Next Article in Journal
Universal or Personalized Mesenchymal Stem Cell Therapies: Impact of Age, Sex, and Biological Source
Next Article in Special Issue
Age and Sex-Related Changes in Retinal Function in the Vervet Monkey
Previous Article in Journal
Inflammasome Activation in Retinal Pigment Epithelium from Human Donors with Age-Related Macular Degeneration
Previous Article in Special Issue
Computerized Texture Analysis of Optical Coherence Tomography Angiography of Choriocapillaris in Normal Eyes of Young and Healthy Subjects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

High-Fat Diet Alters the Retinal Pigment Epithelium and Choroidal Transcriptome in the Absence of Gut Microbiota

1
Department of Ophthalmology and Visual Science, University of Chicago, Chicago, IL 60637, USA
2
Department of Medicine, University of Chicago, Chicago, IL 60637, USA
3
Animal Resources Center, University of Chicago, Chicago, IL 60637, USA
4
Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
*
Author to whom correspondence should be addressed.
Cells 2022, 11(13), 2076; https://doi.org/10.3390/cells11132076
Submission received: 25 May 2022 / Revised: 22 June 2022 / Accepted: 22 June 2022 / Published: 30 June 2022
(This article belongs to the Collection Retina in Health and Disease)

Abstract

:
Relationships between retinal disease, diet, and the gut microbiome have started to emerge. In particular, high-fat diets (HFDs) are associated with the prevalence and progression of several retinal diseases, including age-related macular degeneration (AMD) and diabetic retinopathy (DR). These effects are thought to be partly mediated by the gut microbiome, which modulates interactions between diet and host homeostasis. Nevertheless, the effects of HFDs on the retina and adjacent retinal pigment epithelium (RPE) and choroid at the transcriptional level, independent of gut microbiota, are not well-understood. In this study, we performed the high-throughput RNA-sequencing of germ-free (GF) mice to explore the transcriptional changes induced by HFD in the RPE/choroid. After filtering and cleaning the data, 649 differentially expressed genes (DEGs) were identified, with 616 genes transcriptionally upregulated and 33 genes downregulated by HFD compared to a normal diet (ND). Enrichment analysis for gene ontology (GO) using the DEGs was performed to analyze over-represented biological processes in the RPE/choroid of GF-HFD mice relative to GF-ND mice. GO analysis revealed the upregulation of processes related to angiogenesis, immune response, and the inflammatory response. Additionally, molecular functions that were altered involved extracellular matrix (ECM) binding, ECM structural constituents, and heparin binding. This study demonstrates novel data showing that HFDs can alter RPE/choroid tissue transcription in the absence of the gut microbiome.

1. Introduction

Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in industrialized countries, affecting 196 million individuals worldwide in 2020 [1]. In recent years, growing evidence has indicated that diet and nutrition may play important roles in the pathogenesis of retinal diseases, including AMD and diabetic retinopathy (DR) [2,3,4,5]. Notably, several studies have found significant associations between high-fat diets (HFDs) with increased prevalence and severity of AMD [6,7,8,9]. The retina is extremely metabolically active and requires a unique lipid composition for visual processing, making this ocular tissue highly susceptible to oxidative stress and metabolic fluctuations [10,11,12,13]. Maintaining retinal homeostasis relies heavily on the adjacent retinal pigment epithelium (RPE) and choroid for barrier protection, nutrient supply, lipid transport, and waste clearance [14]. Consequently, RPE/choroid pathology often precedes signs of retinal dysfunction and may be particularly sensitive to physiologic changes induced by HFDs [14,15,16]. Several mechanisms have been proposed as to how HFDs can reproduce or accelerate retinal disease: fatty acid signaling, metabolic dysregulation, vascular remodeling, and persistent inflammation [15,17,18]. Crucially, HFDs may disrupt the complex equilibrium between the RPE/choroid and retina, subsequently inducing hallmark features of AMD, including the deposition of lipids and proteins beneath the RPE, the thickening of Bruch’s membrane (BM), and choroidal neovascularization (CNV) [16,19,20,21,22,23].
Emerging literature suggests that HFD-induced features of AMD may partially be facilitated by the gut microbiome. Trillions of gut microbes function in diverse ways to impact health homeostasis, including the regulation of inflammation and metabolic signaling [24]. Beyond the gastrointestinal tract, the effects of gut microbiota–host interplay are observed in distant anatomic organs, including the heart, lungs, and brain [25,26,27]. Although research on retinal disease and the gut microbiome is in its nascent stage, several studies have identified strong associations with the intestinal microbiome and AMD [28,29,30]. For example, Andriessen et al. demonstrated that HFDs can induce gut dysbiosis, which in turn exacerbates laser-induced CNV in a model of AMD [31]. Other studies have reported HFD-induced changes to gut microbiota composition and function, suggesting diet and gut microbiota are closely related [32,33].
Considering this, a connection may exist between the gut microbiome, HFDs, and the retina, as well as the RPE/choroid. We recently demonstrated that retinal transcription in germ-free (GF) mice can be altered by an 8-week course of HFD [34]. However, whether and how HFDs affect the adjacent RPE/choroid, which is essential for retinal homeostasis, independent of the gut microbiome, is still unknown. In this study, we investigated changes in RPE/choroid transcriptome induced by HFDs using high-throughput RNA-sequencing in GF mice. We used the framework of AMD pathogenesis to highlight key genes and pathways in the RPE/choroid corresponding to AMD, such as inflammation and angiogenesis, which were significantly altered in response to HFDs in the absence of the microbiome.

2. Materials and Methods

2.1. Animals and Diets

Mouse experiments were approved by the University of Chicago Institutional Animal Care and Use Committee and adhered to research guidelines established by the Association for Research in Vision and Ophthalmology (ARVO). Germ-free (GF) C57BL/6 adult, male mice were bred and housed in the Gnotobiotic Research Animal Facility at the University of Chicago. Starting at 7 weeks of age, GF mice were placed on a normal diet (ND) or high-fat diet (HFD) for 8 consecutive weeks. The HFD (Teklad Custom Diet TD.130135) consisted of 44.9% saturated fat, 14.8% protein, and 40.3% carbohydrate by caloric content (Envigo, Indianapolis, IN, USA). Of the 40.3% carbohydrates, approximately 21% came from sucrose. The normal diet (ND) consisted of 12% fat, 22% protein, and 66% carbohydrate, with an estimated 0.3% derived from sucrose (Envigo, Indianapolis, IN, USA). GF mice lived under a 12-h light cycle, and environmental conditions such as temperature and humidity were in accordance with The Guide for the Care and Use of Laboratory Animals, 8th edition [35]. At 15 weeks of age, mice were euthanized with carbon dioxide and cervical dislocation. RPE/choroid samples were placed on ice and processed for RNA-sequencing.

