Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Circulating Autoantibodies in Age-Related Macular Degeneration Recognize Human Macular Tissue Antigens Implicated in Autophagy, Immunomodulation, and Protection from Oxidative Stress and Apoptosis

  • Alessandro Iannaccone ,

    aiannacc@yahoo.com

    Affiliation Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Francesco Giorgianni,

    Affiliation Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • David D. New,

    Affiliation Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • T. J. Hollingsworth,

    Affiliation Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Allison Umfress,

    Current address: Vanderbilt University School of Medicine, Nashville, TN, United States of America

    Affiliation Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Albert H. Alhatem,

    Affiliation Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Indira Neeli,

    Affiliations Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America, Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Nataliya I. Lenchik,

    Affiliations Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America, Department of Internal Medicine/Endocrinology, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Barbara J. Jennings,

    Affiliation Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Jorge I. Calzada,

    Affiliations Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America, Charles Retina Institute, Memphis, TN, United States of America

  • Suzanne Satterfield,

    Affiliation Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Dennis Mathews,

    Affiliations Eye Specialty Group, Memphis, TN, United States of America, Southern College of Optometry, Memphis, TN, United States of America

  • Rocio I. Diaz,

    Affiliations Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America, Charles Retina Institute, Memphis, TN, United States of America

  • Tamara Harris,

    Affiliation National Institute on Aging, NIH, Bethesda, MD, United States of America

  • Karen C. Johnson,

    Affiliation Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Steve Charles,

    Affiliations Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, United States of America, Charles Retina Institute, Memphis, TN, United States of America

  • Stephen B. Kritchevsky,

    Affiliations Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America, Sticht Center on Aging, Wake Forest University, Winston-Salem, NC, United States of America

  • Ivan C. Gerling,

    Affiliation Department of Internal Medicine/Endocrinology, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Sarka Beranova-Giorgianni,

    Affiliation Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States of America

  • Marko Z. Radic,

    Affiliation Department of Microbiology, Immunology & Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States of America

  •  [ ... ],
  • Health ABC study

    Membership of the Health ABC study is provided in the Acknowledgments.

  • [ view all ]
  • [ view less ]

Abstract

Background

We investigated sera from elderly subjects with and without age-related macular degeneration (AMD) for presence of autoantibodies (AAbs) against human macular antigens and characterized their identity.

Methods

Sera were collected from participants in the Age-Related Maculopathy Ancillary (ARMA) Study, a cross-sectional investigation ancillary to the Health ABC Study, enriched with participants from the general population. The resulting sample (mean age: 79.2±3.9 years old) included subjects with early to advanced AMD (n = 131) and controls (n = 231). Sera were tested by Western blots for immunoreactive bands against human donor macular tissue homogenates. Immunoreactive bands were identified and graded, and odds ratios (OR) calculated. Based on these findings, sera were immunoprecipitated, and subjected to 2D gel electrophoresis (GE). Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to identify the targets recognized by circulating AAbs seen on 2D-GE, followed by ELISAs with recombinant proteins to confirm LC-MS/MS results, and quantify autoreactivities.

Results

In AMD, 11 immunoreactive bands were significantly more frequent and 13 were significantly stronger than in controls. Nine of the more frequent bands also showed stronger reactivity. OR estimates ranged between 4.06 and 1.93, and all clearly excluded the null value. Following immunoprecipitation, 2D-GE and LC-MS/MS, five of the possible autoreactivity targets were conclusively identified: two members of the heat shock protein 70 (HSP70) family, HSPA8 and HSPA9; another member of the HSP family, HSPB4, also known as alpha-crystallin A chain (CRYAA); Annexin A5 (ANXA5); and Protein S100-A9, also known as calgranulin B that, when complexed with S100A8, forms calprotectin. ELISA testing with recombinant proteins confirmed, on average, significantly higher reactivities against all targets in AMD samples compared to controls.

Conclusions

Consistent with other evidence supporting the role of inflammation and the immune system in AMD pathogenesis, AAbs were identified in AMD sera, including early-stage disease. Identified targets may be mechanistically linked to AMD pathogenesis because the identified proteins are implicated in autophagy, immunomodulation, and protection from oxidative stress and apoptosis. In particular, a role in autophagy activation is shared by all five autoantigens, raising the possibility that the detected AAbs may play a role in AMD via autophagy compromise and downstream activation of the inflammasome. Thus, we propose that the detected AAbs provide further insight into AMD pathogenesis and have the potential to contribute to disease biogenesis and progression.

Introduction

Age-related macular degeneration (AMD) is a highly prevalent, multifactorial, polygenic and complex retinal degenerative disease [1, 2] in which characteristic deposits, termed drusen, develop mostly under the retinal pigment epithelium (RPE) at the interface with subjacent choroidal circulation, and clinically visible RPE changes (i.e., migration and clustering, appearing clinically as macular focal hyperpigmentation, and loss, resulting in focal drop-out, appearing clinically as hypopigmentation) occur [36]. It is estimated that approximately one in three elderly over the age of 75 develops early AMD, which progresses to advanced AMD and affects approximately 10% of the elderly in this age range. With early AMD, vision loss is still minimal [1]. In advanced AMD, choroidal neovascularization (nvAMD) and/or patches of RPE loss [geographic atrophy (GA)] develop, leading to photoreceptor and severe central vision loss, which often results in legal blindness and significant compromise of the quality of life of affected patients. Characterization of drusen and RPE changes helps predict likelihood of disease progression [711], but the underlying mechanisms leading up to these changes and to AMD progression remain incompletely understood. Thus, identification of factors and mechanisms that favor the development of AMD and its progression from early to advanced AMD would prove invaluable in mitigating the impact of AMD on the elderly.

It is now widely accepted that inflammation and the immune system play important roles in AMD pathogenesis [1226]. In AMD, the choriocapillaris and the RPE are the object of antibody (Ab)-mediated complement deposition, and accumulating immune complexes have been shown to contribute to RPE degeneration, and drusen formation [3, 27]. Dendritic cells (DCs) infiltrate the Bruch’s membrane (BM), project processes inside drusen cores, and break the blood-retinal barrier, which leads to RPE loss due to inflammatory damage. RPE loss coincides with, and is proportional to drusen formation [3]. Drusen may act as a reservoir of autoantigens that, upon presentation by DC and other immunocompetent cells, may drive autoimmune-mediated damage to macular tissues [3]. Thus, inflammatory and immune-mediated events are likely at play in drusen biogenesis and RPE loss in AMD.

Accordingly, genetic factors linked to inflammation have been clearly associated both with probability of having AMD and progressing to advanced AMD [2848]. Thus, genetic evidence argues that AMD can be considered at least in part as a congenital predisposition to defective modulation of inflammation with late-onset manifestations, likely modulated by a number of intervening factors. While statistical approaches have consistently confirmed the role of genetic factors in AMD [39, 4951], when population samples without clear-cut, preexisting advanced AMD are included in the analyses, these methods do not achieve ideal discrimination of AMD even when modifiable environmental factors are accounted for [44]. Therefore, AMD routine genotyping is not presently recommended [52]. Gene-environment interactions have also been demonstrated [37, 5355], highlighting the complexity of AMD. Consistent with this complexity, studies have shown that genomic biomarkers in combination with autoimmune serum biomarkers such as anti-carboxyethylpyrrole (CEP) auto-Abs (AAbs) are better predictors of AMD than genomic biomarkers alone [56]. Thus, there is much value in characterizing further the autoimmune component of AMD.

We have proposed that AAbs could be very useful biomarkers and play a mechanistic role in AMD [57]–a view that is shared by others [1214, 17, 24, 56, 58, 59]. Here, we present evidence for AAbs against human macular tissue antigens in AMD sera also in participants with early disease stages, and we identify five macular autoantigens, all of which are potentially related to AMD pathogenesis. These findings raise the possibility that these AAbs are not mere after-the-fact biomarkers but, in fact, active contributors to disease development and progression.

Methods

Participant Population

All investigations were conducted in compliance with the Declaration of Helsinki and following approval by the Institutional Review Board of the University of Tennessee Health Science Center. Our study involved primarily participants in the Health ABC Age-Related Maculopathy Ancillary (ARMA) Study, and details about its participants have been published [6063]. In brief, the resulting study population included 362 participants (mean age: 79.2 ± 3.9 years old; range: 63–91 years old; 54% females; 81% self-reported Whites), 131 of whom with AMD and 231 unaffected. Based on the original AREDS classification [7], 92 AMD cases were early to intermediate stage (category 3), and 39 advanced (category 4), 70% of which nvAMD. All participants were chosen by design to be free of diabetes >2 year duration and of diabetic retinopathy, glaucoma, inflammatory eye diseases, retinal conditions other than AMD that could confound the study (e.g., retinal detachment), systemic autoimmune conditions, or any other condition that was deemed a potential confounder. Further details are provided in S1 Supporting Information.

After obtaining written informed consent, serum samples were collected from all eligible participants, stored at –80°C according to standard methods, and subsequently analyzed by Western blots (WBs). A subset of serum aliquots also underwent immunoprecipitation (IP) and 2D gel electrophoresis (2D-GE). Putative IP autoantigens were identified via liquid chromatography tandem mass spectrometry (LC-MS/MS) and confirmed by ELISA.

