Next Article in Journal
Motif2Mol: Prediction of New Active Compounds Based on Sequence Motifs of Ligand Binding Sites in Proteins Using a Biochemical Language Model
Next Article in Special Issue
Emerging Role of Adiponectin/AdipoRs Signaling in Choroidal Neovascularization, Age-Related Macular Degeneration, and Diabetic Retinopathy
Previous Article in Journal
Lipoxin A4 (LXA4) Reduces Alkali-Induced Corneal Inflammation and Neovascularization and Upregulates a Repair Transcriptome
Previous Article in Special Issue
The Autoimmune Rheumatic Disease Related Dry Eye and Its Association with Retinopathy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Complement in the Onset of Age-Related Macular Degeneration

1
Department of Ophthalmology, School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
2
Departments of Ophthalmology and Biochemistry & Molecular Biology, School of Medicine, Saint Louis University, St. Louis, MO 63104, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2023, 13(5), 832; https://doi.org/10.3390/biom13050832
Submission received: 18 March 2023 / Revised: 9 May 2023 / Accepted: 11 May 2023 / Published: 13 May 2023
(This article belongs to the Special Issue New Discoveries in Retinal Cell Degeneration and Retinal Diseases)

Abstract

:
Age-related macular degeneration (AMD) is a progressive degenerative disease of the central retina and the leading cause of severe loss of central vision in people over age 50. Patients gradually lose central visual acuity, compromising their ability to read, write, drive, and recognize faces, all of which greatly impact daily life activities. Quality of life is significantly affected in these patients, and there are worse levels of depression as a result. AMD is a complex, multifactorial disease in which age and genetics, as well as environmental factors, all play a role in its development and progression. The mechanism by which these risk factors interact and converge towards AMD are not fully understood, and therefore, drug discovery is challenging, with no successful therapeutic attempt to prevent the development of this disease. In this review, we describe the pathophysiology of AMD and review the role of complement, which is a major risk factor in the development of AMD.

1. Introduction

Age-related macular degeneration (AMD) is the leading cause of severe loss of central vision in people over age 50 [1]. It is estimated that nearly 20 million Americans have some form of AMD [2]. Currently, over 200 million people are affected worldwide, and it is estimated that this number will double by 2040 [3,4,5]. In Western countries, nearly 22% of people over 70 and 34% over 80 suffer from AMD in at least one eye [5]. Patients gradually lose central visual acuity, compromising their ability to read, write, drive, and recognize faces, all of which greatly impair their daily life activities [6]. Studies have also shown that quality of life is significantly affected in AMD patients, with less life satisfaction, more stress, less physical activity, and worse depression levels [7,8].

1.1. Anatomy of AMD

The pathology of AMD is restricted to the outer retinal layers and the retinal pigment epithelium (RPE) of the macula [9]. The central macula, or central posterior retina, measures 5 mm in diameter, with the highest density of cone photoreceptors located in the fovea, a 1500-micron area responsible for central visual acuity and color vision [10] (Figure 1). The fovea has a unique anatomical structure, both because of the high density of cone photoreceptors and the absence of retinal blood vessels [11]. The cells in the fovea receive nutrition from the underlying choroidal circulation and the adjacent perifoveal capillary network. The absence of an inner retinal vasculature allows improved light absorption by the cone photoreceptor cells, providing high-resolution vision [12].
RPE cells are a polarized monolayer of cuboidal cells between the neurosensory retina and Bruch’s membrane (BM). They are of vital importance for the homeostasis of the entire retina, but especially the fovea. Nutrients and oxygen are supplied to the macula by the choroidal circulation through Bruch’s membrane and the RPE. Metabolic waste products of the retina are also transported through the RPE/BM complex into the choroidal circulation [11]. RPE cells are also important in the phagocytosis of shed photoreceptor outer segments, which is an essential part of the visual cycle [13] and have an important role in protecting the retina from oxidative stress due to large numbers of melanosomes that absorb light [14]. RPE is separated from the underlying choroid by Bruch’s membrane (BM), which is composed of five layers—the basement membrane of RPE cells, the inner collagenous layer, the middle elastic layer, the outer collagenous layer, and the basement membrane of the choriocapillaris endothelial cells [15]. In addition to being a structural support for the RPE, BM also contributes to the outer blood–retinal barrier and the selective transport of molecules and proteins. Therefore, any changes in the choroid–BM–RPE complex will affect the natural homeostasis of photoreceptors. These changes may contribute to the development of AMD [12].

1.2. Pathophysiology of AMD

Major changes in BM contribute to macular damage, including increased thickness, which is a result of increased deposition and the cross-linking of collagen fibers, and decreased permeability, resulting in decreased RPE metabolic function [12,16,17]. It has also been observed that with aging, there is a deposition of advanced glycated end products (AGEs), i.e., glycated or oxidized lipoproteins [18], which can trigger inflammation, as well as the presence of leukocytes in the choroid underneath the macula in AMD.
RPE cells lose melanosomes, and mitochondria become more prone to oxidative stress with aging [19,20]. This is associated with major changes in the choroid. The normal choroid has high blood flow and can change rapidly, depending on the physiologic demand of the RPE and neurosensory retina. Aging leads to decreased blood flow, as well as the thinning of the choroid [21]. Therefore, flexibility decreases, and the choroid cannot rapidly respond to changes in blood flow required for the physiologic demand. Decreased blood flow results in a decreased support of oxygen and nutrients to the retina. The resultant hypoxia and lack of sufficient nutrient support drives RPE cells to extreme metabolic stress. Additionally, reduced blood flow impairs the removal of photoreceptor and RPE waste products, which are now deposited in the BM, leading to drusen formation [22].
“Drusen” are extracellular deposits between the basal lamina of the RPE and the inner collagenous layer of BM [23,24]. They consist of many different components, and the composition varies with the disease stage. More than 40% of its contents include neutral lipids and esterified and unesterified cholesterol [22]. In addition, more than 129 proteins have been identified in drusen [25], including inflammatory and complement factors, such as vitronectin, serum beta amyloid protein, tissue inhibitor of metalloproteinase 3 (TIMP3), apolipoproteins (E, B, A-I, C-I, and C-II), immunoglobulin light chains, complement proteins (C5 and the C5b-9 complex), proteins involved in complement regulation, and zinc and iron ions [22,25,26].