2.2. Sterility Monitoring

To ensure sterility, GF mice were housed in positive-pressure incubators and fed diets that had been irradiated and autoclaved (250 °F for 30 min). Germ-free status was evaluated as described previously [36]. Briefly, fecal samples were collected every week and cultured aerobically at 37 °C and 42 °C, as well as anaerobically at 37 °C. Cultures were assessed after 1, 2, 3, and 5 days. No positive cultures were identified throughout the study. Additionally, DNA extraction and quantitative real-time polymerase chain reaction (RT-PCR) were performed on fecal samples to screen for contamination using bacterial primers for the 16 S RNA-encoding gene (IDT, 8 F was 5′-AGA GTT TGA TCC TGG CTC AG-3′, and 338 R was 5′-TGC TGC CTC CCG TAG GAG T-3′).

2.3. RNA Extraction

Eyes were enucleated, and RPE/choroid tissue was isolated on ice with all equipment, surfaces, and tubes treated with RNase decontamination solution (Thermo Fisher Scientific, Waltham, MA, USA) prior to use. Samples were stored at −80 °C in RNAlater solution (Thermo Fisher Scientific, Waltham, MA, USA) until RNA was extracted using the RNeasy kit from Qiagen (Qiagen, Hilden, Germany). Concentrations were quantified using a Nanodrop 2000c (Thermo Fisher Scientific, Waltham, MA, USA) prior to sequencing.

2.4. RNA Sequencing

RNA from eight samples from GF-ND (n = 4) and GF-HFD (n = 4) was used for analysis. A Bioanalyzer at the University of Chicago Genomics Core was used to determine that RNA quality met appropriate RNA integrity numbers (RIN). Next, cDNA libraries were constructed using TruSeq RNA Sample Prep kits (Illumina, San Diego, CA, USA) to generate 100bp paired-end reads, which were indexed for multiplexing and sequenced using PE100bp on the NovaSeq 6000 System (Illumina, San Diego, CA, USA). Data was provided in FASTQ format and analyzed in R.

2.5. Statistical Analysis

The secondary analysis of RNA-sequencing data was conducted in Globus Genomics, an enhanced, cloud-based analytical platform that provides Next-Generation Sequence analysis tools and workflow capabilities. Tools such as STAR [37], featureCounts [38], and Limma [39,40] were run from within the Globus Genomics platform. We used STAR (version 2.4.2 a, Stanford University, Stanford, CA, USA) aligner default parameters to align the RNA-sequencing reads to the reference mouse genome (GRCm38) for all samples. A raw gene expression count matrix was generated with featureCounts (version subread-1.4.6-p1), and gene annotation was acquired from Gencode vM23 [41]. The STAR default parameter for the maximum mismatches was 10, which was optimized based on mammalian genomes and recent RNA-sequencing data.
Genes with low expression (count-per-million < 10) were filtered using edgeR [42,43]. Significant DEGs estimated by Limma with an adjusted p-value < 0.05 and LogFC > 1.5 were selected for further downstream analysis. Enrichment analysis in Lynx suite took both the upregulated and downregulated DEGs in GF and extracted the over-represented gene ontology functional classification (molecular functions, biological processes, and cellular components) [44]. The enrichment gene ratio was measured using the number of input DEGs that mapped to the pathway divided by the total number of genes in that pathway. A list of all DEGs is available in Supplementary Table S1.

3. Results

3.1. High-Fat Diet Is Associated with Changes in the Rpe/Choroid Transcriptome

In order to study the effects of a HFD on the RPE/choroid in the absence of the microbiome, we performed high-throughput RNA-sequencing (RNA-seq) on RPE/choroid tissue in GF-ND and GF-HFD mice (n = 4 per group). After the removal of pseudogenes and uncharacterized cDNA using the National Center for Biotechnology Information (NCBI) database and applying a false discovery rate (FDR) < 0.05 with an absolute LogFC > 1.5 threshold, 649 differentially expressed genes (DEGs) were identified for downstream analysis. Of the 649 DEGs identified, the majority of the genes were transcriptionally upregulated by HFD, with only 33 genes downregulated. A list of all DEGs is available in Supplementary Table S1. The top 30 overexpressed genes are shown in Table 1, which include genes involved in natural killer (NK) T-cell functioning, such as Natural killer cell receptor 2B4 (Cd244a) and Natural cytotoxicity triggering receptor 1 (Ncr1), as well as inflammatory markers, including Tumor necrosis factor receptor superfamily member 13B (Tnfrsf13b), C-C motif chemokine ligand 19 (Ccl19), and Prostaglandin-endoperoxide synthase 2 (Ptgs2 or Cox-2). The top 30 overexpressed genes also included mediators of endothelial adhesion and vessel permeability, notably Selectin E (Sele) and Lysophosphatidic acid receptor 3 (Lpar3). Additionally, a heatmap was generated using an FDR < 0.01 and absolute LogFC > 1.5 threshold to demonstrate the hierarchical clustering of the DEGs by experimental groups, indicating that the changes observed in RPE/choroid transcription likely could be attributed to HFD in the absence of microbiota (Figure 1).

3.2. High-Fat Diet Upregulates Multiple Biological Processes and Genes Related to Inflammation and Angiogenesis

After identifying 649 DEGs, we performed enrichment analysis using the Lynx suite for gene ontology (GO) in order to identify over-represented biological processes in the RPE/choroid of GF-HFD mice relative to GF-ND mice [44]. This unbiased approach identified angiogenesis as the most significantly upregulated biological process (adjusted p-value = 5.51 × 10−10) due to HFD intervention (Table 2, Figure 2). Among the angiogenic genes affected were Vascular endothelial growth factor C (Vegfc); angiopoietin genes Angpt1, Angpt2, and Angptl2; their respective receptors Tie1 and Tie2 (Tek); and platelet derived growth factors Pdgfc and Pdgfd, which together are involved in angiogenesis, maturation, and vascular remodeling. Additionally, the GF-HFD group demonstrated upregulation of GO biological processes related to the inflammatory response and immune response, such as Complement factor H (Cfh) and TNF superfamily genes Tnfaip2, Tnfrsf11b, Tnfrsf13b, Tnfrsf19, Tnfrsf1b, Tnfsf10, and Tnfsf14.
Our analysis also indicated significant alterations induced by HFD in molecular processes such as extracellular matrix (ECM) binding, ECM structural constituents, and heparin binding (Table 3, Figure 3). As the RPE/choroid has multiple functions in addition to providing nutrient exchange to the retina, the connective tissue cells types such as fibroblasts, melanocytes, pericytes, and immune cells are active players in maintaining homeostasis [45]. Genes involved in ECM interactions that were upregulated include Apolipoprotein E (Apoe); the matrix metalloproteinase Adamts9; collagens Col2a1, Col8a1, and Col10a1; and fibronectins such as Fbln1 and Fbln5.

