biol. Chem. 286, 15543–15555 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. West, X. Z. et al. Oxidative stress induces angiogenesis by activating TLR2 with novel endogenous ligands. Nature 467, 972–976 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Fujimoto, T. et al. Choroidal neovascularization enhanced by Chlamydia pneumoniae via Toll-like receptor 2 in the retinal pigment epithelium. Invest. Ophthalmol. Vis. Sci. 51, 4694–4702 (2010).

    Article  PubMed  Google Scholar 

  74. Patel, N. et al. Circulating anti-retinal antibodies as immune markers in age-related macular degeneration. Immunology 115, 422–430 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Penfold, P. L., Provis, J. M., Furby, J. H., Gatenby, P. A. & Billson, F. A. Autoantibodies to retinal astrocytes associated with age-related macular degeneration. Graefes Arch. Clin. Exp. Ophthalmol. 228, 270–274 (1990). References 57 and 75 provided the first evidence, in mice and humans, that autoantibodies are involved in AMD.

    Article  CAS  PubMed  Google Scholar 

  76. Cherepanoff, S., Mitchell, P., Wang, J. J. & Gillies, M. C. Retinal autoantibody profile in early age-related macular degeneration: preliminary findings from the Blue Mountains Eye Study. Clin. Exp. Ophthalmol. 34, 590–595 (2006).

    Article  PubMed  Google Scholar 

  77. Zhou, J., Jang, Y. P., Kim, S. R. & Sparrow, J. R. Complement activation by photooxidation products of A2E, a lipofuscin constituent of the retinal pigment epithelium. Proc. Natl Acad. Sci. USA 103, 16182–16187 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Fernandes, A. F. et al. Oxidative inactivation of the proteasome in retinal pigment epithelial cells. A potential link between oxidative stress and up-regulation of interleukin-8. J. Biol. Chem. 283, 20745–20753 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Bian, Q. et al. Lutein and zeaxanthin supplementation reduces photooxidative damage and modulates the expression of inflammation-related genes in retinal pigment epithelial cells. Free Radic. Biol. Med. 53, 1298–1307 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sparrow, J. R., Zhou, J. & Cai, B. DNA is a target of the photodynamic effects elicited in A2E-laden RPE by blue-light illumination. Invest. Ophthalmol. Vis. Sci. 44, 2245–2251 (2003).

    Article  PubMed  Google Scholar 

  81. Sparrow, J. R., Nakanishi, K. & Parish, C. A. The lipofuscin fluorophore A2E mediates blue light-induced damage to retinal pigmented epithelial cells. Invest. Ophthalmol. Vis. Sci. 41, 1981–1989 (2000).

    CAS  PubMed  Google Scholar 

  82. Radu, R. A. et al. Complement system dysregulation and inflammation in the retinal pigment epithelium of a mouse model for Stargardt macular degeneration. J. Biol. Chem. 286, 18593–18601 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Iriyama, A. et al. A2E, a component of lipofuscin, is pro-angiogenic in vivo. J. Cell. Physiol. 220, 469–475 (2009).

    Article  CAS  PubMed  Google Scholar 

  84. Catala, A. Lipid peroxidation of membrane phospholipids in the vertebrate retina. Front. Biosci. (Schol Ed) 3, 52–60 (2011).

    Article  Google Scholar 

  85. Hoppe, G., O'Neil, J., Hoff, H. F. & Sears, J. Products of lipid peroxidation induce missorting of the principal lysosomal protease in retinal pigment epithelium. Biochim. Biophys. Acta 1689, 33–41 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Krohne, T. U., Stratmann, N. K., Kopitz, J. & Holz, F. G. Effects of lipid peroxidation products on lipofuscinogenesis and autophagy in human retinal pigment epithelial cells. Exp. Res. 90, 465–471 (2010).