1.3. Clinical Presentation

Upon clinical examination of the fundus, drusen present as small, yellowish-white spots underneath the retina in the central macula. They are classified clinically in the Wisconsin grading system as hard or soft drusen. Hard drusen are defined as well-defined yellow spots that are 1–63 microns in diameter. Soft drusen are larger than 125 microns, or if they are 63–125 microns, they have visible thickness. Soft drusen can have either distinct or indistinct borders (Figure 2) [27]. They are linked with a higher risk of choroidal neovascularization (CNV) because they include C3a and C5a and can trigger the overexpression of the vascular endothelial growth factor (VEGF) in RPE [28].
Hard drusen are often present in small numbers with aging but increase in AMD patients. This implies that other mechanisms underlie AMD pathogenesis beyond the simple presence of drusen themselves [12]. As the disease progresses from the early to the intermediate stage of AMD, the number and size of drusen increase, and RPE changes occur. With further progression, two late sight-threatening stages are recognized: geographic atrophy (GA, or end-stage dry AMD) and neovascular AMD (nAMD or wet AMD). GA is characterized by the death of RPE cells, the overlying photoreceptors, and the underlying choriocapillaris, whereas nAMD is most frequently characterized by the growth of abnormal choroidal blood vessels into the sub-RPE or subretinal space (CNV) [12,29]. GA clinically starts as horseshoe patches of RPE atrophy in the perifoveal area and gradually enlarges to involve the fovea. It starts in an area with drusen, and as the disease progresses, regression of drusen occurs [30,31].

1.4. Risk Factors for AMD

The pathogenesis of AMD is multifactorial, involving a combination of genetic and environmental factors in its development. The most important modifiable environmental factor is cigarette smoking, which contributes significantly to the development, as well as the progression, of the disease and oxidative damage [32]. Smoking increases the risk of AMD 2–4-fold. The risk is dose dependent, and after cessation of smoking, the risk of AMD decreases. It has been demonstrated that 20 years after quitting smoking, the risk is minimal [33]. Other environmental risk factors include cataract surgery, diet, cardiovascular disease, hypertension, and body mass index (BMI). The Mediterranean diet has been shown to decrease the risk of AMD, presumably because of the high content of antioxidants in vegetables and fruits, which are integral components of this diet [12,34,35,36]. It has also been observed that eating fish protects against AMD [37]. The Age-Related Eye Disease Study (AREDS), a large, multicenter clinical trial, demonstrated the most convincing evidence for a connection between oxidative stress and AMD [38,39]. They showed that certain vitamins and antioxidants decreased the rate of progression of intermediate-stage AMD by 25% in an average follow-up of 6.3 years.
Studies into the molecular components of drusen already hinted that AMD might have an immune component before any particular gene or biological pathway was definitively connected to the condition. This hypothesis was proposed after proteins related to inflammation and/or other immune-related molecules, such as complement system components, were discovered in drusen [40].
Twin and family studies provided evidence that AMD has a significant hereditary component. Twin studies found that monozygotic pairs had a high concordance of AMD, even double that of dizygotic pairs, and they predicted that the heredity component of AMD could be as high as 45 to 70% [40,41,42,43].
Genetic associations with AMD are well documented in different genome-wide association analyses (GWAS). The strongest genetic contributors to AMD with the highest odds ratios are variants associated with complement factor H (CFH)—complement factor H-related (CFHR) 5 on chromosome 1q32 (Chr1 locus), and age-related maculopathy susceptibility 2 (ARMS2) and high-temperature requirement factor A1 (HTRA1), two tightly-linked genes located on chromosome 10q26 (Chr10 locus) [44,45,46,47]. The most well-known genetic risk factor for AMD is the complement factor H Y402H (Tyr402His) variation. While another missense mutation of CFH, I62V (Ile62Val), is more common in Asian populations, this mutation is specifically connected to AMD susceptibility in Caucasians [48,49,50]. The primary function of the glycoprotein CFH, which has 20 domains called short consensus repeats (SCR), is to prevent the activation of the alternative complement pathway [51].
Complement FH, a significant complement system inhibitor, not only competes with CFB for binding to C3b, speeding up the dissociation of the resultant alternative system C3 convertase (C3bBb), it also functions as a cofactor to facilitate the factor I (FI)-mediated inactivation of C3b [52,53].
In addition to CFH, several complement genes are involved, including CFB, CFI, C3, and C2 [54]. Since the latter discoveries, a genetic association between AMD and the complement system was further strengthened. At least 34 genomic loci are also associated with AMD pathogenesis, including genes related to cholesterol metabolism, extracellular matrix remodeling, and collagen structure [55].
Many of these genes are involved in either enhancing activation or decreasing deactivation of the active complement factors, therefore resulting in the dysregulation of complement control, with ultimate damage to the host tissue.
These genetic association studies led to the identification of a dysregulated alternative complement pathway as a main driver of AMD, and more recently, they served as useful tools for proposing associated candidate therapeutic targets [12]. Immunohistochemical studies supported this conclusion [56,57]. Clearly, inflammation plays a role in AMD pathogenesis and is referred to by some investigators as an example of para-inflammation [11,54].
In this study, we outlined potential scenarios for how AMD might develop and worsen as a result of complement system malfunction.

