Abstract Autoimmune uveitis (AU) is a group of autoimmune-driven diseases characterized by intraocular inflammation, often leading to severe vision loss. Ferroptosis, a recently discovered form of programmed cell death, has not yet been fully explored in the pathogenesis of AU. This study aims to investigate the role of ferroptosis-related key genes in AU, providing a theoretical foundation for further mechanistic studies. We downloaded [29]GSE198533 data set from the Gene Expression Omnibus (GEO). Through differential gene expression (DEG) analysis, weighted gene coexpression network analysis (WGCNA), and two machine learning models, TGFBR1 and ZFAS1 were identified as critical hub genes. Additionally, we validated the expression of TGFBR1 in retinal inflammation in a mouse model of experimental autoimmune uveitis (EAU) and explored its functional role. The results showed that TGFBR1 expression was significantly downregulated in EAU. Functional experiments demonstrated that TGFBR1 overexpression exacerbated retinal inflammation by promoting lipid peroxidation, downregulating GPX4 and xCT, and altering immune homeostasis by suppressing FOXP3 and enhancing IL-17A expression. Conversely, TGFBR1 inhibition via Galunisertib alleviated retinal inflammation and reversed ferroptosis- and immunity-related protein expression. These findings suggest that TGFBR1 contributes to AU pathogenesis by linking ferroptosis and immune imbalance and may serve as a potential biomarker and therapeutic target, particularly in BD-associated uveitis. __________________________________________________________________ graphic file with name ao5c00595_0013.jpg __________________________________________________________________ graphic file with name ao5c00595_0011.jpg 1. Introduction Autoimmune uveitis (AU) is a general name for a group of ocular autoimmune diseases, including conditions such as Vogt-Koyanagi-Harada (VKH) disease, Behçet’s disease (BD)-associated uveitis, HLA-B27 positive uveitis, and sympathetic ophthalmia. These diseases may be confined to the eye or may manifest alongside systemic symptoms. This common intraocular inflammatory disease, which predominantly affects individuals of working age, frequently involves the uvea, retina, retinal vasculature, and vitreous body, and is one of the leading causes of vision impairment and blindness. Recent bibliometric analyses have systematically summarized global research trends in uveitis over the past decade, highlighting significant progress and the increasing attention this disease has garnered from multidisciplinary research fields, including immunology, ophthalmology, and bioinformatics. AU is characterized by a complex pathogenesis, is difficult to cure, and tends to recur. Evidence suggests that uveitis triggered by autoinflammation and autoimmunity may share common molecular and immunopathogenic mechanisms. Previous research has found that type 1 helper T (Th1) cells and the primary cytokine produced by Th1, interferon-γ (IFN-γ), play a central role in the pathogenesis of uveitis. However, as research into AU has advanced, it has been found that various lymphocyte subpopulationsTh1, Th2, Th17, and Tregmay contribute significantly to the autoimmune aspects of uveitis. Experimental autoimmune uveitis (EAU) is a noninfectious uveitis model and the best animal model of AU to date. This model is induced by the injection of purified retinal proteins or their derived peptides. The immunopathogenesis of EAU is thought to be driven by aberrantly activated T cells, such as Th1 cells, which primarily secrete IFN-γ, and Th17 cells, which secrete IL-17A. These cells cross the blood-retinal barrier, recruiting inflammatory cells like macrophages and monocytes, leading to intraocular inflammation and destruction of the retina. Furthermore, Tregs characterized by FOXP3 expression also play a crucial role in maintaining immune tolerance and preventing excessive inflammation. Dysfunction of FOXP3^+ Tregs may contribute to the exacerbation of uveitis by weakening their suppressive effects on Th1 and Th17 cells. , Ferroptosis is a form of iron-dependent programmed cell death driven by the overload of lipid peroxides on cellular membranes, first reported in 2012, and is characterized by the accumulation of lipid peroxides. When phospholipids containing polyunsaturated fatty acids (PUFA–PLs) undergo peroxidation catalyzed by iron, and if this process exceeds the buffering capacity of the ferroptosis defense system, lipid peroxides accumulate to lethal levels on the cell membrane, leading to membrane rupture and ultimately resulting in cell death through ferroptosis. The defense mechanisms against ferroptosis include the GPX4-GSH system, FSP1-CoQH2 system, DHODH-CoQH2 system, and GCH1-BH4 system. The GPX4-GSH system is regarded as the principal defense mechanism against ferroptosis. Glutathione peroxidase 4 (GPX4) is the only member of the GPX family capable of reducing lipid peroxides; due to this distinctive feature, it is always considered a key enzyme in this pathway and a critical marker of ferroptosis. , Cells utilize the transport protein complex known as system Xc- (composed of the transport subunit SLC7A11 and the regulatory subunit SLC3A2) to reversibly transport cystine and glutamate in a 1:1 ratio across the cell membrane. Cystine imported into the cell is subsequently reduced to cysteine by glutathione or thioredoxin-2, which then participates in glutathione synthesis; the synergistic action of glutathione and GPX4 can eliminate lipid peroxides effectively. At present, ferroptosis has been involved in the onset and progression of various autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and autoimmune hepatitis (AIH). In autoimmune uveitis, previous studies have shown that lipid peroxidation levels are elevated in mouse models of EAU. , Furthermore, in investigations into BD, higher levels of lipid peroxides have been reported in patient serum compared to healthy controls. These findings suggest that lipid peroxidation may play a significant role in EAU, and that ferroptosis could contribute to its pathogenesis. Moreover, with the widespread application of bioinformatics and artificial intelligence in the medical field, machine learning algorithms have emerged as important tools for identifying novel retinal biomarkers and developing precise diagnostic and predictive models, and have already shown utility in Behçet’s disease-associated uveitis. Utilizing data from the Gene Expression Omnibus (GEO) and the Ferrdb database, we employed Weighted Gene Coexpression Network Analysis (WGCNA) and machine learning algorithms to identify two hub genes, transforming growth factor-β receptor 1 (TGFBR1) and ZFAS1, associated with ferroptosis in BD. Subsequently, we validated the expression of these ferroptosis-related genes in the EAU mouse model and explored the role of TGFBR1 in the pathogenesis of uveitis. This study provides novel insights and potential targets for understanding the pathological mechanisms of uveitis and developing therapeutic strategies. 2. Result 2.1. Weighted Gene Co-Expression Network Construction and Screening of Differentially Expressed Ferroptosis-Related Genes In this study, we utilized the [30]GSE198533 data set, which comprises sample data from 9 patients with BD and 10 healthy controls, in conjunction with ferroptosis-related genes from the Ferrdb database. Based on the filtering criteria of |log FC| > 0.5 and an adjusted p-value <0.05, we identified 42 upregulated and 11 downregulated genes ([31]Figure A). Subsequently, we performed a heatmap analysis on the top 30 upregulated and downregulated genes, where red represents upregulated genes and blue indicates downregulated genes ([32]Figure B). 1. [33]1 [34]Open in a new tab Weighted gene co-expression network construction and screening of cDEGs. (A) Volcano plot of DEGs after integration of the [35]GSE198533 data set and the Ferrdb database. (B) Heatmap of the DEGs. (C) Network topology analysis under various soft-threshold powers. (D) Dendrogram of clustered genes based on topological overlap and module color assignment. (E) Module-trait correlation analysis. (F) Venn diagram showing the intersection of DEGs and turquoise module genes. To identify gene modules significantly associated with clinical traits, we constructed a gene co-expression network for the training group using WGCNA. First, a similarity matrix was calculated between genes, and the selection of a soft threshold ensured the network conformed to the scale-free topology. To achieve an optimal network topology, a soft threshold of 9 was selected, indicating high average connectivity within the network ([36]Figure C). Subsequently, the adjacency matrix was transformed into a Topological Overlap Matrix (TOM), and hierarchical clustering was performed using the dissimilarity matrix (dissTOM = 1-TOM) derived from the TOM. The Dynamic Tree Cut method was then applied to divide the genes into distinct modules ([37]Figure D). These modules were then correlated with BD, and the genes of the most positively correlated modules were identified. Ultimately, the turquoise module (MEturquoise) was found to be strongly positively correlated with BD (R = 0.91, p < 0.001), comprising 180 genes ([38]Figure E). To further investigate the role of ferroptosis in BD, we performed an intersection analysis between the differentially expressed genes (DEGs) and the genes within the turquoise module, resulting in the identification of 51 candidate differentially expressed genes (cDEGs) ([39]Figure F). 2.2. Functional Enrichment Analysis of Characteristic Genes and Construction of Protein–Protein Interaction (PPI) Network Subsequently, we performed a functional enrichment analysis of the identified key genes to investigate the ferroptosis-related biological functions and potential pathways involved in BD. Gene Ontology (GO) enrichment analysis revealed that these genes were significantly enriched in biological processes and molecular functions, such as oxidative stress, TGF-β receptor binding, and ubiquitin protein ligase binding ([40]Figure A,B). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis indicated that ferroptosis, human T-cell leukemia virus type 1 infection, arachidonic acid metabolism, and hepatitis B, as well as signaling pathways including the TGF-β, relaxin, and FoxO signaling pathways, were closely associated with these genes ([41]Figure C,D). 2. [42]2 [43]Open in a new tab Functional enrichment analysis of characteristic genes. (A, B) GO functional annotation of characteristic genes. (C, D) KEGG signaling pathway annotation of characteristic genes. For all enriched GO and KEGG terms, p < 0.05. To further investigate the potential mechanisms, we constructed a PPI network based on the STRING database ([44]Figure D). 3. [45]3 [46]Open in a new tab Identification of hub genes via machine learning and hub jene validation of expression patterns. (A) Seven hub genes identified by LASSO, along with the cross-validation curve of the LASSO regression. (B) Six hub genes identified by SVM-RFE. (C) Venn diagram showing the overlap of hub genes identified by LASSO and SVM-RFE. (D) PPI network constructed using the STRING database. (E) Expression levels of TGFBR1 in [47]GSE198533. (F) Expression levels of ZFAS1 in [48]GSE198533. 2.3. Identification of Hub Genes To narrow down the set of characteristic genes, we employed two machine learning algorithms for feature gene identification. First, the Least Absolute Shrinkage and Selection Operator (LASSO) regression algorithm was used to extract seven genes from the feature set, including ZFAS1, CHMP6, OIP5-AS1, TGFBR1, FH, PIEZO1 and MMD ([49]Figure A). Next, the Support Vector Machine–Recursive Feature Elimination (SVM-RFE) algorithm identified six genes: ZFAS1, MLST8, TGFBR1, HSF1, SRC and OTUB1 ([50]Figure B). Finally, TGFBR1 and ZFAS1 were identified as overlapping hub genes by both the LASSO and SVM-RFE algorithms ([51]Figure C). To assess whether TGFBR1 and ZFAS1 could serve as diagnostic biomarkers, we analyzed their mRNA expression levels. Box plots were used to illustrate the expression levels of these two hub genes in BD patients compared to healthy individuals ([52]Figure E,F). 2.4. Immune Cell Infiltration and Its Correlation with Hub Genes We further explored the differences in immune cell infiltration between BD patients and healthy controls in the [53]GSE198533 data set. A total of 17 immune cell types were identified in the data set and are presented using heatmaps and boxplots. The heatmap revealed a significantly higher infiltration of monocytes, eosinophils, M1 macrophages, and M2 macrophages in BD samples ([54]Figure A). The boxplots also displayed significant differences in the levels of γδ T cells, eosinophils, M1 macrophages, and M2 macrophages between healthy individuals and BD patients ([55]Figure B). Additionally, correlation analysis between immune cells and hub genes showed that, in BD samples, γδ T cells, NK cells, memory CD4+ T cells, and CD4+ T cells were positively correlated with TGFBR1 ([56]Figure C), while eosinophils, M1 macrophages, M2 macrophages, and neutrophils were positively correlated with ZFAS1 ([57]Figure D). 4. [58]4 [59]Open in a new tab Immune cell infiltration and correlation with hub genes. (A, B) Heatmap and boxplots of the 17 identified immune cell types in the [60]GSE198533 data set. (C) Correlation between immune cell infiltration and TGFBR1. (D) Correlation between immune cell infiltration and ZFAS1. 2.5. EAU Model Construction and Hub Gene Expression Verification The EAU model is a well-established model used to study autoimmune uveitis. In this study, 6–8 weeks old B10.RIII mice were selected, and an EAU model was induced by subcutaneously injecting a mixture of interphotoreceptor retinoid-binding protein (IRBP) and complete Freund’s adjuvant (CFA). On the 14th day postimmunization, the inflammatory manifestations in the anterior segment of the mice’s eyes were observed and evaluated using image scoring. The results showed that EAU mice exhibited significant inflammatory characteristics in the anterior segment, such as corneal edema and conjunctival hyperemia ([61]Figure A), with clinical scores markedly higher than those of the control group ([62]Figure B). Furthermore, histopathological examination on day 14 confirmed these anterior segment findings. HE staining of the retina revealed a pronounced increase in inflammatory cell infiltration and retinal folds in EAU mice ([63]Figure C), with histology scores notably elevated compared to the CON group ([64]Figure D). These results confirm the successful establishment of the EAU inflammatory model. 5. [65]5 [66]Open in a new tab Construction of the EAU model, validation of hub genes, and retinal MDA expression. (A, B) Representative anterior segment images and clinical scores of the CON and EAU groups on day 14. Red arrows indicate conjunctival hyperemia; black arrows indicate posterior synechiae and inflammatory exudates. (C, D) Representative histological staining images and histopathological scores of tissue sections from the CON and EAU groups. Red arrows indicate vitreous cell infiltration; black arrows indicate retinal folds; yellow arrows indicate neovascularization and hemorrhage. (GCL, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer). (E, F) qRT-PCR results for TGFBR1 and ZFAS1 in the retinas of the CON and EAU groups. (G) Measurement of MDA levels in the retinas of the CON and EAU groups. All qRT-PCR data were normalized to the mean value of the CON group. Data are presented as mean ± SD, n = 3–4. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant. In our previous bioinformatics analysis, we identified two ferroptosis-related genes, ZFAS1 and TGFBR1, exhibiting substantial expression differences between BD patients and healthy controls. To further validate these findings, we assessed their expression in the EAU mouse model. On the 14th day after injection, retinal samples from the mice were collected, and quantitative real-time polymerase chain reaction (qRT-PCR) was employed to observe the relative expression changes of ZFAS1 and TGFBR1 mRNA. The results showed that, compared to CON, TGFBR1 was downregulated while ZFAS1 was upregulated in the retinas of EAU ([67]Figure E, F). Similarly, Western blot (WB) analysis confirmed a significant downregulation in the relative expression of TGFBR1 protein ([68]Figure A,D). 6. [69]6 [70]Open in a new tab Protein expression in the CON and EAU groups. (A, D) WB images and quantitative analysis of TGFBR1 in the retinas of the CON and EAU groups. (B, E) WB images and quantitative analysis of GPX4 in the retinas of the CON and EAU groups. (C, F) WB images and quantitative analysis of xCT in the retinas of the CON and EAU groups. All WB quantification values were normalized to the mean value of the CON group. Data are presented as mean ± SD, n = 4–5. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant. 2.6. Measurement of Lipid Peroxidation Levels and Ferroptosis Markers in the EAU Model Next, we assessed the levels of ferroptosis in the retinas of the CON and EAU groups. To evaluate lipid peroxidation, we measured the levels of malondialdehyde (MDA), a key byproduct of lipid peroxidation, in both groups. The results showed that MDA levels were significantly elevated in the retinas of EAU mice, indicating increased lipid peroxidation in the EAU group ([71]Figure G). Given the crucial role of GPX4 in scavenging lipid peroxides, it is recognized as a critical component in ferroptosis defense and a pivotal marker of this process.We analyzed GPX4 protein expression levels in the retinas of EAU and CON mice, finding a significant increase in GPX4 expression in the EAU group ([72]Figure B,E). Additionally, the expression of xCT (also known as SLC7A11), a key component of the cystine/glutamate antiporter involved in ferroptosis, exhibited a mild increase in the EAU group, though this result did not reach statistical significance ([73]Figure C,F). These findings indicate that while lipid peroxidation is heightened in the retinas of EAU mice, extensive ferroptosis may not occur. 2.7. Upregulation of TGFBR1 Exacerbates Retinal Inflammation in the EAU Model To investigate the role of TGFBR1 in EAU, we utilized the peptide KRFK derived from TSP-1 as a TGFBR1 agonist. , Initially, we assessed the retinal toxicity of this peptide. KRFK was diluted with PBS to a concentration of 5 mg/mL. Mice aged 6 to 8 weeks were randomly assigned to two groups: one group received intraperitoneal injections of KRFK at a dose of 0.08 mL per mouse per day, while the CON group was administered an equivalent volume of PBS daily. Both groups underwent continuous intraperitoneal injections for seven consecutive days. On the seventh day, retinal function was evaluated using electroretinograms (ERG) and optokinetic response (Optomotor) tests ([74]Figure A). ERG data indicated no significant changes in the amplitudes of the A-wave and B-wave between the two groups ([75]Figure B,C). Similarly, the Optomotor results showed comparable visual acuity in both groups ([76]Figure D). These findings suggest that there are no significant differences in visual electrophysiology or function between the KRFK and CON groups, indicating that KRFK does not exhibit evident retinal toxicity. Additionally, Additionally, WB analysis revealed that the protein expression level of TGFBR1 in the retinas of KRFK-treated EAU mice was significantly elevated compared to the untreated EAU group and CON group ([77]Figure A,D). Clinically and histologically, the KRFK treatment group exhibited exacerbated anterior segment inflammation, including corneal edema and neovascularization ([78]Figure E,G), along with more pronounced retinal detachment and other pathological changes ([79]Figure F,H). These results indicate that intraperitoneal injection of KRFK does not damage the mouse retina, while it enhances the protein levels of TGFBR1, thus exacerbating the inflammatory response in the EAU model. 