4. Discussion

Diet and nutrition are significant risk factors in retinal disease pathobiology, including AMD and DR [46,47]. Specifically, several studies have emphasized an association between HFDs and the increased prevalence and progression of AMD [8,9]. Using animal models of neovascular AMD, our team previously has demonstrated that HFDs can increase laser-induced CNV lesion size, vascular leakage, and the formation of sub-RPE deposits [19]. Other studies, too, have recapitulated AMD-like features in mice fed HFDs [22,23]. Given the role of the gut microbiome in immunomodulation, nutrition, and energy metabolism, there is a growing body of literature connecting the gut microbiome and AMD [2,29,30,48]. Gut microbiota may be key mediators of HFDs in retinal disease, whereby HFDs induce gut dysbiosis, increase intestinal permeability, and induce chronic inflammation in AMD models irrespective of total energy intake [31]. We previously studied HFD-induced changes in retinal transcription independent of gut microbiota; however, its impact at the level of the RPE/choroid is unknown [34].
In this study, we sought to uncouple the impact of HFDs on RPE/choroidal biology from the gut microbiome. In both dry and wet AMD, pathological changes typically occur first in the supporting tissue of the RPE/choroid before damage is observed to underlying retinal cells [14]. In addition, there is evidence in the RPE/choroid of lipid accumulation that contain fats derived exclusively from diet, which helps corroborate the role of diet in RPE/choroid biology [49]. To the best of our knowledge, this is the first study to explore the transcriptional changes induced by HFDs in the RPE/choroid in the absence of the gut microbiome by using germ-free mice. After analyzing and filtering the data, we identified 649 DEGs and performed GO enrichment analysis to highlight changes in important biological pathways.

4.1. High-Fat Diet Affects Gene Expression in Angiogenic Pathways in Germ-Free Mice

Angiogenesis is a hallmark feature of wet AMD, which may account for up to 90% of cases of AMD-related severe vision loss [50]. During this process, abnormal blood vessels from the choroid infiltrate the sub-RPE space, resulting in vascular leakage, bleeding, scarring, and damage to the macula [51]. Consequently, therapeutics that limit angiogenesis are widely-used to delay AMD progression, such as anti-VEGF therapy [52]. In the absence of the microbiome, HFD upregulated pathways are involved in angiogenesis, as well as in its positive regulation (Figure 1). In addition to Vegfc, HFD significantly elevated the angiopoietin genes Angpt1, Angpt2, and Angptl2. Angiopoietins are selective growth factors for vascular endothelium. The ANG signaling pathway is heavily involved in vascular development in both the choroid and retina [53]. Specifically, choroidal neovascularization occurs when Vegf and Angpt2 are elevated in conjunction with the disruption of Bruch’s membrane and the RPE [51]. This relationship is substantiated by other studies showing that Angpt2 is necessary for ischemia-induced neovascularization in mice lacking Angpt2 [53]. Recently, the FDA approved faricimab, a biologic targeting both VEGF and ANGPT2, in the treatment of AMD and DR. We also detected the concomitant upregulation of the respective angiopoietin receptors, Tie1 and Tek (Tie2), which are tyrosine kinases that transduce the signaling pathways for vessel maturation [54]. VEGF and ANG drive complementary angiogenic pathways, with VEGF inducing early vessel sprouting and growth, whereas ANG1 mediates vascular remodeling, maturation, and protection [55,56]. Similarly, angiopoietin-like 2 protein (Angptl2) is involved in angiogenesis and vasculogenesis [57]. Studies have found that excess ANGPTL2 signaling results in chronic inflammation and irreversible tissue remodeling [58]. Consequently, VEGF-inhibition may be only effective so long as neovascularization is in its nascent stage, which may explain why an estimated nearly 50% of patients with neovascular eye disease do not respond to anti-VEGF treatments such as bevacizumab or ranibizumab [53,59,60,61]. Clinical trials have begun that either target ANG2 alone or concurrently with VEGF for treatment of AMD [62,63].
In addition to the ANG pathway, we found that HFDs elevated the transcription of Platelet-derived growth factors C and D (Pdgfc and Pdgfd), which are angiogenic factors that play critical roles in several ocular neovascular diseases, including AMD [64]. PDGF levels are normally low or undetectable, but they become elevated in numerous vascular and cardiovascular pathologies [65]. Crucially, PDGF ligands help recruit and maintain choroidal fibroblasts and pericytes, which serve as scaffolds for vascular endothelium. In mouse models of wet AMD, both Pdgfc and Pdgfd expression is upregulated. PDGFC also positively regulates other pro-angiogenic factors, such as VEGF and PDGFB [64]. PDGFs are targets for AMD treatment both in preclinical trials and in clinical trials, where dual anti-PDGF and anti-VEGF therapy has demonstrated superior efficacy to anti-VEGF monotherapy [66,67].
Additional genes involved in angiogenesis that were transcriptionally upregulated with HFD included Lpar3 and Tnfaip2, as well as Cyp1b1 and Cxcr3. Previous studies have linked Cxcr3 dysregulation with wet AMD, though the nature of this pathway is not clearly understood [68]. Cyp1b1 is another gene required for the neovascular response to ischemic retinopathy as it plays roles in angiogenesis and capillary morphogenesis [69,70].

