Skip to main content

Systematic review and meta-analysis of mass spectrometry proteomics applied to ocular fluids to assess potential biomarkers of age-related macular degeneration

Abstract

Background

Age-related macular degeneration (AMD) is a significant cause of severe vision loss. The main purpose of this study was to identify mass spectrometry proteomics-based potential biomarkers of AMD that contribute to understanding the mechanisms of disease and aiding in early diagnosis.

Methods

This study retrieved studies that aim to detect differences relate to proteomics in AMD patients and healthy control groups by mass spectrometry (MS) proteomics approaches. The search process was accord with PRISMA guidelines (PROSPERO database: CRD42023388093). Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes Pathway Analysis (KEGG) were performed on differentially expressed proteins (DEPs) in the included articles using the DAVID database. DEPs were included in a meta-analysis when their effect size could be computed in at least two research studies. The effect size of measured proteins was transformed to the log2-fold change. Protein‒protein interaction (PPI) analysis was conducted on proteins that were statistically significant in the meta-analysis using the String online database.

Results

Eleven studies fulfilled the inclusion criteria, and 161 DEPs were identified. The GO analysis showed that AMD is significantly related to proteolysis, extracellular exosome and protein binding. In KEGG, the most significant pathway was the complement and coagulation cascades. Meta-analysis results suggested that eight proteins were statistically significant, and according to PPI results, the most significant four proteins were serotransferrin (TF), apolipoprotein A1 (APOA1), complement C3 (C3) and lipocalin-1 (LCN1).

Conclusions

Four possible biomarkers, TF, APOA1, C3 and LCN1, were found to be significant in the pathogenesis of AMD and need to be further validated. Further studies should be performed to evaluate diagnostic and therapeutic value of these proteins.

Peer Review reports

Introduction

Age-related macular degeneration, which accounts for 6–9% of legal blindness globally, is the leading cause of severe vision loss among individuals over 55 years of age in developed countries [1, 2]. Despite differences in the prevalence of AMD between eastern and western countries [3], the aging of the global population indicates an inevitable surge in the absolute number of AMD patients worldwide, with a projected increase from 196 million in 2020 to 288 million by 2040 [1].

Although there are powerful diagnostic tools such as optical coherence tomography and fundus autofluorescence imaging for AMD, the diagnosis of AMD may be delayed. One of the main challenges lies in the fact that early AMD is often asymptomatic [4]. Additionally, if only one eye is affected, symptoms may not be apparent until the visual function of the other eye is compromised. Late-stage AMD is characterized by geographic atrophy (GA) and/or neovascular AMD (nvAMD) [5]. GA is manifested as retinal pigment epithelial cells lost, overlying photoreceptors, and underlying choroidal capillaries, which does not involve blood or serum leakage [6]. Unlike GA, nvAMD is characterized by choroidal neovascularization complex which involve blood or serum leakage [4]. In the process of wet AMD, overexpression of VEGF is a crucial risk factor [7]. Meanwhile, several mechanisms are involved in both dry and wet AMD, including RPE cell senescence, oxidative stress, lipid metabolism, inflammation and immunity [8]. As advanced forms of AMD, GA and nvAMD can cause loss of central visual acuity, leading to severe and permanent visual impairment and ultimately resulting in legal blindness, which has a major impact on quality of life and functional independence [4]. In recent years, significant achievements have been made in the therapeutic strategies of nvAMD [9], such as the availability of several anti-VEGF agents, including ranibizumab [10] and aflibercept.[11] However, there is still a lack of effective strategies to prevent photoreceptor loss in the context of de novo development and enlargement of GA. Moreover, the treatment effect of AMD patients is not optimistic, primarily due to the treatment burden or potential final outcomes such as atrophy and fibrosis [12]. Hence, there is an urgent need for ongoing efforts to reduce treatment burden and enhance existing treatment options.

The etiology and pathogenesis of AMD remain unclarified. Given that biomarkers can reveal the dysregulation of molecular expression profiles, the identification of biomarkers holds great significance and provides insight for the early diagnosis, mechanism and treatment of AMD. Intraocular fluid has been proven to be suitable for the evaluation of relevant biomarkers for posterior segment disorders [13]. In recent years, many studies have demonstrated molecular differences in aqueous humor (AH) or vitreous humor (VH) between AMD patients and controls [14,15,16]. These findings have made significant contributions toward the identification of potential biomarkers and have shed light on the underlying mechanisms involved in AMD.

Proteomics is a powerful platform for studying both single proteins and complex protein samples [17]. In recent years, MS technologies have become an effective tool to discover biomarker, and quantitative MS proteomics such as liquid chromatography combined with tandem MS (LC‒MS/MS) approaches show fine specificity and sensitivity [18], which can detect the composition, structure, and function of proteins. In recent decades, with advancements in MS, multiple proteins have been discovered in AH and VH samples of AMD patients, which significantly contribute to uncovering potential biomarkers and shedding light on the underlying mechanisms of AMD.

While significant progress has been made in the field of AMD biomarker research using proteomic techniques, there is still a lack of well-established and validated biomarkers for AMD. Additionally, the precise mechanisms underlying AMD development and progression remain incompletely understood. This article provides a comprehensive summary of the literature on the analysis of AH and VH samples from AMD patients using MS proteomics. The objective of the study was to conduct a meta-analysis to identify potential biomarkers of AMD that could shed light on the disease's underlying mechanisms and aid in early diagnosis.

Method

This study accorded PRISMA guidelines and registered in the PROSPERO database (CRD42021274183). Studies were included in this systematic review if they match the search keywords.

Search strategy

The researchers conducted a comprehensive search for relevant studies through three independent databases: PUBMED, Web of Science (WOS) and EMBASE. Keywords of search process were used the MeSH or Emtree terms as well as free words, until March 2023: [(“age-related macular degeneration” OR “AMD”) AND (“aqueous humor” OR “vitreous Humor”)] AND “proteomics”. Researchers also screened references of relevant studies. The process to select the included studies in this study, which followed PRISMA 2020, was summarized in Fig. 1.

Fig. 1
figure 1

Flow chart of the selection process of the studies included in this systematic review of intraocular fluid mass spectrometry-based proteomics in age-related macular degeneration, following PRISMA 2020

Eligibility criteria

The inclusion of articles complied with the following criteria: (a) study involved proteome profiling and/or quantification based on mass spectrometry techniques; (b) research performed on human aqueous humor or vitreous humor samples; and (c) a group of AMD patients and a healthy control group were valid in research.

Proteomics studies only assessing the effects of specific treatment in AMD patients or only having samples of treated AMD patients will be excluded.

Quality of evidence

QUADOMICS methodology criteria was used to assess the quality of included studies (Table S1). Two authors evaluated the included studies independently (Figure S1). As a quality evaluation tool, QUADOMICS is appropriative for studies used omics techniques [19, 20].

Data extraction

Two authors, Hanmu Guo and Jianqing Li, followed fixed protocol to extract data from the eligible studies (Tables 1 and 2): (1) first author; (2) publication year; (3) participants’ characteristics; (4) analytical technique; and (5) altered proteins in AMD patients as measured against controls. Additional information will request from the authors of included studies when necessary.

Table 1 Demographic summary of all the studies included in the systematic review and meta-analysis
Table 2 Proteomic studies of AMD and biomarkers discovery using MS-based method in human intraocular fluids

Statistical analysis

GO analysis and KEGG were performed using the DAVID database. The effect size in this meta-analysis was determined to the log2-fold change of measured proteins. Proteins will be included in the meta-analysis if their effect sizes were valid in at least two research studies. There are two types of effect sizes and corresponding significance groups appeared in the studies, one is log(ratio) or ratio correspond with p value, the other is group averages correspond with standard deviations, they were all standardized to log2-fold change and corresponding p values. Only the statistics of samples from untreated AMD patients will be calculated. The meta-analysis was completed in R (version 4.2.2) combined with R Studio by using the “meta” [32], “metafor” [33], and “dmetar” R packages [34]. String online database and Cytoscape software was utilized to conduct PPI analysis [35].

Results

Characteristics of the included articles

The search strategy of included studies in this study was summarized in Fig. 1. As shown in the search results of EMBASE, WOS and PUBMED databases, totally 83 studies were identified after excluding duplicates. 11 studies which fulfilled the eligibility criteria were finally included [21,22,23,24,25,26,27,28,29,30,31]. The demographic characteristics are summarized in Table 1.

Bias analysis

The quality evaluation results of the studies included in this study are summarized in Figure 1. Items 1(2 studies), 11(5 studies), and 16 (4 studies) were least fulfilled QUADOMICS quality criteria. Most of studies could not assess items 6 and 12 because of insufficient data.

Analytical technique

AH were dominant sample type used in the eligible studies, in which 8 studies analyzed AH and 3 studies analyzed VH (Table 1) [29,30,31]. Regarding the MS-based method, 9 out of the 11 studies used LC‒MS/MS analysis [22,23,24,25, 27,28,29,30,31]. The other two studies applied MALDI TOF/TOF and intensity-based label-free quantification and were published in 2013 and 2021 [21, 26]. Most studies further applied other techniques to validate protein expression patterns. The most commonly used two approaches were enzyme-linked immunosorbent assay (ELISA) [21, 22, 25, 26, 29, 31], and western blotting [27, 28, 30].

GO and KEGG analysis

GO analysis, which included biological process (BP), cell components (CC) and molecular function (MF), was performed on all the DEPs in the AMD group. Through UNIPROT ACCESSION, 161 proteins were recognized by DAVID Bioinformatics Resources in this ontological analysis, and 10 proteins were not included because no accession number was available. Based on the protein counts, each top 10 categories of BP, CC, and MF were calculated and summarized in Fig. 2. In the BP analysis, the majority of obtained proteins were involved in proteolysis, innate immune response and negative regulation of endopeptidase activity. In terms of CC analysis, most proteins were located in the extracellular exosome, extracellular region, and extracellular space. For the analysis of MF, the results indicated that protein binding, identical protein binding and calcium ion binding were the most important functions.

Fig. 2
figure 2

Gene ontology analysis of proteins that were altered in studies were included in the systematic review (top 10 categories based on the protein counts); red represents biological process, green represents cellular component, and blue represents molecular function

KEGG enrichment highlighted seven significantly accumulated pathways involving the DEPs (Fig. 3). Twenty DEPs were enriched in the complement and coagulation cascade pathways, nine DEPs were accumulated in lysosome, and six DEPs were enriched in the hypoxia-inducible factor 1 (HIF-1) signaling pathway.

Fig. 3
figure 3

Kyoto encyclopedia of genes and genomes pathway analysis pathway analysis of proteins that were altered in studies was included in the systematic review

Meta-analysis

In all included studies, eight studies [21,22,23,24,25,26,27,28] analyzed AH and three studies analyzed VH [29,30,31]. The data of DEPs between AMD and control were used to perform the meta-analysis. The effect size (defined as log2-fold change) of proteins will be computed when it could be identified in at least two studies.

The meta-analysis of the DEPs in AH between AMD patients and healthy controls is shown in Fig. 4: apolipoprotein A1 (APOA1, P02647), apolipoprotein A4 (APOA4, P06727), complement C3 (C3, P01024), clusterin (CLU, P10909), cathepsin D (CTSD, P07339), lipocalin-1 (LCN1, P31025), pigment epithelium-derived factor (PEDF, P36955), prostaglandin-H2 D-isomerase (PTGDS or PH2D, P41222) and kallistatin (SERPINA4, P29622) (gene names and accession numbers, respectively). The meta-analysis results suggested increased expression of APOA1, C3, and LCN1 and decreased expression of PTGDS in dry and wet AMD patients compared with healthy control subjects.

Fig. 4
figure 4

Forest plot from the meta-analysis of proteins identified as altered in AMD vs. control studies analyzed aqueous humor in at least two studies (95% CI, confidence intervals). Squares (whiskers represent 95% CI) indicate the effect sizes of the individual studies. The size of the squares reflects the sample size of each individual study. Diamonds represent summary statistics

The meta-analysis looking at the DEPs in VH between wet AMD patients and healthy controls was performed on five proteins (Fig. 5): Ig gamma − 1 chain C region (IGHG1, P01857), Ig kappa chain C region (IGKC, P01834), Ig lambda − 2 chain C regions (IGLC2, P0DOY2), PTGDS (P41222) and serotransferrin (TF, P02787). All the proteins were more highly expressed in the AMD group.

Fig. 5
figure 5

Forest plot from the meta-analysis of proteins identified as altered in AMD vs. control studies analyzed vitreous humor in at least two studies (95% CI, confidence intervals). Squares (whiskers represent 95% CI) indicate the effect sizes of the individual studies. The size of the squares reflects the sample size of each individual study. Diamonds represent summary statistics

PPI analysis

PPIs was performed by using the STRING database and Cytoscape software in this research [35]. Through the String database, 8 proteins that showed statistical significance in the meta-analysis were filtered into the analysis, finally resulting in 5 nodes and 5 edges (three proteins could not be found in the String database). These statistics are processed by Cytoscape software, and the results are shown in Fig. 6. The figure was created by statistics. The degree dictated the size and color, and the combined score determined the edge size, therefore a low value led to smaller sizes and darker colors than a high value. As shown in the figure, the most significant proteins were TF (P02787), C3 (P01024) and APOA1 (P02647).

Fig. 6
figure 6

Protein–protein interaction analysis conducted by the String database and Cytoscape software

Discussion

To assess protein expression differences between AMD patients and controls, identify biomarkers and related biological pathways of AMD, a comprehensive systematic review and meta-analysis was performed based on mass spectrometry-based proteomics of human intralocular fluids. 161 altered proteins were identified between the AMD and control groups.

GO analysis and KEGG analysis were performed on all the DEPs. GO analysis showed a connection between AMD and the proteolysis, innate immune response, and negative regulation of endopeptidase activity, which mainly occurred in extracellular exosomes, extracellular regions and the extracellular space. In terms of MF, identical protein binding and calcium ion binding were the most important functions. KEGG showed that the most significant pathway was complement and coagulation cascades. The results of GO and KEGG analyses were generally consistent with the known pathogenic mechanism of AMD [9].

The meta-analysis on DEPs in AH found upregulated APOA1, C3, LCN1 and downregulated PTGDS. In separate pooled analysis of these proteins, a consistent trend was shown in both dry and wet AMD patients. The meta-analysis on VH showed IGHG1, IGKC, IGLC2, PTGDS and TF to be overexpressed in wet AMD patients.

PTGDS is downregulated in the AH [22, 24, 25] but upregulated in the VH of AMD patients [29, 30]. PTGDS is an enzyme that converts prostaglandin H2 to prostaglandin D2 and acts as a transport protein for lipophilic substances such as retinoic acid and bilirubin [36]. The relationship between PTGDS and AMD was confirmed by analysis of blood samples [37]. PTGDS is considered a protective factor because it can prevent oxidative stress and apoptosis-related neurodegenerative diseases [38]. The reason for the different trends in PTGDS may be the various degrees of AMD. The retinal pigment epithelium (RPE) is the main source of intraocular PTGDS [39], and mild AMD patients with normal RPE functions may exert protective effects by increasing the level of PTGDS. When RPE functions are damaged to a certain extent in advanced AMD, the level of PTGDS may decrease.

Further study on significant proteins was conducted through PPI analysis. The results indicated that TF, APOA1 and C3 and LCN1 may be the most important proteins, with the former 3 proteins interacting with each other. Based on the results of KEGG analysis, TF was involved in the HIF-1 signaling pathway. C3 was involved in complement and coagulation cascades and Staphylococcus aureus infection pathways. However, APOA1 and LCN1 were not found in the KEGG pathway results.

As iron binding transport proteins, TFs play a role in transporting iron to sites of storage and utilization after absorption or heme degradation. Cell proliferation may also be stimulated by serum transferrin [40, 41]. The serum level of transferrin was found to be higher in the AMD group [42]. Transferrin receptor and variability of its gene might also influence AMD risk [43, 44]. Recent studies discovered that hypoxia may aggravate ferroptosis in RPE cells and then affect the pathophysiology of AMD [45, 46], which provided novel insight into hypoxia, oxidative stress and iron metabolism in AMD pathophysiology. Furthermore, transferrin nonviral gene therapy also showed preliminary effectiveness for the treatment of dry AMD [47]. The above findings indicate that TF may be a promising biomarker of AMD.

APOs are significant proteins for maintaining lipid homeostasis, not only playing an important role in transporting and metabolizing lipids, APOs also relating to regulation of inflammatory and immune response [48, 49]. APOA1 is one of the major components of high-density lipoprotein (HDL) and is considered for regulating levels of free fatty acids in the plasma, involving in metabolism of HDL and triglyceride-rich lipoprotein by the reverse cholesterol transport pathway [50]. Research has shown that elevated HDL cholesterol levels may contribute to formation of drusen in process of AMD [51]. Three meta-analyses have shown elevated APOA1 levels in the AH of AMD patients [22, 24, 25]. Recent studies also showed plasma APOA1 and HDL level may relate to the risk of AMD formation [52,53,54]. These results indicating that APOA1 may be a potential biomarker in AMD patients, and may participate in formation of AMD. The complement system is considered to play a central role in AMD pathogenesis, and overactivation of the alternative complement pathway is one of the main drivers of diseases and is related to multiple pathogenic factors of AMD, such as inflammation, oxidative stress and lipid accumulation [55, 56]. Before exposing binding sites to the surface of pathogenic cell or other complement components, the C3 protein usually stating biologically inactive [57]. The complement system is frequently activated in many inflammatory diseases, including AMD [55, 58]. As the first line of defense against the innate response, the complement system could recognize and mediate the process of pathogens, debris, and dead cells removal to protect the human organism; hence, therapies targeting complement C3 still need to be considered carefully in AMD patients [55, 59].

It is worth noting that LCN1 also demonstrated importance in PPI. LCN1 is stimulated by oxidative stress, and is considered to be a scavenger of potentially harmful lipid peroxidation products [60]. At present, pathological process of AMD is supposed to relate with oxidative stress, although the specific mechanisms remain unclear [61], and oxidative stress-related genes are also associated with AMD risk [62, 63]. Therefore, LCN1 is considered to have a protective role in the progression of AMD. Except for elevating in the aqueous humor, LCN1 was also found in humans and animals tears in early researches [64]. In a recent study, LCN1 in tears demonstrated its potential as a biomarker in screening diabetic retinopathy [65], which brings expectations for LCN1 to become a biomarker of AMD.

The primary limitation was the heterogeneity among enrolled studies. For example, different studies had different definitions of DEPs, which mainly reflected in the minimum fold change values of proteins to be displayed and the indicators for measuring the accuracy of results (p value or adjusted p value). Heterogeneity like type of AMD, age or gender was also observed in the cohorts’ characteristics. Limited number of original studies included in each pooled analysis is also a limitation of this study. Although this study included observational and randomized controlled trial (RCT) studies, it did not bring any additional heterogeneity because only statistics before intervention of RCT studies could be included in the meta-analysis, and the criteria for selecting patients were similar in the included RCT and observational studies.

Conclusions

To sum up, various pathways associated with AMD have been elucidated, including lipid metabolism, the complement system, oxidative stress, inflammation, immunology, iron metabolism and ferroptosis. Four possible biomarkers, TF, APOA1, C3 and LCN1, were found to be significant in the pathogenesis of AMD and need to be further validated. Further studies should be performed to evaluate diagnostic and therapeutic value of these proteins.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AMD:

Age-related macular degeneration

GO:

Gene ontology

KEGG:

Kyoto encyclopedia of genes and genomes pathway analysis

DEPs:

Differentially expressed proteins

PPI:

Protein‒protein interaction

GA:

Geographic atrophy

nvAMD:

Neovascular age-related macular degeneration

AH:

Aqueous humor

VH:

Vitreous humor

LC‒MS/MS:

Liquid chromatography coupled to tandem mass spectrometry

WOS:

Web of Science

ELISA:

Enzyme-linked immunosorbent assay

BP:

Biological process

CC:

Cell components

MF:

Molecular function

MS:

Mass spectrometry

APOA1:

Apolipoprotein A1

APOA4:

Apolipoprotein A4

C3:

Complement C3

CLU:

Clusterin

CTSD:

Cathepsin D

LCN1:

Lipocalin-1

PEDF:

Pigment epithelium-derived factor

PTGDS:

Prostaglandin-H2 D-isomerase

SERPINA4:

Kallistatin

HIF-1:

Hypoxia-inducible factor 1

IGHG1:

Ig gamma − 1 chain C region

IGKC:

Ig kappa chain C region

IGLC2:

Ig lambda − 2 chain C regions

TF:

Serotransferrin

HDL:

High-density lipoprotein

RCT:

Randomized controlled trial

References

  1. Wong WL, Su X, Li X, Cheung CM, Klein R, Cheng CY, et al. 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(2):e106–16.

    Article  PubMed  Google Scholar 

  2. Jonas JB, Cheung CMG, Panda-Jonas S. Updates on the Epidemiology of Age-Related Macular Degeneration. Asia Pac J Ophthalmol (Phila). 2017;6(6):493–7.

    PubMed  Google Scholar 

  3. Rim TH, Kawasaki R, Tham YC, Kang SW, Ruamviboonsuk P, Bikbov MM, et al. Prevalence and Pattern of Geographic Atrophy in Asia: The Asian Eye Epidemiology Consortium. Ophthalmology. 2020;127(10):1371–81.

    Article  PubMed  Google Scholar 

  4. Mitchell P, Liew G, Gopinath B, Wong TY. Age-related macular degeneration. Lancet. 2018;392(10153):1147–59.

    Article  PubMed  Google Scholar 

  5. Ferris FL 3rd, Wilkinson CP, Bird A, Chakravarthy U, Chew E, Csaky K, et al. Clinical classification of age-related macular degeneration. Ophthalmology. 2013;120(4):844–51.

    Article  PubMed  Google Scholar 

  6. McLeod DS, Grebe R, Bhutto I, Merges C, Baba T, Lutty GA. Relationship between RPE and choriocapillaris in age-related macular degeneration. Invest Ophthalmol Vis Sci. 2009;50(10):4982–91.

    Article  PubMed  Google Scholar 

  7. Huang L, Zhang H, Cheng CY, Wen F, Tam PO, Zhao P, et al. A missense variant in FGD6 confers increased risk of polypoidal choroidal vasculopathy. Nat Genet. 2016;48(6):640–7.

    Article  CAS  PubMed  Google Scholar 

  8. Deng Y, Qiao L, Du M, Qu C, Wan L, Li J, et al. Age-related macular degeneration: Epidemiology, genetics, pathophysiology, diagnosis, and targeted therapy. Genes Dis. 2022;9(1):62–79.

    Article  CAS  PubMed  Google Scholar 

  9. Fleckenstein M, Keenan TDL, Guymer RH, Chakravarthy U, Schmitz-Valckenberg S, Klaver CC, et al. Age-related macular degeneration. Nat Rev Dis Primers. 2021;7(1):31.

    Article  PubMed  Google Scholar 

  10. Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, Chung CY, et al. Ranibizumab for neovascular age-related macular degeneration. N Engl J Med. 2006;355(14):1419–31.

    Article  CAS  PubMed  Google Scholar 

  11. Heier JS, Brown DM, Chong V, Korobelnik JF, Kaiser PK, Nguyen QD, et al. Intravitreal aflibercept (VEGF trap-eye) in wet age-related macular degeneration. Ophthalmology. 2012;119(12):2537–48.

    Article  PubMed  Google Scholar 

  12. Jaffe GJ, Ying GS, Toth CA, Daniel E, Grunwald JE, Martin DF, et al. Macular Morphology and Visual Acuity in Year Five of the Comparison of Age-related Macular Degeneration Treatments Trials. Ophthalmology. 2019;126(2):252–60.

    Article  PubMed  Google Scholar 

  13. Tamhane M, Cabrera-Ghayouri S, Abelian G, Viswanath V. Review of Biomarkers in Ocular Matrices: Challenges and Opportunities. Pharm Res. 2019;36(3):40.

    Article  PubMed Central  PubMed  Google Scholar 

  14. Keles A, Erol YO, Ayyildiz SN, Karaman SK, Ogus E. Aqueous Humor Levels of Vascular Endothelial Growth Factor and Stromal Cell-Derived Factor-1α in Age-Related Macular Degeneration. Beyoglu Eye J. 2021;6(4):298–303.

    PubMed Central  PubMed  Google Scholar 

  15. Mundo L, Tosi GM, Lazzi S, Pertile G, Parolini B, Neri G, et al. LRG1 Expression Is Elevated in the Eyes of Patients with Neovascular Age-Related Macular Degeneration. Int J Mol Sci. 2021;22(16):8879.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Murthy KR, Goel R, Subbannayya Y, Jacob HK, Murthy PR, Manda SS, et al. Proteomic analysis of human vitreous humor. Clin Proteomics. 2014;11(1):29.

    Article  PubMed Central  PubMed  Google Scholar 

  17. Rozanova S, Barkovits K, Nikolov M, Schmidt C, Urlaub H, Marcus K. Quantitative Mass Spectrometry-Based Proteomics: An Overview. Methods Mol Biol. 2021;2228:85–116.

    Article  CAS  PubMed  Google Scholar 

  18. Cunningham R, Ma D, Li L. Mass Spectrometry-based Proteomics and Peptidomics for Systems Biology and Biomarker Discovery. Front Biol (Beijing). 2012;7(4):313–35.

    Article  CAS  PubMed  Google Scholar 

  19. Lumbreras B, Porta M, Márquez S, Pollán M, Parker LA, Hernández-Aguado I. QUADOMICS: An adaptation of the Quality Assessment of Diagnostic Accuracy Assessment (QUADAS) for the evaluation of the methodological quality of studies on the diagnostic accuracy of ‘-omics’-based technologies. Clin Biochem. 2008;41(16):1316–25.

    Article  PubMed  Google Scholar 

  20. Parker LA, Gómez Saez N, Lumbreras B, Porta M, Hernández-Aguado I. Methodological deficits in diagnostic research using “-omics” technologies: evaluation of the QUADOMICS tool and quality of recently published studies. PLoS One. 2010;5(7):e11419.

    Article  PubMed Central  Google Scholar 

  21. Rinsky B, Beykin G, Grunin M, Amer R, Khateb S, Tiosano L, et al. Analysis of the Aqueous Humor Proteome in Patients With Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci. 2021;62(10):18.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Baek JH, Lim D, Park KH, Chae JB, Jang H, Lee J, et al. Quantitative proteomic analysis of aqueous humor from patients with drusen and reticular pseudodrusen in age-related macular degeneration. BMC Ophthalmol. 2018;18(1):289.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Kim TW, Kang JW, Ahn J, Lee EK, Cho KC, Han BN, et al. Proteomic analysis of the aqueous humor in age-related macular degeneration (AMD) patients. J Proteome Res. 2012;11(8):4034–43.

    Article  CAS  PubMed  Google Scholar 

  24. Tsai CY, Chen CT, Wu HH, Liao CC, Hua K, Hsu CH, et al. Proteomic Profiling of Aqueous Humor Exosomes from Age-related Macular Degeneration Patients. Int J Med Sci. 2022;19(5):893–900.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  25. Qu SC, Xu D, Li TT, Zhang JF, Liu F. iTRAQ-based proteomics analysis of aqueous humor in patients with dry age-related macular degeneration. Int J Ophthalmol. 2019;12(11):1758–66.

    Article  PubMed Central  PubMed  Google Scholar 

  26. Yao J, Liu X, Yang Q, Zhuang M, Wang F, Chen X, et al. Proteomic analysis of the aqueous humor in patients with wet age-related macular degeneration. Proteomics Clin Appl. 2013;7(7–8):550–60.

    Article  CAS  PubMed  Google Scholar 

  27. Kang GY, Bang JY, Choi AJ, Yoon J, Lee WC, Choi S, et al. Exosomal proteins in the aqueous humor as novel biomarkers in patients with neovascular age-related macular degeneration. J Proteome Res. 2014;13(2):581–95.

    Article  CAS  PubMed  Google Scholar 

  28. Lee H, Choi AJ, Kang GY, Park HS, Kim HC, Lim HJ, et al. Increased 26S proteasome non-ATPase regulatory subunit 1 in the aqueous humor of patients with age-related macular degeneration. BMB Rep. 2014;47(5):292–7.

    Article  PubMed Central  PubMed  Google Scholar 

  29. Nobl M, Reich M, Dacheva I, Siwy J, Mullen W, Schanstra JP, et al. Proteomics of vitreous in neovascular age-related macular degeneration. Exp Eye Res. 2016;146:107–17.

    Article  CAS  PubMed  Google Scholar 

  30. Koss MJ, Hoffmann J, Nguyen N, Pfister M, Mischak H, Mullen W, et al. Proteomics of vitreous humor of patients with exudative age-related macular degeneration. PLoS One. 2014;9(5):e96895.

    Article  PubMed Central  PubMed  Google Scholar 

  31. Schori C, Trachsel C, Grossmann J, Zygoula I, Barthelmes D, Grimm C. The Proteomic Landscape in the Vitreous of Patients With Age-Related and Diabetic Retinal Disease. Invest Ophthalmol Vis Sci. 2018;59(4):Amd31–amd40.

    Article  CAS  PubMed  Google Scholar 

  32. Balduzzi S, Rücker G, Schwarzer G. How to perform a meta-analysis with R: a practical tutorial. Evid Based Ment Health. 2019;22(4):153–60.

    Article  PubMed Central  PubMed  Google Scholar 

  33. Viechtbauer W. Conducting Meta-Analyses in R with the metafor Package. J Stat Softw. 2010;36(3):1–48.

    Article  Google Scholar 

  34. Harrer M, Cuijpers, P., Furukawa, T., Ebert, D. Doing Meta-Analysis with R: A Hands-On Guide (1st ed.). Chapman and Hall/CRC. 2021.

  35. Guo Y, Bao Y, Ma M, Yang W. Identification of Key Candidate Genes and Pathways in Colorectal Cancer by Integrated Bioinformatical Analysis. Int J Mol Sci. 2017;18(4):722.

    Article  PubMed Central  PubMed  Google Scholar 

  36. Mathurin K, Gallant MA, Germain P, Allard-Chamard H, Brisson J, Iorio-Morin C, et al. An interaction between L-prostaglandin D synthase and arrestin increases PGD2 production. J Biol Chem. 2011;286(4):2696–706.

    Article  CAS  PubMed  Google Scholar 

  37. 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.

    Article  CAS  PubMed  Google Scholar 

  38. Fukuhara A, Yamada M, Fujimori K, Miyamoto Y, Kusumoto T, Nakajima H, et al. Lipocalin-type prostaglandin D synthase protects against oxidative stress-induced neuronal cell death. Biochem J. 2012;443(1):75–84.

    Article  CAS  PubMed  Google Scholar 

  39. Wei Q, Zhang T, Fan J, Jiang R, Chang Q, Hong J, et al. Pathological myopia-induced antioxidative proteins in the vitreous humor. Ann Transl Med. 2020;8(5):193.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  40. Gomme PT, McCann KB, Bertolini J. Transferrin: structure, function and potential therapeutic actions. Drug Discov Today. 2005;10(4):267–73.

    Article  CAS  PubMed  Google Scholar 

  41. Elsayed ME, Sharif MU, Stack AG. Transferrin Saturation: A Body Iron Biomarker. Adv Clin Chem. 2016;75:71–97.

    Article  CAS  PubMed  Google Scholar 

  42. Wysokinski D, Danisz K, Blasiak J, Dorecka M, Romaniuk D, Szaflik J, et al. An association of transferrin gene polymorphism and serum transferrin levels with age-related macular degeneration. Exp Eye Res. 2013;106:14–23.

    Article  CAS  PubMed  Google Scholar 

  43. Wysokinski D, Danisz K, Pawlowska E, Dorecka M, Romaniuk D, Robaszkiewicz J, et al. Transferrin receptor levels and polymorphism of its gene in age-related macular degeneration. Acta Biochim Pol. 2015;62(2):177–84.

    Article  CAS  PubMed  Google Scholar 

  44. Wysokinski D, Blasiak J, Dorecka M, Kowalska M, Robaszkiewicz J, Pawlowska E, et al. Variability of the transferrin receptor 2 gene in AMD. Dis Markers. 2014;2014:507356.

    Article  PubMed Central  PubMed  Google Scholar 

  45. Henning Y, Blind US, Larafa S, Matschke J, Fandrey J. Hypoxia aggravates ferroptosis in RPE cells by promoting the Fenton reaction. Cell Death Dis. 2022;13(7):662.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Čolak E, Žorić L, Radosavljević A, Ignjatović S. The Association of Serum Iron-Binding Proteins and the Antioxidant Parameter Levels in Age-Related Macular Degeneration. Curr Eye Res. 2018;43(5):659–65.

    Article  PubMed  Google Scholar 

  47. Bigot K, Gondouin P, Bénard R, Montagne P, Youale J, Piazza M, et al. Transferrin Non-Viral Gene Therapy for Treatment of Retinal Degeneration. Pharmaceutics. 2020;12(9):836.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  48. Georgila K, Vyrla D, Drakos E. Apolipoprotein A-I (ApoA-I), Immunity, Inflammation and Cancer. Cancers (Basel). 2019;11(8):1097.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Woods AG, Sokolowska I, Taurines R, Gerlach M, Dudley E, Thome J, et al. Potential biomarkers in psychiatry: focus on the cholesterol system. J Cell Mol Med. 2012;16(6):1184–95.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  50. Boiko AS, Mednova IA, Kornetova EG, Semke AV, Bokhan NA, Loonen AJM, et al. Apolipoprotein serum levels related to metabolic syndrome in patients with schizophrenia. Heliyon. 2019;5(7):e02033.

    Article  PubMed Central  PubMed  Google Scholar 

  51. Wang L, Clark ME, Crossman DK, Kojima K, Messinger JD, Mobley JA, et al. Abundant lipid and protein components of drusen. PLoS One. 2010;5(4):e10329.

    Article  PubMed Central  PubMed  Google Scholar 

  52. Han X, Ong JS, Hewitt AW, Gharahkhani P, MacGregor S. The effects of eight serum lipid biomarkers on age-related macular degeneration risk: a Mendelian randomization study. Int J Epidemiol. 2021;50(1):325–36.

    Article  PubMed  Google Scholar 

  53. Nordestgaard LT, Tybjærg-Hansen A, Frikke-Schmidt R, Nordestgaard BG. Elevated Apolipoprotein A1 and HDL Cholesterol Associated with Age-related Macular Degeneration: 2 Population Cohorts. J Clin Endocrinol Metab. 2021;106(7):e2749–58.

    Article  PubMed  Google Scholar 

  54. Sivagurunathan S, Selvan LDN, Khan AA, Parameswaran S, Bhattacharjee H, Gogoi K, et al. Proteomics-based approach for differentiation of age-related macular degeneration sub-types. Indian J Ophthalmol. 2021;69(3):647–54.

    Article  PubMed Central  PubMed  Google Scholar 

  55. Armento A, Ueffing M, Clark SJ. The complement system in age-related macular degeneration. Cell Mol Life Sci. 2021;78(10):4487–505.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. 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.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Wagner EK, Raychaudhuri S, Villalonga MB, Java A, Triebwasser MP, Daly MJ, et al. Mapping rare, deleterious mutations in Factor H: Association with early onset, drusen burden, and lower antigenic levels in familial AMD. Sci Rep. 2016;6:31531.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. Kim BJ, Mastellos DC, Li Y, Dunaief JL, Lambris JD. Targeting complement components C3 and C5 for the retina: Key concepts and lingering questions. Prog Retin Eye Res. 2021;83:100936.

    Article  PubMed  Google Scholar 

  59. Park YG, Park YS, Kim IB. Complement System and Potential Therapeutics in Age-Related Macular Degeneration. Int J Mol Sci. 2021;22(13):6851.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  60. Lechner M, Wojnar P, Redl B. Human tear lipocalin acts as an oxidative-stress-induced scavenger of potentially harmful lipid peroxidation products in a cell culture system. Biochem J. 2001;356(Pt 1):129–35.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  61. Datta S, Cano M, Ebrahimi K, Wang L, Handa JT. The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Prog Retin Eye Res. 2017;60:201–18.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. SanGiovanni JP, Chen J, Sapieha P, Aderman CM, Stahl A, Clemons TE, et al. DNA sequence variants in PPARGC1A, a gene encoding a coactivator of the ω-3 LCPUFA sensing PPAR-RXR transcription complex, are associated with NV AMD and AMD-associated loci in genes of complement and VEGF signaling pathways. PLoS One. 2013;8(1):e53155.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Canter JA, Olson LM, Spencer K, Schnetz-Boutaud N, Anderson B, Hauser MA, et al. Mitochondrial DNA polymorphism A4917G is independently associated with age-related macular degeneration. PLoS One. 2008;3(5):e2091.

    Article  PubMed Central  PubMed  Google Scholar 

  64. Holzfeind P, Merschak P, Wojnar P, Redl B. Structure and organization of the porcine LCN1 gene encoding Tear lipocalin/von Ebner’s gland protein. Gene. 1997;202(1–2):61–7.

    Article  CAS  PubMed  Google Scholar 

  65. Wang JY, Kwon JS, Hsu SM, Chuang HS. Sensitive tear screening of diabetic retinopathy with dual biomarkers enabled using a rapid electrokinetic patterning platform. Lab Chip. 2020;20(2):356–62.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

Supported by Jiangsu Provincial Medical Innovation Team (grant No. CXTDA2017039) and the National Natural Science Foundation in China (grant No. 81671641, 82271113) to PL.

Author information

Authors and Affiliations

Authors

Contributions

HG and PL designed study; HG and JL conducted research and wrote the paper; HG performed statistical analysis; PL had primary responsibility for final content. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Peirong Lu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1: Table S1.

QUADOMICS criteria to evaluate the quality of the -omics research reports included in a systematic review.

Additional file 2:

 Figure S1. QUADOMICS evaluation of the quality of the proteomics studies included in the systematic review.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, H., Li, J. & Lu, P. Systematic review and meta-analysis of mass spectrometry proteomics applied to ocular fluids to assess potential biomarkers of age-related macular degeneration. BMC Ophthalmol 23, 507 (2023). https://doi.org/10.1186/s12886-023-03237-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12886-023-03237-0

Keywords