7. [80]7 [81]Open in a new tab Upregulation of TGFBR1 exacerbates anterior segment inflammation and increases retinal MDA levels in EAU. (A) Illustration of the optomotor system. (B) A-wave amplitudes recorded at 3.0 cd s/m^2 in the CON and KRFK groups. (C) B-wave amplitudes recorded at 3.0 cd s/m^2 in the CON and KRFK groups. (D) Quantification of visual acuity in the CON and KRFK-treated mice. (E, G) Representative anterior segment images and clinical scores in the CON group, EAU group, and EAU + KRFK group on day 14. Red arrows indicate conjunctival hyperemia; black arrows indicate posterior synechiae and inflammatory exudates. (F, H) Representative histological staining images and histopathological scores of tissue sections from the CON group, EAU group, and EAU + KRFK group. Red arrows indicate vitreous cell infiltration; black arrows indicate retinal folds; yellow arrows indicate neovascularization and hemorrhage. (GCL, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer). (I) Measurement of MDA levels in the retinas of the CON group, EAU group, and EAU + KRFK group. Data are presented as mean ± SD, n = 3–5. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant. 8. [82]8 [83]Open in a new tab Protein expression and serum ELISA analysis results in the CON, EAU, and EAU + KRFK groups. (A, D) WB images and quantitative analysis of TGFBR1 in the retinas of the CON, EAU, and EAU + KRFK groups. (B, E) WB images and quantitative analysis of GPX4 in the retinas of the CON, EAU, and EAU + KRFK groups. (C, F) WB images and quantitative analysis of xCT in the retinas of the CON, EAU, and EAU + KRFK groups. (G, I) WB images and quantitative analysis of FOXP3 in the retinas of the CON, EAU, and EAU + KRFK groups. (H, J) WB images and quantitative analysis of IL-17A in the retinas of the CON, EAU, and EAU + KRFK groups. (K) Serum IL-17A levels in the EAU and EAU + KRFK groups. All WB quantification values were normalized to the mean value of the CON group. Data are presented as mean ± SD, n = 3–4. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant. 2.8. TGFBR1 Induces Retinal Ferroptosis in EAU Mice by Downregulating GPX4 and xCT Expression In a study on acute liver failure (ALF), TGFBR1 was identified as a driver of ferroptosis, with TGFBR1 knockout leading to a significant increase in xCT and GPX4 expression, thereby reducing ferroptosis in liver tissues. Given that excessive lipid peroxidation is a hallmark of ferroptosis, we assessed retinal MDA levels in CON, EAU, and KRFK-injected EAU mice. Our results indicated a notable elevation of MDA in the retinas of KRFK-treated EAU mice compared to the other two groups ([84]Figure I). Next, we conducted WB analysis to assess the expression levels of GPX4 and xCT, two critical proteins involved in ferroptosis defense systems. Both proteins were significantly downregulated in the retinas of KRFK-injected EAU mice ([85]Figure B,C,E,F). These findings suggest that TGFBR1 may induce retinal ferroptosis in EAU mice by downregulating GPX4 and xCT expression, thereby exacerbating retinal inflammation. 2.9. TGFBR1 Exacerbates Retinal Inflammation in EAU Mice by Modulating the Expression Of Immune Cell-Related Cytokines In addition to inducing ferroptosis, TGFBR1 has been reported to regulate immune responses via cytokine signaling. TGFBR1 is a critical component of the TGF-β receptor complex and plays a pivotal role in the TGF-β dual receptor signaling pathway. The TGF-β pathway is crucial for the transcriptional regulation of FOXP3 expression and the suppressive function of regulatory T cells (Tregs). , To investigate the cytokine signaling regulation by TGFBR1 in the context of EAU, we measured the expression levels of key immune cell-related cytokines in the retinas of EAU mice. WB analysis revealed that FOXP3 expression was lower in the retinas of EAU mice compared to CON, and its expression was significantly reduced following KRFK treatment ([86]Figure G,I). Retinal IL-17A levels were assessed in CON, EAU, and KRFK-treated EAU mice, with results showing that IL-17A levels were elevated in the EAU group compared to CON but were significantly higher in the KRFK-treated group ([87]Figure H,J). The enzyme-linked immunosorbent assay (ELISA) results also demonstrated that the serum IL-17A levels in EAU mice were lower than those in the KRFK-treated EAU mice ([88]Figure K). These findings suggest that increased TGFBR1 expression may not enhance Treg cell numbers. Instead, TGFBR1 upregulation reduces FOXP3 levels and leads to increased IL-17A production in both the retina and serum, thereby exacerbating ocular inflammation. 9. [89]9 [90]Open in a new tab Inhibition of TGFBR1 alleviates anterior segment inflammation and reduces retinal MDA levels in EAU mice. (A, C) Representative anterior segment images and clinical scores in the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group on day 14. Red arrows indicate conjunctival hyperemia; black arrows indicate posterior synechiae and inflammatory exudates. (B, D) Representative histological staining images and histopathological scores of tissue sections from the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group. Red arrows indicate vitreous cell infiltration; black arrows indicate retinal folds. (GCL, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer). (E) Measurement of MDA levels in the retinas of the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group. Data are presented as mean ± SD, n = 4–5. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant. 2.10. TGFBR1 Inhibition Attenuates Retinal Inflammation in EAU Mice by Regulating Ferroptosis and Immune-Related Protein Expression To further validate the role of TGFBR1, we subsequently administered the TGFBR1 inhibitor Galunisertib (LY2157299) via oral gavage to mice in the EAU group and those treated with KRFK. Each mouse received Galunisertib at a dose of 150 mg/kg twice daily, starting from day 7 after EAU induction, and continued for 7 consecutive days. , Anterior segment photography and HE staining revealed a significant reduction in inflammation in both the anterior segment and retina of mice in the Galunisertib-treated group compared to the EAU group. Conversely, mice treated with KRFK alone exhibited notably aggravated ocular inflammation. Importantly, ocular inflammation in mice receiving combined treatment with KRFK and Galunisertib was significantly alleviated compared with that in mice treated with KRFK alone ([91]Figure A–D). Additionally, assessment of retinal MDA levels revealed that Galunisertib treatment led to a pronounced reduction in MDA content relative to the EAU group, whereas treatment with KRFK alone resulted in a clear elevation of retinal MDA levels. The combined treatment with KRFK and Galunisertib further attenuated retinal MDA levels compared to treatment with KRFK alone ([92]Figure E). The WB results further demonstrated that, compared to the EAU group, retinal GPX4 and xCT protein levels were significantly elevated in mice treated with Galunisertib alone, whereas these protein levels were markedly decreased in mice treated with KRFK alone. Combined treatment with KRFK and Galunisertib further increased retinal GPX4 and xCT protein expression levels compared to the group treated with KRFK alone ([93]Figure A–D). Additionally, we evaluated the expression of immune-related proteins FOXP3 and IL-17A in retinal tissues. Compared to the EAU group, Galunisertib treatment alone notably increased FOXP3 protein levels and reduced IL-17A protein levels, whereas KRFK treatment alone resulted in decreased FOXP3 expression and increased IL-17A expression. Furthermore, compared with the group treated with KRFK alone, combined treatment with KRFK and Galunisertib markedly increased FOXP3 expression and decreased IL-17A expression in retinal tissues ([94]Figure E–H). These findings indicate that inhibition of TGFBR1 function may alleviate retinal inflammatory injury in EAU mice by enhancing retinal GPX4 and xCT protein levels, reducing MDA content, and concurrently regulating immune responses by upregulating FOXP3 expression and suppressing IL-17A levels. 10. [95]10 [96]Open in a new tab TGFBR1 inhibition restores ferroptosis- and immunity-related protein expression in the retina of EAU mice. (A, C) WB images and quantitative analysis of GPX4 in the retinas of the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group. (B, D) WB images and quantitative analysis of xCT in the retinas of the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group. (E, G) WB images and quantitative analysis of FOXP3 in the retinas of the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group. (F, H) WB images and quantitative analysis of IL-17A in the retinas of the EAU group, Galunisertib group, KRFK group and KRFK + Galunisertib group. All WB quantification values were normalized to the mean value of the EAU group. Data are presented as mean ± SD, n = 4–6. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001; ns = not significant. 3. Discussion An increasing body of evidence suggests a close association between ferroptosis and the pathogenesis of autoimmune diseases. For instance, Luo et al. discovered reduced GPX4 levels and elevated MDA levels in a lipopolysaccharide (LPS)-induced synovitis cell model, providing preliminary evidence for the link between ferroptosis and synovitis. A recent study has demonstrated that, compared to normal mice, mice with SLE exhibit lower GPX4 expression alongside significantly elevated MDA levels and Th1/Th2 ratios. Treatment with ferroptosis inhibitors markedly increased GPX4 expression while decreasing MDA levels and the Th1/Th2 ratio, suggesting that decreased GPX4 may modulate T cell ferroptosis, consequently leading to an increased Th1/Th2 ratio that may trigger SLE. Another study reported similar findings in an experimental autoimmune encephalomyelitis (EAE) mouse model. Notable lipid peroxidation was observed in oligodendrocytes of the EAE mice, accompanied by significantly reduced levels of GPX4 and xCT in the spinal cord. Importantly, these changes could be reversed by treatment with ferroptosis inhibitors. These investigations indicate that ferroptosis significantly impacts the inflammation and organ damage associated with autoimmune diseases and plays a crucial role in their pathogenesis. These findings support our hypothesis that ferroptosis may be closely related to the onset and progression of autoimmune uveitis. This study initially employed bioinformatics techniques to explore the association between ferroptosis and autoimmune uveitis. We gained transcriptomic data sets for Behçet’s disease (BD) and healthy controls from the GEO database and utilized WGCNA, machine learning, and enrichment analysis to identify two hub genes related to ferroptosis: TGFBR1 and ZFAS1. Furthermore, we analyzed the expression differences of these genes in the data set, finding that TGFBR1 expression was significantly lower in BD patients compared to healthy individuals, while ZFAS1 expression was higher. Additionally, these gene expressions were validated in the EAU model, with qPCR results demonstrating downregulation of TGFBR1 in EAU mice and upregulation of ZFAS1. The Western blot results for TGFBR1 were consistent with those obtained from qPCR. These findings suggest that TGFBR1 expression is closely associated with disease severity and may contribute to EAU pathogenesis by regulating ferroptosis, supporting its potential as a therapeutic target in EAU and a prospective biomarker in BD. Given that the identification of hub genes in this study was based on peripheral immune transcriptomic data, we further evaluated the feasibility and validity of this strategy. The [97]GSE198533 data set was originally designed to investigate the immunopathogenesis of Behçet’s disease by analyzing transcriptomic differences in peripheral blood mononuclear cells (PBMCs) between patients and healthy controls, with a particular focus on the potential relationship between COVID-19 and BD. Although PBMCs are fundamentally different in tissue origin from retinal tissues in EAU mice, they play a central role in systemic immune responses and have been widely used in studies of autoimmune diseases, including ocular conditions such as AU and Sjögren’s syndrome-related keratoconjunctivitis sicca. , In contrast to previous studies using this data set that primarily focused on immune cell infiltration and inflammatory signaling pathways, our study introduces ferroptosis as a novel perspective. By integrating WGCNA with multiple machine learning algorithms for hub gene selection and validation, we propose a new hypothesis that TGFBR1 may serve as a potential regulator of ferroptosis in AU. TGFBR1 has previously been implicated in the regulation of ferroptosis in several disease models, prompting us to further explore its mechanistic role in EAU. TGFBR1 has been identified as a driver of ferroptosis in previous studies. For instance, in an ALF mouse model, it was found that the deletion of TGFBR1 in the liver effectively alleviated ALF symptoms. Another study demonstrated that calcipotriol mitigated osteoarthritis (OA) by inhibiting GPX4-mediated ferroptosis in chondrocytes through blocking the TGF-β1 pathway. Similarly, Liu C and colleagues observed a comparable role of TGFBR1 in diabetic kidney disease (DKD). Our study demonstrated that TGFBR1 similarly acts as a driver of ferroptosis in the EAU mouse model. In this study, the upregulation of TGFBR1 significantly elevated MDA levels in the retinas of EAU mice, while also increasing the protein expression of GPX4 and xCT. This suggests that TGFBR1 upregulation may promote ferroptosis in retinal cells by enhancing the expression of GPX4 and xCT, thereby exacerbating retinal inflammation. In addition to its aforementioned functions, as a crucial component of the TGF-β receptor complex, TGFBR1 plays a pivotal role in the TGF-β dual receptor system. Upon ligand binding, TGFBR1 is phosphorylated by TGFBR2, which is a critical step for activating downstream signaling pathways, essential for both SMAD-dependent and SMAD-independent signal transduction. Recent studies have revealed that the TGF-β signaling pathway exerts significant regulatory effects on immune cells, particularly those involved in autoimmune processes. TGF-β promotes the differentiation of Tregs and regulates FOXP3 expression, thereby maintaining immune tolerance and preventing the onset of autoimmune diseases. Additionally, TGF-β can synergize with cytokines such as interleukin-6 (IL-6) to promote the development of Th17 cells. However, in our study, we observed that upregulating TGFBR1 did not lead to an increase in FOXP3 expression or a reduction in EAU-associated inflammation, but rather produced the opposite results. It is possible that the divergence in our results, compared to earlier studies, is due to altered expression of GPX4, which could impact the activity of FOXP3^+ Tregs. Xu et al. reported that the specific deletion of GPX4 in Tregs impaired their survival in tumor environments, increased T cell infiltration into tumors, and enhanced antitumor immune responses, indicating that GPX4 plays an important role in maintaining Treg survival and their immunosuppressive functions, thus facilitating tumor immune evasion. Moreover, our study found that IL-17A levels in the retina and serum of EAU mice significantly increased with the upregulation of TGFBR1, consistent with previous reports. These observations suggest that TGFBR1 may reduce FOXP3 levels by downregulating GPX4 expression while simultaneously enhancing Th17 cell activity, leading to elevated IL-17A secretion, which exacerbates ocular inflammation in EAU. The limitations of this study are mainly reflected in the following aspects. First, the results are primarily based on transcriptomic data from human PBMCs and the EAU animal model, and therefore require further validation in clinical samples to determine their applicability to patients with AU. Second, although we confirmed that TGFBR1 can induce ferroptosis in the EAU model, the specific mechanisms by which it regulates GPX4 and other related molecules remain unclear and warrant further investigation. In addition, this study focused on Behçet’s disease-associated uveitis and the EAU mouse model. While Behçet’s disease-associated uveitis represents a major subtype of AU, it does not encompass all forms of the disease. Other AU subtypes, such as VKH disease and HLA-B27-associated anterior uveitis, may involve distinct immunopathogenic mechanisms. Therefore, the findings of this study should not be directly extrapolated to all AU subtypes and should be further validated in diverse clinical contexts. Future research should expand to include a broader range of AU subtypes and incorporate patient-derived ocular tissues or retinal organoid models to comprehensively evaluate the clinical relevance and translational potential of TGFBR1 as a ferroptosis-related biomarker and therapeutic target. 4. Conclusions This study employed a combination of WGCNA, machine learning, and other bioinformatics approaches to identify TGFBR1 and ZFAS1 as core ferroptosis-related genes in AU. Subsequently, their expression patterns in retinal inflammation were validated using the EAU model, and the mechanistic role of TGFBR1 in the disease was further investigated. The findings indicate that TGFBR1 and ZFAS1 hold potential as therapeutic targets and diagnostic biomarkers for AU. Specifically, TGFBR1 was found to exacerbate retinal ferroptosis by regulating the expression of GPX4 and xCT, while also downregulating FOXP3 and upregulating IL-17A, thereby intensifying ocular inflammation in mice. This research provides new insights into the development of diagnostic and therapeutic strategies for BD. A deeper understanding of the mechanisms underlying TGFBR1 could facilitate the design of more effective interventions for BD. Future studies should continue to elucidate the role of TGFBR1 and its associated pathways to develop more targeted therapeutic strategies for BD and other immune-related diseases. 5. Materials and Methods 5.1. Data Download and Preprocessing The BD data set used in this study, [98]GSE198533, was downloaded from the GEO public database (GEO, [99]https://www.ncbi.nlm.nih.gov/geo/), which contains data from 9 BD patients and 10 healthy controls. The ferroptosis-related genes were sourced from the Ferrdb database ([100]http://www.zhounan.org/ferrdb). The data were analyzed and processed using R software (version 4.1.1). First, Ensembl IDs were converted to gene symbols and annotated for subsequent research and analysis. The R package “limma” (3.60.6) was then employed to normalize and merge the data sets, using adjusted p-values of <0.05 and |log FC| > 0.5 as cutoff thresholds to identify DEGs. The results were visualized using heatmaps and volcano plots generated by the ‘pheatmap’ (1.0.12) and ‘ggplot2′ R packages (3.5.1), respectively. 5.2. Construction of Weighted Gene Coexpression Networks and Identification of cDEGs WGCNA was performed to identify modules of highly correlated genes, elucidate the interconnections among these modules, and assess their associations with external sample traits, thereby identifying potential biomarkers or therapeutic targets. The R package “WGCNA” (1.73) was utilized to construct a weighted gene coexpression network and perform correlation analyses among genes. To construct a weighted gene coexpression network, we selected the top 5000 genes ranked by variance. The optimal soft-thresholding power (β) was determined using the pickSoftThreshold function in the WGCNA R package, which evaluates scale-free topology fit (R ^2) and mean connectivity across a range of candidate powers (1–30). According to the scale-free topology criterion, a power value of β = 9 was chosen, as it was the lowest power for which the scale-free topology model fit index exceeded 0.9 while maintaining sufficient mean connectivity. A minimum module size of 30 and merge cut height of 0.25 were set. Gene modules were identified using hierarchical clustering based on the topological overlap matrix, and their correlation with the BD/control phenotype was evaluated. The module most strongly associated with ferroptosis was selected as the key module for subsequent analysis. The ‘venn’ package (1.12) was employed to extract intersecting feature genes between DEGs and WGCNA, which were visualized in a Venn diagram. Genes overlapping between DEGs and WGCNA were selected as cDEGs for further investigation. 5.3. Function Enrichment Analysis and PPI Network Construction The ‘clusterProfiler’ package (4.12.6) in R was implemented to perform GO and KEGG functional enrichment analysis on the cDEGs, assessing the associated biological processes (BP), molecular functions (MF), cellular components (CC), and gene-related signaling pathways. A p-value of <0.05 was considered indicative of a statistically significant level of enrichment analysis. Bubble plots were generated using the “ggplot2” R package to visualize the enrichment findings. To further investigate the interactions among the identified cDEGs, a PPI network was constructed using the STRING database (version 11.0b, [101]https://www.string-db.org/). The PPI network was visualized using Cytoscape software to better understand the interaction dynamics. 5.4. Screening Hub Genes by Machine Learning To identify optimal feature genes among cDEGs, we applied two supervised machine learning techniques: LASSO logistic regression and SVM-RFE. LASSO regression was conducted using the “glmnet” package (version 4.1) in R. This method applies an L1 penalty (lambda) to shrink the coefficients of less informative variables to zero, thereby performing variable selection and reducing overfitting risk. The input gene expression matrix was constructed from the [102]GSE198533 data set, filtered to include BD and control samples. Binary phenotype labels (“1” for BD, “0” for control) were assigned accordingly. All gene expression data were log-transformed and standardized prior to analysis. LASSO regression was performed with the following settings: family = “binomial” to reflect binary classification, α = 1 for LASSO penalty, nlambda = 1000 to explore a wide range of regularization parameters, and nfolds = 10 for 10-fold cross-validation. The optimal penalty parameter (lambda.min) was selected based on the minimum mean cross-validated error. Genes with nonzero coefficients at lambda.min were retained as the final LASSO-selected features. SVM-RFE analysis is a supervised machine learning technique utilized to identify the optimal hub genes by eliminating feature vectors generated by SVM. The SVM-RFE procedure was implemented using the svmRFE function sourced from a custom script (msvmRFE.R), along with the e1071, caret, and sigFeature R packages. The analysis was conducted using 10-fold cross-validation (k = 10) and recursive elimination was applied with halve.above = 100, meaning the feature set was halved when more than 100 features remained in each iteration. The procedure was repeated across folds to ensure stability, and the top-ranked features were averaged based on cross-validated performance. The top 40 features were further used to construct the SVM model, and classification performance was assessed by plotting the error rate and accuracy curves. The optimal number of features corresponding to the lowest classification error was selected and retained as the final SVM-RFE output. The “venn” package was used to identify and visualize the intersecting hub genes shared between LASSO and SVM-RFE results. These overlapping genes were designated as final hub genes for subsequent validation and mechanistic investigation. 5.5. Differential Expression Analysis The R packages “limma” and “ggpubr” (0.6.0) were used to analyze and compare the expression levels of the final hub genes between the BD group and the control group, which were visualized using boxplots. 5.6. Immune Infiltration Analysis The CIBERSORT algorithm is based on linear support vector regression (SVR) to deconvolve transcriptomic expression matrices, estimating the composition and abundance of immune cells within mixed cellular populations based on transcriptomic data. Immune cell infiltration was evaluated using the CIBERSORT algorithm, which deconvolutes bulk tissue gene expression data into proportions of 22 immune cell types. The analysis was performed by uploading the normalized gene expression matrix from the [103]GSE198533 data set to the CIBERSORT web portal ([104]https://cibersort.stanford.edu/), using the LM22 signature matrix. Quantile normalization was disabled (QN = FALSE) to accommodate RNA-seq data, and 1000 permutations were performed to ensure robust statistical inference. Only samples with a deconvolution p-value <0.05 were retained for downstream analysis. Bar plots and heatmaps of immune cell proportions were generated using the ggplot2 and pheatmap packages. Spearman correlation analysis between hub genes and immune cell proportions was conducted using the ggpubr and ggExtra packages. Immune cell types were plotted against hub gene expression values, and correlation strength and significance were visualized using lollipop plots generated from the correlation matrix. 5.7. Construction of Mouse Models This study was reported in accordance with ARRIVE guidelines. All methods were carried out following the relevant guidelines and regulations. The ethical and legal approval with the number WDRM20240702D was obtained from the Ethics Committee of the Renmin Hospital of Wuhan University prior to the commencement of the study. B10.RIII mouse parents were sourced from The Jackson Laboratory (Bar Harbor, ME), and all experimental mice were bred and maintained under specific pathogen-free (SPF) conditions with a 12-h light/dark cycle with free access to standard normal diet and water. Mice aged 6–8 weeks were selected for the experiment and anesthetized with sodium pentobarbital. An emulsion was prepared by mixing 50 μg of IRBP with an equal volume of CFA containing 1.0 mg/mL of Mycobacterium tuberculosis strain (MTB). Each B10.RIII mouse was subcutaneously injected with 100 μL of the emulsion at the base of the tail and 50 μL at the subcutaneous tissue of each inner thigh to establish the EAU model. 5.8. Clinical and Histopathologic Assessment On the 14th day after injection, after anesthetizing the mice with sodium pentobarbital, the anterior segment inflammation of the eyes was observed, and images were captured for scoring. The clinical scoring of uveitis in EAU mice was based on five independent criteria described in previous studies, with scores ranging from 0 to 5. On the 14th day following IRBP peptide immunization, the mice’s eyes were enucleated and fixed in 4% buffered paraformaldehyde for 1 h at room temperature. The tissues were then embedded in paraffin and sectioned into slices 4 to 6 μm thick, followed by hematoxylin and eosin (H&E) staining. The histopathological scoring of the EAU mouse ocular tissues was performed under a microscope, according to the criteria proposed by Caspi R. 5.9. Determination of MDA levels MDA is a metabolite of cellular lipid peroxidation. Fourteen days after immunization, we isolated the retinas and analyzed MDA levels. MDA assay kits were used to quantify MDA levels in retinal tissues according to the manufacturer’s instructions (Servicebio, China). The absorbance of samples was measured at a wavelength of 450 nm. We subsequently normalized the MDA levels by the total protein concentration, which was determined utilizing the BCA (Servicebio, China) assay kit. 5.10. RNA Extraction and qRT-PCR and ELISA Fresh mouse retinas were collected, and total RNA was extracted using Trizol reagent (Vazyme, China). After measuring the RNA concentration with a NanoDrop spectrophotometer (Thermo, America), RNA was reverse transcribed into cDNA using ABScript III RT Master Mix (ABclonal, China). Universal SYBR Green Fast qPCR Mix (ABclonal, China) was used for quantitative real-time PCR. The mRNA levels were normalized to β-actin, and the 2^–ΔΔCT method was applied to calculate relative mRNA expression. All primers used in this study are listed in [105]Table . 1. Gene Specific Primer. primers forward (5′-3′) reverse (5′-3′) TGFBR1 TGCTCCAAACCACAGAGTAGGC CCCAGAACACTAAGCCCATTGC ZFAS1 AGCGTTTGCTTTGTTCCC CTCCCTCGATGCCCTTCT B-actin GAAGTCCCTCACCCTCCCAA GGCATGGACGCGACCA [106]Open in a new tab On the 14th day after emulsion injection, blood was collected from the mouse eyeball and centrifuged to obtain serum. Serum IL-17A levels in the different groups were measured using the Mouse IL-17A ELISA Kit (Servicebio, China) according to the manufacturer’s instructions. 5.11. WB Analysis Mouse retinas were harvested and lysed in ice-cold lysis buffer. The samples were then sonicated and centrifuged to obtain the protein supernatant. Proteins of different molecular weights were separated using a 12.5% Bis-Tris polyacrylamide gel and transferred onto a poly(vinylidene fluoride) (PVDF) membrane. After blocking with 5% skim milk, the membrane was incubated overnight at 4 °C with the following primary antibodies: GAPDH (1:10000, HUABIO, China), TGFBR1 (1:500, ZENBIO, China), GPX4 (1:10000, HUABIO, China), xCT (1:500, ZENBIO, China), and IL-17A (1:500, HUABIO, China). Subsequently, the membrane was incubated with horseradish peroxidase (HRP)-conjugated secondary antibody for 1 h, and protein bands were detected using enhanced chemiluminescence (ECL) reagent (BIOPRIMACY, China). The absorbance of each band was measured using ImageJ 1.54 ([107]https://imagej.net/ij/). Relative protein expression was quantified as the ratio of the target band to the GAPDH band absorbance. 5.12. Optomotor The grating visual stimuli were scripted in MATLAB (version 2020b) and generated utilizing the Psychtoolbox suite. After 12 h of adaptation in the dark, the mice were placed on a platform surrounded by four screens, creating a box. These screens presented the animals with moving vertical sinusoidal gratings, displaying increasing spatial frequencies in both clockwise and counterclockwise directions (including 0.1, 0.15, 0.2, 0.25, 0.3, 0.35, 0.4, 0.45, 0.5, and 0.6 cycles/degree, with a contrast of 100%). The head movements of the mice were scored only if the angular speed of the head corresponded to that of the screen rotation. This data was monitored via a camera positioned at the top of the enclosure and assessed by an independent, blinded researcher. The highest spatial frequency at which a visually evoked response was observed was recorded as the visual acuity of the mice. 5.13. ERG ERG stimulation and recording of electrical responses were performed using the visual electrophysiological system (RetiMINER-C, China). Mice were subjected to dark adaptation for over 12 h. They were then anesthetized with intraperitoneal injection of sodium pentobarbital, following which the mice were secured on a mobile animal experimental platform. Afterward, the recording electrode, a circular platinum electrode, was positioned at the corneal limbus of both eyes. The reference electrode and ground electrode were positioned on the forehead and tail, respectively. ERG measurements were conducted under dark-adapted conditions at stimulus intensities of 3.0 cd·s/m^2, and all procedures were conducted under dim red light. The amplitudes of the a-wave and b-wave were recorded for subsequent analysis. 5.14. Statistical Analysis All statistical analyses, including heatmaps, volcano plots, WGCNA, LASSO, SVM-RFE, and immune infiltration analyses, were conducted using R version 4.1.1 ([108]https://www.r-project.org/). Additional statistical analyses were performed using GraphPad Prism version 10.0.2 (GraphPad Software, La Jolla, California). The one-sample Kolmogorov–Smirnov test was used to assess the distribution of parametric variables. For normally distributed data, comparisons between two groups were made using an independent samples t test, while one-way analysis of variance (ANOVA) was applied for multiple group comparisons. For non-normally distributed data, the Kruskal–Wallis rank-sum test or Mann–Whitney U test was employed. All results are expressed as mean ± standard deviation (SD), with p < 0.05 considered statistically significant. Each group included at least three mice, and the data represent the mean ± SD from three independent experiments. Statistical significance levels are denoted as follows: ns = not significant, * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001. Acknowledgments