4.2. High-Fat Diet Alters Gene Expression Involved in Inflammatory and Immune Response Pathways in Germ-Free Mice

Inflammation is present during every stage of AMD pathology, beginning with drusen formation; drusen are deposits of cellular debris that serve as nodes for inflammatory processes [71]. Inflammatory signaling molecules, macrophages, and activated resident microglial cells localize to sites of drusen deposits, Bruch’s membrane degeneration, and CNV [72,73]. Serum levels of inflammatory markers such as C-reactive protein (CRP), as well as retinal autoantibodies, are significantly associated with AMD advancement [74,75]. As a result, there are a host of preclinical and clinical trials using anti-inflammatory agents to treat AMD [72,76]. Current literature supports the notion that HFDs can promote the pathogenesis of diseases in multiple organ sites by means of inducing chronic, low-grade inflammation and accelerating age-related cellular processes [77]. Part of this “inflammaging” is thought to be mediated by gut microbiota that can release inflammatory products, signal to other organ sites, and regulate circadian rhythm [78,79].
In this study, we found that HFDs in the absence of the microbiome altered the gene transcription of inflammatory pathways in the RPE/choroid. One of the top 10 biological processes upregulated by HFDs was the inflammatory response (Figure 1). Transcriptional upregulation was observed in multiple genes involved in the chemoattraction, activation, and functioning of natural killer T (NKT) cells, such as Cd244a, Cd48, Cxcl10, Granzyme A (Gzma), Perforin (Prf1), and Il12b. NKT cells act at the interface of innate and adaptive immunity, acting rapidly to immunogenic stimuli and possessing powerful cytotoxic capabilities [80]. NKT cell activity has been associated with a number of neurodegenerative/neuroinflammatory diseases such as Alzheimer’s and multiple sclerosis [81,82]. Immunohistological studies have identified several immune cell-types in the subretinal space in AMD patients, including natural killer lymphocytes, suggesting that NKT cells may play a role in its pathogenesis [83]. NKT cells are reported to accumulate during laser-induced CNV. In support of this, two different NKT-deficient mouse strains demonstrated decreased CNV severity and Vegf expression. Furthermore, co-culturing RPE with NKT cells confirmed the ability of NKT cells to produce VEGF, potentially driving further angiogenesis [84]. Haplotypes of killer cell immunoglobulin-like receptors (KIRs) found on NKT cells are associated with AMD in certain populations [85].
Cd244a, a risk factor for inflammatory diseases such as rheumatoid arthritis, was the most highly upregulated gene in our data set [86]. Cd244a is a cell-surface receptor on NKT cells that mediates their expansion, activation, and cytotoxicity [87]. In addition, its principal binding partner, Cd48, was also upregulated in GF mice fed a HFD. Cd48 is also involved in other immunoregulatory functions, including immune cell adhesion and the co-stimulation of antigen-presenting cells [88]. Gzma and Prf1, which encode proteins underlying the main mechanisms by which NKT cells induce cytotoxicity, were also upregulated by HFDs [89]. The additional upregulated genes included Cxcl10, a activator and recruiter of NKT cells, and Il12b, a cytokine that serves as a growth factor for NKT cells, enhances their cytolytic activity, and induces interferon-gamma production [90,91]. Several studies have identified IL-12 as a potential driver of chronic inflammation in the context of AMD [92,93]. In addition to NKT cells, T cell-related genes such as Ccl19, Ccl22, and Cd28 were upregulated, which are involved in activation and chemoattraction [94].
Several other inflammatory genes upregulated included Ccl4, which has been shown to mediate inflammation in response to retinal damage, as well as prostaglandin and TNF families [95]. Tumor necrosis factors (TNF), particularly TNF-alpha, are pro-inflammatory cytokines whose signaling is thought to play a role in the neovascularization of the RPE/choroid and AMD pathogenesis [96,97,98]. Many members of the TNF superfamily were upregulated in the GF-HFD group, including Tnfaip2, Tnfrsf11b, Tnfrsf13b, Tnfrsf1b, Tnfsf10, and Tnfsf14. Genetic variations of Tnfsf10 in particular have been associated with AMD [99]. Anti-TNF therapies may be effective in treating AMD and reducing the frequency of anti-VEGF therapy [100,101]. Finally, we noted the elevation of Prostaglandins D, E, and I (Ptgds, Ptges, and Ptgis, respectively), along with Cox-1 and Cox-2 (Ptgs1 and Ptgs2), which synthesize prostaglandins. Prostaglandins regulate vascular permeability and vasodilation and are induced in the inflammatory response. Elevated transcripts of these genes may contribute to the bridge between inflammation and aberrant blood flow and vascular leakage; however, the role of prostaglandins in the retina and RPE/choroid is unclear, with only sparse evidence suggesting prostaglandins are implicated in the pathogenesis of AMD and DR [102]. In a rat model of CNV, ketorolac, a type of anti-inflammatory NSAID that inhibits COX enzymes, was shown to significantly reduce CNV leakage and vascular budding [103].

4.3. High-Fat Diet Affects Gene Expression Involved in the Complement System

One specific immune pathway that is highly implicated in AMD is the complement system, with existing reviews that detail how complement activation may influence AMD pathogenesis [104]. The complement system is important for the removal of immune complexes, apoptotic cells, and adaptive immunity. With over 40 proteins involved in the cascade, alterations at multiple steps can impart significant differences in the overall inflammatory response. In our transcriptional analysis, we identified several DEGs involved in the complement cascade, such as C1qb, C2, C4b, and Cfh. CFH was the first complement protein associated with AMD in genetic studies and can be directly synthesized by RPE cells [105,106,107]. The CFH Y402H polymorphism is thought to increase AMD risk by up to sevenfold [108]. CFH binds to polyanionic moieties, in particular sulfated glycosaminoglcyans (GAGs). The CFH Y402H polymorphism is thought to alter the binding property of CFH to GAGs, such as heparan sulfate [109]. Interestingly, one of the top 10 molecular functions upregulated by HFD was heparin binding, which is a well-established mechanistic function of CFH (Figure 2) [110]. In addition to CFH, variations in C2 have been linked to differential risk for AMD by several groups [111,112]. We also identified elevated expression of C4b, whose variants may also play a role in AMD [99].
While the precise role of different components of the complement system in AMD pathology is unclear, it appears that the RPE/choroid is an important hub for complement activity. The membrane attack complex (MAC) is the endproduct of the complement cascade, and studies report MAC localization to the choriocapillaris of the choroid, as opposed to RPE or retinal tissue [113,114]. After comparing the presence of MAC in aged human tissues across multiple organs, Chirco et al. determined that the MAC selectively accumulates in the choroid, which may partly explain the tight association between AMD and the complement system [115]. In vitro studies confirm that direct exposure of the MAC to choroidal endothelial cells promoted death and upregulated pro-angiogenic factors, ultimately leading to CNV [116]. HFDs have been shown, in addition to promoting chronic inflammation, to specifically induce complement activation in animal models and upregulate levels in the blood [117]. One study found that aged Cfh+/− mice fed a high-fat, high-cholesterol diet developed features of AMD, including complement dysregulation, sub-RPE deposits, and impaired visual function changes resulting from changes in RPE morphology [118]. Further studies looking into the interactions between age, diet, and complement dysregulation are required. A number of targets in the complement cascade are being targeted for the potential therapeutic benefit of AMD [119,120].

4.4. Additional Genes and Pathways Are Differentially Represented by High-Fat Diet in Germ-Free Mice

Several other pathways that were affected by HFD were related to ECM interactions and RPE function. Changes in the ECM, including sub-RPE deposits and the thickening of Bruch’s membrane, are often the initial clinical symptoms of AMD. These deposits consist of many different substances, including ECM proteins, complement, lipids, and other cellular debris. GF-HFD mice demonstrated elevated transcriptional levels in various ECM components, such as Apoe, Adamts9, Esm1, Col2a1, Col8a1, Col10a1, Fbln1, and Fbln5. As a key player in lipid, vitamin, and cholesterol transport, variants in APOE are found to be associated with AMD [121]. In AMD, unbalanced lipid exchange may drive RPE decline and impair the exchange of lipids across from the choroid, which may potentiate pathological changes in the eye [122]. APOE is the most abundant lipoprotein component of drusen, which can stress the RPE, hinder nutrient exchange, and serve as a focal point for generating inflammation and CNV [108,123]. Additionally, APOE has been found to interact with complement, co-localizing with the MAC in human eyes [124]. Within the apolipoprotein family, Apold1 and Apol7c also were identified DEGs in GF-HFD mice. Apold1 (Apolipoprotein L Domain Containing 1) is a protein that regulates endothelial and vascular functioning. Its expression has been found to be elevated in the RPE/choroid of AMD patients compared to controls [125].
Our analysis also identified the elevated expression of other matrix proteins, including collagens, metalloproteinases, and other secreted proteins. Of note, collagen Col8a1 and Col10a1 expression increased, with both genes having genetic associations with AMD [126]. In addition, Col2a1 previously has been linked with ocular disorders such as retinal detachment [127]. Fibulins Fbln1 and Fbln5 are differentially expressed ECM glycoproteins, and of note, one study has associated mutations in FBLN5 with AMD [128]. Adamts9 is a metalloproteinase that was identified by the AMD Gene Consortium as one of seven new loci associated with AMD [99]. Endothelial cell-specific molecule 1 (Esm1) is a secreted protein that is a highly overexpressed gene in oxygen-induced retinopathy. Esm1 modulates Vegf bioavailability and leukocyte extravastion [129]. In addition, ESM1 variants are associated with increased levels of advanced glycation products (AGEs), which are significant components of macula drusen and basal lamina deposits [130,131]. Patients fed a high-fat diet had elevations in blood levels of AGEs [130].
Given the functional relevance and spatial proximity, the RPE is thought to be the principal driver of debris accumulation and deposition in both aging and AMD eyes [108,132]. A HFD altered the transcription of multiple genes related to RPE function and the visual cycle, including Lrat, Rpe65, Rdh5, and Rdh10. These genes were also found to be upregulated in aged, human RPE [133]. LRAT is an enzyme that catalyzes vitamin A esterification into all-trans-retinyl esters, which are converted to 11-cis-retinol via RPE65 [134]. By-products of the RPE visual cycle are common constituents of lipofuscin, which are toxic to RPE cells and may impact AMD pathogenesis [135]. These by-products positively regulate LRAT expression, and thus a positive feedback mechanism has been suggested that links visual cycle by-product accumulation with the RPE visual cycle [133]. HFD-fed mice tend to have thicker Bruch’s membranes, RPE dysfunction, and greater accumulation of basal laminar deposits, suggesting HFDs may alter the ECM in AMD pathobiology [22,23].
Across HFD research, the composition of HFDs in animal studies ranges greatly, spanning from 8.3–80% fat content [136]. The selection of HFD, therefore, deserves further discussion. The HFD used in these experiments approximates a “Western diet”, which, according to reports from the National Health and Nutrition Examination Survey (NHANES), is around 35% fat by energy content and 23% by simple carbohydrates such as sucrose [137,138]. Whereas diet duration did not vary, the composition was chosen to parallel human dietary patterns and thus included a high sucrose component of around 21% by energy content. Additionally, we wanted to examine how our results compared to other HFD RPE/choroid transcription data in mice with intact gut microbiota. In this context, Andriessen et al. detected elevated levels of mRNA of Il6, Tnf, and Vegfa in the choroids of mice fed HFD compared to mice fed a regular-chow diet, suggesting an increase in endotoxemia and systemic inflammation [31]. Separately, Zhang and colleagues demonstrated increased Vegfa, as well as a non-significant rise in ApoE mRNA in the RPE/retina in mice fed HFD [22]. That Andriessen et al. and Zhang et al. demonstrated elevated Vegfa expression, in conjunction with our GF model of elevated Vegfc, suggests the HFD may impact angiogenesis with or without microbiota [22,31]. Additionally, our experiments with GF mice detected the relative overexpression of numerous Tnf superfamily genes and ApoE, which were also implicated in these other studies. Nevertheless, due to study design differences with respect to HFD composition and the mode of gut microbiota depletion, it is difficult to directly compare these results.
Within our model, when contrasting these results with previously identified changes in retinal transcription of GF mice, HFDs induced a greater shift in transcription in the RPE/choroid, with little overlap in the genes affected. In fact, the only two overlapping genes were Ms4a6b (membrane spanning 4-domains A6A) and Hbb-bs (hemoglobin subunit beta), both of which were upregulated in the retina and in the RPE/choroid [34]. These transcriptional differences may partly be due to the distinct functions of the retina and RPE/choroid within the visual system, with the RPE/choroid providing barrier protection, nutrients, substrates, and waste clearance for the neural retina [14]. Additionally, the separate cell types and lack of vascular barrier of the choroid compared to the immune-privileged retina could further contribute to discrepancies in transcriptional effects caused by systemic, metabolic changes induced by diet in the absence of gut microbiome. As changes in RPE/choroid biology often precede overt signs of retinal dysfunction, these findings also suggest tissue-specific sensitivity and a response to dietary changes over a fixed duration. One recent study found that mice fed a HFD with 45% fat for 12 months demonstrated no retinal function abnormalities as measured by ERG; however, evidence of lipid deposition, RPE distortion, and endothelial vacuolization was present in the HFD-fed group [139]. Interestingly, an unbiased pathway analysis of transcriptional changes in the retina and RPE/choroid in GF-HFD mice both point toward alterations in angiogenic and inflammatory pathways in response to the dietary intervention. Overall, however, these results indicate that the diet-microbiome-transcriptome interactions could be distinct in the RPE/choroid compared to the retina. Because these experiments were performed using GF mice, the transcriptional pathways affected by HFD in the RPE/choroid could either be attenuated or exacerbated by the presence of the gut microbiome. Gut microbiota can significantly change dietary metabolism and the resultant small molecule profiles of the host organism, potentially influencing the transcriptional responses of the retina and RPE/choroid. Likewise, HFDs have the capacity to alter the gut microbiome, a process that itself may affect retinal health and homeostasis. As a result, the microbiome-dependent and microbiome-independent effects of HFD on the RPE/choroidal transcriptome are complex and require further investigation.
While the pathobiology between HFDs and retinal disease is studied mostly in rodent models, its connection with human pathways and disease still holds biological plausibility, though proof of causality is absent. Nevertheless, there are numerous epidemiologic associations of AMD with high-fat or Western diets, with some reporting up to a threefold greater incidence of late AMD [9,140]. In addition to low-grade inflammation caused by HFDs, its potential role in aberrant lipid homeostasis may further contribute to AMD pathology, especially given that about 40% of drusen is composed of lipid-containing particles [141]. The genetic components of lipid metabolism and transport, such as APOE alleles, along with serum lipid levels, have also been identified as differentiators for AMD risk [142,143]. Beyond AMD, saturated fatty acid intake has been linked with increased prevalence and severity of DR, with some studies reporting an odds ratio of 2.37 [5,144]. In rhesus monkeys, which are more biologically similar to humans, a high-fat Western diet for 2–4 years resulted in retinal changes characteristic of human DR, including venous engorgement, macular exudates, and hemorrhages [145]. Further studies are warranted to bridge HFD animal studies with human manifestations of AMD and DR.

5. Conclusions and Limitations

This study presents data that suggests diet can impact the RPE/choroid transcriptome in the absence of gut microbiota. We use the framework of AMD pathogenesis to highlight gene expression and biological pathways that contrast greatly between GF-HFD and GF-ND mice. In particular, HFDs altered the transcription of genes involved in angiogenesis, inflammation, complement, and RPE function. As an unbiased exploration of the RPE/choroid transcriptome, the major limitation of this study is its reliance on RNA-sequencing. Future studies should include the quantitative PCR of specific genes, protein expression/proteomics, and functional assays in order to better elucidate the putative role of HFDs in RPE/choroid biology. In this current investigation, we did not address the effects of HFD-induced changes in GF models of AMD; future studies, including laser-induced CNV and other AMD animal models and aging studies, should be directed to assess potential relationships between HFDs and the pathogenesis of retinal disease, as well as to explore the dietary effects in the absence of gut microbiota. Additionally, emerging evidence suggests a more granular approach towards dietary fat and retinal disease may be warranted, as specific types of fat, such as saturated fats and oleic acid, may confer different risk profiles for AMD [6,146,147,148].
AMD is a multifactorial, vision-threatening disease whose prevalence is expected to increase dramatically as other factors, including obesity, diabetes, and average population age, continue to rise [1,149]. Diet is one of the primary modifiable risk factors for AMD progression. However, interventions targeting AMD through diet are rare. Current examples, such as AREDS nutritional supplementation, have limited efficacy and are found to reduce the risk of AMD progression in only certain sub-populations [150,151]. Therefore, investigating the complex interactions between diet, the gut microbiome, and retinal health may potentially unlock new insights for AMD prevention, as well as develop the foundation for interventions that are minimally invasive and cost-effective.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cells11132076/s1, Table S1: provides all 649 identified DEGs.