    CAS  Google Scholar 

  87. Weismann, D. et al. Complement factor H binds malondialdehyde epitopes and protects from oxidative stress. Nature 478, 76–81 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Sharma, A. et al. 4-Hydroxynonenal induces p53-mediated apoptosis in retinal pigment epithelial cells. Arch. Biochem. Biophys. 480, 85–94 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Dentchev, T., Milam, A. H., Lee, V. M., Trojanowski, J. Q. & Dunaief, J. L. Amyloid-β is found in drusen from some age-related macular degeneration retinas, but not in drusen from normal retinas. Mol. Vision 9, 184–190 (2003).

    CAS  Google Scholar 

  90. Ding, J. D. et al. Anti-amyloid therapy protects against retinal pigmented epithelium damage and vision loss in a model of age-related macular degeneration. Proc. Natl Acad. Sci. USA 108, e279–e287 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Liu, X. C., Liu, X. F., Jian, C. X., Li, C. J. & He, S. Z. IL-33 is induced by amyloid-β stimulation and regulates inflammatory cytokine production in retinal pigment epithelium cells. Inflammation 35, 776–784 (2012).

    Article  CAS  PubMed  Google Scholar 

  92. Ambati, J. & Fowler, B. J. Mechanisms of age-related macular degeneration. Neuron 75, 26–39 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Killingsworth, M. C., Sarks, J. P. & Sarks, S. H. Macrophages related to Bruch's membrane in age-related macular degeneration. Eye (Lond.) 4, 613–621 (1990).

    Article  Google Scholar 

  94. Lopez, P. F. et al. Pathologic features of surgically excised subretinal neovascular membranes in age-related macular degeneration. Am. J. Ophthalmol. 112, 647–656 (1991).

    Article  CAS  PubMed  Google Scholar 

  95. Oh, H. et al. The potential angiogenic role of macrophages in the formation of choroidal neovascular membranes. Invest. Ophthalmol. Vis. Sci. 40, 1891–1898 (1999).

    CAS  PubMed  Google Scholar 

  96. Seregard, S., Algvere, P. V. & Berglin, L. Immunohistochemical characterization of surgically removed subfoveal fibrovascular membranes. Graefes Arch. Clin. Exp. Ophthalmol. 232, 325–329 (1994).

    Article  CAS  PubMed  Google Scholar 

  97. Tsutsumi-Miyahara, C. et al. The relative contributions of each subset of ocular infiltrated cells in experimental choroidal neovascularisation. Br. J. Ophthalmol. 88, 1217–1222 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Cherepanoff, S., McMenamin, P., Gillies, M. C., Kettle, E. & Sarks, S. H. Bruch's membrane and choroidal macrophages in early and advanced age-related macular degeneration. Br. J. Ophthalmol. 94, 918–925 (2010).

    Article  CAS  PubMed  Google Scholar 

  99. Grossniklaus, H. E. et al. Correlation of histologic 2-dimensional reconstruction and confocal scanning laser microscopic imaging of choroidal neovascularization in eyes with age-related maculopathy. Arch. Ophthalmol. 118, 625–629 (2000).

    Article  CAS  PubMed  Google Scholar 

  100. Apte, R. S., Richter, J., Herndon, J. & Ferguson, T. A. Macrophages inhibit neovascularization in a murine model of age-related macular degeneration. PLoS Med. 3, e310 (2006). References 36, 37, 99 and 100 show the diverging roles of macrophages in neovascular AMD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Cao, X. et al. Macrophage polarization in the maculae of age-related macular degeneration: a pilot study. Pathol. Int. 61, 528–535 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hasegawa, E. et al. IL-27 inhibits pathophysiological intraocular neovascularization due to laser burn. J. Leukoc. Biol. 91, 267–273 (2012).

    Article  CAS  PubMed  Google Scholar 

  103. Matsumura, N. et al. Low-dose lipopolysaccharide pretreatment suppresses choroidal neovascularization via IL-10 induction. PLoS ONE 7, e39890 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Kelly, J., Ali Khan, A., Yin, J., Ferguson, T. A. & Apte, R. S. Senescence regulates macrophage activation and angiogenic fate at sites of tissue injury in mice. J. Clin. Invest. 117, 3421–3426 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Zhou, J. et al. Neutrophils promote experimental choroidal neovascularization. Mol. Vision 11, 414–424 (2005).

    CAS  Google Scholar 

  106. Zhou, J. et al. Neutrophils compromise retinal pigment epithelial barrier integrity. J. Biomed. Biotechnol. 2010, 289360 (2010).

    PubMed  PubMed Central  Google Scholar 

  107. Penfold, P. L., Provis, J. M. & Billson, F. A. Age-related macular degeneration: ultrastructural studies of the relationship of leucocytes to angiogenesis. Graefes Arch. Clin. Exp. Ophthalmol. 225, 70–76 (1987).

    Article  CAS  PubMed  Google Scholar 

  108. Luo, L. et al. Targeted intraceptor nanoparticle therapy reduces angiogenesis and fibrosis in primate and murine macular degeneration. ACS Nano. 7, 3264–3275 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Martin, D. F. et al. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. N. Engl. J. Med. 364, 1897–1908 (2011).

    Article  CAS  PubMed  Google Scholar 

  110. Jonas, J. B., Degenring, R. F., Kreissig, I., Friedemann, T. & Akkoyun, I. Exudative age-related macular degeneration treated by intravitreal triamcinolone acetonide. A prospective comparative nonrandomized study. Eye (Lond.) 19, 163–170 (2005).

    Article  CAS  Google Scholar 

  111. Gillies, M. C. et al. A randomized clinical trial of a single dose of intravitreal triamcinolone acetonide for neovascular age-related macular degeneration: one-year results. Arch. Ophthalmol. 121, 667–673 (2003).

    Article  CAS  PubMed  Google Scholar 

  112. Smailhodzic, D. et al. Cumulative effect of risk alleles in CFH, ARMS2, and VEGFA on the response to ranibizumab treatment in age-related macular degeneration. Ophthalmology 119, 2304–2311 (2012).

    Article  PubMed  Google Scholar 

  113. Hagstrom, S. A. et al. Pharmacogenetics for genes associated with age-related macular degeneration (AMD) in the comparison of AMD treatments trials (CATT). Ophthalmology 120, 593–599 (2013).

    Article  PubMed  Google Scholar 

  114. Scholl, H. P. et al. CFH, C3 and ARMS2 are significant risk loci for susceptibility but not for disease progression of geographic atrophy due to AMD. PLoS ONE 4, e7418 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Klein, M. L. et al. Progression of geographic atrophy and genotype in age-related macular degeneration. Ophthalmology 117, 1554–1559.e1 (2010).

    Article  PubMed  Google Scholar 

  116. Nozaki, M. et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc. Natl Acad. Sci. USA 103, 2328–2333 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Curcio, C. A. et al. Subretinal drusenoid deposits in non-neovascular age-related macular degeneration: morphology, prevalence, yopography, and biogenesis model. Retina 33, 265–276 (2013).

    Article  PubMed  Google Scholar 

  118. Johnson, L. V. et al. Cell culture model that mimics drusen formation and triggers complement activation associated with age-related macular degeneration. Proc. Natl Acad. Sci. USA 108, 18277–18282 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Wang, A. L. et al. Autophagy and exosomes in the aged retinal pigment epithelium: possible relevance to drusen formation and age-related macular degeneration. PLoS ONE 4, e4160 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  120. Munch, I. C. et al. Small, hard macular drusen and peripheral drusen: associations with AMD genotypes in the Inter99 Eye Study. Invest. Ophthalmol. Vis. Sci. 51, 2317–2321 (2010).

    Article  PubMed  Google Scholar 

  121. Seddon, J. M., Reynolds, R. & Rosner, B. Peripheral retinal drusen and reticular pigment: association with CFHY402H and CFHrs1410996 genotypes in family and twin studies. Invest. Ophthalmol. Vis. Sci. 50, 586–591 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  122. Hageman, G. S. & Mullins, R. F. Molecular composition of drusen as related to substructural phenotype. Mol. Vis. 5, 28 (1999).

    CAS  PubMed  Google Scholar 

  123. Crabb, J. W. et al. Drusen proteome analysis: an approach to the etiology of age-related macular degeneration. Proc. Natl Acad. Sci. USA 99, 14682–14687 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Hollyfield, J. G., Salomon, R. G. & Crabb, J. W. Proteomic approaches to understanding age-related macular degeneration. Adv. Exp. Med. Biol. 533, 83–89 (2003).

    Article  CAS  PubMed  Google Scholar 

  125. Johnson, L. V. et al. The Alzheimer's Aβ -peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration. Proc. Natl Acad. Sci. USA 99, 11830–11835 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Klein, M. L. et al. Retinal precursors and the development of geographic atrophy in age-related macular degeneration. Ophthalmology 115, 1026–1031 (2008).

    Article  PubMed  Google Scholar 

  127. Friese, M. A. et al. FHL-1/reconectin and factor H: two human complement regulators which are encoded by the same gene are differently expressed and regulated. Mol. Immunol. 36, 809–818 (1999).

    Article  CAS  PubMed  Google Scholar 

  128. Spencer, K. L. et al. Deletion of CFHR3 and CFHR1 genes in age-related macular degeneration. Hum. Mol. Genet. 17, 971–977 (2008).

    Article  CAS  PubMed  Google Scholar 

  129. Heurich, M. et al. Common polymorphisms in C3, factor B, and factor H collaborate to determine systemic complement activity and disease risk. Proc. Natl Acad. Sci. USA 108, 8761–8766 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Takeda, A. et al. CCR3 is a target for age-related macular degeneration diagnosis and therapy. Nature 460, 225–230 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wang, H. et al. Upregulation of CCR3 by age-related stresses promotes choroidal endothelial cell migration via VEGF-dependent and -independent signaling. Invest. Ophthalmol. Vis. Sci. 52, 8271–8277 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Mo, F. M., Proia, A. D., Johnson, W. H., Cyr, D. & Lashkari, K. Interferon γ-inducible protein-10 (IP-10) and eotaxin as biomarkers in age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 51, 4226–4236 (2010).

    Article  PubMed  Google Scholar 

  133. Sharma, N. K. et al. New biomarker for neovascular age-related macular degeneration: eotaxin-2. DNA Cell Biol. 31, 1618–1627 (2012).

    Article  CAS  PubMed  Google Scholar 

  134. Mizutani, T., Ashikari, M., Tokoro, M., Nozaki, M. & Ogura, Y. Suppression of laser-induced choroidal neovascularization by a CCR3 antagonist. Invest. Ophthalmol. Vis. Sci. 54, 1564–1572 (2013).

    Article  CAS  PubMed  Google Scholar 

  135. Seddon, J. M., 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. 123, 774–782 (2005).

    Article  PubMed  Google Scholar 

  136. Dankbar, B. et al. Vascular endothelial growth factor and interleukin-6 in paracrine tumor-stromal cell interactions in multiple myeloma. Blood 95, 2630–2636 (2000).

    CAS  PubMed  Google Scholar 

  137. Izumi-Nagai, K. et al. Interleukin-6 receptor-mediated activation of signal transducer and activator of transcription-3 (STAT3) promotes choroidal neovascularization. Am. J. Pathol. 170, 2149–2158 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Leung, K. W., Barnstable, C. J. & Tombran-Tink, J. Bacterial endotoxin activates retinal pigment epithelial cells and induces their degeneration through IL-6 and IL-8 autocrine signaling. Mol. Immunol. 46, 1374–1386 (2009).

    Article  CAS  PubMed  Google Scholar 

  139. Fujimura, S. et al. Angiostatic effect of CXCR3 expressed on choroidal neovascularization. Invest. Ophthalmol. Vis. Sci. 53, 1999–2006 (2012).

    Article  CAS  PubMed  Google Scholar 

  140. Arakawa, S. et al. Genome-wide association study identifies two susceptibility loci for exudative age-related macular degeneration in the Japanese population. Nature Genet. 43, 1001–1004 (2011).

    Article  CAS  PubMed  Google Scholar 

  141. Nakata, I. et al. Association of genetic variants on 8p21 and 4q12 with age-related macular degeneration in Asian populations. Invest. Ophthalmol. Vis. Sci. 53, 6576–6581 (2012).

    Article  CAS  PubMed  Google Scholar 

  142. Lichtlen, P., Lam, T. T., Nork, T. M., Streit, T. & Urech, D. M. Relative contribution of VEGF and TNF-α in the cynomolgus laser-induced CNV model: comparing the efficacy of bevacizumab, adalimumab, and ESBA105. Invest. Ophthalmol. Vis. Sci. 51, 4738–4745 (2010).

    Article  PubMed  Google Scholar 

  143. Olson, J. L., Courtney, R. J. & Mandava, N. Intravitreal infliximab and choroidal neovascularization in an animal model. Arch. Ophthalmol. 125, 1221–1224 (2007).

    Article  CAS  PubMed  Google Scholar 

  144. Shi, X. et al. Inhibition of TNF-α reduces laser-induced choroidal neovascularization. Exp. Res. 83, 1325–1334 (2006).

    CAS  Google Scholar 

  145. de Oliveira Dias, J. R. et al. Cytokines in neovascular age-related macular degeneration: fundamentals of targeted combination therapy. Br. J. Ophthalmol. 95, 1631–1637 (2011).

    Article  PubMed  Google Scholar 

  146. Sengupta, N. et al. Preventing stem cell incorporation into choroidal neovascularization by targeting homing and attachment factors. Invest. Ophthalmol. Vis. Sci. 46, 343–348 (2005).

    Article  PubMed  Google Scholar 

  147. Lee, E. & Rewolinski, D. Evaluation of CXCR4 inhibition in the prevention and intervention model of laser-induced choroidal neovascularization. Invest. Ophthalmol. Vis. Sci. 51, 3666–3672 (2010).

    Article  PubMed  Google Scholar 

  148. Guerin, E. et al. SDF1-α is associated with VEGFR-2 in human choroidal neovascularisation. Microvasc. Res. 75, 302–307 (2008).

    Article  CAS  PubMed  Google Scholar 

  149. Bhutto, I. A., McLeod, D. S., Merges, C., Hasegawa, T. & Lutty, G. A. Localisation of SDF-1 and its receptor CXCR4 in retina and choroid of aged human eyes and in eyes with age related macular degeneration. Br. J. Ophthalmol. 90, 906–910 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Mevorach, D. Clearance of dying cells and systemic lupus erythematosus: the role of C1q and the complement system. Apoptosis 15, 1114–1123 (2010).

    Article  CAS  PubMed  Google Scholar 

  151. Wang, Q. et al. Identification of a central role for complement in osteoarthritis. Nature Med. 17, 1674–1679 (2011).

    Article  CAS  PubMed  Google Scholar 

  152. Wu, G. et al. Complement regulator CD59 protects against atherosclerosis by restricting the formation of complement membrane attack complex. Circ. Res. 104, 550–558 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Szeplaki, G., Varga, L., Fust, G. & Prohaszka, Z. Role of complement in the pathomechanism of atherosclerotic vascular diseases. Mol. Immunol. 46, 2784–2793 (2009).

    Article  CAS  PubMed  Google Scholar 

  154. Hansson, G. K. & Hermansson, A. The immune system in atherosclerosis. Nature Immunol. 12, 204–212 (2011).

    Article  CAS  Google Scholar 

  155. Fritsche, L. G. et al. Seven new loci associated with age-related macular degeneration. Nature Genet. 45, 433–439 (2013).