Human Tissue Harvesting, Western Blotting and Immunoreactivity Analysis

Sera were assessed for reactivity against homogenates comprising neuroretina (nRet), RPE, BM and choroid (Ch), harvested from the macular region of normal human donor eyes (≥60 yo) [64], as detailed in S1 Supporting Information and in S1 Fig. Human donor eyes used in these experiments were obtained from cadaver eyes by the Mid-South Eye Bank, Memphis, TN, and by the National Disease Research Interchange (NDRI), Philadelphia, PA, and provided to us as anonymous specimens. Homogenates from various donor eyes were pooled, and used as source of antigens for WBs. Details on the WB methodology are also provided in S1 Supporting Information. In brief, three films were generated for each blot with exposure times of 5, 15, and 30 sec. Films were then evaluated to determine the presence of bound Ab in a semi-quantitative manner and graded. WBs were graded by trained lab staff for presence and intensity of immunoreactive (IR) bands. To minimize bias, staff was masked to disease status of the samples. Details on the methods and technique used to grade the WB gels are provided in S1 Supporting Information. The method was developed and tested for reproducibility, based on a conceptually similar scale previously developed to assess other images [65] (see S1 Table and S2 Fig). IR values were recorded in a database and analyzed as described in S1 Supporting Information.

Immunoprecipitation (IP), 2D Gel Electrophoresis (2D-GE) and Liquid Chromatography Tandem Mass Spectrometry (LC-MS/MS)

A representative subset of AMD (n = 18) and control samples (n = 15) was used to identify candidate protein autoantigens that are preferentially targeted by AAbs from AMD sera. All AMD subjects in this subset had at least one eye with AREDS stage 3 disease [7], and half of them had at least one eye with advanced AMD. Control subjects included AREDS stages 0–2 [7]. Macular tissue homogenates were immunoprecipitated with AMD and control sera. The human macular antigens pulled down by IP from these AMD and control sera were separated on 2D-GE gels, stained by SYPRO-Ruby Gel Stain (BioRad) and analyzed with Progenesis SameSpots software (Nonlinear Dynamics, USA). This software not only aligns images, matches the same spots, and finds differentially expressed protein spots, but also adjusts for the loading differences across the set of gels by normalizing spot intensities to total gel stain intensity. Normalized spot volumes (quantity) are also calculated automatically by the software. 2DGE is a semi-quantitative method that determines the absolute levels of auto-reactivity. This semi-quantitative information from this discovery experiment was used to guide decisions on which proteins should be evaluated by quantitative assays (ELISA, see below) performed on the proteins identified by LC-MS/MS.

The proteins of interest were digested with trypsin as described previously [66] [67], and the digests were analyzed with LC-MS/MS performed on an ion trap tandem mass spectrometer. The proteins were identified via searches of the UniProt protein sequence database. Further details are provided below and in S1 Supporting Information.

Enzyme-Linked Immunosorbent Assays (ELISA) for confirmation of specific autoreactivities against detected antigens by circulating auto-antibodies

Aliquots of the serum samples used in the 2D-GE and LC-MS/MS experiments were used in ELISA to test their reactivity against recombinant proteins corresponding to the candidate target proteins identified by LC-MS/MS (see results). Immobilon IV 96-well plates (Millipore) were coated with recombinant human proteins and probed in duplicate with serum from either normal or AMD subjects for 2 hrs at room temperature. Plates were then washed and probed with HRP-conjugated anti-human IgG. Again, control subjects included AREDS stages 0–2 and all sera were from AMD participants grade ≥3 based on the original AREDS grading [7]. This selected sub-group of AMD participants had an AMD score of ≥4 based on the new simplified AREDS scoring system [8], and of 7–9 or higher (central GA and/or neovascular AMD) based on the new AREDS severity scale [9]. ELISA reactivities were measured with a microQuant Spectrophotometer (BioTek) and expressed as optical density values (OD450).

Statistical analyses

Each serum sample generated 100 data points (thus, 13,100 data points for the AMD serum sample group, and 23,100 for the control group). We accounted for multiple comparisons using the Benjamini-Hochberg (B-H) multiple test correction and p<0.05 [68, 69], as shown by Thissen et al [16]. In brief, the B-H correction limits the type-I error, or false discovery rate (FDR). Starting from an initial α = 0.05 cut-off, the B-H method allows for application of sequentially more stringent α values for each of the comparisons made until the observed p-value exceeds the critical B-H value (which, for our dataset was 0.0065). This method, utilized with large data arrays [70], avoids the overcorrection (and, thus, the risk of Type-II error) caused when classical methods (e.g., Bonferroni) are applied to exploratory datasets that require multiple comparisons.

Two levels of preplanned analysis were performed by comparing the entire AMD and control dataset via unpaired t-test for samples with unequal variances and by constructing 2x2 tables and computing odds ratios (ORs), 95% confidence intervals (CI), χ2 statistic, and p-values, for each significant IR band. Differences were ranked from the highest χ2 statistic to the lowest and the B-H multiple test correction method was applied until the significance cut-off defined by this method was no longer significant. Only more frequent and/or more intense IR bands meeting the B-H criteria were considered significant.

Sera exhibiting IR bands meeting these criteria were considered more likely to contain AAbs and were further analyzed by IP, 2D-GE and LC-MS/MS to identify potential autoantigens. Differences in 2D-GE spot intensity between control and AMD sera were quantified automatically by the Progenesis SameSpots software via one-way analysis of variance (ANOVA). To identify differentially abundant proteins, LC-MS/MS datasets were used to interrogate the UniProt protein sequence database (subset of human proteins) using the SEQUEST HT search engine (Proteome Discoverer 1.4 software suite, Thermo Scientific). Significance of the peptide spectrum matches was assigned via Percolator, a standard algorithm for LC-MS/MS data, using a threshold value of q = 0.01, which corresponds to an FDR of 1% for peptide match assignment. Additional details about this procedure are provided in S1 Supporting Information.

Lastly, ELISA reactivities were compared by means of 2-tailed, unpaired Student’s t-test (or equal or unequal variances, as appropriate) with α = 0.05 as cut-off for statistical significance also for these comparisons.

Results

AMD sera recognized a greater number of IR intervals of human macular antigens and exhibited stronger autoreactivity compared to control sera. More frequent IR was observed in AMD for the dataset as a whole (p = 0.02 x 10−8) and, after applying the B-H multiple test correction method to our data set, specifically for 11 distinct 2-kDa IR intervals. The more frequent IR bands clustered primarily in two areas, between 26 and 32 kDa (n = 3), between 36 and 50 kDa (n = 7), and at the 98–100 kDa interval. By the same method, greater IR intensity was observed for 13 levels of comparisons among the observed IR bands. Of these, 9 of 11 bands coincided with the intervals in which IR was also seen more frequently. Some bands were significantly more intense for various cut-off levels of comparison (e.g., the 40–42 kDa band showed greater IR for the <3 vs. ≥3 and <4 vs. ≥4 IR score comparisons). Two comparisons were significant uniquely for frequency of the IR band (38–40 kDa) and for IR intensity (88–90 kDa, <2 vs. ≥2 comparison), respectively. Fig 1 illustrates these results, by presenting point estimates of ORs and 95% CIs for the IR bands whose χ2 values met the B-H critical value, ranked by the latter. The lowest significance level that met the B-H critical value was a χ2 of 7.85, p = 0.0051. All IR intervals meeting the B-H criteria for inclusion were highly significant, with the 95% CIs excluding clearly the null (OR = 1.0) value and OR values ranging between 1.93 and 4.56.

thumbnail
Fig 1. Plot of odds ratios (ORs) and 95% CIs for immunoreactive (IR) bands detected by Western blot.

OR data points are ordered, top to bottom, from the highest χ2 statistic (32.46, p = 0.012 x 10−6) to the lowest (7.30, p = 0.0068) meeting the critical B-H value. All IR bands listed were significantly more intense and/or more common in AMD sera.

https://doi.org/10.1371/journal.pone.0145323.g001

AMD sera with the greatest and most frequent IR and controls were used to conduct additional IP, 2D-GE and LC-MS/MS experiments, which identified five potential targets of autoreactivity (Table 1): two members of the heat shock protein 70 (HSP70) family, HSPA8 and HSPA9; another member of the HSP family, HSPB4, also known as alpha-crystallin A chain (CRYAA); Annexin A5 (ANXA5); and Protein S100-A9 (S100A9), also known as calgranulin B that, when complexed with S100A8, forms calprotectin. LC-MS/MS spectra for some of the identified peptides are also illustrated in S3 Fig. A representative pair of gels illustrating an example of the differentially expressed spots on 2D-GE is shown in Fig 2. In this example, three of the aforementioned targets– HSPA8, ANXA5, and HSPB4 (CRYAA)–were confirmed among the immunoprecipitated macular lysate proteins that were upregulated in the AMD serum compared to the control serum and subsequently verified as such by ELISA criteria (see below). Note that additional spots that appear to be upregulated on 2D-GE for this one AMD serum are present. While it is possible that these spots too may represent meaningful autoreactivities, they were not confirmed as significant in the remaining subset of AMD samples subjected to 2D-GE and/or by ELISA testing and, thus, have not been considered further. Note also the different intensity of the autoreactivity for the three targets: HSPA8 is highest in this case of AMD, whereas ANXA5 and HSPB4/CRYAA were weaker. Other AMD sera (see below) exhibited much stronger reactivity for ANXA5 or for other targets. This inter-subject variability emphasizes the concept, proposed by Adamus et al. [59], of different “AAb signatures” that AMD patients may have.

thumbnail
Fig 2. Example of two dimensional gel electrophoresis (2D-GE) analyses comparing control vs. AMD sera.

The left panel shows a gel on which protein from human macular lysate immunoprecipitated by a control serum sample have been separated on two dimensions (by molecular weight and pI), compared to an AMD sample ran identically and simultaneously shown on the right. There are several spots on the 2D-GE AMD gel that appear different compared to the control one. Confirmed antigens recognized by the AAbs present in the serum of this one particular AMD participant included HSPA8, ANXA5 and HSPB4/CRYAA (arrows). See text for further discussion of these findings. Note also how the AMD gel exhibits a stronger “load” compared to the control gel, consistent with the fact that AMD sera tended to be more often autoreactive and thus, even when some autoreactivities were shared, AMD sera exhibited much stronger levels of reactivity, indicative of a larger amount and numbers of proteins pulled down by the IP process.

https://doi.org/10.1371/journal.pone.0145323.g002

The highly significant preferential binding of AMD sera by ELISA conclusively confirmed that HSPA8, HSPA9, HSPB4/CRYAA, ANXA5, and S100A9 are specific AAb targets in AMD (Fig 3): anti-HSPA8 autoreactivity was 0.54±0.04 in AMD samples (mean±SE) and 0.25±0.02 in control samples (p = 0.000003, 2-sided test for unequal variances), anti-HSPA9 autoreactivity was 0.52±0.06 in AMD samples and 0.27±0.02 in control samples (p = 0.0003, 2-sided test for unequal variances), anti-HSPB4/CRYAA autoreactivity was 0.43±0.02 in AMD samples and 0.29±0.02 in control samples (p = 0.00007, 2-sided test for equal variances), anti-ANXA5 autoreactivity was 0.44±0.03 in AMD samples and 0.24±0.01 in control samples (p = 0.0000001 2-sided test for unequal variances), and anti-S100A9 autoreactivity was 0.52±0.06 in AMD samples and 0.26±0.02 in control samples (p = 0.001, 2-sided test for unequal variances).

thumbnail
Fig 3. Scatterplots and bar graphs summarizing ELISA results.

All autoreactivities against the five antigens identified by LC-MS/MS (HSPA8, HSPA9, HSPB4/CRYAA, ANXA5, and S100A9) were confirmed as specifically directed against these targets, and were all significantly higher in AMD than in controls.

https://doi.org/10.1371/journal.pone.0145323.g003

Discussion

Our finding that multiple AAbs in AMD sera recognize human macular tissue antigens dovetails nicely with evidence that has accumulated over the past two decades [7175] for a role of autoimmunity in AMD. Following seminal studies that shed light on the inflammatory and immune-mediated component of drusen formation [3, 76, 77] and proteomics studies of drusen composition [78, 79], and following the discovery linking AMD to genetic variants involved in abnormal regulation of complement-mediated inflammatory processes [2848], the role of inflammation and of the immune system in AMD has been repeatedly confirmed [56, 58, 59, 8087] and has become widely accepted [1526]. It has been recently suggested that distinct AAb signatures may exist between early AMD, GA and nvAMD [59]. In summary, the concept of AMD as a disease that is at least in part immunologically driven has emerged [1214, 24].

The partial breakdown of the blood-retinal barrier that characterizes AMD eyes also at the early stages of the diseases, the accumulation of inflammatory debris and effectors at the sub-RPE levels, and the infiltration of DCs in drusen cores all concur to creating a favorable milieu to both an autoimmune response and an active role in situ of AAbs directed against macular antigens. With regard to the specific autoreactivities that we identified, there is a large body of evidence that supports the possibility that AAbs against HSPA8, HSPA9, HSPB4/CRYAA, ANXA5 and S100A9 could be of relevance to AMD pathogenesis. The many roles and functions of these five antigens in and outside the eye for these proteins that are directly relevant to our findings and to our working hypothesis are summarized in Table 2, based on both information reported in the UniProt database (http://www.uniprot.org/) and additional specific references from the literature [78, 88138]. HSPA8, HSPA9, HSPB4/CRYAA, ANXA5 and S100A9 are of particular interest due to their expression in human macular tissues, their upregulation in AMD donor eyes, their known role in autophagy, protection from oxidative stress and apoptosis, and/or immunomodulation.

thumbnail
Table 2. Summary of known roles and functions of the identified autoantigensa

https://doi.org/10.1371/journal.pone.0145323.t002

The potential importance of compromised autophagy in AMD and its role in causing age-related RPE damage and loss and sub- and intra-RPE deposit accumulation has been elegantly demonstrated in recent years in rats and mice with genetically compromised small HSP-dependent autophagy pathways [131, 132, 136, 138]. In brief, autophagy is a key regulator of intracellular nutrient and energy homeostasis, affects local immune responses, and regulates both endogenous activators as well as expression of components of the inflammasome. When autophagy is compromised, a build-up in reactive oxygen species and aggregates occurs, which in turn causes NLRP3 inflammasome activation as well. The NLRP3 inflammasome is part of the pattern recognition receptors (PRRs) family devoted to recognizing pathogen-associated and danger-associated molecular patterns (DAMPs and PAMPs, respectively), and has recently emerged as a likely role player in AMD pathogenesis [131]. In conditional knock-out mice with compromised autophagy in the RPE, Yao et al. have shown a multitude of age-related AMD-like phenotypic changes, as well intraretinal and sub-RPE inflammation, Bruch’s membrane disruption, and eventual damage and loss of overlying photoreceptors [132]. Thus, AAbs directed against targets implicated in autophagy control could exacerbate AMD at least in part also via a direct autophagy-mediated mechanism.

The interplay and crosstalk between protein homeostasis, autophagy, the proteasome, and HSPs in the pathogenesis of AMD has become increasingly appreciated over the past few years [139141] and has been recently reviewed in detail [142]. The particular role of HSPs as gatekeepers of proteolytic pathways in the RPE and the implications of the disruption of the HSP-mediated chaperone functions in the aging RPE with regard to regulation of autophagy, accumulation of oxidative stress-induced damage, protein aggregation, lipofuscinogenesis, and AMD etiology have also been reviewed [88]. In brief, HSPA8 and HSPA9 are members of the HSP70 family that is a key effector of chaperone-mediated autophagy, in the RPE as well as in neurons [88, 89]. In addition, HSP70s act as well as direct downregulators of the inflammatory response mediated by DCs and other innate immune system cells [130]. Anti-HSPA8 AAbs are implicated in the pathogenesis of both cancer-associated retinopathy (CAR) [94] and autoimmune hepatitis [95]. In neurons, HSPA8 is known to be involved in the lysosomal degradation of α-synuclein, which accumulates in Parkinson's disease and other neurodegenerative diseases [96], and in the degradation of the amyotrophic lateral sclerosis-linked mutant SOD1 protein [97, 98]. Genetic depletion of SOD1 is implicated in a murine model of AMD [99, 100], in which our group has very recently shown that anti-HSPA8 AAbs develop before overt AMD-like changes develop but at a stage when marked intraretinal inflammation is already present [New et al. Invest. Ophthalmol. Vis. Sci. 2015; 56: E-Abstract 3986]. Of direct relevance to AMD, HSPA8 is expressed in the retina and the RPE, in which it declines with non-pathologic aging [105], but is markedly upregulated in nvAMD tissues [106]. The specific role of HSPA8 as a chaperone-mediated autophagy regulator [88, 89] has been characterized recently, showing that it is mediated by its ATPase domain and that it is associated with induction of Akt and mTOR phosporylation [133, 134]. Elevated HSP levels have also been documented in the RPE of human AMD donor eyes [107]. Thus, AAbs against these HSPs could lead to compromised regulation of autophagy.

HSPB4, also known as CRYAA, has anti-apoptotic functions. In addition to the lens, HSPB4/CRYAA expression in the eye has been demonstrated also in nRet, RPE, BM and Ch tissues [101103]. An elevation of retinal HSPB4/CRYAA expression that protects photoreceptors from degeneration has been reported in the early stages of experimental autoimmune uveitis [104]. Both alphaA- (CRYAA) and alphaB- (CRYAB) crystallins, two members of the small HSP family, accumulate in the BM and subjacent choroidal connective tissue from human AMD tissues, and are abundant in drusen [78, 101, 108]. Compromised HSPB4/CRYAA function is predicted to exacerbate oxidative stress and apoptosis in the RPE, and boosting its function may offer a potential treatment for AMD and other retinal degenerative diseases [102, 103, 109111]. In Parkinson’s disease (PD), HSPB4 has been shown to be the most potent inhibitor, in an autophagy-independent fashion, of disease-causing protein aggregates induced by the C289G parkin E3-ubiquitin protein ligase (PARK2) mutation [135]. Thus, it can be envisioned how, in AMD, AAbs directed against HSPB4/CRYAA could compromise its functions and contribute to AMD-promoting RPE damage and loss, possibly by promoting formation of the intra- and/or extra-cellular aggregates that are seen in AMD and that can lead, independently of autophagy compromise, to NLRP3 inflammasome activation [136].

Annexin A5 (ANXA5) is an anti-apoptotic protein involved in the modulation of immune response repeatedly implicated in autoimmune diseases such as antiphospholipid syndrome, rheumatoid arthritis, lupus, type-1 diabetes, and autoimmune myocharditis [112115]. Of specific relevance to AMD, ANXA5 is expressed in retina and RPE, is upregulated in nvAMD tissues and, like HSPB4, is a documented component of drusen in AMD [106, 116]. Furthermore, AAbs against a related protein, Annexin II, have been found in cynomolgus monkeys with AMD [117]. Interestingly, ANXA5 has been shown to be an autophagy stimulator [137]. Thus, much like the anti-HSPA8, A9 and B4 AAbs, also anti-ANXA5 AAbs could in theory contribute to AMD pathogenesis at least in part via an autophagy-mediated mechanism.