Author Contributions

Conceptualization and methodology was carried out by D.S. (Dimitra Skondra) and E.B.C. Bioinformatics and the formal validation of data were performed by B.X., J.X., M.D. and D.S. (Dinanath Sulakhe). Experimental procedures were carried out by B.T. and M.S. Original manuscript draft preparation was done by J.X., B.X. and D.D., with review and editing by D.S. (Dimitra Skondra). Funding acquisition was from D.S. (Dimitra Skondra), with contributions by E.B.C., S.M.H. and J.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by BrightFocus Foundation “Role of high fat diet and gut microbiome in macular degeneration” (D.S. (Dimitra Skondra), M2018042), NIDDK P30 (E.B.C., DK42086), The University of Chicago Women’s Board (Dimitra Skondra), FORE-I Foundation (Dimitra Skondra) and the Illinois Society for the Prevention of Blindness (D.S. (Dimitra Skondra), FP067271-01-PR and J.X., FP105447).

Institutional Review Board Statement

All animal experiments were performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health and approved by University of Chicago IACUC (#72557).

Informed Consent Statement

Not applicable.

Data Availability Statement

The complete dataset of identified DEGs is available in Supplementary Table S1.

Acknowledgments

We thank the University of Chicago Core Genomics team, especially Pieter Faber and Mikayla Marchuk, for their assistance with RNA sequencing; the University of Chicago Gnotobiotic Research Animal Facility (GRAF) staff, especially Donna Arvans, for care of the germ-free animals; and the members of the Chang lab, including Candace Cham, Xiaorong Zhu, Jason Koval, and Amal Kambal.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Wong, W.L.; Su, X.; Li, X.; Cheung, C.M.G.; Klein, R.; Cheng, C.-Y.; Wong, T.Y. Global Prevalence of Age-Related Macular Degeneration and Disease Burden Projection for 2020 and 2040: A Systematic Review and Meta-Analysis. Lancet Glob. Health 2014, 2, e106–e116. [Google Scholar] [CrossRef] [Green Version]
  2. Rinninella, E.; Mele, M.C.; Merendino, N.; Cintoni, M.; Anselmi, G.; Caporossi, A.; Gasbarrini, A.; Minnella, A.M. The Role of Diet, Micronutrients and the Gut Microbiota in Age-Related Macular Degeneration: New Perspectives from the Gut–Retina Axis. Nutrients 2018, 10, 1677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Weikel, K.A.; Chiu, C.-J.; Taylor, A. Nutritional Modulation of Age-Related Macular Degeneration. Mol. Asp. Med. 2012, 33, 318–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Chapman, N.A.; Jacobs, R.J.; Braakhuis, A.J. Role of Diet and Food Intake in Age-Related Macular Degeneration: A Systematic Review. Clin. Exp. Ophthalmol. 2019, 47, 106–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Cundiff, D.K.; Nigg, C.R. Diet and Diabetic Retinopathy: Insights From the Diabetes Control and Complications Trial (DCCT). MedGenMed 2005, 7, 3. [Google Scholar]
  6. Parekh, N.; Voland, R.P.; Moeller, S.M.; Blodi, B.A.; Ritenbaugh, C.; Chappell, R.J.; Wallace, R.B.; Mares, J.A. CAREDS Research Study Group. Association between Dietary Fat Intake and Age-Related Macular Degeneration in the Carotenoids in Age-Related Eye Disease Study (CAREDS): An Ancillary Study of the Women’s Health Initiative. Arch. Ophthalmol. 2009, 127, 1483–1493. [Google Scholar] [CrossRef] [Green Version]
  7. Chiu, C.-J.; Chang, M.-L.; Zhang, F.F.; Li, T.; Gensler, G.; Schleicher, M.; Taylor, A. The Relationship of Major American Dietary Patterns to Age-Related Macular Degeneration. Am. J. Ophthalmol. 2014, 158, 118–127.e1. [Google Scholar] [CrossRef] [Green Version]
  8. Cho, E.; Hung, S.; Willett, W.C.; Spiegelman, D.; Rimm, E.B.; Seddon, J.M.; Colditz, G.A.; Hankinson, S.E. Prospective Study of Dietary Fat and the Risk of Age-Related Macular Degeneration. Am. J. Clin. Nutr. 2001, 73, 209–218. [Google Scholar] [CrossRef]
  9. Seddon, J.M.; Cote, J.; Rosner, B. Progression of Age-Related Macular Degeneration. Arch. Ophthalmol. 2003, 121, 1728–1737. [Google Scholar] [CrossRef] [Green Version]
  10. Wong-Riley, M. Energy Metabolism of the Visual System. Eye Brain 2010, 2, 99–116. [Google Scholar] [CrossRef] [Green Version]
  11. Berry, K.A.Z.; Gordon, W.C.; Murphy, R.C.; Bazan, N.G. Spatial Organization of Lipids in the Human Retina and Optic Nerve by MALDI Imaging Mass Spectrometry. J. Lipid Res. 2014, 55, 504–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Duda, M.; Kawula, K.; Pawlak, A.; Sarna, T.; Wisniewska-Becker, A. EPR Studies on the Properties of Model Photoreceptor Membranes Made of Natural and Synthetic Lipids. Cell Biochem. Biophys. 2017, 75, 433–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Fu, Z.; Chen, C.T.; Cagnone, G.; Heckel, E.; Sun, Y.; Cakir, B.; Tomita, Y.; Huang, S.; Li, Q.; Britton, W.; et al. Dyslipidemia in Retinal Metabolic Disorders. EMBO Mol. Med. 2019, 11, e10473. [Google Scholar] [CrossRef] [PubMed]
  14. Bhutto, I.; Lutty, G. Understanding Age-Related Macular Degeneration (AMD): Relationships between the Photoreceptor/Retinal Pigment Epithelium/Bruch’s Membrane/Choriocapillaris Complex. Mol. Asp. Med. 2012, 33, 295–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Clarkson-Townsend, D.A.; Douglass, A.J.; Singh, A.; Allen, R.S.; Uwaifo, I.N.; Pardue, M.T. Impacts of High Fat Diet on Ocular Outcomes in Rodent Models of Visual Disease. Exp. Eye Res. 2021, 204, 108440. [Google Scholar] [CrossRef]
  16. Miceli, M.V.; Newsome, D.A.; Tate, D.J.; Sarphie, T.G. Pathologic Changes in the Retinal Pigment Epithelium and Bruch’s Membrane of Fat-Fed Atherogenic Mice. Curr. Eye Res. 2000, 20, 8–16. [Google Scholar] [CrossRef]
  17. Lee, J.-J.; Wang, P.-W.; Yang, I.-H.; Huang, H.-M.; Chang, C.-S.; Wu, C.-L.; Chuang, J.-H. High-Fat Diet Induces Toll-like Receptor 4-Dependent Macrophage/Microglial Cell Activation and Retinal Impairment. Investig. Ophthalmol. Vis. Sci. 2015, 56, 3041–3050. [Google Scholar] [CrossRef] [Green Version]
  18. Albouery, M.; Buteau, B.; Grégoire, S.; Martine, L.; Gambert, S.; Bron, A.M.; Acar, N.; Chassaing, B.; Bringer, M.-A. Impact of a High-Fat Diet on the Fatty Acid Composition of the Retina. Exp. Eye Res. 2020, 196, 108059. [Google Scholar] [CrossRef]
  19. Skondra, D.; She, H.; Zambarakji, H.J.; Connolly, E.; Michaud, N.; Chan, P.; Kim, I.K.; Gragoudas, E.S.; Miller, J.W.; Hafezi-Moghadam, A. Effects of ApoE Deficiency, Aging and High Fat Diet on Laser-Induced Choroidal Neovascularization and Bruch’s Membrane-RPE Interface Morphology. Investig. Ophthalmol. Vis. Sci. 2007, 48, 1768. [Google Scholar]
  20. Biswas, L.; Ibrahim, K.S.; Li, X.; Zhou, X.; Zeng, Z.; Craft, J.; Shu, X. Effect of a TSPO Ligand on Retinal Pigment Epithelial Cholesterol Homeostasis in High-Fat Fed Mice, Implication for Age-Related Macular Degeneration. Exp. Eye Res. 2021, 208, 108625. [Google Scholar] [CrossRef]
  21. Cousins, S.W.; Espinosa-Heidmann, D.G.; Alexandridou, A.; Sall, J.; Dubovy, S.; Csaky, K. The Role of Aging, High Fat Diet and Blue Light Exposure in an Experimental Mouse Model for Basal Laminar Deposit Formation. Exp. Eye Res. 2002, 75, 543–553. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, M.; Chu, Y.; Mowery, J.; Konkel, B.; Galli, S.; Theos, A.C.; Golestaneh, N. Pgc-1α Repression and High-Fat Diet Induce Age-Related Macular Degeneration-like Phenotypes in Mice. Dis Model. Mech. 2018, 11, dmm032698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Dithmar, S.; Sharara, N.A.; Curcio, C.A.; Le, N.-A.; Zhang, Y.; Brown, S.; Grossniklaus, H.E. Murine High-Fat Diet and Laser Photochemical Model of Basal Deposits in Bruch Membrane. Arch. Ophthalmol. 2001, 119, 1643–1649. [Google Scholar] [CrossRef] [Green Version]
  24. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Reddy, D.N. Role of the Normal Gut Microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef] [PubMed]
  25. Schroeder, B.O.; Bäckhed, F. Signals from the Gut Microbiota to Distant Organs in Physiology and Disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef]
  26. Fujimura, K.E.; Lynch, S.V. Microbiota in Allergy and Asthma and the Emerging Relationship with the Gut Microbiome. Cell Host Microbe 2015, 17, 592–602. [Google Scholar] [CrossRef] [Green Version]
  27. Mohajeri, M.H.; La Fata, G.; Steinert, R.E.; Weber, P. Relationship between the Gut Microbiome and Brain Function. Nutr. Rev. 2018, 76, 481–496. [Google Scholar] [CrossRef]
  28. Rowan, S.; Taylor, A. The Role of Microbiota in Retinal Disease. Adv. Exp. Med. Biol. 2018, 1074, 429–435. [Google Scholar] [CrossRef]
  29. Zysset-Burri, D.C.; Keller, I.; Berger, L.E.; Largiadèr, C.R.; Wittwer, M.; Wolf, S.; Zinkernagel, M.S. Associations of the Intestinal Microbiome with the Complement System in Neovascular Age-Related Macular Degeneration. NPJ Genom. Med. 2020, 5, 34. [Google Scholar] [CrossRef]
  30. Zinkernagel, M.S.; Zysset-Burri, D.C.; Keller, I.; Berger, L.E.; Leichtle, A.B.; Largiadèr, C.R.; Fiedler, G.M.; Wolf, S. Association of the Intestinal Microbiome with the Development of Neovascular Age-Related Macular Degeneration. Sci. Rep. 2017, 7, 40826. [Google Scholar] [CrossRef] [Green Version]
  31. Andriessen, E.M.; Wilson, A.M.; Mawambo, G.; Dejda, A.; Miloudi, K.; Sennlaub, F.; Sapieha, P. Gut Microbiota Influences Pathological Angiogenesis in Obesity-driven Choroidal Neovascularization. EMBO Mol. Med. 2016, 8, 1366–1379. [Google Scholar] [CrossRef] [PubMed]
  32. Murphy, E.A.; Velazquez, K.T.; Herbert, K.M. Influence of High-Fat-Diet on Gut Microbiota: A Driving Force for Chronic Disease Risk. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 515–520. [Google Scholar] [CrossRef] [PubMed]
  33. Xiao, L.; Sonne, S.B.; Feng, Q.; Chen, N.; Xia, Z.; Li, X.; Fang, Z.; Zhang, D.; Fjære, E.; Midtbø, L.K.; et al. High-Fat Feeding Rather than Obesity Drives Taxonomical and Functional Changes in the Gut Microbiota in Mice. Microbiome 2017, 5, 43. [Google Scholar] [CrossRef] [PubMed]
  34. Dao, D.; Xie, B.; Nadeem, U.; Xiao, J.; Movahedan, A.; D’Souza, M.; Leone, V.; Hariprasad, S.M.; Chang, E.B.; Sulakhe, D.; et al. High-Fat Diet Alters the Retinal Transcriptome in the Absence of Gut Microbiota. Cells 2021, 10, 2119. [Google Scholar] [CrossRef]
  35. National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. The National Academies Collection: Reports funded by National Institutes of Health. In Guide for the Care and Use of Laboratory Animals, 8th ed.; National Academies Press (US): Washington, DC, USA, 2011; ISBN 978-0-309-15400-0. [Google Scholar]
  36. Theriault, B.; Wang, Y.; Chen, L.; Vest, A.; Bartman, C.; Alegre, M.-L. Long-Term Maintenance of Sterility After Skin Transplantation in Germ-Free Mice. Transplant. Direct 2015, 1, e28. [Google Scholar] [CrossRef]
  37. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast Universal RNA-Seq Aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
  38. Liao, Y.; Smyth, G.K.; Shi, W. FeatureCounts: An Efficient General Purpose Program for Assigning Sequence Reads to Genomic Features. Bioinformatics 2014, 30, 923–930. [Google Scholar] [CrossRef] [Green Version]
  39. Ritchie, M.E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C.W.; Shi, W.; Smyth, G.K. Limma Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015, 43, e47. [Google Scholar] [CrossRef]