    Article  CAS  PubMed  Google Scholar 

  156. Landa, G., Butovsky, O., Shoshani, J., Schwartz, M. & Pollack, A. Weekly vaccination with Copaxone (glatiramer acetate) as a potential therapy for dry age-related macular degeneration. Curr. Res. 33, 1011–1013 (2008).

    CAS  Google Scholar 

  157. Nussenblatt, R. B. et al. A randomized pilot study of systemic immunosuppression in the treatment of age-related macular degeneration with choroidal neovascularization. Retina 30, 1579–1587 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  158. Gomi, F., Sawa, M., Tsujikawa, M. & Nishida, K. Topical bromfenac as an adjunctive treatment with intravitreal ranibizumab for exudative age-related macular degeneration. Retina 32, 1804–1810 (2012).

    Article  CAS  PubMed  Google Scholar 

  159. Ahmadieh, H. et al. Intravitreal bevacizumab versus combined intravitreal bevacizumab and triamcinolone for neovascular age-related macular degeneration: six-month results of a randomized clinical trial. Retina 31, 1819–1826 (2011).

    Article  CAS  PubMed  Google Scholar 

  160. Lachmann, P. J. The amplification loop of the complement pathways. Adv. Immunol. 104, 115–149 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

J.A. is supported by US National Institutes of Health (NIH) grants (R01EY018836, R01EY020672 and R01EY022238), the Doris Duke Charitable Foundation, USA, the Ellison Medical Foundation, USA, the Burroughs Wellcome Fund, USA, the Reeves Foundation, USA, a Dr. E. Vernon Smith and Eloise C. Smith Endowment and a Research to Prevent Blindness Unrestricted Grant, USA. J.P.A. is supported by NIH grants (AI041592, AR007279, AR0483335, GM099111 and HL112303), the Edward N. and Della L. Thome Memorial Foundation, USA, and Alexion Pharmaceuticals, USA. B.D.G. is suported by the National Center for Advancing Translational Sciences, USA, grants UL1TR000117 and UL1TR000117. The content of this article is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jayakrishna Ambati.

Ethics declarations

Competing interests

J.A. is named as an inventor on patent applications filed by his employer (University of Kentucky, USA) on technologies related to AMD diagnosis and therapy, and he is a co-founder of iVeena, Inc., USA. which is involved in the commercial development of AMD therapies. J.P.A. is involved in Compliment Corporation, USA (Scientific Advisory Board, 2010–present); Kypha, Inc., USA (Scientific Advisory Board, 2012); Genentech Inc., USA (Consultant, 2007–present); Idera Pharmaceuticals, USA (Consultant, 2007–present); KEREOS Incorporated, USA (Consultant, 2009–present); Alnylam Pharmaceuticals, Inc., USA (Consultant, 2010–present); Celldex Therapeutics, USA, formerly Avant Immunotherapeutics, Inc. (Consultant, 2008–present); and Alexion Pharmaceuticals, Inc., USA (Consultant, 2011–present).

Related links

FURTHER INFORMATION

Jayakrishna Ambati's homepage

PowerPoint slides

Glossary

Retina

A highly organized and specialized neural network where light is converted into electrical impulses. Diseases of the retina such as age-related macular degeneration are leading causes of blindness in the developed world.

Macula

A specialized region of the retina densely populated with cone photoreceptors, which is responsible for fine visual acuity. Degeneration of the macular photoreceptors following either atrophy of the retinal pigmented epithelium (geographic atrophy) or fluid leakage from choroidal neovessels (neovascular age-related macular degeneration (AMD)) is the cause of vision loss in AMD.

Complement factor H

(CFH). A negative regulator of alternative complement pathway activation. Single nucleotide polymorphisms in CFH that reduce its inhibitory potential are responsible for a substantial proportion of the genetic risk for the development of age-related macular degeneration.

Neovascular AMD

(also known as exudative or 'wet' age-related macular degeneration). Characterized by degeneration of the macula following fluid leakage from choroidal neovessels that have invaded the retina. The use of vascular endothelial growth factor A-targeted therapies has revolutionized the management of this disease, which accounts for the majority of blindness that results from AMD.

Photoreceptors

Specialized neurons that are responsible for the conversion of light into biochemical signals.

Blood–retinal barrier

A tightly controlled transport barrier that is maintained by tight junctions in the retinal capillary endothelium (comprising the inner blood–retinal barrier), and by the Bruch membrane and the retinal pigmented epithelium monolayer (comprising the outer blood–retinal barrier). Integrity of the blood–retinal barrier is important for control of fluid leakage, solute transport and immune quiescence, all of which support the functional homeostasis of the retina.

Retinal pigmented epithelium

(RPE). A monolayer of epithelial cells that has multiple essential roles in visual function, including recycling components of the visual cycle, secreting trophic factors and maintaining the outer blood–retinal barrier. The RPE is widely considered to be the focal point of age-related macular degeneration pathogenesis, in which breakdown of the RPE leads to secondary photoreceptor degeneration.

Drusen

Discrete extracellular deposits that commonly precede the development of age-related macular degeneration, and that are comprised of numerous cellular and inflammatory factors.

Geographic atrophy

(also known as end-stage 'dry' age-related macular degeneration (AMD) or atrophic AMD). A disease affecting the macula in which the retinal pigmented epithelium can no longer support photoreceptor function owing to spontaneous degeneration of large confluent regions. Although geographic atrophy occurs less frequently than neovascular AMD (approximately 50% as common), there are no currently approved therapies.

Immune privilege

The property of a tissue being tolerant to antigen. Retinal immune privilege is maintained by blood–retinal barrier integrity and the absence of functional lymphatic circulation.

Fundus photography

A common method for visualizing the retinal pigmented epithelium, retina and retinal circulation photographically. Funduscopy is used by ophthalmologists to diagnose retinal disorders such as age-related macular degeneration.

Para-inflammation

A state in which tissue homeostasis is maintained by low-grade inflammatory-based clearance of noxious stimuli. In the retina, para-inflammation may persist and ultimately contribute to age-related macular degeneration through increased immune cell infiltration and activation at sites of tissue damage.

Genome-wide association studies

(GWASs). The process by which genetic variations and disease phenotypes are statistically correlated. Linkage studies carried out with markers located across the entire genome that were traditionally carried out with approximately 300 markers of simple sequence-length repeats but that have been more recently carried out with approximately 1–5 million single nucleotide polymorphisms.

Alternative pathway of complement activation

An evolutionarily ancient innate immune process by which microorganisms are destroyed through opsonization and activation of the membrane attack complex. According to the 'complement hypothesis', misactivation of, and/or the inability to appropriately inhibit, the alternative pathway results in retinal tissue damage and drives age-related macular degeneration pathology.

Carboxyethylpyrrole-adducted proteins

Proteins that are modified through the oxidation of the fatty acid docosahexaenoic acid. These adducts are abundant in the retina. They can have direct pro-inflammatory effects through pattern recognition, and autoantibodies that recognize them are abundant in the circulation of patients with age-related macular degeneration.

M1 macrophages

A pro-inflammatory or 'classically activated' subset of macrophages, which are characterized by phagocytic activity and expression of particular inflammatory cytokines (such as tumour necrosis factor) and inflammatory mediators (such as inducible nitric oxide synthase).

M2 macrophages

A pro-angiogenic or 'alternatively activated' subset of macrophages, which are characterized by the expression of particular angiogenic cytokines (such as vascular endothelial growth factor A) and anti-inflammatory mediators (such as arginase).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ambati, J., Atkinson, J. & Gelfand, B. Immunology of age-related macular degeneration. Nat Rev Immunol 13, 438–451 (2013). https://doi.org/10.1038/nri3459

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri3459

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing