Abstract Choroidal neovascularization (CNV) is a severe pathological outcome of age-related macular degeneration (AMD) which could lead to blindness. The present study was aimed at exploring the mechanisms of CNV, hoping to provide new clues in finding the treatment target of CNV. ITRAQ approach was applied to analyze the proteins in choroidal neovascularization of CNV and normal rats. KEGG and GO annotation were used to analyze the differently-expressed proteins. Western Blotting was used as the method to identify the differently-expressed proteins. TAB1, one of the differently-expressed proteins, was chosen as the study objective in the following research. AAV-TAB1 over-expression vehicle was constructed to investigate the function of TAB1 in CNV. IL-6 and IL-18 were tested with the use of ELISA. Western Blotting was used to test the expression of NF-kB pathway molecules. In terms of the results, expressions of 49 kinds of proteins were up-regulated in CNV rats while expressions of 241 kinds of proteins were down-regulated among the 4380 identified proteins. Overexpression of TAB1 significantly reduced areas of CNV lesions in vivo. Cell proliferation significantly increased in the TAB1 over-expressed group in RPE. IL-6 expression significantly increased in the TAB1-overexpression group while IL-18 expression significantly reduced. Western blotting results showed NF-kB pathway molecules were involved in it. The present study suggests TAB1 may play a protective role in CNV progression and the relative pathway owns the potential to be the treating target of CNV. Supplementary Information The online version contains supplementary material available at 10.1038/s41598-025-15134-1. Keywords: Proteomics, ITRAQ, CNV, TAB1, Inflammation Subject terms: Cell growth, Mechanisms of disease Introduction Choroidal neovascularization (CNV) is one of the main severe pathological outcomes of age-related macular degeneration (AMD), and is a main cause leading to blindness of the elderly over 50 years in European countries^[34]1,[35]2. Choroidal neovascularization originates from the micro-vessels of choroid, breaks through Bruch’s membrane, and stretches into the space under RPE or the space under the neuroepithelium layer. It leads to vision loss eventually. Current golden standard therapy of anti-VEGF has gained great success^[36]3,[37]4, however, some drawbacks remain such as a subset of patients having a low response to the therapy, which alludes to finding new mechanisms of CNV and searching for new treating targets of it. Protein, as we all know, is the function executor of life. Proteomics is defined as the genome expression of one kind of cell, tissue, or whole-life entity. As the onset and progression of one disease involve various kinds of protein, the application of proteomics in the exploration of disease progression or diagnosis and treatment is getting hot. The isobaric tag for relative and absolute quantification (ITRAQ) technology was developed by ABI company in 2004. It was widely applied in exploring disease mechanisms and searching for new disease markers. The present research was conducted in an attempt to identify differently-expressed proteins, aiming to find potential mechanisms of CNV and new treatment targets. TAK1(TGF-βactivated kinase-1) is a member of the MAPK family and also plays a role in TGF-βsignaling pathway. TAK1 lies in the point of the intersection of two pathways. The activation of TAK1 relies on the binding of TAB1 to it. The interaction of TAK1 and TAB1 functions in growth and development, cell survival, immune response, and tumor growth^[38]3. Results The number of total protein species identified by iTRAQ labeling technology and the number of differently-expressed protein types By analyzing and comparing the labeled peptides, a total of 4380 types of protein were identified in the samples of the CNV group and the control group (Table [39]1). Differently-expressed proteins are defined as proteins whose up-regulation ratio is greater than 1.2 times or whose down-regulation ratio is less than 0.83 times, and P < 0.05. A total of 290 types of differently-expressed proteins were identified in this study, including 49 kinds of up-regulated proteins and 241 kinds of down-regulated proteins (Table [40]2). The statistical results of protein quantification were presented in the form of a Volcano Plot (Fig. [41]1). Table 1. Statistic result of protein identification. Database Total spectra Spectra (PSM) Peptides Unique peptides Protein groups Rat 376,515 51,216 22,228 20,348 4380 [42]Open in a new tab Table 2. Statistic results of protein quantification. Comparisons (CNV/control) Up- Down- All Number of protein types 49 241 290 [43]Open in a new tab Fig. 1. [44]Fig. 1 [45]Open in a new tab The volcano plot. The volcano plot was made with the fold change of protein expression between the CNV group and the control group and the P value obtained by the T-test. Volcano plot was used to show the significant difference between the two groups. The horizontal coordinate indicated the fold change (logarithmic transformation with base 2), and the vertical coordinate indicated the significant P value of the fold change (logarithmic transformation with base 10). The red dots in the picture represented the significantly differently-expressed proteins (the fold change is greater than 1.2 times and the P value is less than 0.05), and the black dots represented the proteins with no significant change. Gene ontology analysis of differently-expressed proteins We first analyzed gene ontology (GO) terms for biological process, cellular components, and molecular function with the use of the software Blat2Go, then performed GO functional enrichment analysis of the differentially-expressed proteins using Fisher’s exact test. The analysis results showed that the differently-expressed proteins took part in the biological process of mitotic nuclear division, immune response-regulating cell surface receptor signaling pathway, negative regulation of transcription, DNA-templated, negative regulation of cell death and cytokine production (Fig. [46]2A, D, E). Besides, they were involved in cellular components like extracellular matrix component, COPII vesicle coat, nuclear exosome (RNase complex), endopeptidase Clp complex, and anchored component of membrane (Fig. [47]2B, D, E). Additionally, the differently-expressed proteins played a role in molecular functions like chromatin binding, transmembrane receptor activity, helicase activity, kinase regulator activity, and enzyme inhibitor activity (Fig. [48]2C–E); Fig. 2. [49]Fig. 2 [50]Open in a new tab Gene ontology of differently-expressed proteins in control and CNV groups. Analysis results of GO terms for biological process (A), cellular component (B), and molecular function (C) of the differently-expressed proteins in CNV and control group based on the Blast2Go software. Analysis of biological processes, cellular components, and molecular functions were fused as one graph (D). The significance enrichment analysis of GO functional annotations is to evaluate the significance level of protein enrichment of a certain GO functional term through Fisher’s Exact Test (E). The horizontal axis in the picture showed the enriched GO function classification, which was divided into three categories: Biological Process (BP), Molecular Function (MF), and Cellular Component (CC). The vertical ordinate represented the number of different proteins in each functional classification; The color of the bar chart represented the significance of the enriched GO functional classification, that is, the P value calculated based on Fisher’s Exact Test. The color of the bar changed from orange to red, representing the gradient of the P value. The closer the color is to red, the smaller the P value is. The label above the bar showed the enrichment factor (Rich Factor ≤ 1), which represented the proportion of the number of differentially-expressed proteins annotated to the GO functional class in all identified proteins annotated to that GO functional class. Enrichment analysis of KEGG pathways of the differentially expressed proteins Pathway analysis is a direct and necessary way to systematically understand the biological processes of cells, the pathogenesis of diseases, and the mechanisms of drug action. The present results of the KEGG pathway annotation of significantly differently-expressed proteins revealed a series of significant changes in the proteins from the cell surface to the nucleus, biological events, and acting factors involved in these processes. Besides, the top 20 KEGG pathways were shown (Fig. [51]3A), including the PI3K-AKT signaling pathway, human papillomavirus infection, herpes simplex infection, and so forth. Furthermore, KEGG pathway enrichment analysis was performed on differently-expressed proteins by Fisher’s exact test. The results showed that pathways of human papillomavirus infection, PI3K-Akt signaling, herpes simplex infection, tuberculosis, and significant changes that occurred in important pathways in cancer were enriched (Fig. [52]3B). Fig. 3. [53]Fig. 3 [54]Open in a new tab Top 20 KEGG pathways and the enriched KEGG pathways (A) Top 20 KEGG pathways including PI3K-AKT signaling pathway, human papillomavirus infection, and herpes simplex infection were shown in this picture. The horizontal axis showed the signaling pathways and the vertical axis showed the number of the protein types which were enriched in the corresponding signaling pathway. (B) The horizontal coordinate represents the number of differentially-expressed protein types contained in each KEGG pathway and the vertical axis in this picture represents significantly-enriched KEGG pathways. The color gradient of the bar graph showed the significance of the enriched KEGG pathway, namely the P value calculated based on Fisher’s exact test. Specifically, the color changed from orange to red. The closer the color was to red, the higher the significance level of the KEGG pathway enrichment was; The label beside the bar showed the enrichment factor (Rich Factor ≤ 1), which represented the proportion of the number of differentially expressed proteins involved in a KEGG pathway to the number of proteins involved in that pathway among all identified proteins. Validation of candidate differentially-expressed proteins with the use of western blotting A total of eight types of protein, namely MCM7, YES1, SEPT9, p27 kip1, p57 kip2, TRIM32, RPIA, and TAB1 were chosen for validation and the total protein in the RPE-choroidal-sclera was detected with the use of western blotting method. The results showed the proteins