2. AMD and the Complement System

The complement system is a crucial component of the innate immune response that protects the host against invading pathogens, such as bacteria. It has three pathways: classic (antigen–antibody complex), lectin-dependent (mannose polysaccharides on microorganisms), and alternative (pathogen cell surfaces) [58]. Each pathway has its own distinctive factors, but they all converge and ultimately lead to the cleavage of complement factor 3 (C3) to C3a and C3b. C3a is anaphylatoxin and induces inflammation, and C3b opsonizes cells and labels them for phagocytosis. The complement cascade continues and ultimately forms the membrane attack complex (MAC), which includes (C5b-C9). The MAC penetrates the cell membrane and results in cell death [58,59,60]. The formation of the MAC can be inhibited by complement inhibitors, such as CD59 [61]. Under normal conditions, host cell regulatory proteins deactivate active complement components and prevent damage to host cells [59](Figure 3).
The complement system can be considered a double-edged sword. On one side, under normal conditions, it protects the body against foreign pathogens and modifies the surveillance of immune responses; on the other side, under abnormal conditions, its dysregulation or dysfunction can lead to uncontrolled inflammatory responses and can damage host tissue, resulting in disease [62].
The alternative pathway is activated in two different ways. The first one is called “tickover”, which is the spontaneous hydrolysis of C3 into C3 (H2O) in the fluid phase. The second one is by C4b2a (C3 convertase), which results in the cleavage of C3 to C3a and C3b in the solid phase [63]. The alternative pathway is responsible for the majority (80–90%) of the terminal pathway activation [64]; therefore, overactivation of this pathway may have a major role in disease pathogenesis. Early investigations initially debated whether systemic or local complement activation caused disease in the eye. However, it may well be that complement-mediated molecular processes causing AMD are a combination of systemic complement proteins, as well as locally synthesized complement proteins [12]. Experimental studies in animal models suggested the idea above.
The most commonly used animal model mimics wet AMD and is induced by retinal laser photocoagulation in rodents. Laser-induced CNV can be suppressed by blocking complement activation via local or systemic mechanisms. Inhibiting C3a, C5a, CFB, and MAC or administering the complement regulating molecules CD59 and CFH can prevent the formation of CNV in animal models [65,66,67]. In CFH-deficient mice, excessive alternative pathway activation resulted in the enlargement of the induced CNV [68]. It was also shown that mice lacking the alternative complement pathway (CFB-deficient) had decreased rates of CNV, compared to animals lacking other complement pathway constituents [67,69].
In individuals with AMD, several complement system activation products have been identified to be increased systemically [70]. Patients with intermediate or late dry AMD (i.e., central GA or inactive CNV) have higher levels of systemic complement activation, compared to both controls and early AMD [12]. In contrast, the level of systemic complement activation in wet AMD is relatively low [12,70]. Study of donor eyes from patients with the CFH gene showed increased deposits of local C3b before clinical manifestations of the disease. Thus, poorly controlled complement turnover may start earlier than clinical disease manifestation within the retina [70].
Five AMD risk alleles have been recognized involving the alternative complement pathway, implying a role for innate immunity in disease development. Additionally, the pathogenesis of early AMD may be different than that of late-stage AMD [35]. A recent GWAS identified a gene variant near CD46 almost exclusively associated with early AMD [71].
Interestingly, complement products and the membrane attack complex (MAC) have been found in the choroids of eyes from young donors, as well as those without genetic risks for AMD. This suggests that low-grade complement activation is likely required for the normal homeostasis of the retina/RPE complex [12,72,73,74], similar to observations made in the anterior chamber of the eye [75]. The complement system may assist in the removal of waste products, such as the shed photoreceptor outer segments. MAC levels increase with aging, possibly leading to bystander complement-mediated injury of the tissue [12,72,73,74].
A frequent consequence of complement activation is the recruitment and activation of immune cells and the release of the anaphylatoxins C3a and C5a [12]. The immune cells involved in AMD comprise resident microglial cells, as well as circulating lymphocytes, monocytes/macrophages, and mast cells. Stimulation of monocytes by C3a leads to the secretion of multiple cytokines, possibly leading to tissue damage. Mast cell degranulation by C3a and C5a can lead to the release of proteases, tryptase, and chymase, with resultant damage to the extracellular matrix (ECM) [76].
Lipid deposition in the retina has also been linked to both systemic and local complement systems dysregulation [77]. Studies on the serum of AMD patients demonstrated a substantial correlation between elevated large and extra-large high-density lipoprotein (HDL) levels and decreased, very low-density lipoprotein (VLDL) and amino acid levels. These changes were associated with elevated serum complement activation [78].
CFH has been shown to be present in large HDL particles but not in small or medium HDL molecules [79]. Whether this is pro-inflammatory or suppressive for inflammatory responses is not yet clear. Entrapment by HDL might decrease the availability of circulating CFH to deactivate the complement cascade [79]. On the other hand, the presence of CFH in drusen might suppress inflammation locally in the retina. The latter has supportive evidence, since it has been shown that factor H binds the native low-density lipoprotein (LDL) and the oxidized LDL; this binding affinity is reduced in patients with the CFH high-risk 402H variant. As a result, increased levels of oxidized LDL may lead to the upregulation of inflammatory cytokines and the start of an inflammatory cycle that results in tissue damage [80].
It was also observed that disturbed RPE metabolism is important in the pathogenesis of AMD. Compared to RPE cells from healthy controls, primary RPE cells isolated from AMD patients were shown to have severely impaired energy metabolism [81].
Complement dysregulation associated with the CFH variant is associated with reduced energy metabolism in RPE cells [82]. Complement dysregulation was associated with mitochondrial damage in RPE cells [83]. Compared to RPE cells from donors with the low-risk 402Y genetic variation, RPE cells from AMD donors harboring the high-risk 402H variant exhibited greater mtDNA damage [83]. In the CFH (cfh-/-) knock-out mouse model, abnormally large mitochondria were shown in the RPE and photoreceptors. At the same time, the amount of mitochondrial DNA (mt DNA) decreased, which is a sign of energy dysregulation confirmed by lower adenosine tri-phosphate (ATP) production [82]. Regarding an oxidative stress response, there seems to be a reciprocal interaction between the complement system control and energy metabolism [12]. In a cytoplasmic hybrid (cybrid) model, with mitochondria from AMD patients and ARPE19 devoid of mitochondria, changes in complement components were demonstrated. Complement activators, including CFB, were found to be increased, and complement inhibitors, including CFH, were found to be decreased [84].
Another crucial component of RPE homeostasis is the lysosome–autophagy machine, which is involved in recycling metabolites and degrading damaged organelles, including shed photoreceptor outer segments [85,86]. If the lysosome–autophagy machine in RPE is affected, resultant waste products within the cell will accumulate, rather than being recycled/cleared, with the possible onset of AMD [86]. Figure 3 is a simplified diagram of complement pathways and the major regulators involved in AMD.

3. Therapeutic Strategies

There is no effective treatment to prevent the development of either type of AMD, and although intravitreal injection of anti-VEGF medications can stabilize and improve vision in the majority of patients with wet AMD, the treatment is costly, it requires monthly treatments for several years, and up to 58% of Medicare patients discontinue treatment within the first year [87]. Additionally, anti-VEGF treatments remain ineffective for some patients, and a significant proportion of these patients still develop severe visual loss and progress to legal blindness over time [88].
On 17th February 2023, the FDA approved the first intravitreal injection therapy for geographic atrophy. Pegcetacoplan is a drug that targets C3, and the 2-year Oak and Derby studies leading to FDA approval demonstrated efficacy, compared to sham injections and efficacy, over time, with the greatest benefit resulting in a 36% growth reduction in the GA lesion from 18–24 months in the Derby study [SYFOVRE (pegcetacoplan injection) [package insert]. Waltham, MA: Apellis Pharmaceuticals, Inc.; 2023]
Given the strong genetic and biological evidence that dysregulation of the complement system is a major driver of AMD pathogenesis, it is not surprising that there is increasing interest in complement therapeutic targets to prevent progression of the disease [12].
Early clinical trials with intravenous administration of eculizumab, which targets C5 and is FDA approved for use in paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (HUS), failed to show any effects in GA patients during a phase II clinical trial [89].
Several factors may have possibly prevented successful eculizumab treatment of GA, including the lack of stratification of patients based on their genetic risk, intervening too late in the disease process, or low drug dosage at the site of disease because of systemic administration rather than at the extracellular matrix of the choriocapillaris. Certainly, local administration of drugs directly into the eye holds multiple possible benefits and is now the preferred route of delivery in several current clinical trials for retinal disorders [12].
On the other hand, local administration is associated with several caveats. Introducing drugs to an immune-privileged site, such as the eye, with the inhibition of complement components may increase the risk of endophthalmitis, particularly with the requirement for frequent, regular intravitreal injections [12,90].
More than 14 complement inhibitors have been developed so far that target major components of the complement system (C3, C5) or regulators of the complement system (CFD, CFI). More than 40 clinical trials are either completed or on-going in the treatment of dry AMD with complement inhibitors [62]. Most of these trials are using the enlargement of the GA lesion or loss of visual function as therapeutic endpoints.
APL-2 and Zimura, respectively, target C3 and C5 and are the two most advanced treatments, since both are in phase III trials. Both drugs are administered locally via intravitreal injection. They stop the formation of C5a and MAC and, ultimately, cell lysis. APL2 also prevents the formation of C3a, since it works more upstream. In phase II trials, both APL-2 and Zimura were associated with decreased GA growth rates—29 and 27.8%, respectively. At the same time, both showed a small increased incidence of choroidal neovascularization in the treated eyes, compared with the sham [91,92]. The latter may be related to polyethylene glycol (PEG) in the formulation of both drugs. PEG has been used in the past to induce CNV in mice [93]. As mentioned earlier, on 17th Feb 2023, the FDA approved the first intravitreal drug for the treatment of geographic AMD, whether involving the fovea or not, after the evaluation of the two-year results. Pegcetcoplan/APL-2 works by targeting C3. The drug will be available shortly to slow down the progression and growth of the GA lesion, but there is no reversal effect.
Interestingly, none of the current studies address the downstream effects related to the modulation of the complement pathway, such as the effect on the immune response, RPE cell metabolism, and RPE attachment to the ECM and BM [12]. In the future, a combined mechanism that addresses both upstream and downstream genes in target cells might be a more effective way to treat and/or prevent the development of AMD.

4. Conclusions

In conclusion, age-related macular degeneration is a complex disease that leads to central vision loss. Genetic and environmental factors are involved in the development of the disease. The mechanisms by which these factors interact to develop the disease are not fully understood; however, it is presumed that the complement system has a major role in the onset and development of the disease. Focusing future studies on the complement system in the retina and the RPE may further elucidate molecular changes at a cellular level that lead to disease development.

Author Contributions

Conceptualization, H.J.K. and N.P.; methodology N.P. and H.J.K.; writing—original draft preparation, N.P.; writing—review and editing, N.P. and H.J.K.; visualization, N.P.; supervision, H.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lim, L.S.; Mitchell, P.; Seddon, J.M.; Holz, F.G.; Wong, T.Y. Age-related macular degeneration. Lancet 2012, 379, 1728–1738. [Google Scholar] [CrossRef]
  2. Rein, D.B.; Wittenborn, J.S.; Burke-Conte, Z.; Gulia, R.; Robalik, T.; Ehrlich, J.R.; Lundeen, E.A.; Flaxman, A.D. Prevalence of Age-Related Macular Degeneration in the US in 2019. JAMA Ophthalmol. 2022, 140, 1202–1208. [Google Scholar] [CrossRef]
  3. Li, J.Q.; Welchowski, T.; Schmid, M.; Mauschitz, M.M.; Holz, F.G.; Finger, R.P. Prevalence and incidence of age-related macular degeneration in Europe: A systematic review and meta-analysis. Br J. Ophthalmol. 2020, 104, 1077–1084. [Google Scholar] [CrossRef] [PubMed]
  4. Colijn, J.M.; Buitendijk, G.H.S.; Prokofyeva, E.; Alves, D.; Cachulo, M.L.; Khawaja, A.P.; Cougnard-Gregoire, A.; Merle, B.M.J.; Korb, C.; Erke, M.G.; et al. Prevalence of Age-Related Macular Degeneration in Europe: The Past and the Future. Ophthalmology 2017, 124, 1753–1763. [Google Scholar] [CrossRef] [PubMed]
  5. Wong, W.L.; Su, X.; Li, X.; Cheung, C.M.; Klein, R.; Cheng, C.Y.; Wong, T.Y. Global prevalence of age-related macular degeneration and disease burden projection for 2020 and 2040: A systematic review and meta-analysis. Lancet Glob. Health 2014, 2, e106-116. [Google Scholar] [CrossRef] [PubMed]
  6. Coleman, A.L.; Yu, F.; Ensrud, K.E.; Stone, K.L.; Cauley, J.A.; Pedula, K.L.; Hochberg, M.C.; Mangione, C.M. Impact of age-related macular degeneration on vision-specific quality of life: Follow-up from the 10-year and 15-year visits of the Study of Osteoporotic Fractures. Am. J. Ophthalmol. 2010, 150, 683–691. [Google Scholar] [CrossRef] [PubMed]
  7. Brody, B.L.; Gamst, A.C.; Williams, R.A.; Smith, A.R.; Lau, P.W.; Dolnak, D.; Rapaport, M.H.; Kaplan, R.M.; Brown, S.I. Depression, visual acuity, comorbidity, and disability associated with age-related macular degeneration. Ophthalmology 2001, 108, 1893–1900; discussion 1900. [Google Scholar] [CrossRef]
  8. Mitchell, P.; Liew, G.; Gopinath, B.; Wong, T.Y. Age-related macular degeneration. Lancet 2018, 392, 1147–1159. [Google Scholar] [CrossRef]
  9. Coleman, H.R.; Chan, C.C.; Ferris, F.L., 3rd; Chew, E.Y. Age-related macular degeneration. Lancet 2008, 372, 1835–1845. [Google Scholar] [CrossRef]
  10. Curcio, C.A.; Medeiros, N.E.; Millican, C.L. Photoreceptor loss in age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 1996, 37, 1236–1249. [Google Scholar]
  11. Chen, M.; Xu, H. Parainflammation, chronic inflammation, and age-related macular degeneration. J. Leukoc. Biol. 2015, 98, 713–725. [Google Scholar] [CrossRef] [PubMed]
  12. Armento, A.; Ueffing, M.; Clark, S.J. The complement system in age-related macular degeneration. Cell. Mol. Life Sci. 2021, 78, 4487–4505. [Google Scholar] [CrossRef]
  13. Bok, D. The retinal pigment epithelium: A versatile partner in vision. J. Cell Sci. Suppl. 1993, 17, 189–195. [Google Scholar] [CrossRef]
  14. Bhutto, I.; Lutty, G. Understanding age-related macular degeneration (AMD): Relationships between the photoreceptor/retinal pigment epithelium/Bruch’s membrane/choriocapillaris complex. Mol. Aspects Med. 2012, 33, 295–317. [Google Scholar] [CrossRef] [PubMed]
  15. Hogan, M.J.; Alvarado, J. Studies on the human macula. IV. Aging changes in Bruch’s membrane. Arch. Ophthalmol. 1967, 77, 410–420. [Google Scholar] [CrossRef] [PubMed]
  16. Ramrattan, R.S.; van der Schaft, T.L.; Mooy, C.M.; de Bruijn, W.C.; Mulder, P.G.; de Jong, P.T. Morphometric analysis of Bruch’s membrane, the choriocapillaris, and the choroid in aging. Invest. Ophthalmol. Vis. Sci. 1994, 35, 2857–2864. [Google Scholar]
  17. Hjelmeland, L.M.; Cristofolo, V.J.; Funk, W.; Rakoczy, E.; Katz, M.L. Senescence of the retinal pigment epithelium. Mol. Vis. 1999, 5, 33. [Google Scholar]
  18. Booij, J.C.; Baas, D.C.; Beisekeeva, J.; Gorgels, T.G.; Bergen, A.A. The dynamic nature of Bruch’s membrane. Prog. Retin. Eye Res. 2010, 29, 1–18. [Google Scholar] [CrossRef] [PubMed]
  19. Feher, J.; Kovacs, I.; Artico, M.; Cavallotti, C.; Papale, A.; Balacco Gabrieli, C. Mitochondrial alterations of retinal pigment epithelium in age-related macular degeneration. Neurobiol. Aging 2006, 27, 983–993. [Google Scholar] [CrossRef]
  20. Brown, E.E.; DeWeerd, A.J.; Ildefonso, C.J.; Lewin, A.S.; Ash, J.D. Mitochondrial oxidative stress in the retinal pigment epithelium (RPE) led to metabolic dysfunction in both the RPE and retinal photoreceptors. Redox Biol. 2019, 24, 101201. [Google Scholar] [CrossRef]
  21. Wakatsuki, Y.; Shinojima, A.; Kawamura, A.; Yuzawa, M. Correlation of Aging and Segmental Choroidal Thickness Measurement using Swept Source Optical Coherence Tomography in Healthy Eyes. PLoS ONE 2015, 10, e0144156. [Google Scholar] [CrossRef] [PubMed]
  22. Curcio, C.A.; Johnson, M.; Huang, J.D.; Rudolf, M. Aging, age-related macular degeneration, and the response-to-retention of apolipoprotein B-containing lipoproteins. Prog. Retin. Eye Res. 2009, 28, 393–422. [Google Scholar] [CrossRef]
  23. Hageman, G.S.; Luthert, P.J.; Victor Chong, N.H.; Johnson, L.V.; Anderson, D.H.; Mullins, R.F. 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. [Google Scholar] [CrossRef] [PubMed]
  24. Hageman, G.S.; Mullins, R.F. Molecular composition of drusen as related to substructural phenotype. Mol. Vis. 1999, 5, 28. [Google Scholar] [PubMed]
  25. Rudolf, M.; Clark, M.E.; Chimento, M.F.; Li, C.M.; Medeiros, N.E.; Curcio, C.A. Prevalence and morphology of druse types in the macula and periphery of eyes with age-related maculopathy. Invest. Ophthalmol. Vis. Sci. 2008, 49, 1200–1209. [Google Scholar] [CrossRef]
  26. Mullins, R.F.; Russell, S.R.; Anderson, D.H.; Hageman, G.S. 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. [Google Scholar] [CrossRef]
  27. Klein, R.; Davis, M.D.; Magli, Y.L.; Segal, P.; Klein, B.E.; Hubbard, L. The Wisconsin age-related maculopathy grading system. Ophthalmology 1991, 98, 1128–1134. [Google Scholar] [CrossRef]
  28. Miao, H.; Tao, Y.; Li, X.X. Inflammatory cytokines in aqueous humor of patients with choroidal neovascularization. Mol. Vis. 2012, 18, 574–580. [Google Scholar]
  29. McLeod, D.S.; Grebe, R.; Bhutto, I.; Merges, C.; Baba, T.; Lutty, G.A. Relationship between RPE and choriocapillaris in age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 2009, 50, 4982–4991. [Google Scholar] [CrossRef]
  30. Toy, B.C.; Krishnadev, N.; Indaram, M.; Cunningham, D.; Cukras, C.A.; Chew, E.Y.; Wong, W.T. Drusen regression is associated with local changes in fundus autofluorescence in intermediate age-related macular degeneration. Am. J. Ophthalmol. 2013, 156, 532–542.e531. [Google Scholar] [CrossRef]
  31. McHarg, S.; Clark, S.J.; Day, A.J.; Bishop, P.N. Age-related macular degeneration and the role of the complement system. Mol. Immunol. 2015, 67, 43–50. [Google Scholar] [CrossRef] [PubMed]
  32. Velilla, S.; García-Medina, J.J.; García-Layana, A.; Dolz-Marco, R.; Pons-Vázquez, S.; Pinazo-Durán, M.D.; Gómez-Ulla, F.; Arévalo, J.F.; Díaz-Llopis, M.; Gallego-Pinazo, R. Smoking and age-related macular degeneration: Review and update. J. Ophthalmol. 2013, 2013, 895147. [Google Scholar] [CrossRef] [PubMed]
  33. Risk Factors Associated with Age-Related Macular Degeneration. A case-control study in the age-related eye disease study: Age-Related Eye Disease Study Report Number 3. Ophthalmology 2000, 107, 2224–2232. [Google Scholar] [CrossRef]
  34. Mozaffarieh, M.; Sacu, S.; Wedrich, A. The role of the carotenoids, lutein and zeaxanthin, in protecting against age-related macular degeneration: A review based on controversial evidence. Nutr. J. 2003, 2, 20. [Google Scholar] [CrossRef] [PubMed]
  35. Merle, B.M.J.; Colijn, J.M.; Cougnard-Gregoire, A.; de Koning-Backus, A.P.M.; Delyfer, M.N.; Kiefte-de Jong, J.C.; Meester-Smoor, M.; Feart, C.; Verzijden, T.; Samieri, C.; et al. Mediterranean Diet and Incidence of Advanced Age-Related Macular Degeneration: The EYE-RISK Consortium. Ophthalmology 2019, 126, 381–390. [Google Scholar] [CrossRef] [PubMed]
  36. Chakravarthy, U.; Wong, T.Y.; Fletcher, A.; Piault, E.; Evans, C.; Zlateva, G.; Buggage, R.; Pleil, A.; Mitchell, P. Clinical risk factors for age-related macular degeneration: A systematic review and meta-analysis. BMC Ophthalmol. 2010, 10, 31. [Google Scholar] [CrossRef] [PubMed]
  37. Merle, B.M.; Delyfer, M.N.; Korobelnik, J.F.; Rougier, M.B.; Malet, F.; Féart, C.; Le Goff, M.; Peuchant, E.; Letenneur, L.; Dartigues, J.F.; et al. High concentrations of plasma n3 fatty acids are associated with decreased risk for late age-related macular degeneration. J. Nutr. 2013, 143, 505–511. [Google Scholar] [CrossRef]
  38. Age-Related Eye Disease Study Research Group. A randomized, placebo-controlled, clinical trial of high-dose supplementation with vitamins C and E, beta carotene, and zinc for age-related macular degeneration and vision loss: AREDS report no. 8. Arch. Ophthalmol. 2001, 119, 1417–1436. [Google Scholar] [CrossRef]
  39. SanGiovanni, J.P.; Chew, E.Y.; Clemons, T.E.; Ferris, F.L., 3rd; Gensler, G.; Lindblad, A.S.; Milton, R.C.; Seddon, J.M.; Sperduto, R.D. The relationship of dietary carotenoid and vitamin A, E, and C intake with age-related macular degeneration in a case-control study: AREDS Report No. 22. Arch. Ophthalmol. 2007, 125, 1225–1232. [Google Scholar] [CrossRef]
  40. Geerlings, M.J.; de Jong, E.K.; den Hollander, A.I. The complement system in age-related macular degeneration: A review of rare genetic variants and implications for personalized treatment. Mol. Immunol. 2017, 84, 65–76. [Google Scholar] [CrossRef]
  41. Hammond, C.J.; Webster, A.R.; Snieder, H.; Bird, A.C.; Gilbert, C.E.; Spector, T.D. Genetic influence on early age-related maculopathy: A twin study. Ophthalmology 2002, 109, 730–736. [Google Scholar] [CrossRef] [PubMed]
  42. Meyers, S.M.; Greene, T.; Gutman, F.A. A twin study of age-related macular degeneration. Am. J. Ophthalmol. 1995, 120, 757–766. [Google Scholar] [CrossRef]
  43. Seddon, J.M.; Cote, J.; Page, W.F.; Aggen, S.H.; Neale, M.C. The US twin study of age-related macular degeneration: Relative roles of genetic and environmental influences. Arch. Ophthalmol. 2005, 123, 321–327. [Google Scholar] [CrossRef]
  44. Chen, H.; Yu, K.-D.; Xu, G.-Z. Association between variant Y402H in age-related macular degeneration (AMD) susceptibility gene CFH and treatment response of AMD: A meta-analysis. PLoS ONE 2012. [Google Scholar] [CrossRef] [PubMed]
  45. Tong, Y.; Liao, J.; Zhang, Y.; Zhou, J.; Zhang, H.; Mao, M. LOC387715/HTRA1 gene polymorphisms and susceptibility to age-related macular degeneration: A HuGE review and meta-analysis. Mol. Vis. 2010, 16, 1958. [Google Scholar] [PubMed]
  46. Rivera, A.; Fisher, S.A.; Fritsche, L.G.; Keilhauer, C.N.; Lichtner, P.; Meitinger, T.; Weber, B.H.F. Hypothetical LOC387715 is a second major susceptibility gene for age-related macular degeneration, contributing independently of complement factor H to disease risk. Hum. Mol. Genet. 2005, 14, 3227–3236. [Google Scholar] [CrossRef]
  47. Jakobsdottir, J.; Conley, Y.P.; Weeks, D.E.; Mah, T.S.; Ferrell, R.E.; Gorin, M.B. Susceptibility genes for age-related maculopathy on chromosome 10q26. Am. J. Hum. Genet. 2005, 77, 389–407. [Google Scholar] [CrossRef]
  48. Edwards, A.O.; Ritter, R., 3rd; Abel, K.J.; Manning, A.; Panhuysen, C.; Farrer, L.A. Complement factor H polymorphism and age-related macular degeneration. Science 2005, 308, 421–424. [Google Scholar] [CrossRef]
  49. Horie-Inoue, K.; Inoue, S. Genomic aspects of age-related macular degeneration. Biochem. Biophys. Res. Commun. 2014, 452, 263–275. [Google Scholar] [CrossRef]
  50. Hageman, G.S.; Anderson, D.H.; Johnson, L.V.; Hancox, L.S.; Taiber, A.J.; Hardisty, L.I.; Hageman, J.L.; Stockman, H.A.; Borchardt, J.D.; Gehrs, K.M.; et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc. Natl. Acad. Sci. USA 2005, 102, 7227–7232. [Google Scholar] [CrossRef]
  51. Okemefuna, A.I.; Nan, R.; Miller, A.; Gor, J.; Perkins, S.J. Complement factor H binds at two independent sites to C-reactive protein in acute phase concentrations. J. Biol. Chem. 2010, 285, 1053–1065. [Google Scholar] [CrossRef]
  52. Forneris, F.; Wu, J.; Xue, X.; Ricklin, D.; Lin, Z.; Sfyroera, G.; Tzekou, A.; Volokhina, E.; Granneman, J.C.; Hauhart, R.; et al. Regulators of complement activity mediate inhibitory mechanisms through a common C3b-binding mode. Embo. J. 2016, 35, 1133–1149. [Google Scholar] [CrossRef]
  53. Sofat, R.; Mangione, P.P.; Gallimore, J.R.; Hakobyan, S.; Hughes, T.R.; Shah, T.; Goodship, T.; D’Aiuto, F.; Langenberg, C.; Wareham, N.; et al. Distribution and determinants of circulating complement factor H concentration determined by a high-throughput immunonephelometric assay. J. Immunol. Methods 2013, 390, 63–73. [Google Scholar] [CrossRef] [PubMed]
  54. Zipfel, P.F.; Lauer, N.; Skerka, C. The role of complement in AMD. Adv. Exp. Med. Biol. 2010, 703, 9–24. [Google Scholar] [CrossRef] [PubMed]
  55. Fritsche, L.G.; Chen, W.; Schu, M.; Yaspan, B.L.; Yu, Y.; Thorleifsson, G.; Zack, D.J.; Arakawa, S.; Cipriani, V.; Ripke, S.; et al. Seven new loci associated with age-related macular degeneration. Nat. Genet. 2013, 45, 433–439, 439e431-432. [Google Scholar] [CrossRef] [PubMed]
  56. 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. 2002, 134, 411–431. [Google Scholar] [CrossRef]
  57. Donoso, L.A.; Kim, D.; Frost, A.; Callahan, A.; Hageman, G. The role of inflammation in the pathogenesis of age-related macular degeneration. Surv. Ophthalmol. 2006, 51, 137–152. [Google Scholar] [CrossRef]
  58. Park, Y.G.; Park, Y.S.; Kim, I.B. Complement System and Potential Therapeutics in Age-Related Macular Degeneration. Int. J. Mol. Sci. 2021, 22, 6851. [Google Scholar] [CrossRef]
  59. Boyer, D.S.; Schmidt-Erfurth, U.; van Lookeren Campagne, M.; Henry, E.C.; Brittain, C. The Pathophysiology of Geographic Atrophy Secondary to Age-Related Macular Degeneration and the Complement Pathway as a Therapeutic Target. Retina 2017, 37, 819–835. [Google Scholar] [CrossRef]
  60. Schramm, E.C.; Clark, S.J.; Triebwasser, M.P.; Raychaudhuri, S.; Seddon, J.; Atkinson, J.P. Genetic variants in the complement system predisposing to age-related macular degeneration: A review. Mol. Immunol. 2014, 61, 118–125. [Google Scholar] [CrossRef]
  61. Morgan, B.P.; Walters, D.; Serna, M.; Bubeck, D. Terminal complexes of the complement system: New structural insights and their relevance to function. Immunol. Rev. 2016, 274, 141–151. [Google Scholar] [CrossRef] [PubMed]
  62. Qin, S.; Dong, N.; Yang, M.; Wang, J.; Feng, X.; Wang, Y. Complement Inhibitors in Age-Related Macular Degeneration: A Potential Therapeutic Option. J. Immunol. Res. 2021, 2021, 9945725. [Google Scholar] [CrossRef] [PubMed]
  63. Kim, B.J.; Mastellos, D.C.; Li, Y.; Dunaief, J.L.; Lambris, J.D. Targeting complement components C3 and C5 for the retina: Key concepts and lingering questions. Prog. Retin. Eye Res. 2021, 83, 100936. [Google Scholar] [CrossRef] [PubMed]
  64. Harboe, M.; Ulvund, G.; Vien, L.; Fung, M.; Mollnes, T.E. The quantitative role of alternative pathway amplification in classical pathway induced terminal complement activation. Clin. Exp. Immunol. 2004, 138, 439–446. [Google Scholar] [CrossRef]
  65. Kim, S.J.; Kim, J.; Lee, J.; Cho, S.Y.; Kang, H.J.; Kim, K.Y.; Jin, D.K. Intravitreal human complement factor H in a rat model of laser-induced choroidal neovascularisation. Br J. Ophthalmol. 2013, 97, 367–370. [Google Scholar] [CrossRef]
  66. Bora, N.S.; Jha, P.; Lyzogubov, V.V.; Kaliappan, S.; Liu, J.; Tytarenko, R.G.; Fraser, D.A.; Morgan, B.P.; Bora, P.S. Recombinant membrane-targeted form of CD59 inhibits the growth of choroidal neovascular complex in mice. J. Biol. Chem. 2010, 285, 33826–33833. [Google Scholar] [CrossRef]
  67. Nozaki, M.; Raisler, B.J.; Sakurai, E.; Sarma, J.V.; Barnum, S.R.; Lambris, J.D.; Chen, Y.; Zhang, K.; Ambati, B.K.; Baffi, J.Z.; et al. Drusen complement components C3a and C5a promote choroidal neovascularization. Proc. Natl. Acad. Sci. USA 2006, 103, 2328–2333. [Google Scholar] [CrossRef]
  68. Lundh von Leithner, P.; Kam, J.H.; Bainbridge, J.; Catchpole, I.; Gough, G.; Coffey, P.; Jeffery, G. Complement factor h is critical in the maintenance of retinal perfusion. Am. J. Pathol. 2009, 175, 412–421. [Google Scholar] [CrossRef]
  69. Rohrer, B.; Long, Q.; Coughlin, B.; Wilson, R.B.; Huang, Y.; Qiao, F.; Tang, P.H.; Kunchithapautham, K.; Gilkeson, G.S.; Tomlinson, S. A targeted inhibitor of the alternative complement pathway reduces angiogenesis in a mouse model of age-related macular degeneration. Invest. Ophthalmol. Vis. Sci. 2009, 50, 3056–3064. [Google Scholar] [CrossRef]
  70. Heesterbeek, T.J.; Lechanteur, Y.T.E.; Lorés-Motta, L.; Schick, T.; Daha, M.R.; Altay, L.; Liakopoulos, S.; Smailhodzic, D.; den Hollander, A.I.; Hoyng, C.B.; et al. Complement Activation Levels Are Related to Disease Stage in AMD. Invest. Ophthalmol. Vis. Sci. 2020, 61, 18. [Google Scholar] [CrossRef]
  71. Winkler, T.W.; Grassmann, F.; Brandl, C.; Kiel, C.; Günther, F.; Strunz, T.; Weidner, L.; Zimmermann, M.E.; Korb, C.A.; Poplawski, A.; et al. Genome-wide association meta-analysis for early age-related macular degeneration highlights novel loci and insights for advanced disease. BMC Med. Genom. 2020, 13, 120. [Google Scholar] [CrossRef]
  72. Mullins, R.F.; Schoo, D.P.; Sohn, E.H.; Flamme-Wiese, M.J.; Workamelahu, G.; Johnston, R.M.; Wang, K.; Tucker, B.A.; Stone, E.M. The membrane attack complex in aging human choriocapillaris: Relationship to macular degeneration and choroidal thinning. Am. J. Pathol. 2014, 184, 3142–3153. [Google Scholar] [CrossRef] [PubMed]
  73. Keenan, T.D.; Toso, M.; Pappas, C.; Nichols, L.; Bishop, P.N.; Hageman, G.S. Assessment of Proteins Associated With Complement Activation and Inflammation in Maculae of Human Donors Homozygous Risk at Chromosome 1 CFH-to-F13B. Invest. Ophthalmol. Vis. Sci. 2015, 56, 4870–4879. [Google Scholar] [CrossRef] [PubMed]
  74. Whitmore, S.S.; Sohn, E.H.; Chirco, K.R.; Drack, A.V.; Stone, E.M.; Tucker, B.A.; Mullins, R.F. Complement activation and choriocapillaris loss in early AMD: Implications for pathophysiology and therapy. Prog. Retin. Eye Res. 2015, 45, 1–29. [Google Scholar] [CrossRef]
  75. Sohn, J.H.; Kaplan, H.J.; Suk, H.J.; Bora, P.S.; Bora, N.S. Chronic low level complement activation within the eye is controlled by intraocular complement regulatory proteins. Invest. Ophthalmol. Vis. Sci. 2000, 41, 3492–3502. [Google Scholar] [PubMed]
  76. Lohman, R.-J.; Hamidon, J.K.; Reid, R.C.; Rowley, J.A.; Yau, M.-K.; Halili, M.A.; Nielsen, D.S.; Lim, J.; Wu, K.-C.; Loh, Z.; et al. Exploiting a novel conformational switch to control innate immunity mediated by complement protein C3a. Nat. Commun. 2017, 8, 351. [Google Scholar] [CrossRef]
  77. Landowski, M.; Kelly, U.; Klingeborn, M.; Groelle, M.; Ding, J.D.; Grigsby, D.; Bowes Rickman, C. Human complement factor H Y402H polymorphism causes an age-related macular degeneration phenotype and lipoprotein dysregulation in mice. Proc. Natl. Acad. Sci. USA 2019, 116, 3703–3711. [Google Scholar] [CrossRef]
  78. Acar İ, E.; Lores-Motta, L.; Colijn, J.M.; Meester-Smoor, M.A.; Verzijden, T.; Cougnard-Gregoire, A.; Ajana, S.; Merle, B.M.J.; de Breuk, A.; Heesterbeek, T.J.; et al. Integrating Metabolomics, Genomics, and Disease Pathways in Age-Related Macular Degeneration: The EYE-RISK Consortium. Ophthalmology 2020, 127, 1693–1709. [Google Scholar] [CrossRef]
  79. Zhang, Y.; Gordon, S.M.; Xi, H.; Choi, S.; Paz, M.A.; Sun, R.; Yang, W.; Saredy, J.; Khan, M.; Remaley, A.T.; et al. HDL subclass proteomic analysis and functional implication of protein dynamic change during HDL maturation. Redox Biol. 2019, 24, 101222. [Google Scholar] [CrossRef]
  80. Shaw, P.X.; Zhang, L.; Zhang, M.; Du, H.; Zhao, L.; Lee, C.; Grob, S.; Lim, S.L.; Hughes, G.; Lee, J.; et al. Complement factor H genotypes impact risk of age-related macular degeneration by interaction with oxidized phospholipids. Proc. Natl. Acad. Sci. USA 2012, 109, 13757–13762. [Google Scholar] [CrossRef]
  81. Ferrington, D.A.; Ebeling, M.C.; Kapphahn, R.J.; Terluk, M.R.; Fisher, C.R.; Polanco, J.R.; Roehrich, H.; Leary, M.M.; Geng, Z.; Dutton, J.R.; et al. Altered bioenergetics and enhanced resistance to oxidative stress in human retinal pigment epithelial cells from donors with age-related macular degeneration. Redox Biol. 2017, 13, 255–265. [Google Scholar] [CrossRef]
  82. Sivapathasuntharam, C.; Hayes, M.J.; Shinhmar, H.; Kam, J.H.; Sivaprasad, S.; Jeffery, G. Complement factor H regulates retinal development and its absence may establish a footprint for age related macular degeneration. Sci. Rep. 2019, 9, 1082. [Google Scholar] [CrossRef] [PubMed]
  83. Ferrington, D.A.; Kapphahn, R.J.; Leary, M.M.; Atilano, S.R.; Terluk, M.R.; Karunadharma, P.; Chen, G.K.; Ratnapriya, R.; Swaroop, A.; Montezuma, S.R.; et al. Increased retinal mtDNA damage in the CFH variant associated with age-related macular degeneration. Exp. Eye Res. 2016, 145, 269–277. [Google Scholar] [CrossRef] [PubMed]
  84. Nashine, S.; Chwa, M.; Kazemian, M.; Thaker, K.; Lu, S.; Nesburn, A.; Kuppermann, B.D.; Kenney, M.C. Differential Expression of Complement Markers in Normal and AMD Transmitochondrial Cybrids. PLoS One 2016, 11, e0159828. [Google Scholar] [CrossRef] [PubMed]
  85. Sinha, D.; Valapala, M.; Shang, P.; Hose, S.; Grebe, R.; Lutty, G.A.; Zigler, J.S., Jr.; Kaarniranta, K.; Handa, J.T. Lysosomes: Regulators of autophagy in the retinal pigmented epithelium. Exp. Eye Res. 2016, 144, 46–53. [Google Scholar] [CrossRef] [PubMed]
  86. Hyttinen, J.M.T.; Viiri, J.; Kaarniranta, K.; Błasiak, J. Mitochondrial quality control in AMD: Does mitophagy play a pivotal role? Cell Mol. Life Sci. 2018, 75, 2991–3008. [Google Scholar] [CrossRef] [PubMed]
  87. Mulligan, K.; Seabury, S.A.; Dugel, P.U.; Blim, J.F.; Goldman, D.P.; Humayun, M.S. Economic Value of Anti-Vascular Endothelial Growth Factor Treatment for Patients With Wet Age-Related Macular Degeneration in the United States. JAMA Ophthalmol. 2020, 138, 40–47. [Google Scholar] [CrossRef] [PubMed]
  88. Rasmussen, A.; Sander, B. Long-term longitudinal study of patients treated with ranibizumab for neovascular age-related macular degeneration. Curr. Opin. Ophthalmol. 2014, 25, 158–163. [Google Scholar] [CrossRef]
  89. Yehoshua, Z.; de Amorim Garcia Filho, C.A.; Nunes, R.P.; Gregori, G.; Penha, F.M.; Moshfeghi, A.A.; Zhang, K.; Sadda, S.; Feuer, W.; Rosenfeld, P.J. Systemic complement inhibition with eculizumab for geographic atrophy in age-related macular degeneration: The COMPLETE study. Ophthalmology 2014, 121, 693–701. [Google Scholar] [CrossRef]
  90. Clark, S.J.; Bishop, P.N. The eye as a complement dysregulation hotspot. Semin. Immunopathol. 2018, 40, 65–74. [Google Scholar] [CrossRef]
  91. Liao, D.S.; Grossi, F.V.; El Mehdi, D.; Gerber, M.R.; Brown, D.M.; Heier, J.S.; Wykoff, C.C.; Singerman, L.J.; Abraham, P.; Grassmann, F.; et al. Complement C3 Inhibitor Pegcetacoplan for Geographic Atrophy Secondary to Age-Related Macular Degeneration: A Randomized Phase 2 Trial. Ophthalmology 2020, 127, 186–195. [Google Scholar] [CrossRef] [PubMed]
  92. Jaffe, G.J.; Westby, K.; Csaky, K.G.; Monés, J.; Pearlman, J.A.; Patel, S.S.; Joondeph, B.C.; Randolph, J.; Masonson, H.; Rezaei, K.A. C5 Inhibitor Avacincaptad Pegol for Geographic Atrophy Due to Age-Related Macular Degeneration: A Randomized Pivotal Phase 2/3 Trial. Ophthalmology 2021, 128, 576–586. [Google Scholar] [CrossRef] [PubMed]
  93. Lyzogubov, V.V.; Bora, N.S.; Tytarenko, R.G.; Bora, P.S. Polyethylene glycol induced mouse model of retinal degeneration. Exp. Eye Res. 2014, 127, 143–152. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clinical fundus photograph of the macula and fovea and their dimensions.
Figure 1. Clinical fundus photograph of the macula and fovea and their dimensions.
Biomolecules 13 00832 g001
Figure 2. (Left) Color fundus photograph of a patient with intermediate AMD demonstrating soft drusen, with indistinct borders. (Right) Optical coherence tomography (OCT) imaging of the central macula in the same patient demonstrating an optical section of all retinal layers. Notice the hyperreflective deposits between the RPE (top arrow)and the BM (bottom arrow), which represent drusen from the left panel.
Figure 2. (Left) Color fundus photograph of a patient with intermediate AMD demonstrating soft drusen, with indistinct borders. (Right) Optical coherence tomography (OCT) imaging of the central macula in the same patient demonstrating an optical section of all retinal layers. Notice the hyperreflective deposits between the RPE (top arrow)and the BM (bottom arrow), which represent drusen from the left panel.
Biomolecules 13 00832 g002
Figure 3. Complement pathways. The alternative pathway is important in AMD. Green triangles demonstrate regulators/inhibitors of the specific pathways. Malfunction will lead to overactivation of complement and, ultimately, cell damage.
Figure 3. Complement pathways. The alternative pathway is important in AMD. Green triangles demonstrate regulators/inhibitors of the specific pathways. Malfunction will lead to overactivation of complement and, ultimately, cell damage.
Biomolecules 13 00832 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Piri, N.; Kaplan, H.J. Role of Complement in the Onset of Age-Related Macular Degeneration. Biomolecules 2023, 13, 832. https://doi.org/10.3390/biom13050832

AMA Style

Piri N, Kaplan HJ. Role of Complement in the Onset of Age-Related Macular Degeneration. Biomolecules. 2023; 13(5):832. https://doi.org/10.3390/biom13050832

Chicago/Turabian Style

Piri, Niloofar, and Henry J. Kaplan. 2023. "Role of Complement in the Onset of Age-Related Macular Degeneration" Biomolecules 13, no. 5: 832. https://doi.org/10.3390/biom13050832

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop