Open Access
Retina  |   November 2024
Potential Drug Targets for Diabetic Retinopathy Identified Through Mendelian Randomization Analysis
Author Affiliations & Notes
  • Huan Liu
    Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, PR China
  • Feiyan Wang
    Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, PR China
  • Ziqing Hu
    Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
  • Jing Wei
    Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan, PR China
  • Correspondence: Jing Wei, Department of Ophthalmology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, No. 24 Jinghua Rd., Luoyang, Henan 471003, PR China. e-mail: 18567635881@163.com 
Translational Vision Science & Technology November 2024, Vol.13, 17. doi:https://doi.org/10.1167/tvst.13.11.17
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Huan Liu, Feiyan Wang, Ziqing Hu, Jing Wei; Potential Drug Targets for Diabetic Retinopathy Identified Through Mendelian Randomization Analysis. Trans. Vis. Sci. Tech. 2024;13(11):17. https://doi.org/10.1167/tvst.13.11.17.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

Purpose: This study aimed to investigate the causal effect of plasma proteins on diabetic retinopathy (DR) risk and identify potential drug targets for this disease.

Methods: Two-sample Mendelian randomization was performed to explore potential drug targets for DR. A total of 734 proteins were selected as instrumental variables. The Steiger filtering test and colocalization analysis were conducted to determine the causal direction and genetic pleiotropy. Plasma proteins from the decode study were used to validate the findings.

Results: Eleven plasma proteins were associated with DR risk. Genetically predicted high levels of CCL3L1 (odds ratio [OR] = 0.582; 95% confidence interval [CI], 0.343–0.986; P = 0.044), PAM (OR = 0.782; 95% CI, 0.652–0.937; P = 0.008), GP1BA (OR = 0.793; 95% CI, 0.632–0.994; P = 0.044), GALNT16 (OR = 0.832; 95% CI, 0.727–0.952; P = 0.008), POGLUT1 (OR = 0.836; 95% CI = 0.703–0.995; P = 0.043), and DKK3 (OR = 0.859; 95% CI, 0.777–0.950; P = 0.003) have the protective effect on DR risk. Genetically predicted high levels of GFRA2 (OR = 1.104; 95% CI, 1.028–1.187; P = 0.007), PATE4 (OR = 1.405; 95% CI, 1.060-1.860; P = 0.018), GSTA1 (OR = 1.464; 95% CI, 1.163–1.842; P = 0.001), SIRPG (OR = 1.600, 95% CI, 1.244–2.057; P = 2.51E-04), and MAPK13 (OR = 1.731; 95% CI, 1.233–2.431; P = 0.002) were associated with an increased risk of DR. However, the colocalization analysis results suggested that SIRPG and GP1BA have a shared causal variant with DR.

Conclusions: CCL3L1, PAM, GALNT16, POGLUT1, DKK3, GFRA2, PATE4, GSTA1, and MAPK13 were associated with DR risk and were identified as potential drug targets for DR.

Translational Relevance: The present study has highlighted the role of CCL3L1, PAM, GALNT16, POGLUT1, DKK3, GFRA2, PATE4, GSTA1, and MAPK13 in the development of DR.

Introduction
Diabetic retinopathy (DR) is prevalent microvascular complication of diabetes and a leading cause of blindness in working-age individuals globally.1 DR is a prevalent retinal vascular disease characterized by increased vascular permeability, retinal ischemia and edema, new blood vessel formation, and retinal inflammation.2 In 2020, it was estimated that more than 103 million individuals globally were affected by DR, with projections suggesting an increase to 130 million by 2030 and 161 million by 2045.3 It is highlighted that screening for DR is crucial for identifying eyes at high risk of vision loss, because it enables appropriate management and timely treatment.4 
The DR etiology is complex, and its pathogenesis is not fully understood. Previous studies have demonstrated that prolonged hyperglycemia is a major risk factor for the development of DR.5 Genetic susceptibility, inflammation and immune cell activation, dietary and lifestyle habits, blood pressure, and plasma lipid levels are the primary risk factors for DR development.68 DR progresses through two clinical stages: nonproliferative DR (NPDR) and proliferative DR (PDR).9 Currently, no effective treatment is clinically available for DR. Therapies for early-stage NPDR focus on controlling blood glucose, blood pressure, and lipid levels.10 The mainstay treatment for PDR is laser photocoagulation and surgery.8,10 Intraocular injections of steroids and anti-vascular endothelial growth factor (VEGF) were the primary treatment for macular edema (ME) caused by diabetes mellitus.11,12 Previous studies have demonstrated that the overexpression of VEGF contributed to the hyperpermeability of retinal vessels, the breakdown of the blood-retinal barrier, and neovascularization, resulting in ME in DR.8 VEGF-targeting drugs have been developed as a first-line treatment for diabetic macular edema (DME).11 Although current treatment strategies may slow DR progression, developing new therapies to prevent the onset or slow DR progression remains a high priority. 
Proteins play a critical role in the development and progression of human diseases and serve as primary drug targets.13,14 Proteomic changes in the serum, plasma, vitreous humor, aqueous humor, and tears of DR patients have been identified through the use of advanced high-throughput proteomic technologies.15,16 These findings provide insight into the pathophysiological mechanisms underlying DR.15,16 Furthermore, proteins, including VEGF, interleukin-6, and alpha-1-acid glycoprotein, have emerged as potential diagnostic biomarkers and therapeutic targets for DR.11,17 However, the clinical value of these potential drug targets requires further studies and validation. 
Mendelian randomization (MR) analysis is widely used to assess the potential causal relationships between proteins and diseases and for new drug development or drug repurposing.18 MR is a genetic epidemiological method that uses single nucleotide polymorphisms (SNPs) as instrumental variables (IVs) to assess the causal effect of exposure on outcome.19 Compared to traditional observational studies, the MR method reduces the effect of confounders or reverse causation bias by leveraging the random allocation of genetic variants at conception.20 MR analyses have identified glucagon-like peptide-1 receptor agonists as potential therapeutic targets for DR.21 However, other proteins with potential applications as therapeutic drugs for DR have rarely been studied. This study aimed to investigate the causal effect of proteins on DR and identify the potential drug targets for the disease using a two-sample MR analysis. 
Material and Methods
Study Design
A two-sample MR study was designed to identify potential drug targets for DR. This study was based on three critical assumptions: (i) The genetic variants selected as IVs must be associated with plasma proteins; (ii) Genetic variants used as IVs for plasma proteins must not be associated with known confounding factors; and (iii) Genetic variants serving as IVs should influence the risk of DR exclusively through plasma proteins. The general design of this study is presented in Figure 1
Figure 1.
 
The overview of the two-sample MR study design. Assumption 1: The genetic variants selected as IVs must be associated with plasma proteins; Assumption 2: Genetic variants used as IVs for plasma proteins must not be associated with known confounding factors. Assumption 3: Genetic variants serving as IVs should influence the risk of DR exclusively through plasma proteins.
Figure 1.
 
The overview of the two-sample MR study design. Assumption 1: The genetic variants selected as IVs must be associated with plasma proteins; Assumption 2: Genetic variants used as IVs for plasma proteins must not be associated with known confounding factors. Assumption 3: Genetic variants serving as IVs should influence the risk of DR exclusively through plasma proteins.
Data Source and the Selection of Instrumental Variables
The source of protein quantitative trait loci (pQTL) data used in the study was retrieved from published genome-wide association studies (GWAS) conducted by Zheng et al.14 (http://www.epigraphdb.org/pqtl/). Zheng et al.14 integrated data from five previously published GWAS, which were downloaded from the IEU database (https://gwas.mrcieu.ac.uk/). These five recently published studies included 3301, 3200, 1000, 3394, and 6861 participants of European descent, respectively.2226 A total of 3606 pQTLs associated with 2656 proteins (1981 instruments for 1478 proteins, 539 instruments for 284 proteins, 80 instruments for 58 proteins, 131 instruments for 60 proteins, and 875 instruments for 776 proteins) were obtained from the five previously published GWAS at a P value threshold of ≤5 × 10−8. After removing linkage disequilibrium (LD) within a 500-kb window from either side of the leading pQTL of the protein and r2 < 0.001 threshold and SNPs located within the human major histocompatibility complex region (chr6: from 26-34 Mb), 2113 instruments for 1699 proteins were retained for further analysis. Additionally, 987 instruments were classified as nonspecific because they were associated with more than five proteins and were excluded from further analysis. Consequently, 1136 pQTLs were selected as IVs, including 783 cis-acting and 343 trans-acting pQTLs. In our study, 738 cis-acting SNPs for 734 proteins were selected as IVs (Supplementary Table S1). The detailed analysis workflow for the IVs selected was described in Zheng et al.14 A summary of the samples of pQTLs used in this study can be found in Supplementary Table S1
Data for Outcome
Data on DR were obtained from the FinnGen database (R9 release), which included 6818 cases and 344,569 controls of European descent (https://storage.googleapis.com/finngen-public-data-r9/summary_stats/finngen_R9_H7_RETINOPATHYDIAB.gz). The FinnGen database includes samples sourced from Finnish biobanks and phenotype data gathered from national health registers.27 The Helsinki and Uusimaa Hospital District Coordinating Ethics Committee reviewed and approved the FinnGen study project. The project complies with current legislation, including the Biobank Act and the Personal Data Act. The official data controller for this study is the University of Helsinki. 
MR Analysis
A two-sample MR approach was used to assess the effects of plasma proteins on DR risk. If only one SNP was available, the Wald ratio method was used to examine causal effect.28 If two or more SNPs were available, the inverse variance weighted (IVW) method was used to assess the causal effect of plasma proteins on DR.29 For plasma proteins with three or more SNPs, the MR Egger, weighted median, weighted mode, and simple mode methods were used as supplementary analyses.30,31 The MR pleiotropy residual sum and outlier test was applied to detect outliers and correct for widespread horizontal pleiotropy.32 If there is significant heterogeneity, the random effects IVW model will be used as the primary analysis; otherwise, the fixed effects IVW model will be used as the primary analysis. 
A threshold P value < 0.05 was considered statistically significant for the MR analysis. The associations between plasma proteins and DR risk were measured by odds ratios (ORs) per standard deviation (SD). The corresponding 95% confidence intervals (CIs) and ORs for increased DR risk were expressed per SD increase in plasma protein levels. For analyses with two or more IVs, the MR results were presented as scatter plots. 
Sensitivity Analysis
PhenoScanner (http://www.phenoscanner.medschl.cam.ac.uk/) was used to minimize the influence of confounding factors and enhance the robustness of the causal findings. The Steiger directionality test was conducted to assess the causal relationship between plasma proteins and DR, as well as to reduce the potential impact of reverse causality.33 Proxy and palindromic and ambiguous SNPs were excluded from subsequent analyses. The F-statistics and explained variance were calculated to assess the strength of the instruments. An F-statistic greater than 10 was considered indicative of a sufficiently strong instrument, whereas R2 represented the proportion of variation in each plasma protein explained by the instrument. 
Bioinformatics Analysis
Bioinformatic analysis was performed to investigate the biological functions of plasma proteins (P < 0.05) associated with DR risk. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment pathway analyses were performed on the protective and risk plasma proteins. Protein-protein interaction (PPI) network diagrams were generated using the protective and risk proteins to gain deeper insight into their interactions. 
External Validation of Potential Drug Targets for DR
External validation was performed to assess the effect of plasma proteins on DR and to verify the reliability of the MR results. In the external validation, 4719 unique proteins corresponding to 4907 aptamer targets were measured in 35,559 individuals of European ancestry from the deCODE study (https://download.decode.is/form/folder/proteomics).34 
Colocalization Analysis
Colocalization analysis, a Bayesian-based method, was performed to assess whether plasma proteins and DR share a common causal variant in a given region. The colocalization analysis was based on posterior probabilities for the five hypotheses: PPH0, no causal variants for either trait; PPH1, a causal variant for trait 1; PPH2, a causal variant for trait 2; PPH3, two different causal variants for trait 1 and 2; and PPH4, a shared causal variant between two traits. PPH4 > 80% was considered to provide strong evidence of colocalization. 
Potential Therapeutic Drugs Prediction and Molecular Docking
To further identify potential therapeutic drugs (small molecule drugs) for drug targets, we searched each drug target to obtain the small molecule drugs and drug target interactions using the PubChem database. The compound library used for the molecular docking analysis was sourced from Interactions and Pathways in the PubChem database. For example, small molecule drugs that interact with GALNT16 can be found at https://pubchem.ncbi.nlm.nih.gov/gene/57452#section=Interactions-and-Pathways. These small molecule drugs were identified by searching the database for plasma proteins and determining their interactions. For proteins associated with increased risk, we identified small-molecule drugs that reduce their expression levels. Conversely, for protective proteins, we identified small-molecule drugs that enhance their expression levels. Information on the effects of small molecule drugs that increase or decrease protein expression was obtained from the Action section of Interactions and Pathways in the PubChem database. Evidence concerning the effects of small molecule drugs that increase or decrease protein expression was derived from published literature, which was also obtained from the Evidence IDs section of Interactions and Pathways in the PubChem database (Supplementary Table S5). We further explored whether small molecule drugs (ligands) could interact with drug targets (grids) using molecular docking analysis. Two-dimensional structures of small molecule ligands were obtained from the PubChem database. The three-dimensional protein structures were obtained from the RCSB Protein Data Bank (https://www.rcsb.org/). Molecular docking analysis was performed using AutoDock Vina 1.1.2, and the docking results were visualized using PyMOL software 2.5.0. Binding affinity between ligands and drug targets was evaluated based on binding energy (kcal/mol). Binding energies of less than −5.0 and −7.0 kcal/mol were considered indicative of good and strong binding affinities, respectively. 
Statistical Analysis
The results of this study were reported in accordance with the Strengthening the Reporting of Observational Studies in Epidemiology Using Mendelian Randomization (Supplementary Table S2).35 MR analysis was performed using TwoSampleMR.32 Colocalization analysis was performed using the Coloc package.36 GO and KEGG enrichment analyses were performed using the R package “clusterProfler” (version 3.18.1).37 PPI was constructed using the online tool STRING (version 11.5, https://cn.string-db.org/). 
Results
Causal Effects of Plasma Proteins on DR
Thirty-eight SNPs could not be obtained from the GWAS summary data for DR, and 12 SNPs with palindromic and ambiguous allele frequencies were excluded in the subsequent two‐sample MR analysis. One SNP with the potential reverse causal was excluded through Steiger filtering. A total of 687 SNPs were used to assess the effects of 684 plasma proteins on DR risk (Supplementary Table S1). We identified that 55 plasma proteins were independently associated with greater risk of DR (Table 1). Among these, 23 plasma proteins were protective against DR risk, while 32 were positively associated with increased DR risk. The results are visualized in a volcano plot (Fig. 2A), and the F-statistics and variance for each association are provided in Supplementary Table S1. The scatter plot for plasma protein PLXNC1 is displayed in Supplementary Figure S1A. 
Table 1.
 
The Causal Effect of Plasma Proteins on DR Risk in Zheng's Study Cohort
Table 1.
 
The Causal Effect of Plasma Proteins on DR Risk in Zheng's Study Cohort
Figure 2.
 
Bioinformatics analysis results. (A) Volcano plot. (B) GO enrichment analysis for risk proteins. (C) GO enrichment analysis for protective proteins. (D) Protein-protein interaction. (1) The points above the black dashed horizontal line and log (OR) > 0 represent that the plasma proteins were positively associated with increased DR risk, on the contrary, the points above the black dashed horizontal line and log (OR) < 0 represent that the plasma proteins were protective against DR risk (A). (2) Count represents the number of plasma proteins enriched in this pathway (B, C). (3) The black solid line represents the interaction relationship between plasma proteins (D).
Figure 2.
 
Bioinformatics analysis results. (A) Volcano plot. (B) GO enrichment analysis for risk proteins. (C) GO enrichment analysis for protective proteins. (D) Protein-protein interaction. (1) The points above the black dashed horizontal line and log (OR) > 0 represent that the plasma proteins were positively associated with increased DR risk, on the contrary, the points above the black dashed horizontal line and log (OR) < 0 represent that the plasma proteins were protective against DR risk (A). (2) Count represents the number of plasma proteins enriched in this pathway (B, C). (3) The black solid line represents the interaction relationship between plasma proteins (D).
GO and KEGG Pathway Analysis and PPI Network
The potential biological functions of plasma proteins associated with DR were explored using GO and KEGG pathway analyses. These analyses were conducted separately for protective and risk-related proteins. The top 10 enriched GO biological process terms (P < 0.05) for risk proteins included negative regulation of immune response, adaptive immune response via immunoglobulin superfamily domains, membrane protein proteolysis, lymphocyte-mediated immunity, heparin metabolic process, surfactant homeostasis, synapse pruning, regulation of humoral immune response mediated by circulating immunoglobulin, chemokine-mediated signaling pathway, and response to chemokines (Fig. 2B). The top 10 GO biological process terms (P < 0.05) for protective proteins included monocyte chemotaxis, leukocyte migration, leukocyte chemotaxis, response to interleukin-1, lymphocyte chemotaxis, cell chemotaxis, mononuclear cell migration, chemokine-mediated signaling pathway, eosinophil chemotaxis, and response to chemokines (Fig. 2C). No significant KEGG terms were identified for either protective or risk-related proteins. The PPI network for plasma proteins associated with DR risk is presented in Figure 2D. 
Validation of Causal Effects of Plasma Proteins on DR Risk
Replication analysis validated the association of 11 out of 55 plasma proteins in an independent cohort. Genetically predicted higher levels of C-C motif chemokine 3-like 1 (CCL3L1) (OR = 0.582; 95% CI, 0.343–0.986; P = 0.044), peptidyl-glycine alpha-amidating monooxygenase (PAM) (OR = 0.782; 95% CI, 0.652–0.937; P = 0.008), platelet glycoprotein Ib alpha chain (GP1BA) (OR = 0.793; 95% CI, 0.632–0.994; P = 0.044), polypeptide N-acetylgalactosaminyltransferase 16 (GALNT16) (OR = 0.832; 95% CI, 0.727–0.952; P = 0.008), protein O-glucosyltransferase 1 (POGLUT1) (OR = 0.836; 95% CI = 0.703–0.995; P = 0.043), and dickkopf-related protein 3 (DKK3) (OR = 0.859; 95% CI, 0.777–0.950; P = 0.003) were found to be protective against DR (Table 2). Conversely, higher levels of glial cell line-derived neurotrophic factor family receptor alpha 2 (GFRA2) (OR = 1.104; 95% CI, 1.028–1.187; P = 0.007), prostate and testis-expressed protein 4 (PATE4) (OR = 1.405; 95% CI, 1.060–1.860; P = 0.018), glutathione S-transferase A1 (GSTA1) (OR = 1.464; 95% CI, 1.163–1.842; P = 0.001), signal-regulatory protein gamma (SIRPG) (OR = 1.600; 95% CI, 1.244–2.057; P = 2.51E-04), and mitogen-activated protein kinase 13 (MAPK13) (OR = 1.731; 95% CI, 1.233–2.431; P = 0.002) were associated with increased DR risk (Table 2). Supplementary MR analysis results for DKK3, GFRA2, and GALNT16 are shown in Supplementary Table S3, and scatter plots for these proteins are presented in Supplementary Figures S1B–D. Sensitivity analysis further confirmed the reliability of the results (Supplementary Table S3). 
Table 2.
 
The Causal Effect of Plasma Proteins on DR Risk in deCODE Cohort
Table 2.
 
The Causal Effect of Plasma Proteins on DR Risk in deCODE Cohort
Reverse MR Analysis
A reverse MR study was performed to investigate whether there is a reverse causal effect between DR and plasma proteins. Based on the IVW method, genetically predicted DR was not associated with CCL3L1 (OR = 1.04 [1.00–1.09], P = 0.080), DKK3 (OR = 1.05 [0.99–1.11], P = 0.109), GALNT16 (OR = 1.02 [0.97–1.06], P = 0.434), GFRA2 (OR = 1.00 [0.94–1.07], P = 0.995), GP1BA (OR = 1.03 [0.99–1.08], P = 0.133), GSTA1 (OR = 1.01 [0.95–1.07], P = 0.770), MAPK13 (OR = 0.99 [0.95–1.04], P = 0.729), PAM (OR = 1.01 [0.95–1.07], P = 0.687), PATE4 (OR = 0.99 [0.94–1.03], P = 0.516), POGLUT1 (OR = 1.00 [0.96–1.05], P = 0.983), SIRPG (OR = 1.04 [0.99–1.09], P = 0.096) (Fig. 3). Complementary analyses were consistent with the IVW results (Supplementary Table S4). 
Figure 3.
 
The causal effect of DR on plasma proteins risk based on the IVW method. (1) IVW_FE: the fixed-effect IVW model was used to assess the causal effect of plasma proteins on DR. IVW_MRE: the random-effects IVW model was used to assess the causal effect of plasma proteins on DR.
Figure 3.
 
The causal effect of DR on plasma proteins risk based on the IVW method. (1) IVW_FE: the fixed-effect IVW model was used to assess the causal effect of plasma proteins on DR. IVW_MRE: the random-effects IVW model was used to assess the causal effect of plasma proteins on DR.
Genetic Colocalization Analysis
Bayesian colocalization analysis suggested that SIRPG (coloc.abf-PPH4 = 0.837), and GP1BA (coloc.abf-PPH4 = 0.94) shared the same variant with DR. CCL3L1 (coloc.abf-PPH4 = 0.004), GALNT16 (coloc.abf-PPH4 = 0.010), PATE4 (coloc.abf-PPH4 = 0.038), GFRA2 (coloc.abf-PPH4 = 0.087), POGLUT1 (coloc.abf-PPH4 = 0.090), DKK3 (coloc.abf-PPH4 = 0.197), GSTA1(coloc.abf-PPH4 = 0.395), and DR were unlikely to share a causal variant within the same locus (Table 3 and Supplementary Fig. S2). 
Table 3.
 
Colocalization Analysis of Plasma Proteins With DR
Table 3.
 
Colocalization Analysis of Plasma Proteins With DR
Molecular Docking Results
For risk proteins, small-molecule drugs were predicted to reduce the expression of target proteins, whereas for protective proteins, these drugs were predicted to increase their expression. Our analysis showed that dronabinol and doxorubicin could enhance the expression of CCL3L1. Acetaminophen, aristolochic acid, temozolomide, doxorubicin, tretinoin, decitabine, halofuginone, and fenretinide can increase the expression of DKK3. Valproic acid, triptonide, and ketamine can increase the expression of GALNT16. Bisphenol A, coumestrol, dexamethasone, doxorubicin, pirinixic acid, reserpine, tretinoin, and triptonide can increase the expression of PAM. In addition, dexamethasone, fenretinide, genistein, resveratrol, rotenone, and acetaminophen can decrease the expression of GFRA2. Acetaminophen, abrine, artemisinin, bardoxolone methyl, ciprofibrate, clotrimazole, curcumin, cyclosporine, deoxycholic acid, eugenol, idoxifene, and methyleugenol can decrease the expression of GSTA1. Bisphenol A, fostamatinib, genistein, minocycline, resveratrol, and vancomycin can reduce the expression of MAPK13. Bisphenol A and Genistein can reduce the expression of PATE4 (Table 4). The binding energy < −7.0 kcal/mol between the small molecule ligands and drug targets was plotted the molecular docking diagram (Fig. 4). 
Table 4.
 
The Binding Energy Between Drug Targets and the Predicted Drugs
Table 4.
 
The Binding Energy Between Drug Targets and the Predicted Drugs
Figure 4.
 
Result of molecular docking. (A–X) Action mode of small-molecule drugs with small-molecule drugs. (1) The term before the “_” refers to plasma proteins, whereas the term after the “_” refers to the docking small molecule drugs in the Figure legends (A–X). (2) Molecular docking results between the small molecule drugs (ligands, Cyan molecular structure) and receptor (plasma proteins) in the enlarged figures. The numbers represent the binding energy (for example, “2.7” in A), and “GLY” et al. was the abbreviation for amino acids (A).
Figure 4.
 
Result of molecular docking. (A–X) Action mode of small-molecule drugs with small-molecule drugs. (1) The term before the “_” refers to plasma proteins, whereas the term after the “_” refers to the docking small molecule drugs in the Figure legends (A–X). (2) Molecular docking results between the small molecule drugs (ligands, Cyan molecular structure) and receptor (plasma proteins) in the enlarged figures. The numbers represent the binding energy (for example, “2.7” in A), and “GLY” et al. was the abbreviation for amino acids (A).
Discussion
This study used pQTL and GWAS summary data to investigate the causal effects of proteins on DR risk through two-sample MR. Sensitivity and colocalization analyses identified CCL3L1, PAM, GP1BA, GALNT16, POGLUT1, DKK3, GFRA2, PATE4, GSTA1, SIRPG, and MAPK13 as potential drug targets for DR. Colocalization analysis indicated that SIRPG and GP1BA likely share a causal variant with DR, suggesting possible bias due to LD. 
In this study, genetically predicted high levels of CCL3L1, PAM, GALNT16, POGLUT1, and DKK3 were protective against DR. CCL3L1, also known as LD78β, encodes the MIP-1αP protein, and is a potent chemoattractant for monocytes and lymphocytes.38 CCL3L1 has a high affinity for CCR5 and is a potent HIV-1 inhibitor. CCR5+ CD11b+ monocyte-macrophages are involved in retinal microvascular occlusion.39 PAM, an amidate neuroendocrine peptide, was highly expressed in human islets.40 Studies have demonstrated that SNPs within PAM (rs78408340 and rs35658696) were associated with type 2 diabetes risk in Europeans.41,42 PAM is a key amidating enzyme in the beta cell-regulated secretory pathway. PAM deficiency decreases insulin content and changes insulin secretion dynamics in a human beta cell model and primary islets from cadaveric donors.40 These reports are consistent with our results. GALNT16, a paralog of GALNT2, is a glycosyltransferase.43 Down-regulation of GALNT2 has been observed in obese patients with type 2 diabetes.44 Decreased GALNT2 expression impaired insulin signaling and action in HepG2 cells.45 Moreover, the decreased GALNT2 expression has been associated with insulin resistance and atherogenic dyslipidemia.46 However, the biological role of GALNT16 has not been investigated in DR. To the best of our knowledge, this study is the first to demonstrate that elevated levels of GALNT16 have a protective effect against DR risk. POGLUT1, a protein-O-glucosyltransferase, recognizes a diverse set of Notch epidermal growth factor-like domain substrates and plays a distinct role in modulating the Notch signaling pathway.47 Activation of the Notch1 signaling pathway was significantly enriched in Müller glia, astrocytes, and pericytes, and was associated with retinal barrier function in DR.48 Dickkopf-3 (Dkk3), a secreted glycoprotein, acts as a negative regulator of the Wnt/β-catenin signaling pathway.49 Aberrant activation of this pathway is a leading cause of pathological ocular neovascularization in DR.50 Additionally, Dkk3 is crucial for constitutive VEGF expression. Elevated levels of Dkk3 in the aqueous humor have been observed in DME patients.51 
Genetically predicted high levels of GFRA2, PATE4, GSTA1, and MAPK13 were associated with an increased risk of DR. GFRA2, a cell-surface receptor, is the preferential co-receptor for neurturin. GFRA2 belongs to the glial cell line-derived neurotrophic factor family ligands.52 A SNP within GFRA2 is associated with DR.53 In addition, high GFRA2 expression has been previously reported in epiretinal membranes after human DR.54 GSTA1, an antioxidant, detoxifies reactive oxygen species by conjugating with glutathione and plays an essential role in cellular detoxification and in protecting cells from oxidative damage.55 SNPs within the GSTA1 were associated with smoking-related type 2 diabetes in Japanese.56 High GSTA1 levels were associated with elevated markers of systemic inflammation in the urine of juvenile type 1 diabetes.57 In addition, proteomic studies have demonstrated that higher levels of GSTA1 are associated with an increased risk of diabetes. MAPK13, a member of the MAP kinase family, plays an essential role in the cellular response cascade evoked by proinflammatory cytokines.58 The lack of MAPK13 protected against insulin resistance and pancreatic beta cell failure in mice.59 MAPK13 was also involved in angiogenesis during early DR.60 
The results of GO and KEGG enrichment analyses highlighted that the cytokine signaling pathway and chemokine signaling pathway are involved in the occurrence of DR. The role of immune response, cytokine and chemokine in the development of DR has been well established.61 Immune response, cytokine, and chemokine are associated with the destruction of the blood-retinal barrier and the onset and progression of DR.62,63 We provide further evidence that immune response, cytokine and chemokine play a role in the pathogenesis of DR. The protein interaction network was applied to explore the mutual regulation between proteins. Our results confirmed that these proteins were independent of each other. 
Molecular docking was a powerful tool for drug discovery. Our study found several therapeutic drugs (such as cyclosporine, dexamethasone, and doxorubicin) for drug targets by molecular docking. Cyclosporine, dexamethasone, and doxorubicin were commonly used to inhibit immune and inflammatory responses. Iglicki et al.64 have demonstrated that the intravitreal dexamethasone implant (DEX) has the potential to both delay the progression of DR and improve its severity over 24 months. Iglicki et al.64 also demonstrated DEX could effectively and safely reduce central retinal thickness using optical coherence tomography (OCT) with spectral-domain OCT (Heidelberg Engineering, Heidelberg, Germany) and improve the best-corrected visual acuity in DME.65 Besides, swept-source OCT (SS-OCT) and spectral-domain OCT (SD-OCT) are important diagnostic tools for diagnosing and managing patients with DME by measuring the central retinal thickness.66 Compared to SD-OCT, SS-OCT uses a longer wavelength and provides significantly superior performance in evaluating the choroid with enhanced resolution and greater depth.67 The clinical applications of SD-OCT in enhanced depth imaging mode and SS-OCT may further contribute to studying choroid's role in the pathophysiology of DME and investigating choroidal biomarkers in DR patients with DME. However, further studies are needed to validate the role of these therapeutic drugs in DR. 
The study had several strengths. First, we used the large-scale plasma proteins GWAS summary data and DR GWAS summary data to investigate the causal effect of plasma proteins on DR risk and identify potential drug targets for this disease. Our findings have important implications for clinical research and the further development of these drugs for DR. Besides, the current study introduced a new conceptual framework and provided valuable clinical insights into this disease. For example, high levels of MAPK13 were associated with an increased risk of DR. Targeting MAPK13 could be beneficial for DR treatment. Additionally, the MR study mitigated the influence of confounding factors and provided a fresh perspective for exploring the pathogenesis of DR. Moreover, the restriction of the study sample to individuals of European descent helped reduce stratification bias, and the use of an independent external dataset for validation ensured more reliable results. 
This study also had several limitations. First, our study only included participants of European ancestry, and the results may not be generalizable to other ethnic groups. Second, most plasma proteins are associated with only one cis-acting SNP, which restricts the application of other MR algorithms, such as heterogeneity and pleiotropy tests. Third, we did not validate our results in animal models or at the molecular level. Moreover, our study is exploratory to identify more potential drug targets for DR, and the Bonferroni correction was not used in the current study. This may be a limitation of our study. However, we used an external validation dataset to validate our results. This may compensate for the limitations of the study. In future research, we will conduct experiments to verify our results and compensate for these limitations. Additionally, due to the complexity of genomic data and the challenges of multiple comparisons for MR study, P-value correction is an important step. However, we also acknowledge that in large-scale genomic data analyses, the large sample size and the high number of hypothesis tests typically result in a more stringent P-value threshold, which reduces the likelihood of finding significant results. In some previously published genomic MR studies, P-values were also uncorrected.68,69 
Conclusions
This study demonstrated for the first time that high levels CCL3L1, PAM, GALNT16, POGLUT1, and DKK3 are associated with a protective effect against DR risk, Conversely, genetically predicted high levels of GFRA2, PATE4, GSTA1, and MAPK13 were associated with an increased risk of DR. These proteins are potential drug targets for DR. 
Acknowledgments
The author thank the participants and investigators of the FinnGen study, the deCODE study, and the Zheng study. 
Supported by the Medical Science and Technology Project of Henan province of China (Grant No. LHGJ20230471). 
Author Contributions: JW and HL designed the study. HL collected and analyzed data. HL and FYW conducted the literature search. HL wrote the first draft of the paper. ZQH and JW supervised the study. 
Data Availability: Supplemental data are available in Translational Vision Science and Technology. The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request. 
Disclosure: H. Liu, None; F. Wang, None; Z. Hu, None; J. Wei, None 
References
Cheung N, Mitchell P, Wong TY. Diabetic retinopathy. Lancet (London, England). 2010; 376: 124–136. [CrossRef] [PubMed]
Do DV, Wang X, Vedula SS, et al. Blood pressure control for diabetic retinopathy. Cochrane Database Syst Rev. 2015; 1: Cd006127. [PubMed]
Teo ZL, Tham YC, Yu M, et al. Global prevalence of diabetic retinopathy and projection of burden through 2045: systematic review and meta-analysis. Ophthalmology. 2021; 128: 1580–1591. [CrossRef] [PubMed]
Tang F, Luenam P, Ran AR, et al. Detection of diabetic retinopathy from ultra-widefield scanning laser ophthalmoscope images: a multicenter deep learning analysis. Ophthalmology Retina. 2021; 5: 1097–1106. [CrossRef] [PubMed]
Wong TY, Cheung CM, Larsen M, Sharma S, Simó R. Diabetic retinopathy. Nat Rev Dis Primers. 2016; 2: 16012. [CrossRef] [PubMed]
Bryl A, Mrugacz M, Falkowski M, Zorena K. The effect of diet and lifestyle on the course of diabetic retinopathy—a review of the literature. Nutrients. 2022; 14: 1252. [CrossRef] [PubMed]
Han J, Lando L, Skowronska-Krawczyk D, Chao DL. Genetics of diabetic retinopathy. Curr Diabetes Rep. 2019; 19: 67. [CrossRef]
Stitt AW, Curtis TM, Chen M, et al. The progress in understanding and treatment of diabetic retinopathy. Prog Retin Eye Res. 2016; 51: 156–186. [CrossRef] [PubMed]
Nawaz IM, Rezzola S, Cancarini A, et al. Human vitreous in proliferative diabetic retinopathy: characterization and translational implications. Prog Retin Eye Res. 2019; 72: 100756. [CrossRef] [PubMed]
Simó R, Hernández C. New insights into treating early and advanced stage diabetic retinopathy. Int J Mol Sci. 2022; 23: 8513. [CrossRef] [PubMed]
Iglicki M, González DP, Loewenstein A, Zur D. Next-generation anti-VEGF agents for diabetic macular oedema. Eye. 2022; 36: 273–277. [CrossRef] [PubMed]
Zur D, Iglicki M, Loewenstein A. The role of steroids in the management of diabetic macular edema. Ophthalmic Res. 2019; 62: 231–236. [CrossRef] [PubMed]
Imming P, Sinning C, Meyer A. Drugs, their targets and the nature and number of drug targets. Nat Rev Drug Discov. 2006; 5: 821–834. [CrossRef] [PubMed]
Zheng J, Haberland V, Baird D, et al. Phenome-wide Mendelian randomization mapping the influence of the plasma proteome on complex diseases. Nat Genet. 2020; 52: 1122–1131. [CrossRef] [PubMed]
Csősz É, Deák E, Kalló G, Csutak A, Tőzsér J. Diabetic retinopathy: proteomic approaches to help the differential diagnosis and to understand the underlying molecular mechanisms. J Proteomics. 2017; 150: 351–358. [CrossRef] [PubMed]
Youngblood H, Robinson R, Sharma A, Sharma S. Proteomic biomarkers of retinal inflammation in diabetic retinopathy. Int J Mol Sci. 2019; 20: 4755. [CrossRef] [PubMed]
Wang H, Feng L, Hu J, Xie C, Wang F. Differentiating vitreous proteomes in proliferative diabetic retinopathy using high-performance liquid chromatography coupled to tandem mass spectrometry. Exp Eye Res. 2013; 108: 110–119. [CrossRef] [PubMed]
Trajanoska K, Bhérer C, Taliun D, Zhou S, Richards JB, Mooser V. From target discovery to clinical drug development with human genetics. Nature. 2023; 620: 737–745. [CrossRef] [PubMed]
Lawlor DA, Harbord RM, Sterne JA, Timpson N, Davey Smith G. Mendelian randomization: using genes as instruments for making causal inferences in epidemiology. Stat Med. 2008; 27: 1133–1163. [CrossRef] [PubMed]
Evans DM, Davey Smith G. Mendelian randomization: new applications in the coming age of hypothesis-free causality. Ann Rev Genomics Hum Genet. 2015; 16: 327–350. [CrossRef]
Zheng D, Li N, Hou R, et al. Glucagon-like peptide-1 receptor agonists and diabetic retinopathy: nationwide cohort and Mendelian randomization studies. BMC Med. 2023; 21: 40. [CrossRef] [PubMed]
Sun BB, Maranville JC, Peters JE, et al. Genomic atlas of the human plasma proteome. Nature. 2018; 558: 73–79. [CrossRef] [PubMed]
Emilsson V, Ilkov M, Lamb JR, et al. Co-regulatory networks of human serum proteins link genetics to disease. Science. 2018; 361: 769–773. [CrossRef] [PubMed]
Suhre K, Arnold M, Bhagwat AM, et al. Connecting genetic risk to disease end points through the human blood plasma proteome. Nat Commun. 2017; 8: 14357. [CrossRef] [PubMed]
Folkersen L, Fauman E, Sabater-Lleal M, et al. Mapping of 79 loci for 83 plasma protein biomarkers in cardiovascular disease. PLoS Genetics. 2017; 13: e1006706. [CrossRef] [PubMed]
Yao C, Chen G, Song C, et al. Genome-wide mapping of plasma protein QTLs identifies putatively causal genes and pathways for cardiovascular disease. Nat Commun. 2018; 9: 3268. [CrossRef] [PubMed]
Kurki MI, Karjalainen J, Palta P, et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature. 2023; 613: 508–518. [CrossRef] [PubMed]
Teumer A. Common methods for performing mendelian randomization. Front Cardiovasc Med. 2018; 5: 51. [CrossRef] [PubMed]
Zheng J, Baird D, Borges MC, et al. Recent developments in mendelian randomization studies. Curr Epidemiol Rep. 2017; 4: 330–345. [CrossRef] [PubMed]
Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016; 40: 304–314. [CrossRef] [PubMed]
Bowden J, Del Greco MF, Minelli C, Davey Smith G, Sheehan NA, Thompson JR. Assessing the suitability of summary data for two-sample Mendelian randomization analyses using MR-Egger regression: the role of the I2 statistic. Int J Epidemiol. 2016; 45: 1961–1974. [CrossRef] [PubMed]
Verbanck M, Chen CY, Neale B, Do R. Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nat Genet. 2018; 50: 693–698. [CrossRef] [PubMed]
Hemani G, Tilling K, Davey Smith G. Orienting the causal relationship between imprecisely measured traits using GWAS summary data. PLoS Genet. 2017; 13: e1007081. [CrossRef] [PubMed]
Ferkingstad E, Sulem P, Atlason BA, et al. Large-scale integration of the plasma proteome with genetics and disease. Nat Genet. 2021; 53: 1712–1721. [CrossRef] [PubMed]
Skrivankova VW, Richmond RC, Woolf BAR, et al. Strengthening the reporting of observational studies in epidemiology using mendelian randomisation (STROBE-MR): explanation and elaboration. BMJ Clin Res. 2021; 375: n2233.
Giambartolomei C, Vukcevic D, Schadt EE, et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 2014; 10: e1004383. [CrossRef] [PubMed]
Yu G, Wang LG, Han Y, He QY. ClusterProfiler: an R package for comparing biological themes among gene clusters. Omics. 2012; 16: 284–287. [CrossRef] [PubMed]
Struyf S, Menten P, Lenaerts JP, et al. Diverging binding capacities of natural LD78beta isoforms of macrophage inflammatory protein-1alpha to the CC chemokine receptors 1, 3 and 5 affect their anti-HIV-1 activity and chemotactic potencies for neutrophils and eosinophils. Eur J Immunol. 2001; 31: 2170–2178. [CrossRef] [PubMed]
Forrester JV, Kuffova L, Delibegovic M. The role of inflammation in diabetic retinopathy. Front Immunol. 2020; 11: 583687. [CrossRef] [PubMed]
Thomsen SK, Raimondo A, Hastoy B, et al. Type 2 diabetes risk alleles in PAM impact insulin release from human pancreatic β-cells. Nat Genet. 2018; 50: 1122–1131. [CrossRef] [PubMed]
Fuchsberger C, Flannick J, Teslovich TM, et al. The genetic architecture of type 2 diabetes. Nature. 2016; 536: 41–47. [CrossRef] [PubMed]
Steinthorsdottir V, Thorleifsson G, Sulem P, et al. Identification of low-frequency and rare sequence variants associated with elevated or reduced risk of type 2 diabetes. Nat Genet. 2014; 46: 294–298. [CrossRef] [PubMed]
Wu H, He G, Song T, et al. Evaluation of GALNT16 polymorphisms to breast cancer risk in Chinese population. Mol Genet Genom Med. 2019; 7: e848. [CrossRef]
Marucci A, di Mauro L, Menzaghi C, et al. GALNT2 expression is reduced in patients with Type 2 diabetes: possible role of hyperglycemia. PloS One. 2013; 8: e70159. [CrossRef] [PubMed]
Marucci A, Cozzolino F, Dimatteo C, et al. Role of GALNT2 in the modulation of ENPP1 expression, and insulin signaling and action: GALNT2: a novel modulator of insulin signaling. Biochim Biophys Acta. 2013; 1833: 1388–1395. [CrossRef] [PubMed]
Antonucci A, Marucci A, Trischitta V, Di Paola R. Role of GALNT2 on insulin sensitivity, lipid metabolism and fat homeostasis. Int J Mol Sci. 2022; 23: 929. [CrossRef] [PubMed]
Li Z, Fischer M, Satkunarajah M, Zhou D, Withers SG, Rini JM. Structural basis of Notch O-glucosylation and O-xylosylation by mammalian protein-O-glucosyltransferase 1 (POGLUT1). Nat Commun. 2017; 8: 185. [CrossRef] [PubMed]
Miloudi K, Oubaha M, Ménard C, et al. NOTCH1 signaling induces pathological vascular permeability in diabetic retinopathy. Proc Natl Acad Sci U S A. 2019; 116: 4538–4547. [CrossRef] [PubMed]
Cheng WL, Yang Y, Zhang XJ, et al. Dickkopf-3 ablation attenuates the development of atherosclerosis in ApoE-deficient mice. J Am Heart Assoc. 2017; 6(2): e004690. [CrossRef] [PubMed]
Wang Z, Liu CH, Huang S, Chen J. Wnt signaling in vascular eye diseases. Prog Retin Eye Res. 2019; 70: 110–133. [CrossRef] [PubMed]
Ji B, Lim D, Kim J, Kim HC, Chung H. Increased levels of Dickkopf 3 in the aqueous humor of patients with diabetic macular edema. Invest Ophthalmol Vis Sci. 2016; 57: 2296–2304. [CrossRef] [PubMed]
Pottier C, Zhou X, Perkerson RB, 3rd, et al. Potential genetic modifiers of disease risk and age at onset in patients with frontotemporal lobar degeneration and GRN mutations: a genome-wide association study. Lancet Neurol. 2018; 17: 548–558. [CrossRef] [PubMed]
Politi C, Ciccacci C, D'Amato C, Novelli G, Borgiani P, Spallone V. Recent advances in exploring the genetic susceptibility to diabetic neuropathy. Diabetes Res Clin Pract. 2016; 120: 198–208. [CrossRef] [PubMed]
Harada T, Harada C, Mitamura Y, et al. Neurotrophic factor receptors in epiretinal membranes after human diabetic retinopathy. Diabetes Care. 2002; 25: 1060–1065. [CrossRef] [PubMed]
Matsuoka TA, Kaneto H, Kawashima S, et al. Preserving Mafa expression in diabetic islet β-cells improves glycemic control in vivo. J Biol Chem. 2015; 290: 7647–7657. [CrossRef] [PubMed]
Oniki K, Umemoto Y, Nagata R, et al. Glutathione S-transferase A1 polymorphism as a risk factor for smoking-related type 2 diabetes among Japanese. Toxicol Lett. 2008; 178: 143–145. [CrossRef] [PubMed]
Holmquist P, Liuba P. Urine α-glutathione S-transferase, systemic inflammation and arterial function in juvenile type 1 diabetes. J Diabetes Its Complications. 2012; 26: 199–204. [CrossRef]
Anitha A, Nakamura K, Thanseem I, et al. Downregulation of the expression of mitochondrial electron transport complex genes in autism brains. Brain Pathol. 2013; 23: 294–302. [CrossRef] [PubMed]
Sumara G, Formentini I, Collins S, et al. Regulation of PKD by the MAPK p38delta in insulin secretion and glucose homeostasis. Cell. 2009; 136: 235–248. [CrossRef] [PubMed]
Gu C, Lhamo T, Zou C, et al. Comprehensive analysis of angiogenesis-related genes and pathways in early diabetic retinopathy. BMC Med Genom. 2020; 13: 142. [CrossRef]
Yue T, Shi Y, Luo S, Weng J, Wu Y, Zheng X. The role of inflammation in immune system of diabetic retinopathy: molecular mechanisms, pathogenetic role and therapeutic implications. Front Immunol. 2022; 13: 1055087. [CrossRef] [PubMed]
Wang X, Antony V, Wang Y, Wu G, Liang G. Pattern recognition receptor-mediated inflammation in diabetic vascular complications. Med Res Rev. 2020; 40: 2466–2484. [CrossRef] [PubMed]
Abu El-Asrar AM, Mohammad G, Nawaz MI, et al. The chemokine platelet factor-4 variant (PF-4var)/CXCL4L1 inhibits diabetes-induced blood-retinal barrier breakdown. Invest Ophthalmol Vis Sci. 2015; 56: 1956–1964. [CrossRef] [PubMed]
Iglicki M, Zur D, Busch C, Okada M, Loewenstein A. Progression of diabetic retinopathy severity after treatment with dexamethasone implant: a 24-month cohort study the “DR-Pro-DEX Study.” Acta Diabetol. 2018; 55: 541–547. [CrossRef] [PubMed]
Mello Filho P, Andrade G, Maia A, et al. Effectiveness and safety of intravitreal dexamethasone implant (Ozurdex) in patients with diabetic macular edema: a real-world experience. Ophthalmologica. 2019; 241: 9–16. [CrossRef] [PubMed]
Zur D, Iglicki M, Sala-Puigdollers A, et al. Disorganization of retinal inner layers as a biomarker in patients with diabetic macular oedema treated with dexamethasone implant. Acta Ophthalmol. 2020; 98: e217–e223. [CrossRef] [PubMed]
Lee MW, Park HJ, Shin YI, Lee WH, Lim HB, Kim JY. Comparison of choroidal thickness measurements using swept source and spectral domain optical coherence tomography in pachychoroid diseases. PloS One. 2020; 15: e0229134. [CrossRef] [PubMed]
Liu K, Zou J, Yuan R, et al. Exploring the effect of the gut microbiome on the risk of age-related macular degeneration from the perspective of causality. Invest Ophthalmol Vis Sci. 2023; 64: 22. [CrossRef] [PubMed]
Wang T, Chen J, Wang J, Zhang Y, Mao W, Yi Q. 91 Circulating inflammatory proteins and the risk of age-related macular degeneration: a bidirectional Mendelian randomization study. Int Immunopharmacol. 2024; 139: 112678. [CrossRef] [PubMed]
Figure 1.
 
The overview of the two-sample MR study design. Assumption 1: The genetic variants selected as IVs must be associated with plasma proteins; Assumption 2: Genetic variants used as IVs for plasma proteins must not be associated with known confounding factors. Assumption 3: Genetic variants serving as IVs should influence the risk of DR exclusively through plasma proteins.
Figure 1.
 
The overview of the two-sample MR study design. Assumption 1: The genetic variants selected as IVs must be associated with plasma proteins; Assumption 2: Genetic variants used as IVs for plasma proteins must not be associated with known confounding factors. Assumption 3: Genetic variants serving as IVs should influence the risk of DR exclusively through plasma proteins.
Figure 2.
 
Bioinformatics analysis results. (A) Volcano plot. (B) GO enrichment analysis for risk proteins. (C) GO enrichment analysis for protective proteins. (D) Protein-protein interaction. (1) The points above the black dashed horizontal line and log (OR) > 0 represent that the plasma proteins were positively associated with increased DR risk, on the contrary, the points above the black dashed horizontal line and log (OR) < 0 represent that the plasma proteins were protective against DR risk (A). (2) Count represents the number of plasma proteins enriched in this pathway (B, C). (3) The black solid line represents the interaction relationship between plasma proteins (D).
Figure 2.
 
Bioinformatics analysis results. (A) Volcano plot. (B) GO enrichment analysis for risk proteins. (C) GO enrichment analysis for protective proteins. (D) Protein-protein interaction. (1) The points above the black dashed horizontal line and log (OR) > 0 represent that the plasma proteins were positively associated with increased DR risk, on the contrary, the points above the black dashed horizontal line and log (OR) < 0 represent that the plasma proteins were protective against DR risk (A). (2) Count represents the number of plasma proteins enriched in this pathway (B, C). (3) The black solid line represents the interaction relationship between plasma proteins (D).
Figure 3.
 
The causal effect of DR on plasma proteins risk based on the IVW method. (1) IVW_FE: the fixed-effect IVW model was used to assess the causal effect of plasma proteins on DR. IVW_MRE: the random-effects IVW model was used to assess the causal effect of plasma proteins on DR.
Figure 3.
 
The causal effect of DR on plasma proteins risk based on the IVW method. (1) IVW_FE: the fixed-effect IVW model was used to assess the causal effect of plasma proteins on DR. IVW_MRE: the random-effects IVW model was used to assess the causal effect of plasma proteins on DR.
Figure 4.
 
Result of molecular docking. (A–X) Action mode of small-molecule drugs with small-molecule drugs. (1) The term before the “_” refers to plasma proteins, whereas the term after the “_” refers to the docking small molecule drugs in the Figure legends (A–X). (2) Molecular docking results between the small molecule drugs (ligands, Cyan molecular structure) and receptor (plasma proteins) in the enlarged figures. The numbers represent the binding energy (for example, “2.7” in A), and “GLY” et al. was the abbreviation for amino acids (A).
Figure 4.
 
Result of molecular docking. (A–X) Action mode of small-molecule drugs with small-molecule drugs. (1) The term before the “_” refers to plasma proteins, whereas the term after the “_” refers to the docking small molecule drugs in the Figure legends (A–X). (2) Molecular docking results between the small molecule drugs (ligands, Cyan molecular structure) and receptor (plasma proteins) in the enlarged figures. The numbers represent the binding energy (for example, “2.7” in A), and “GLY” et al. was the abbreviation for amino acids (A).
Table 1.
 
The Causal Effect of Plasma Proteins on DR Risk in Zheng's Study Cohort
Table 1.
 
The Causal Effect of Plasma Proteins on DR Risk in Zheng's Study Cohort
Table 2.
 
The Causal Effect of Plasma Proteins on DR Risk in deCODE Cohort
Table 2.
 
The Causal Effect of Plasma Proteins on DR Risk in deCODE Cohort
Table 3.
 
Colocalization Analysis of Plasma Proteins With DR
Table 3.
 
Colocalization Analysis of Plasma Proteins With DR
Table 4.
 
The Binding Energy Between Drug Targets and the Predicted Drugs
Table 4.
 
The Binding Energy Between Drug Targets and the Predicted Drugs
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×