of YES1, SEPT9, p27 kip1, and p57 kip2 expressions were not significantly different between the two groups. The expressions of TAB1, MCM7, TRIM32, and RPIA in the CNV model group were significantly lower than those in the normal group (Fig. [55]4). Fig. 4. Fig. 4 [56]Open in a new tab Verification of the eight types of candidate protein by western blotting (n = 3). (A, C, E) The expressions of the candidate differently-expressed proteins, namely MCM7, YES1, SEPT 9, p27 Kip1, p57 Kip2, TRIM 32, RPIA and TAB1. (B, D, F) As the statistical result was shown, expressions of TRIM 32, RPIA, and TAB1 were significantly down-regulated while expressions of MCM7, YES1, p27 kip, SEPT 9, and p57 kip 2 did not significantly differ from the control. (Note, ns = no significance; * = P < 0.05; ** = P < 0.01; *** = P < 0.001). TAB1 over-expression in vivo and TAB1 expression in RPE-choroidal-sclera complex At day 0 (immediately after transfection), day 7, day 14, day 30, and day 60 after transfection with AAV- TAB1 over-expression vector, the total protein of the RPE-choroid-sclera complex was extracted. Expressions of TAB1 at the five points in time were detected (Fig. [57]5). The expressions of TAB1 at day 14, day 30, and day 60 were significantly higher than those at day 0 after transfection (P < 0.001, P < 0.01, P < 0.01), while there was no significant difference between day 7 and day 0 after transfection (P > 0.05). Fig. 5. [58]Fig. 5 [59]Open in a new tab Western Blotting results tested on TAB1 expression after AAV-TAB1 over-expression (A) Fundus photograph of the BN rats hinted that the subretinal injection of AAV-TAB1 vector was successfully performed as the arrow showed the swelling of the retinal neuroepithelium layer. TAB1 in RPE-choroidal-sclera was tested by western Blotting. (B) A representative picture was shown (n = 3). (C) The statistical analysis result showed that TAB1 expression significantly increased at day 14, day 30, and day 60 compared with day 0 (P < 0.001, P < 0.01, P < 0.01), while there was no significant difference between day 7 and day 0 (P > 0.05). (**, P < 0.01; ***, P < 0.01; ns, P > 0.05). Analysis of choroidal neovascularization area Cardiac perfusion of FITC-dextran solution was performed into the hearts of BN rats in the CNV group, CNV + AAV group, and CNV + AAV-TAB1 group at 7, 14, 30, and 60 days after the CNV model was constructed and the RPE-choroid-sclera complexes were taken out for flat mounts. CNV lesions were outlined and the areas of CNV lesions in rats were measured (Fig. [60]6A). The results showed that the lesion areas of the CNV + AAV-TAB1 group significantly decreased compared with CNV group at day 14, day 30 and day 60 after the performance of laser coagulation (P < 0.01, P < 0.001, P < 0.001) while there was no significant change of lesion area in the CNV + AAV group and CNV group (P > 0.05) (Fig. [61]6B). The concentrations of IL-6 and IL-18 in the RPE-choroid-sclera complex homogenate of the rats in the CNV group, CNV + AAV group, and CNV + AAV-TAB1 group were detected by ELISA. The ELISA results showed the expression of IL-6 decreased in the CNV + AAV-TAB1 group at day 14, day 30, and day 60 (Fig. [62]6C). In terms of IL-18, the expression of it was significantly up-regulated in CNV + AAV-TAB1 group compared with the other two groups at day 14, day 30 and day 60 (Fig. [63]6D). Fig. 6. [64]Fig. 6 [65]Open in a new tab Flat mounts of RPE-choroidal-sclera complex and ELISA test of IL-6 and IL-18 (n = 3). (A) The rats in the three groups were sacrificed and the eyeballs were made for RPE-choroid-sclera flat mounts. CNV lesions were sketched and evaluated. The representative CNV lesion pictures are shown in this figure. (B) The statistic results showed that the CNV lesions were significantly smaller in the CNV + AAV-TAB1 group than in the CNV group at day 14, day 30, and day 60 (P < 0.01, P < 0.001, P < 0.001), while there was no statistical difference between CNV and CNV + AAV group at any point in time (note, ns = no significance; *, P < 0.05; **, P < 0.01; ***, P < 0.001). (C, D) The statistical analysis results showed the expression variation of IL-6 and IL-18 in each group. (C) IL-6 expression in the RPE-choroid-sclera complex of rats in the CNV + AAV-TAB1 group significantly decreased compared with that in the CNV control group at day 14, day 30, and day 60. (P < 0.01, P < 0.01, P < 0.01) while there was no significant difference in IL-6 expression between the CNV + AAV group and CNV group at any point in time (P > 0.05). (D) The expression of IL-18 in the RPE-choroid-sclera complex of rats in the CNV + AAV-TAB1 group was significantly up-regulated compared with that in the CNV control group at day 14, day 30, and day 60 (P < 0.01, P < 0.001, P < 0.01). There was no significant difference in IL-18 expressions between the CNV + AAV group and the CNV group at any time point (P > 0.05). Detection of the transfection efficiency of Adv-TAB1 over-expression vector in rat RPE cells by western blotting The transfected cells were placed under a fluorescence microscope to observe the GFP expression after 48 h of transfection, to judge the transfection efficiency of adenovirus vector. The transfection efficiency of adenovirus was measured as 80–90% (Fig. [66]7A). In addition, the total protein of RPE cells in the six-well plate was extracted and the expression of TAB1 was detected by WB method. It was found that the expression of TAB1 in the TAB1-Adv group was significantly higher than that in the control group and the TAB1 expression didn’t significantly change between the Control and Adv group (Fig. [67]7B, C). Fig. 7. [68]Fig. 7 [69]Open in a new tab Transfection efficiency of the adenovirus in the rat primary RPE cells and the expression of TAB1. (A) As the picture showed, the transfection efficiency was around 80–90% (the number of GFP-expressing cells divided by the number of the whole cells in the same visual field). (B, C) The western blotting results showed that the TAB1 expression was significantly higher than that in the control group (P < 0.001) while the TAB1 expressions in control and Adv were not significantly different (n = 3). (P > 0.05) (Note, ns = no significance; *, P < 0.05; **, P < 0.01; ***, P < 0.001). Proliferation of the primary RPE cells, IL-6 and IL-18 concentration in the cell culture supernatant, and the P52 and Rel B expression after TAB-1 overexpression in an anaerobic environment Cell proliferation was measured by CCK-8 assay at 12 h, 24 h, 48 h, and 72 h after treatment of CoCl[2] in both the Adv group and the Adv-TAB1 group. The results of the CCK-8 experiment showed that the cell proliferation of the Adv-TAB1 group was higher than that of the Adv group after CoCl[2] treatment for 24 h, 48 h, and 72 h (P < 0.05, P < 0.01, P < 0.01), while there was no significant difference in cell proliferation between the two groups at 12 h after the treatment (Fig. [70]8A). ELISA data showed that TAB1 over-expression significantly down-regulated IL-6 secretion and up-regulated IL-18 secretion (Fig. [71]8B, C). Western Blotting was used to test the expression of molecules in NF-κB, namely P52 and Rel B. Representative pictures were shown (Fig. [72]8D). Statistical data showed that TAB1 over-expression enhanced the expression of P52 at 24 h, 48 h, and 72 h and also promoted the expression of Rel B at 48 h and 72 h (Fig. [73]8E, F). Fig. 8. [74]Fig. 8 [75]Open in a new tab Cell proliferation, IL-6 & IL-18 secretion, and NF-κB signaling molecules expression. (A) Cell proliferations were tested by CCK-8 assay 12, 24, 48, and 72 h after adenovirus transfection. As is shown in the picture, cell proliferation was up-regulated in the TAB1-Adv group compared with the Adv group at hour 24, hour 48, and hour 72 (P < 0.05, P < 0.01, P < 0.01). (B) IL-6 expression was significantly reduced at hour 24, hour 48, and hour 72 after CoCl[2] treatment, while it didn’t change at hour 12. (Note, ns = no significance; *, P < 0.05; **, P < 0.01; ***, P < 0.001). (C) IL-18 expression was significantly up-regulated in the Adv-TAB1 group at hour 24, hour 48, and hour 72 after CoCl[2] treatment, while it didn’t significantly change at hour 12 after CoCl[2] treatment (P > 0.05). (D) Representative pictures are shown in this figure (n = 3). (E) P52 expression didn’t differ significantly at hour 12 between Adv and Adv-TAB1 groups. P52 expression significantly increased in Adv-TAB1 compared with Adv group at hours 24, 48 and 72 (P < 0.05, P < 0.05, P < 0.01). (F) Rel B expression didn’t differ significantly at hour 12 and hour 24 between the Adv and Adv-TAB1 group (P > 0.05), and it significantly increased in Adv-TAB1 compared with the Adv group at hour 24, 48 and 72 (P < 0.05, P < 0.01). Discussion About 5%-10% of AMD patients will progress to CNV, which causes serious damage to central vision to the patients, making a severe physical and mental burden to the families and a heavy economic burden to society^[76]5,[77]6. Anti-VEGF therapy has become a mainstream treatment of CNV and has gained great success^[78]7–[79]9. However, some patients don’t respond to the drug very well, which hinted us some other factors besides VEGF could cause CNV^[80]10, and it’s urgent to search for new treatment targets for CNV. In the present proteomics study, we applied the iTRAQ method to identify the proteins that the RPE-Choroid-Sclera complex contained and also analyzed proteins that were differently expressed between normal BN rats and CNV BN rats. For some research, it’s important to figure out a specific or the targeted protein’s function, the cellular localization, and the biological process in which the protein takes part. In a high throughput omics research and a macroscopical view, the primary task is to find out what proteins or biological processes were significantly impacted by the biological treatment^[81]11. Therefore, we need to generalize and analyze the protein functions more systematically. Thus, GO annotation and pathway enrichment emerged to analyze the differently-expressed proteins. In the present study, GO analyzation and KEGG pathways enrichment results showed that, in the RPE-choroidal-sclera complex, differently-expressed proteins not only took part in the biological process or cellular functions like chromosome binding, DNA replication, and cell proliferation but also participated in some biological processes like cytokine secretion and immunocompetence such as immune response-regulating cell surface receptor as was shown in our study results. As is known to us, the alteration of pro- or anti-angiogenesis function is associated with CNV development and abnormal cell proliferation of endothelial was involved in the progress of CNV. VEGF was one of the important factors that promote CNV progression among the pro-angiogenesis factors, which was the theoretical foundation of the antibody-based therapy targeting VEGF. Despite the great success the anti-VEGF treatment has gained, there remains the situation that some patients don’t respond positively to this therapy, which implies other factors besides VEGF have probably participated in CNV development^[82]12,[83]13. Some recent research showed an abundance of other factors, such as immune factors, facilitate CNV development^[84]14,[85]15, which was consistent with our present proteomics results that immune-related processes were involved in CNV development. For instance, at the early stage of CNV development, complement factors deposit at the place between Bruch’s membrane and RPE, format a local inflammation situation, which causes direct damage to the retinal photoreceptors and RPEs. On the other hand, innate system cells, such as microglia and macrophages promote the CNV development by triggering VEGF secretion by other cells like RPEs^[86]2. These findings are consistent with our proteomics results, which imply that immune-related processes were accompanied by cell proliferation in CNV progression. In consideration of the change fold of differentially-expressed proteins and their function, eight types of differently-expressed proteins were chosen for the following validation assay, namely MCM7, YES1, SEPT9, p27 kip1, p57 kip2, TRIM 32, RPIA and TAB1, their functions are generally on cell proliferation and immunological function, which are similar to the mechanisms of CNV development. The validation assay result showed that MCM7, TRIM32, RPIA, and TAB1 were differently expressed in RPE-Choroidal-Sclera complexes between Control and CNV groups. TAB1 was set as our study object in the following research since it has not been studied in the CNV development yet and the past research reported TAB1 was a proliferation- and immune-related factor, which is probably involved in CNV since CNV is an over-proliferation and immune-related disease. TGF-β activated kinase-1(TAK1), is a member of the Mitogen-activated Protein Kinase (MAPK) family, and also plays a role in TGF-β associated pathways, participating in cell proliferation, inflammation, and angiogenesis^[87]16,[88]17. Protein TABs, which contain three members, termed TAB1, TAB2, and TAB3, were adapter proteins of TAK1^[89]18. The activation of TAK1 depends on the direct binding of TAB1. TAB1 regulates body growth, cell survival, immune response, and metabolism by triggering MAPK and TGF-β signaling pathways via binding and activating TAK1^[90]19,[91]20. Some research showed that the binding of TAB1 to TAK1 promoted tumor development by facilitating cell survival^[92]21. Meanwhile, some other research showed the binding of TAB1 to TAK1 promotes cell death. In the present research, TAB1 over-expression was performed to explore its function in CNV development in vivo. RPE-choroidal-sclera complex flat mounts showed TAB1 over-expression significantly reduced CNV lesions compared with the control group at day 7, day 14, day 30, and day 60. In addition, TAB1 over-expression significantly reduced IL-6 secretion and increased IL-18 secretion of the RPE-choroidal-sclera complex. The results hinted to us that TAB1 may inhibit CNV development. It is speculated that TAB1 may inhibit CNV progression in a similar way with cancers as angiogenesis shares lots of similar mechanisms with cancers such as over-proliferation of cells, cell migration, and inflammation. In the last research, TAB1 over-expression was demonstrated to inhibit breast cancer cell migration and invasion through TAB1/TAK1/p38 MAPK signaling pathway^[93]22. The TAB1 expression level was positively associated with patients’ prognosis. When TAB1 was knocked down, cell migration and invasion were significantly enhanced. Some other research showed TAB1 was associated with non-small cell lung cancer^[94]23. The evidence above showed the relationship between TAB1 and cancers may be similar to it between TAB1 and CNV. Inflammation was demonstrated in the pathogenesis of CNV progression. Inflammatory factors accompanied by other cytokines regulated CNV onset and progression by impacting vascular formation, vascular leakage, and retina edema. At the late stage of AMD, drusen bodies deposit underneath the retina, release various cytokines and chemokines create a chronic inflammation circumstance inducing angiogenesis, termed choroidal neovascularization, the severe pathological outcome of many diseases including AMD^[95]24,[96]25. It has been demonstrated by a large amount of evidence that IL-6 was a pro-inflammation cytokine that activated and prompted CNV progression. In a recent study, IL-6 was significantly elevated in laser-induced mice compared with normal mice^[97]26. The researchers constructed an IL-6 knockout mice to explore the role of IL-6 in CNV and found that IL-6 deficiency decreased laser-induced CNV area and exogenous IL-6 and increased choroidal sprouting angiogenesis. Lai et al. found that administration of triptolide down-regulated pro-inflammation cytokines including IL-6, along with reduced CNV lesions^[98]26. The study also demonstrated IL-6 expression ties with CNV progression. As a complex biological process, immunoreaction may have complicated functions, including pathogenic and protective effects, to CNV progression. In the past decades, IL-18, a member of the IL-1 family, was demonstrated to play a protective role in CNV onset and progression. Doyle and colleagues administered mature IL-18 to nonhuman primates and found that intravitreally injection of IL-18 significantly reduced CNV lesions in cynomolgus monkeys. It may be applied as an adjuvant immunotherapy-based treatment for CNV in the future^[99]27. In our study, TAB1 over-expression significantly reduced IL-6 expression and upregulated IL-18 expression in RPE-choroidal complex and hinted to us that TAB1 may play an anti-inflammation role in CNV progression and may present a potential therapeutic target to CNV. In the present study, the in vivo study verified the protective role of TAB1 in CNV as it reduced IL-6 expression, upregulated IL-18 expression, and lessened CNV lesions in rats. RPE cells are recognized as the initial site of drusen accumulation during AMD pathogenesis. The excessive build-up of drusen and associated inflammatory microenvironmental damage compromise the integrity of the RPE barrier, thereby facilitating CNV and subsequent retinal damage. Inflammatory mediators secreted by RPE cells contribute significantly to the progression of CNV^[100]2. Second, in our in vivo studies, we utilized the AAV2 subtype, which has been demonstrated to exhibit a high tropism for RPE cells in vivo. Thus, RPE cells were selected as the targeted cells of our in vitro investigations. For the further exploration of the effects and mechanisms of TAB1 in RPE cells in anoxic environment, we constructed adenovirus-mediated TAB1 gene over-expression vehicle to rat RPE cells in vitro to investigate the impact of TAB1 in cell proliferation and inflammation and its related mechanisms. CCK-8 assay was performed and the results showed that TAB1 over-expression significantly enhanced RPE proliferation, reduced IL-6 production, and increased IL-18 secretion in an anoxic environment. In past research, TAB1 was demonstrated to be related to cytokines release in a recent genome-wide association transethnic meta-analyses research. TAB1 was identified as one of 11 genes that were significantly associated with von Willebrand factor (VWF) levels^[101]28, in this study, TAB1-silence increased VWF release in the in vitro functional assay. TAB1, as an intracellular mediator, regulated several intracellular signaling pathways including TGF-β and IL-1, which hinted that TAB1 may function in a similar way to regulate IL-6 and IL-18 secretion in RPE cells in anoxic environment. Recent studies showed that TAB1 not only regulates cytokines secretion but also regulates cell proliferation^[102]28–[103]35. TAB1, as a member of TLR3–MyD88–IRAK1–TRAF6–TAK1 axis, was identified to mediate cell proliferation in breast cancer^[104]36. Besides, it was demonstrated that micro-RNAs participate in the regulation of TAB1. In colorectal cancers, TAB1 was the target gene of miR-873 and was involved in regulating colorectal cancer cells’ proliferation^[105]34. In addition, TAB1 was confirmed to be over-expressing in non-small lung cancers, identified as a direct target gene of miR-889, and regulated the proliferation and invasiveness of non-small lung cancer cells^[106]33. In general, TAB1 was demonstrated to regulate cytokine production and cell proliferation, which followed the findings in our research. TAB1 has been widely demonstrated involved in the NF-κB signaling pathway via binding to TAK1. TAB1 was demonstrated expressed in many kinds of cells. For example, in esophageal cancer cells, activation of the TAB1/TAK1/IKKβ/NF-κB signaling axis induced proliferation, invasion, and migration of the cells^[107]34. In breast cancer cells, stabilization of the XIAP-TAB1-TAK1 complex induced NF-kB activation and thus led to cell death^[108]37. Constitutive EGFR signaling promotes invasion via activation of a TAB1-TAK1-NF-κB-EMP1 pathway, resulting in large tumors and worse survival in glioblastoma orthotopic models^[109]29. This evidence presented the regulation of TAK1-TAB1 complex in NF-κB signaling in different types of cells in various conditions^[110]38–[111]42. So far, the role of TAB1 in CNV has not been studied yet. In the present study, over-expression of TAB1 in rat RPE cells in the anoxic condition increased IL-18 secretion, down-regulated IL-6 secretion, and up-regulated the expressions of Rel B and P52 expressions. TAK1 is the first important part of the Nf-kB cascade signaling in the cell, and the Nf-kB signaling pathway mainly regulates biological functions related to immunity and inflammation^[112]19,[113]20. Overexpression of TAB1 in RPE cells may induce more TAK1 activation, thereby activating downstream relevant signaling pathway molecules and inducing a series of signaling cascades. Therefore, we examined the expression of P52 and Rel B in the Nf-kB signaling pathway to explore the mechanism of inflammation protection and proliferation protection of TAB1 in BN rat RPE cells under a hypoxia environment. The WB results of Nf-KB showed that the expression levels of P52 and Rel B were increased after overexpression of TAB1, which may promote the secretion of IL-18 and inhibit the secretion of IL-6 by triggering the Nf-kB signaling pathway molecules. The results hinted that TAB1 may function as a protective regulator in CNV conditions by impacting the NF-κB signaling pathway. However, we have to admit that further experiments were needed to explain the mechanism of TAB1 overexpression in RPE cells for in-depth exploration. In the present study, the number of sample replicates was set to three, a decision informed by multiple considerations. First, given the highly controllable experimental conditions—wherein cell culture and operations strictly adhere to standardized protocols and the anticipated variation is minimal—three replicate samples are sufficient to reflect the experimental effect accurately. Second, in alignment with conventions within the field and the experimental designs of comparable studies, three replicate samples are widely regarded as acceptable for preliminary investigations. Although the sample size is relatively small, the findings of this study nonetheless offer valuable insights that inform subsequent research. Future studies will further validate these findings by expanding the sample size to ensure the robustness, reliability, and generalizability of the results. In general, the present study was the first proteomics research to analyze rat CNV RPE-choroid-sclera complex. TAB1 was found to play a role in CNV progression and may have a protective role in CNV by impacting the IL-6 and IL-18 production and cell proliferation via NF-κB signaling, which provided a potential target for CNV treatment. Materials and methods Antibodies and reagents The primary antibodies used were as followed: Anti-MCM7(abcam, ab52489), anti-YES1(abcam, ab109744), anti-SEPT9(abnova, H00010801-A01), anti-P27 kip1 (CST, #2552), anti-P57 kip2 (Santa cruz, sc-56341), anti-TRIM 32 (Abcam, ab96612), anti-RPIA (Santa Cruz, SC515328), Anti-TAB1 (Santa Cruz, SC166138), HRP-tagged goat anti-rabbit antibody (Bioss, bs-80295G-HRP); HRP tagged goat anti-mouse antibody (bs-40296G-HRP); anti-NF-κBp52 (Abcam,ab129097), anti-RelB(Abcam, ab309084). The secondary antibodies used were as follows: Anti-β-actin(Bioss, bs-0061R), and anti-GAPDH(Bioss, bs-41373R). Animals Brown Norway male rats (Animal Experiment Center of Chongqing Medical University, Chongqing, China), weighing 150–180 g and 6–8 weeks old, were used for this study. This study was approved by the Ethics Committee of the Second Affiliated Hospital of Chongqing Medical University (Chongqing, China; approval no. ID: 2018-222). All experimental procedures were conducted in conformity with the institutional guidelines issued by the IACUC of NUCM and the ARRIVE guidelines. CNV model construction Rats in the CNV group were conducted with laser photocoagulation on their retina after anesthetization. (Parameters: Wavelength: 532 nm; Power: 85 mW; Spot diameter: 50 um; Photocoagulation time: 0.05 s). Each photocoagulation spot was 1 to 2 PD away from the optic disc and a total of 5 spots were made in each eye of a rat. FFA was performed to evaluate CNV formation 14 days after laser coagulation (Fig [114]S2). Subretinal injection of AAV-TAB1 over-expression and AAV vector The rats were anesthetized using sodium pentobarbital (350 mg/kg, i.p.), pupils were dilated by the compound tropicamide, and the ocular surface was anesthetized by oxybuprocaine hydrochloride. The cornea was flattened by a coverslip with ofloxacin eye ointment as a medium between the coverslip and the cornea for a clear vision of the operator. A tunnel was made at the limbus with a BD needle. Then a needle of the microinjector went into the tunnel until the needle tip penetrated the retinal nerve epithelium when the operator felt a slight empty sense. 6 μl of AAV vector was injected into the subretinal space. FITC-dextran perfusion and RPE-choroidal-sclera flat mount Rats were anesthetized and then perfused with 1 ml of 50 mg/ml FITC-labeled dextran (FD2000S, sigma, the USA) dissolved in phosphate buffer solution (PBS, PH 7.3) through their left ventricles. The successful performance of the perfusion was indicated by the appearance of a slightly yellow color on the conjunctiva, lips, and limbs. After the perfusion, the rats were sacrificed, and their eyeballs were extracted. The eyeballs were fixed in 10% formaldehyde solution for 1 h and then washed slightly with PBS. The corneas, lens, and retina were removed from the eyeballs, while the sclera, choroid, and RPE layers remained for flat mount preparation. Five or six incisions were radically made on the flat mount, and it was finally covered by a small slide. The CNV lesions were outlined, and the areas of the CNV lesions were measured using Image J. Protein extraction and proteolysis Eyeballs of the rats in both groups were taken out after sacrifice. The choroidal-sclera complex is obtained after the cornea, lens, and retina are carefully removed. Then the choroidal-sclera complexes were all stored in EP tubes at − 80 °C until the protein digestion process. Specimens from nine BN rats in each group were randomly divided into 3 samples in three EP tubes, each EP tube contains specimens from 3 BN rats. Samples were smashed by ultrasonic disrupter and proteins were extracted with the use of SDT solution (4% (w/v) SDS, 100 Tris/HCl pH7.6, 0.1 M DTT). The protein then was digested by trypsin in way of filter-aided proteome preparation (FASP), peptides were desalted by a C18 cartridge and were freeze-dried. Each sample was redissolved by 40 μL dissolution buffer and quantified. ITRAQ labeling and scx chromatography Workflow for ITRAQ-labeling and analysis of differentially-expressed proteins (Fig [115]S1). Peptides in each sample were taken out for ITRAQ labeling according to the ITRAQ labeling kit protocol from AB SCIEX company. The labeled peptides were mixed for the following fractionation with AKTA Purifier 100. Buffer solution A contained 10 mM KH2PO4 and 25% ACN, pH 3.0. Buffer solution B contained 10 mM KH2PO4, 500 mM KCl, 25%ACN, pH 3.0. Buffer solution A worked as an equilibrium liquid for the chromatographic column. Samples were loaded in a sample injector and then flowed to the chromatographic column with a velocity of 1 ml/min. For the peptide separation, linear gradients of buffer solution B were as follows: 0% in 25 min; 0–10% from 25 to 32 min; 10–20% from 32 to 42 min; 20–45% from 42 to 47 min; 45–100% from 47 to 52 min; 100% from 52 to 60 min; After 60 min, buffer solution B was reset to 0%. Light absorption value in 214 nm was monitored during the elution process. Elution fractions were collected every 1 min and each fraction was freeze-dried and desalted by C18 Cartridge. LC–MS/MS data gathering Each graded sample was separated by a liquid phase HPLC system, Easy nLC. Buffer solution A was solution contained 0.1% formic acid and buffer solution B was solution that contained 0.1% formic acid acetonitrile. 95% A worked as the equilibrium solution for the chromatographic column. The sample was loaded to the sample column (Thermo Scientific Acclaim PepMap100, 100 μm*2 cm, nanoViper C18) through an automatic sampler, separated when passing through the analytical column (Thermo scientific EASY column, 10 cm, ID75 μm, 3 μm, C18-A2) at a rate of 300 nl/min. Protein identification & quantification analysis The raw data of the mass spectrometry were exported as raw files and they were identified and quantitatively analyzed with software Mascot 2.2 and Proteome Discoverer 1.4. Bioinformatics Analysis, Go Function Annotation: The target proteins were annotated with Blast 2 Go. It includes four steps which are blast, mapping, annotation, and annotation augmentation. KEGG Pathway Annotations: The target proteins were annotated with the use of a KEGG automatic annotation server^[116]43. Enrichment Analysis of GO and KEGG annotation: Fisher’s exact test was used to compare contributions of GO analysis or KEGG pathways in target protein set and total protein set. Then the target protein set was enriched by GO annotation or KEGG pathway annotation. Protein Cluster Analysis: Firstly, the quantitative information of the target protein set was normalized. Then the Complex heatmap R was used for classification in dimensions of protein names and expression levels. Finally, the hierarchical clustering heat map was made. Western blot analysis The RPE-Choroidal sclera complex was taken out after the cornea, lens, and retina were removed. Then they were milled and smashed when dipped in protein lysis buffer on the ice. The RPE-Choroidal sclera complex was homogenized with a RIPA Protein Extraction Kit (Beyotime Biotechnology, C2006), and the protein concentrations were determined using a BCA Protein Concentration Determination Kit (Beyotime Biotechnology, C2006). The total protein was separated by SDS-PAGE and transferred onto polyvinylidene difluoride (PVDF) membranes (Millipore, Bedford, MA, USA). The membranes were blocked with 5% non-fat milk for 1 h at room temperature and subsequently incubated with primary antibodies at 4 °C overnight. After washing with PBST, the membranes were incubated with secondary antibodies conjugated to horseradish peroxidase for 1 h at room temperature. With chemiluminescence, the antibodies were visualized. The linear range of band detection was met. Three independent technical replicates were conducted for each sample. The original WB image is in the supplementary material (Figure [117]S3–S8). ELISA analysis RPE-choroidal-sclera complexes were cut into pieces and smashed in RIPA Protein Extraction Kit (Beyotime Biotechnology, C2006) on ice. Samples and standard samples were added to each hole, and incubated for 90 min at 37 °C. The biotin-marked antibody was added to incubation for 60 min at 37 °C. Then each hole was washed softly three times with 1× buffer. Avidin–biotin-peroxidase complex was added, and incubated for 30 min at 37 °C. Each hole was washed softly 5 times with 1× buffer. TMB (Tetramethylbenzidine) buffer was added and then each hole was incubated for 15 min at room temperature. A stop buffer was finally added. Statistical analysis Statistical analysis was performed using the SPSS 26.0 software. Correlation analysis between groups was conducted using an unpaired t-test and a one-way ANOVA, followed by post-hoc Tukey’s test, to analyze and compare the differences between groups. The data are expressed as the means ± standard (SD) deviations for at least three independent experiments. P-values below 0.05 were considered statistically significant. Supplementary Information Below is the link to the electronic supplementary material. [118]Supplementary Material 1^ (2.4MB, docx) Acknowledgements