Lastly, S100 calcium-binding protein A9 (S100A9), also known as calgranulin B, plays a prominent role in the regulation of inflammatory processes and immune response. S100A9 is most often complexed with S100A8 to form calprotectin, which too is part of a group of DAMP molecules that trigger inflammatory responses and stimulate innate immune cells via PRR binding that is implicated in a plethora on inflammatory and autoimmune conditions [128]. Individually, S100A9 has many intracellular and extracellular functions, including direct selective inflammatory stimulus-dependent S-nitrosylation of multiple targets, one of which is ANXA5, and acting as a potent amplifier of inflammation in autoimmunity [118127]. Of direct relevance to AMD, S100 proteins have been recently identified as a highly expressed macular protein in human donor eyes by differential proteomics studies [129], and are elevated in the BM/Ch complex of human AMD [108]. Of further interest, calprotectin has been shown to induce both apoptosis and autophagy via a ROS-mediated cross-talk between mitochondria and lysosomes, a dual property that is shared also by other factors [122]. Thus, several mechanisms via which anti-S100A9 AAbs could contribute to AMD pathogenesis could be at play.

The hypothesized connection between the autoreactivities that we detected and mechanisms linked to autophagy, NLRP3 inflammasome activation, protection from oxidative stress and apoptosis, and/or immunomodulation is well supported by a large body of published evidence. However, we do not know yet whether the AAbs detected in our study have either partially causal and/or contributory roles in AMD pathogenesis via these potential mechanisms, and/or which of them may be preeminent. With this limitation in mind, the pathogenicity of anti-retinal AAbs has already been established in patients with cancer-associated retinopathy (CAR), a form of secondary autoimmune retinopathy (AIR), as well as other forms of primary AIR and neuro-retinopathy (AINR) [143153]. In these conditions, many distinct AAbs are found, and can coexist in the same patient. The similarity in IR patterns between AMD and AINR/CAR, noted already by Adamus et al. [59], raises the intriguing possibility that AMD may share at least in part some fundamentally similar pathogenic mechanism with these autoimmune retinal conditions [143153].

Another caveat to our study is that the subset of participants we chose to confirm the IDs obtained via LC-MS/MS, and quite possibly even all of the 131 participants with AMD in our study sample, were in all likelihood not fully representative of all the possible reactivities that could be meaningful to AMD. Some other reactivities may have been missed by testing only a subset of samples, and others that did not meet the B-H multiple comparison correction statistical method used in our analyses may have actually achieved significance with an even larger data set. Thus, additional and potentially important reactivities that were not identified in our study likely exist in AMD (see, e.g., Adamus et al. [59]). As part of our ongoing effort to elucidate further the role of autoimmunity in AMD, we will continue to expand and refine our search for additional demonstrable reactivities in larger and/or different participant serum sample collections in the future.

Our study benefited from the fact that all of our subjects were carefully examined and graded with respect to their fundus findings, and were chosen to be free of ocular or systemic conditions of inflammatory and autoimmune nature and/or that could potentially confound our study. Furthermore, the use of human tissue antigens, and specifically macular ones, to test sera for reactivity supports the nature of the detected autoantigens as bona fide macular ones. In addition, the confirmation of our results by ELISA demonstrates the presence of AAbs directed against HSPA8, HSPA9, HSPB4/CRYAA, ANXA5, and S100A9 – antigens that are related to one another via their implication in protection from oxidative stress and apoptosis immunomodulation, and especially autophagy, a common theme shared by all targets identified by our investigation. This finding suggests that the detected AAbs may all contribute, from different angles, to compromised autophagy and contribute to NLRP3 inflammasome activation in AMD [131]. Our study design allowed us to gain information about the role of autoimmunity across the entire AMD disease spectrum. However, by focusing particularly on early AMD, we were able to gain particular insight on the role that AAbs may play early on in AMD pathogenesis. Thus, without suggesting that AMD could be a primary autoimmune condition like CAR and AINR, we can confidently conclude that our findings support the notion that the observed AAbs are unlikely to be mere after-the-fact biomarkers of advanced degeneration that has already occurred. Rather, we suggest that they represent valuable biomarkers of highly plausible pathogenic potential. As it has been already done for CEP [56, 58, 80, 82, 84, 85] and for similar AAb findings in glaucoma [154156], additional studies aimed at verifying the direct pathogenicity of these AAbs and the mechanisms via which the targeted proteins could participate in AMD biogenesis are now necessary, and represent a key ongoing research focus of our laboratories.

Supporting Information

S1 Fig. Schematic step-by-step representation of the methodology used to harvest full-thickness macular tissue punches and prepare the whole-macular lysates.

See S1 Supplemental Methods text for detailed explanations of the individual steps.

https://doi.org/10.1371/journal.pone.0145323.s001

(PDF)

S2 Fig. Example of gel bands captured for analysis in ImageJ and corresponding densitometric band plots generated by ImageJ64.

Bands were measured from “peak darkness” (shown as the trough in the densitometric plots) to baseline. Measured values were expressed as the relative fractional darkness of a reference band (not shown) on the same gel lane, whereby the apparent differences that could at times exist in the background were identical within each gel lane, making it unnecessary to account for different background intensities between different gel lanes. Thus, e.g., a band that appeared as dark as the reference band would usual give a 0.98 darkness ratio. One that seemed about half as dark, would correspond roughly to a 0.50 ratio, and so on. Note how every element in the cropped gel box is analyzed by ImageJ, including a double peak for the hand-written “37” shown above or a sharp, square-looking spike for the white background captured on the right-hand side of each gel. Note also that ImageJ offers the possibility to plot the densitometric data inverted as peaks instead of troughs at the discretion of the user.

https://doi.org/10.1371/journal.pone.0145323.s002

(PDF)

S3 Fig. Representative MS/MS spectra for the peptides matched to proteins identified in this study (see Table 1).

The spectra were produced via collision-induced dissociation (CID) of the corresponding mass-selected precursor ions in the ion trap mass spectrometer. The MS/MS spectra contain sequence-determining product ions of the b- (shown in red) and y-series (shown in blue).

https://doi.org/10.1371/journal.pone.0145323.s003

(PDF)

S1 Table. Immunoreactivity classification criteria used to grade the Western blot bands exposed to chemiluminescent substrate and imaged at 5, 15, 30 seconds (sec).

Scores (0–5) represent IR intensity of increasing intensity.

https://doi.org/10.1371/journal.pone.0145323.s005

(PDF)

Acknowledgments

We gratefully acknowledge the Health ABC Publications Committee and external reviewer, Dr. Grazyna Adamus, PhD, Casey Eye Institute, Oregon Health and Science University, Portland, OR, for their pre-submission peer-reviews and helpful comments of early versions of this manuscript. Portions of the data reported in this manuscript have been presented in poster format at the 2013 and 2015 ARVO meetings.

Author Contributions

Conceived and designed the experiments: AI MZR ICG FG SBG. Performed the experiments: DDN TJH AU AHA IN NIL. Analyzed the data: AI MZR ICG FG SBG DDN TJH AU AHA IN NIL. Contributed reagents/materials/analysis tools: BJJ JIC SS DM RID TH KCJ SC SBK. Wrote the paper: AI MZR ICG FG SBG TJH AHA DDN. Acquisition, analysis, or interpretation of data: AI MZR ICG FG SBG DDN TJH AU AHA IN NIL BJJ JIC SS DM RID TH KCJ SC SBK. Critical revision of the manuscript for important intellectual content: AI MZR ICG FG SBG DDN TJH AU AHA IN NIL BJJ JIC SS DM RID TH KCJ SC SBK. Statistical analysis: AI FG NIL. Obtaining funding: AI FG SBG ICG MZR SBK. Administrative, technical, or material support: AU IN NIL BJJ JIC SS DM RID TH KCJ SC SBK. Supervision AI FG SBG ICG MZR BJJ SS TH KCJ SBK.

References

  1. 1. Friedman DS, O'Colmain BJ, Munoz B, Tomany SC, McCarty C, de Jong PT, et al. Prevalence of age-related macular degeneration in the United States. Arch Ophthalmol. 2004;122(4):564–72. pmid:15078675
  2. 2. Hawkins BS, Bird A, Klein R, West SK. Epidemiology of age-related macular degeneration. Mol Vis. 1999;5:26. pmid:10562650
  3. 3. Hageman GS, Luthert PJ, Victor Chong NH, Johnson LV, Anderson DH, Mullins RF. An integrated hypothesis that considers drusen as biomarkers of immune-mediated processes at the RPE-Bruch's membrane interface in aging and age-related macular degeneration. Prog Retin Eye Res. 2001;20(6):705–32. pmid:11587915
  4. 4. Curcio CA, Messinger JD, Sloan KR, McGwin G, Medeiros NE, Spaide RF. Subretinal drusenoid deposits in non-neovascular age-related macular degeneration: morphology, prevalence, topography, and biogenesis model. Retina. 2013;33(2):265–76. pmid:23266879
  5. 5. Zweifel SA, Spaide RF, Curcio CA, Malek G, Imamura Y. Reticular pseudodrusen are subretinal drusenoid deposits. Ophthalmology. 2010;117(2):303–12 e1. pmid:19815280
  6. 6. Rudolf M, Malek G, Messinger JD, Clark ME, Wang L, Curcio CA. Sub-retinal drusenoid deposits in human retina: organization and composition. Exp Eye Res. 2008;87(5):402–8. pmid:18721807
  7. 7. Age-Related Eye Disease Study Research Group. The Age-Related Eye Disease Study system for classifying age-related macular degeneration from stereoscopic color fundus photographs: the Age-Related Eye Disease Study Report Number 6. Am J Ophthalmol. 2001;132(5):668–81. pmid:11704028
  8. 8. Ferris FL, Davis MD, Clemons TE, Lee L- Y, Chew EY, Lindblad AS, et al. A simplified severity scale for age-related macular degeneration. AREDS Report No. 20. Arch Ophthalmol. 2005;123(11):1570–4. pmid:16286620
  9. 9. Davis MD, Gangnon RE, Lee LY, Hubbard LD, Klein BE, Klein R, et al. The Age-Related Eye Disease Study severity scale for age-related macular degeneration: AREDS Report No. 17. Arch Ophthalmol. 2005;123(11):1484–98. pmid:16286610
  10. 10. Danis RP, Domalpally A, Chew EY, Clemons TE, Armstrong J, SanGiovanni JP, et al. Methods and reproducibility of grading optimized digital color fundus photographs in the Age-Related Eye Disease Study 2 (AREDS2 Report Number 2). Invest Ophthalmol Vis Sci. 2013;54(7):4548–54. pmid:23620429
  11. 11. 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(10):1417–36. pmid:11594942
  12. 12. Penfold PL, Madigan MC, Gillies MC, Provis JM. Immunological and aetiological aspects of macular degeneration. Prog Retin Eye Res. 2001;20(3):385–414. pmid:11286898
  13. 13. Penfold PL. Inflammation and age-related macular degeneration. JAMA. 2004;292(1):43; author reply
  14. 14. Nussenblatt RB, Ferris F 3rd. Age-related macular degeneration and the immune response: implications for therapy. Am J Ophthalmol. 2007;144(4):618–26. pmid:17698021
  15. 15. Bok D. Evidence for an inflammatory process in age-related macular degeneration gains new support. Proc Natl Acad Sci U S A. 2005;102(20):7053–4. pmid:15886281
  16. 16. McGeer EG, Klegeris A, McGeer PL. Inflammation, the complement system and the diseases of aging. Neurobiol Aging. 2005;26 Suppl 1:94–7. pmid:16198446
  17. 17. Donoso LA, Kim D, Frost A, Callahan A, Hageman G. The role of inflammation in the pathogenesis of age-related macular degeneration. Surv Ophthalmol. 2006;51(2):137–52. pmid:16500214
  18. 18. Gehrs KM, Anderson DH, Johnson LV, Hageman GS. Age-related macular degeneration—emerging pathogenetic and therapeutic concepts. Ann Med. 2006;38(7):450–71. pmid:17101537
  19. 19. Marx J. Genetics. A clearer view of macular degeneration. Science. 2006;311(5768):1704–5. pmid:16556817
  20. 20. Petrukhin K. New therapeutic targets in atrophic age-related macular degeneration. Expert Opin Ther Targets. 2007;11(5):625–39. pmid:17465722
  21. 21. Patel M, Chan CC. Immunopathological aspects of age-related macular degeneration. Semin Immunopathol. 2008;30(2):97–110. pmid:18299834
  22. 22. Kaarniranta K, Salminen A. Age-related macular degeneration: activation of innate immunity system via pattern recognition receptors. J Mol Med. 2009;87(2):117–23. pmid:19009282
  23. 23. Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Prog Retin Eye Res. 2009;28(5):348–68. pmid:19560552
  24. 24. Nussenblatt RB, Liu B, Li Z. Age-related macular degeneration: an immunologically driven disease. Curr Opin Investig Drugs. 2009;10(5):434–42. pmid:19431076
  25. 25. Anderson DH, Radeke MJ, Gallo NB, Chapin EA, Johnson PT, Curletti CR, et al. The pivotal role of the complement system in aging and age-related macular degeneration: hypothesis re-visited. Prog Retin Eye Res. 2010;29(2):95–112. pmid:19961953
  26. 26. Gehrs KM, Jackson JR, Brown EN, Allikmets R, Hageman GS. Complement, age-related macular degeneration and a vision of the future. Arch Ophthalmol. 2010;128(3):349–58. pmid:20212207
  27. 27. Anderson DH, Mullins RF, Hageman GS, Johnson LV. A role for local inflammation in the formation of drusen in the aging eye. Am J Ophthalmol. 2002;134(3):411–31. pmid:12208254
  28. 28. Edwards AO, Ritter R 3rd, Abel KJ, Manning A, Panhuysen C, Farrer LA. Complement factor H polymorphism and age-related macular degeneration. Science. 2005;308(5720):421–4. pmid:15761121
  29. 29. Haines JL, Hauser MA, Schmidt S, Scott WK, Olson LM, Gallins P, et al. Complement factor H variant increases the risk of age-related macular degeneration. Science. 2005;308(5720):419–21. pmid:15761120
  30. 30. Klein RJ, Zeiss C, Chew EY, Tsai JY, Sackler RS, Haynes C, et al. Complement factor H polymorphism in age-related macular degeneration. Science. 2005;308(5720):385–9. pmid:15761122
  31. 31. Hageman GS, Anderson DH, Johnson LV, Hancox LS, Taiber AJ, Hardisty LI, 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 U S A. 2005;102(20):7227–32. pmid:15870199
  32. 32. Hughes AE, Orr N, Esfandiary H, Diaz-Torres M, Goodship T, Chakravarthy U. A common CFH haplotype, with deletion of CFHR1 and CFHR3, is associated with lower risk of age-related macular degeneration. Nat Genet. 2006.
  33. 33. Gold B, Merriam JE, Zernant J, Hancox LS, Taiber AJ, Gehrs K, et al. Variation in factor B (BF) and complement component 2 (C2) genes is associated with age-related macular degeneration. Nat Genet. 2006;38(4):458–62. pmid:16518403
  34. 34. Li M, Atmaca-Sonmez P, Othman M, Branham KE, Khanna R, Wade MS, et al. CFH haplotypes without the Y402H coding variant show strong association with susceptibility to age-related macular degeneration. Nat Genet. 2006;38(9):1049–54. pmid:16936733
  35. 35. Maller J, George S, Purcell S, Fagerness J, Altshuler D, Daly MJ, et al. Common variation in three genes, including a noncoding variant in CFH, strongly influences risk of age-related macular degeneration. Nat Genet. 2006;38(9):1055–9. pmid:16936732
  36. 36. Postel EA, Agarwal A, Caldwell J, Gallins P, Toth C, Schmidt S, et al. Complement factor H increases risk for atrophic age-related macular degeneration. Ophthalmology. 2006;113(9):1504–7. pmid:16828512
  37. 37. Seddon JM, George S, Rosner B, Klein ML. CFH gene variant, Y402H, and smoking, body mass index, environmental associations with advanced age-related macular degeneration. Hum Hered. 2006;61(3):157–65. pmid:16816528
  38. 38. Sepp T, Khan JC, Thurlby DA, Shahid H, Clayton DG, Moore AT, et al. Complement factor H variant Y402H is a major risk determinant for geographic atrophy and choroidal neovascularization in smokers and nonsmokers. Invest Ophthalmol Vis Sci. 2006;47(2):536–40. pmid:16431947
  39. 39. Swaroop A, Branham KE, Chen W, Abecasis G. Genetic susceptibility to age-related macular degeneration: a paradigm for dissecting complex disease traits. Hum Mol Genet. 2007;16 Spec No. 2:R174–82. pmid:17911160
  40. 40. Spencer KL, Hauser MA, Olson LM, Schmidt S, Scott WK, Gallins P, et al. Deletion of CFHR3 and CFHR1 Genes in Age-Related Macular Degeneration. Hum Mol Genet. 2007;17: 971–7. pmid:18084039
  41. 41. Spencer KL, Olson LM, Anderson BM, Schnetz-Boutaud N, Scott WK, Gallins P, et al. C3 R102G Polymorphism Increases Risk of Age-related Macular Degeneration. Hum Mol Genet. 2008;17:1821–4. pmid:18325906
  42. 42. Takeda A, Baffi JZ, Kleinman ME, Cho WG, Nozaki M, Yamada K, et al. CCR3 is a target for age-related macular degeneration diagnosis and therapy. Nature. 2009;460(7252):225–30. pmid:19525930
  43. 43. Francis PJ, Hamon SC, Ott J, Weleber RG, Klein ML. Polymorphisms in C2, CFB and C3 are associated with progression to advanced age related macular degeneration associated with visual loss. J Med Genet. 2009;46(5):300–7. pmid:19015224
  44. 44. Spencer KL, Olson LM, Schnetz-Boutaud N, Scott WK, Gallins P, Agarwal A, et al. Using Genetic Variation and Environmental Risk Factor Data to Identify Individuals at High Risk for Age-Related Macular Degeneration. PLoS ONE. 2011;6(3):e17784. pmid:21455292
  45. 45. Liu MM, Chan CC, Tuo J. Genetic mechanisms and age-related macular degeneration: common variants, rare variants, copy number variations, epigenetics, and mitochondrial genetics. Hum Genomics. 2012;6:13. pmid:23244519
  46. 46. Tuo J, Grob S, Zhang K, Chan CC. Genetics of immunological and inflammatory components in age-related macular degeneration. Ocul Immunol Inflamm. 2012;20(1):27–36. pmid:22324898
  47. 47. Fritsche LG, Lauer N, Hartmann A, Stippa S, Keilhauer CN, Oppermann M, et al. An imbalance of human complement regulatory proteins CFHR1, CFHR3 and factor H influences risk for age-related macular degeneration (AMD). Hum Mol Genet. 2010;19(23):4694–704. pmid:20843825
  48. 48. Schaumberg DA, Rose L, Deangelis MM, Semba RD, Hageman GS, Chasman DI. Prospective Study of Common Variants in CX3CR1 and Risk of Macular Degeneration: Pooled Analysis From 5 Long-term Studies. JAMA Ophthalmol. 2014;132(1):84–95.
  49. 49. Hageman GS, Gehrs K, Lejnine S, Bansal AT, Deangelis MM, Guymer RH, et al. Clinical validation of a genetic model to estimate the risk of developing choroidal neovascular age-related macular degeneration. Hum Genomics. 2011;5(5):420–40. pmid:21807600
  50. 50. Grassmann F, Fritsche LG, Keilhauer CN, Heid IM, Weber BH. Modelling the genetic risk in age-related macular degeneration. PLoS One. 2012;7(5):e37979. pmid:22666427
  51. 51. Yu Y, Reynolds R, Rosner B, Daly MJ, Seddon JM. Prospective assessment of genetic effects on progression to different stages of age-related macular degeneration using multistate Markov models. Invest Ophthalmol Vis Sci. 2012;53(3):1548–56. pmid:22247473
  52. 52. Stone EM. Genetic testing for age-related macular degeneration: not indicated now. JAMA Ophthalmol. 2015;133(5):598–600. pmid:25789813
  53. 53. Baird PN, Robman LD, Richardson AJ, Dimitrov PN, Tikellis G, McCarty CA, et al. Gene-environment interaction in progression of AMD: the CFH gene, smoking and exposure to chronic infection. Hum Mol Genet. 2008;17(9):1299–305. pmid:18203751
  54. 54. Meyers KJ, Mares JA, Igo RP Jr., Truitt B, Liu Z, Millen AE, et al. Genetic evidence for role of carotenoids in age-related macular degeneration in the Carotenoids in Age-Related Eye Disease Study (CAREDS). Invest Ophthalmol Vis Sci. 2014;55(1):587–99. pmid:24346170
  55. 55. Lechanteur YT, van de Camp PL, Smailhodzic D, van de Ven JP, Buitendijk GH, Klaver CC, et al. Association of Smoking and CFH and ARMS2 Risk Variants With Younger Age at Onset of Neovascular Age-Related Macular Degeneration. JAMA Ophthalmol. 2015;133(5):533–41. pmid:25695752
  56. 56. Gu J, Pauer GJ, Yue X, Narendra U, Sturgill GM, Bena J, et al. Assessing susceptibility to age-related macular degeneration with proteomic and genomic biomarkers. Mol Cell Proteomics. 2009;8(6):1338–49. pmid:19202148
  57. 57. Iannaccone A, Neeli I, Krishnamurthy P, Lenchik NI, Wan H, Gerling IC, et al. Autoimmune biomarkers in age-related macular degeneration: a possible role player in disease development and progression. Adv Exp Med Biol. 2012;723:11–6. pmid:22183309
  58. 58. Gu J, Pauer GJ, Yue X, Narendra U, Sturgill GM, Bena J, et al. Proteomic and genomic biomarkers for age-related macular degeneration. Adv Exp Med Biol. 2010;664:411–7. pmid:20238042
  59. 59. Adamus G, Chew EY, Ferris FL, Klein ML. Prevalence of anti-retinal autoantibodies in different stages of Age-related macular degeneration. BMC Ophthalmol. 2014;14:154. pmid:25488058
  60. 60. Gallaher KT, Mura M, Todd WA, Harris TL, Kenyon E, Harris T, et al. Estimation of macular pigment optical density in the elderly: test-retest variability and effect of optical blur in pseudophakic subjects. Vision Res. 2007;47(9):1253–9. pmid:17376502
  61. 61. Iannaccone A, Mura M, Gallaher KT, Johnson EJ, Todd WA, Kenyon E, et al. Macular pigment optical density in the elderly: findings in a large biracial Midsouth population sample. Invest Ophthalmol Vis Sci. 2007;48(4):1458–65. pmid:17389471
  62. 62. Spencer KL, Olson LM, Schnetz-Boutaud N, Gallins P, Agarwal A, Iannaccone A, et al. Using genetic variation and environmental risk factor data to identify individuals at high risk for age-related macular degeneration. PLoS One. 2011;6(3):e17784. pmid:21455292
  63. 63. Vishwanathan R, Iannaccone A, Scott TM, Kritchevsky SB, Jennings BJ, Carboni G, et al. Macular pigment optical density is related to cognitive function in older people. Age Ageing. 2014;43(2):271–5. pmid:24435852
  64. 64. Jablonski MM, Iannaccone A, Reynolds DH, Gallaher P, Allen S, Wang X, et al. Age-Related Decline in VIP-Positive Parasympathetic Nerve Fibers in the Human Submacular Choroid. Invest Ophthalmol Vis Sci. 2007;48(2):479–85. pmid:17251439
  65. 65. Jablonski MM, Graney MJ, Kritchevsky SB, Iannaccone A. Reliability assessment of a rod photoreceptor outer segment grading system. Exp Eye Res. 2001;72(5):573–9. pmid:11311049
  66. 66. Pabst MJ, Pabst KM, Handsman DB, Beranova-Giorgianni S, Giorgianni F. Proteome of monocyte priming by lipopolysaccharide, including changes in interleukin-1beta and leukocyte elastase inhibitor. Proteome Sci. 2008;6:13. pmid:18492268
  67. 67. Giorgianni F, Cappiello A, Beranova-Giorgianni S, Palma P, Trufelli H, Desiderio DM. LC-MS/MS analysis of peptides with methanol as organic modifier: improved limits of detection. AnalChem. 2004;76(23):7028–38.
  68. 68. McGeer PL, McGeer EG. Innate immunity, local inflammation, and degenerative disease. Sci Aging Knowledge Environ. 2002;2002(29):re3. pmid:14602998
  69. 69. McGeer PL, McGeer EG. Inflammation and the degenerative diseases of aging. Ann N Y Acad Sci. 2004;1035:104–16. pmid:15681803
  70. 70. Johnson LV, Leitner WP, Rivest AJ, Staples MK, Radeke MJ, Anderson DH. The Alzheimer's A beta -peptide is deposited at sites of complement activation in pathologic deposits associated with aging and age-related macular degeneration. Proc Natl Acad Sci U S A. 2002;99(18):11830–5. pmid:12189211
  71. 71. Penfold PL, Killingsworth MC, Sarks SH. An ultrastructural study of the role of leucocytes and fibroblasts in the breakdown of Bruch's membrane. Aust J Ophthalmol. 1984;12:23–31. pmid:6732655
  72. 72. Penfold PL, Killingsworth MC, Sarks SH. Senile macular degeneration: the involvement of immunocompetent cells. Graefes Arch Clin Exp Ophthalmol. 1985;223(2):69–76. pmid:2408968
  73. 73. Penfold PL, Killingsworth MC, Sarks SH. Senile macular degeneration. The involvement of giant cells in atrophy of the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 1986;27(3):364–71. pmid:3949464
  74. 74. Penfold PL, Provis JM, Billson FA. Age-related macular degeneration: ultrastructural studies of the relationship of leucocytes to angiogenesis. Graefes Arch Clin Exp Ophthalmol. 1987;225(1):70–6. pmid:2436980
  75. 75. Penfold PL, Provis JM, Furby JH, Gatenby PA, Billson FA. Autoantibodies to retinal astrocytes associated with age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol. 1990;228(3):270–4. pmid:2193850
  76. 76. Hageman GS, Mullins RF. Molecular composition of drusen as related to substructural phenotype. Mol Vis. 1999;5:28. pmid:10562652
  77. 77. Mullins RF, Russell SR, Anderson DH, Hageman GS. Drusen associated with aging and age-related macular degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit disease. FASEB J. 2000;14:835–46. pmid:10783137
  78. 78. Crabb JW, Miyagi M, Gu X, Shadrach K, West KA, Sakaguchi H, et al. Drusen proteome analysis: an approach to the etiology of age-related macular degeneration. Proc Natl Acad Sci U S A. 2002;99(23):14682–7. pmid:12391305
  79. 79. Hollyfield JG, Salomon RG, Crabb JW. Proteomic approaches to understanding age-related macular degeneration. Adv Exp Med Biol. 2003;533:83–9. pmid:15180251
  80. 80. Gu X, Meer SG, Miyagi M, Rayborn ME, Hollyfield JG, Crabb JW, et al. Carboxyethylpyrrole protein adducts and autoantibodies, biomarkers for age-related macular degeneration. J Biol Chem. 2003;278(43):42027–35. pmid:12923198
  81. 81. Patel N, Ohbayashi M, Nugent AK, Ramchand K, Toda M, Chau KY, et al. Circulating anti-retinal antibodies as immune markers in age-related macular degeneration. Immunology. 2005;115(3):422–30. pmid:15946260
  82. 82. Ebrahem Q, Renganathan K, Sears J, Vasanji A, Gu X, Lu L, et al. Carboxyethylpyrrole oxidative protein modifications stimulate neovascularization: Implications for age-related macular degeneration. Proc Natl Acad Sci U S A. 2006;103(36):13480–4. pmid:16938854
  83. 83. Cherepanoff S, Mitchell P, Wang JJ, Gillies MC. Retinal autoantibody profile in early age-related macular degeneration: preliminary findings from the Blue Mountains Eye Study. Clin Experiment Ophthalmol. 2006;34(6):590–5. pmid:16925708
  84. 84. Hollyfield JG, Bonilha VL, Rayborn ME, Yang X, Shadrach KG, Lu L, et al. Oxidative damage-induced inflammation initiates age-related macular degeneration. Nat Med. 2008;14(2):194–8. pmid:18223656
  85. 85. Hollyfield JG, Perez VL, Salomon RG. A hapten generated from an oxidation fragment of docosahexaenoic acid is sufficient to initiate age-related macular degeneration. Mol Neurobiol. 2010;41(2–3):290–8. pmid:20221855
  86. 86. Morohoshi K, Ohbayashi M, Patel N, Chong V, Bird AC, Ono SJ. Identification of anti-retinal antibodies in patients with age-related macular degeneration. Exp Mol Pathol. 2012;93(2):193–9. pmid:22465421
  87. 87. Morohoshi K, Patel N, Ohbayashi M, Chong V, Grossniklaus HE, Bird AC, et al. Serum autoantibody biomarkers for age-related macular degeneration and possible regulators of neovascularization. Exp Mol Pathol. 2012;92(1):64–73. pmid:22001380
  88. 88. Kaarniranta K, Salminen A, Eskelinen EL, Kopitz J. Heat shock proteins as gatekeepers of proteolytic pathways-Implications for age-related macular degeneration (AMD). Ageing Res Rev. 2009;8(2):128–39. pmid:19274853
  89. 89. Yang Q, She H, Gearing M, Colla E, Lee M, Shacka JJ, et al. Regulation of neuronal survival factor MEF2D by chaperone-mediated autophagy. Science. 2009;323(5910):124–7. pmid:19119233
  90. 90. Berberian PA, Myers W, Tytell M, Challa V, Bond MG. Immunohistochemical localization of heat shock protein-70 in normal-appearing and atherosclerotic specimens of human arteries. Am J Pathol. 1990;136(1):71–80. pmid:2297051
  91. 91. Tulapurkar ME, Ramarathnam A, Hasday JD, Singh IS. Bacterial lipopolysaccharide augments febrile-range hyperthermia-induced heat shock protein 70 expression and extracellular release in human THP1 cells. PLoS One. 2015;10(2):e0118010. pmid:25659128
  92. 92. Fouqueray B, Philippe C, Amrani A, Perez J, Baud L. Heat shock prevents lipopolysaccharide-induced tumor necrosis factor-alpha synthesis by rat mononuclear phagocytes. Eur J Immunol. 1992;22(11):2983–7. pmid:1425922
  93. 93. Matsumura Y, Sakai J, Skach WR. Endoplasmic reticulum protein quality control is determined by cooperative interactions between Hsp/c70 protein and the CHIP E3 ligase. J Biol Chem. 2013;288(43):31069–79. pmid:23990462
  94. 94. Ohguro H, Ogawa K, Maeda T, Maeda A, Maruyama I. Cancer-associated retinopathy induced by both anti-recoverin and anti-hsc70 antibodies in vivo. Invest Ophthalmol Vis Sci. 1999;40(13):3160–7. pmid:10586938
  95. 95. Tahiri F, Le Naour F, Huguet S, Lai-Kuen R, Samuel D, Johanet C, et al. Identification of plasma membrane autoantigens in autoimmune hepatitis type 1 using a proteomics tool. Hepatology. 2008;47(3):937–48. pmid:18306218
  96. 96. Mak SK, McCormack AL, Manning-Bog AB, Cuervo AM, Di Monte DA. Lysosomal degradation of alpha-synuclein in vivo. J Biol Chem. 2010;285(18):13621–9. pmid:20200163
  97. 97. Urushitani M, Kurisu J, Tateno M, Hatakeyama S, Nakayama K, Kato S, et al. CHIP promotes proteasomal degradation of familial ALS-linked mutant SOD1 by ubiquitinating Hsp/Hsc70. J Neurochem. 2004;90(1):231–44. pmid:15198682
  98. 98. Zetterstrom P, Graffmo KS, Andersen PM, Brannstrom T, Marklund SL. Proteins that bind to misfolded mutant superoxide dismutase-1 in spinal cords from transgenic amyotrophic lateral sclerosis (ALS) model mice. J Biol Chem. 2011;286(23):20130–6. pmid:21493711
  99. 99. Imamura Y, Noda S, Hashizume K, Shinoda K, Yamaguchi M, Uchiyama S, et al. Drusen, choroidal neovascularization, and retinal pigment epithelium dysfunction in SOD1-deficient mice: a model of age-related macular degeneration. Proc Natl Acad Sci U S A. 2006;103(30):11282–7. pmid:16844785
  100. 100. Hashizume K, Hirasawa M, Imamura Y, Noda S, Shimizu T, Shinoda K, et al. Retinal dysfunction and progressive retinal cell death in SOD1-deficient mice. Am J Pathol. 2008;172(5):1325–31. pmid:18372426
  101. 101. Nakata K, Crabb JW, Hollyfield JG. Crystallin distribution in Bruch's membrane-choroid complex from AMD and age-matched donor eyes. Exp Eye Res. 2005;80(6):821–6. pmid:15939038
  102. 102. Thanos S, Bohm MR, Meyer zu Horste M, Prokosch-Willing V, Hennig M, Bauer D, et al. Role of crystallins in ocular neuroprotection and axonal regeneration. Prog Retin Eye Res. 2014;42:145–61. pmid:24998680
  103. 103. Kannan R, Sreekumar PG, Hinton DR. Alpha crystallins in the retinal pigment epithelium and implications for the pathogenesis and treatment of age-related macular degeneration. Biochim Biophys Acta. 2015.
  104. 104. Saraswathy S, Nguyen AM, Rao NA. The role of TLR4 in photoreceptor {alpha}a crystallin upregulation during early experimental autoimmune uveitis. Invest Ophthalmol Vis Sci. 2010;51(7):3680–6. pmid:20207969
  105. 105. Bernstein SL, Liu AM, Hansen BC, Somiari RI. Heat shock cognate-70 gene expression declines during normal aging of the primate retina. Invest Ophthalmol Vis Sci. 2000;41(10):2857–62. pmid:10967038
  106. 106. Lederman M, Weiss A, Chowers I. Association of neovascular age-related macular degeneration with specific gene expression patterns in peripheral white blood cells. Invest Ophthalmol Vis Sci. 2010;51(1):53–8. pmid:19684010
  107. 107. Decanini A, Nordgaard CL, Feng X, Ferrington DA, Olsen TW. Changes in select redox proteins of the retinal pigment epithelium in age-related macular degeneration. Am J Ophthalmol. 2007;143(4):607–15. pmid:17280640
  108. 108. Yuan X, Gu X, Crabb JS, Yue X, Shadrach K, Hollyfield JG, et al. Quantitative proteomics: comparison of the macular Bruch membrane/choroid complex from age-related macular degeneration and normal eyes. Mol Cell Proteomics. 2010;9(6):1031–46. pmid:20177130
  109. 109. Yaung J, Jin M, Barron E, Spee C, Wawrousek EF, Kannan R, et al. alpha-Crystallin distribution in retinal pigment epithelium and effect of gene knockouts on sensitivity to oxidative stress. Mol Vis. 2007;13:566–77. pmid:17438522
  110. 110. Kannan R, Sreekumar PG, Hinton DR. Novel roles for alpha-crystallins in retinal function and disease. Prog Retin Eye Res. 2012;31(6):576–604. pmid:22721717
  111. 111. Sreekumar PG, Chothe P, Sharma KK, Baid R, Kompella U, Spee C, et al. Antiapoptotic properties of alpha-crystallin-derived peptide chaperones and characterization of their uptake transporters in human RPE cells. Invest Ophthalmol Vis Sci. 2013;54(4):2787–98. pmid:23532520
  112. 112. Cederholm A, Frostegard J. Annexin A5 in cardiovascular disease and systemic lupus erythematosus. Immunobiology. 2005;210(10):761–8. pmid:16325495
  113. 113. Iaccarino L, Ghirardello A, Canova M, Zen M, Bettio S, Nalotto L, et al. Anti-annexins autoantibodies: their role as biomarkers of autoimmune diseases. Autoimmun Rev. 2011;10(9):553–8. pmid:21527362
  114. 114. Alessandri C, Conti F, Pendolino M, Mancini R, Valesini G. New autoantigens in the antiphospholipid syndrome. Autoimmun Rev. 2011;10(10):609–16. pmid:21545849
  115. 115. Bakar F, Unluturk U, Baskal N, Nebioglu S. Annexin V expression and anti-annexin V antibodies in type 1 diabetes. J Clin Endocrinol Metab. 2014;99(3):932–7. pmid:24423325
  116. 116. Rayborn ME, Sakaguchi H, Shadrach KG, Crabb JW, Hollyfield JG. Annexins in Bruch's membrane and drusen. Adv Exp Med Biol. 2006;572:75–8. pmid:17249558
  117. 117. Umeda S, Suzuki MT, Okamoto H, Ono F, Mizota A, Terao K, et al. Molecular composition of drusen and possible involvement of anti-retinal autoimmunity in two different forms of macular degeneration in cynomolgus monkey (Macaca fascicularis). FASEB J. 2005;19(12):1683–5. pmid:16099945
  118. 118. Ryckman C, Vandal K, Rouleau P, Talbot M, Tessier PA. Proinflammatory activities of S100: proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion. J Immunol. 2003;170(6):3233–42. pmid:12626582
  119. 119. Nakatani Y, Yamazaki M, Chazin WJ, Yui S. Regulation of S100A8/A9 (calprotectin) binding to tumor cells by zinc ion and its implication for apoptosis-inducing activity. Mediators Inflamm. 2005;2005(5):280–92. pmid:16258195
  120. 120. Li C, Chen H, Ding F, Zhang Y, Luo A, Wang M, et al. A novel p53 target gene, S100A9, induces p53-dependent cellular apoptosis and mediates the p53 apoptosis pathway. Biochem J. 2009;422(2):363–72. pmid:19534726
  121. 121. Bjork P, Bjork A, Vogl T, Stenstrom M, Liberg D, Olsson A, et al. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides. PLoS Biol. 2009;7(4):e97. pmid:19402754
  122. 122. Ghavami S, Eshragi M, Ande SR, Chazin WJ, Klonisch T, Halayko AJ, et al. S100A8/A9 induces autophagy and apoptosis via ROS-mediated cross-talk between mitochondria and lysosomes that involves BNIP3. Cell Res. 2010;20(3):314–31. pmid:19935772
  123. 123. Simard JC, Simon MM, Tessier PA, Girard D. Damage-associated molecular pattern S100A9 increases bactericidal activity of human neutrophils by enhancing phagocytosis. J Immunol. 2011;186(6):3622–31. pmid:21325622
  124. 124. Riva M, Kallberg E, Bjork P, Hancz D, Vogl T, Roth J, et al. Induction of nuclear factor-kappaB responses by the S100A9 protein is Toll-like receptor-4-dependent. Immunology. 2012;137(2):172–82. pmid:22804476
  125. 125. Koike A, Arai S, Yamada S, Nagae A, Saita N, Itoh H, et al. Dynamic mobility of immunological cells expressing S100A8 and S100A9 in vivo: a variety of functional roles of the two proteins as regulators in acute inflammatory reaction. Inflammation. 2012;35(2):409–19. pmid:21487906
  126. 126. Jia J, Arif A, Terenzi F, Willard B, Plow EF, Hazen SL, et al. Target-selective protein S-nitrosylation by sequence motif recognition. Cell. 2014;159(3):623–34. pmid:25417112
  127. 127. Cesaro A, Anceriz N, Plante A, Page N, Tardif MR, Tessier PA. An inflammation loop orchestrated by S100A9 and calprotectin is critical for development of arthritis. PLoS One. 2012;7(9):e45478. pmid:23029038
  128. 128. Dhas DBB, Bhat BV, Gane DB. Role of Calprotectin in Infection and Inflammation. Curr Pediatr Res. 2012;16 (2):83–94.
  129. 129. Skeie JM, Mahajan VB. Proteomic landscape of the human choroid-retinal pigment epithelial complex. JAMA Ophthalmol. 2014;132(11):1271–81. pmid:25058583
  130. 130. Borges TJ, Wieten L, van Herwijnen MJ, Broere F, van der Zee R, Bonorino C, et al. The anti-inflammatory mechanisms of Hsp70. Front Immunol. 2012;3:95. pmid:22566973
  131. 131. Celkova L, Doyle SL, Campbell M. NLRP3 Inflammasome and Pathobiology in AMD. J Clin Med. 2015;4(1):172–92. pmid:26237026
  132. 132. Yao J, Jia L, Khan N, Lin C, Mitter SK, Boulton ME, et al. Deletion of autophagy inducer RB1CC1 results in degeneration of the retinal pigment epithelium. Autophagy. 2015;11(6):939–53. pmid:26075877
  133. 133. Dokladny K, Myers OB, Moseley PL. Heat shock response and autophagy—cooperation and control. Autophagy. 2015;11(2):200–13. pmid:25714619
  134. 134. Dokladny K, Zuhl MN, Mandell M, Bhattacharya D, Schneider S, Deretic V, et al. Regulatory coordination between two major intracellular homeostatic systems: heat shock response and autophagy. J Biol Chem. 2013;288(21):14959–72. pmid:23576438
  135. 135. Minoia M, Grit C, Kampinga HH. HSPA1A-independent suppression of PARK2 C289G protein aggregation by human small heat shock proteins. Mol Cell Biol. 2014;34(19):3570–8. pmid:25022755
  136. 136. Kaarniranta K, Sinha D, Blasiak J, Kauppinen A, Vereb Z, Salminen A, et al. Autophagy and heterophagy dysregulation leads to retinal pigment epithelium dysfunction and development of age-related macular degeneration. Autophagy. 2013;9(7):973–84. pmid:23590900
  137. 137. Ghislat G, Aguado C, Knecht E. Annexin A5 stimulates autophagy and inhibits endocytosis. J Cell Sci. 2012;125(Pt 1):92–107. pmid:22266906
  138. 138. Zigler JS Jr., Zhang C, Grebe R, Sehrawat G, Hackler L Jr., Adhya S, et al. Mutation in the betaA3/A1-crystallin gene impairs phagosome degradation in the retinal pigmented epithelium of the rat. J Cell Sci. 2011;124(Pt 4):523–31. pmid:21266465
  139. 139. Louie JL, Kapphahn RJ, Ferrington DA. Proteasome function and protein oxidation in the aged retina. Exp Eye Res. 2002;75(3):271–84. pmid:12384090
  140. 140. Ethen CM, Hussong SA, Reilly C, Feng X, Olsen TW, Ferrington DA. Transformation of the proteasome with age-related macular degeneration. FEBS Lett. 2007;581(5):885–90. pmid:17289037
  141. 141. Ryhanen T, Hyttinen JM, Kopitz J, Rilla K, Kuusisto E, Mannermaa E, et al. Crosstalk between Hsp70 molecular chaperone, lysosomes and proteasomes in autophagy-mediated proteolysis in human retinal pigment epithelial cells. J Cell Mol Med. 2009;13(9B):3616–31. pmid:19017362
  142. 142. Ferrington DA, Sinha D, Kaarniranta K. Defects in retinal pigment epithelial cell proteolysis and the pathology associated with age-related macular degeneration. Prog Retin Eye Res. 2015.
  143. 143. Adamus G, Machnicki M, Seigel GM. Apoptotic retinal cell death induced by antirecoverin autoantibodies of cancer-associated retinopathy. Invest Ophthalmol Vis Sci. 1997;38(2):283–91. pmid:9040460
  144. 144. Adamus G, Machnicki M, Elerding H, Sugden B, Blocker YS, Fox DA. Antibodies to recoverin induce apoptosis of photoreceptor and bipolar cells in vivo. J Autoimmun. 1998;11(5):523–33. pmid:9802939
  145. 145. Adamus G, Amundson D, Seigel GM, Machnicki M. Anti-enolase-alpha autoantibodies in cancer-associated retinopathy: epitope mapping and cytotoxicity on retinal cells. J Autoimmun. 1998;11(6):671–7. pmid:9878089
  146. 146. Shiraga S, Adamus G. Mechanism of CAR syndrome: anti-recoverin antibodies are the inducers of retinal cell apoptotic death via the caspase 9- and caspase 3-dependent pathway. J Neuroimmunol. 2002;132(1–2):72–82. pmid:12417436
  147. 147. Adamus G. Autoantibody-induced apoptosis as a possible mechanism of autoimmune retinopathy. Autoimmun Rev. 2003;2(2):63–8. pmid:12848960
  148. 148. Adamus G, Ren G, Weleber RG. Autoantibodies against retinal proteins in paraneoplastic and autoimmune retinopathy. BMC Ophthalmol. 2004;4:5. pmid:15180904
  149. 149. Ren G, Adamus G. Cellular targets of anti-alpha-enolase autoantibodies of patients with autoimmune retinopathy. J Autoimmun. 2004;23(2):161–7. pmid:15324934
  150. 150. Magrys A, Anekonda T, Ren G, Adamus G. The role of anti-alpha-enolase autoantibodies in pathogenicity of autoimmune-mediated retinopathy. J Clin Immunol. 2007;27(2):181–92. pmid:17235687
  151. 151. Adamus G. Autoantibody targets and their cancer relationship in the pathogenicity of paraneoplastic retinopathy. Autoimmun Rev. 2009;8(5):410–4. pmid:19168157
  152. 152. Adamus G, Karren L. Autoimmunity against carbonic anhydrase II affects retinal cell functions in autoimmune retinopathy. J Autoimmun. 2009;32(2):133–9. pmid:19269136
  153. 153. Adamus G, Brown L, Schiffman J, Iannaccone A. Diversity in autoimmunity against retinal, neuronal, and axonal antigens in acquired neuro-retinopathy. J Ophthalmic Inflamm Infect. 2011;1(3):111–21. pmid:21744285
  154. 154. Grus FH, Joachim SC, Hoffmann EM, Pfeiffer N. Complex autoantibody repertoires in patients with glaucoma. Mol Vis. 2004;10:132–7. pmid:14990890
  155. 155. Joachim SC, Bruns K, Lackner KJ, Pfeiffer N, Grus FH. Antibodies to alpha B-crystallin, vimentin, and heat shock protein 70 in aqueous humor of patients with normal tension glaucoma and IgG antibody patterns against retinal antigen in aqueous humor. Curr Eye Res. 2007;32(6):501–9. pmid:17612966
  156. 156. Casola C, Schiwek JE, Reinehr S, Kuehn S, Grus FH, Kramer M, et al. S100 alone has the same destructive effect on retinal ganglion cells as in combination with HSP 27 in an autoimmune glaucoma model. J Mol Neurosci. 2015;56(1):228–36. pmid:25577368