  40. Smyth, G.K. Limma: Linear Models for Microarray Data. In Bioinformatics and Computational Biology Solutions Using R and Bioconductor; Gentleman, R., Carey, V.J., Huber, W., Irizarry, R.A., Dudoit, S., Eds.; Statistics for Biology and Health; Springer: New York, NY, USA, 2005; pp. 397–420. ISBN 978-0-387-29362-2. [Google Scholar]
  41. Frankish, A.; Diekhans, M.; Ferreira, A.-M.; Johnson, R.; Jungreis, I.; Loveland, J.; Mudge, J.M.; Sisu, C.; Wright, J.; Armstrong, J.; et al. GENCODE Reference Annotation for the Human and Mouse Genomes. Nucleic Acids Res. 2019, 47, D766–D773. [Google Scholar] [CrossRef] [Green Version]
  42. Chen, Y.; Lun, A.T.L.; Smyth, G.K. From Reads to Genes to Pathways: Differential Expression Analysis of RNA-Seq Experiments Using Rsubread and the EdgeR Quasi-Likelihood Pipeline. F1000Research 2016, 5, 1438. [Google Scholar]
  43. Robinson, M.D.; McCarthy, D.J.; Smyth, K.G. EdgeR: A Bioconductor Package for Differential Expression Analysis of Digital Gene Expression Data. Bioinformatics 2010, 26, 139–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Sulakhe, D.; Balasubramanian, S.; Xie, B.; Feng, B.; Taylor, A.; Wang, S.; Berrocal, E.; Dave, U.; Xu, J.; Börnigen, D.; et al. Lynx: A Database and Knowledge Extraction Engine for Integrative Medicine. Nucleic Acids Res. 2014, 42, D1007–D1012. [Google Scholar] [CrossRef] [PubMed]
  45. Nickla, D.L.; Wallman, J. The multifunctional choroid. Prog. Retin. Eye Res. 2010, 29, 144–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Armstrong, R.A.; Mousavi, M. Overview of Risk Factors for Age-Related Macular Degeneration (AMD). J. Stem Cells 2015, 10, 171–191. [Google Scholar]
  47. Dow, C.; Mancini, F.; Rajaobelina, K.; Boutron-Ruault, M.-C.; Balkau, B.; Bonnet, F.; Fagherazzi, G. Diet and Risk of Diabetic Retinopathy: A Systematic Review. Eur. J. Epidemiol. 2018, 33, 141–156. [Google Scholar] [CrossRef]
  48. Rowan, S. Involvement of a Gut–Retina Axis in Protection against Dietary Glycemia-Induced Age-Related Macular Degeneration. Proc. Natl. Acad. Sci. USA 2017, 114, 4472–4481. [Google Scholar] [CrossRef] [Green Version]
  49. Bretillon, L.; Thuret, G.; Grégoire, S.; Acar, N.; Joffre, C.; Bron, A.M.; Gain, P.; Creuzot-Garcher, C.P. Lipid and Fatty Acid Profile of the Retina, Retinal Pigment Epithelium/Choroid, and the Lacrimal Gland, and Associations with Adipose Tissue Fatty Acids in Human Subjects. Exp. Eye Res. 2008, 87, 521–528. [Google Scholar] [CrossRef]
  50. Shao, J.; Choudhary, M.M.; Schachat, A.P. Neovascular Age-Related Macular Degeneration. Retin. Pharmacother. 2016, 55, 125–136. [Google Scholar] [CrossRef]
  51. Campochiaro, P.A. Molecular Pathogenesis of Retinal and Choroidal Vascular Diseases. Prog. Retin. Eye Res. 2015, 49, 67–81. [Google Scholar] [CrossRef] [Green Version]
  52. Khanna, S.; Komati, R.; Eichenbaum, D.A.; Hariprasad, I.; Ciulla, T.A.; Hariprasad, S.M. Current and Upcoming Anti-VEGF Therapies and Dosing Strategies for the Treatment of Neovascular AMD: A Comparative Review. BMJ Open Ophthalmol. 2019, 4, e000398. [Google Scholar] [CrossRef] [Green Version]
  53. Saharinen, P.; Eklund, L.; Alitalo, K. Therapeutic Targeting of the Angiopoietin–TIE Pathway. Nat. Rev. Drug Discov. 2017, 16, 635–661. [Google Scholar] [CrossRef] [PubMed]
  54. Fagiani, E.; Christofori, G. Angiopoietins in Angiogenesis. Cancer Lett. 2013, 328, 18–26. [Google Scholar] [CrossRef] [PubMed]
  55. Thurston, G. Complementary Actions of VEGF and Angiopoietin-1 on Blood Vessel Growth and Leakage. J. Anat. 2002, 200, 575–580. [Google Scholar] [CrossRef]
  56. Brindle, N.P.J.; Saharinen, P.; Alitalo, K. Signaling and Functions of Angiopoietin-1 in Vascular Protection. Circ. Res. 2006, 98, 1014–1023. [Google Scholar] [CrossRef] [PubMed]
  57. Richardson, M.R.; Robbins, E.P.; Vemula, S.; Critser, P.J.; Whittington, C.; Voytik-Harbin, S.L.; Yoder, M.C. Angiopoietin-like Protein 2 Regulates Endothelial Colony Forming Cell Vasculogenesis. Angiogenesis 2014, 17, 675–683. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Kadomatsu, T.; Endo, M.; Miyata, K.; Oike, Y. Diverse Roles of ANGPTL2 in Physiology and Pathophysiology. Trends Endocrinol. Metab. 2014, 25, 245–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Lux, A.; Llacer, H.; Heussen, F.M.A.; Joussen, A.M. Non-Responders to Bevacizumab (Avastin) Therapy of Choroidal Neovascular Lesions. Br. J. Ophthalmol. 2007, 91, 1318–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Rosenfeld, P.J.; Brown, D.M.; Heier, J.S.; Boyer, D.S.; Kaiser, P.K.; Chung, C.Y.; Kim, R.Y.; MARINA Study Group. Ranibizumab for Neovascular Age-Related Macular Degeneration. N. Engl. J. Med. 2006, 355, 1419–1431. [Google Scholar] [CrossRef] [Green Version]
  61. Yang, S.; Zhao, J.; Sun, X. Resistance to Anti-VEGF Therapy in Neovascular Age-Related Macular Degeneration: A Comprehensive Review. Drug Des. Dev. Ther. 2016, 10, 1857–1867. [Google Scholar] [CrossRef] [Green Version]
  62. Regeneron Pharmaceuticals. An Open-Label, Dose-Escalation Study of the Safety and Tolerability of Intravitreal (IVT) REGN910-3 and IVT REGN910 in Patients with Either Neovascular AMD or DME. 2016. Available online: https://clinicaltrials.gov (accessed on 6 May 2022).
  63. Patel, S.S.; Sahni, J.; Sadikhov, S.; Pauly-Evers, M.; Szczesny, P.; Weikert, R. Anti-VEGF/Anti-Angiopoietin-2 Bispecific Antibody RG7716 in Diabetic Macular Edema: Complete 36-Week Results from the Phase 2, Multicenter, Randomized, Active Treatment-Controlled BOULEVARD Clinical Trial. Investig. Ophthalmol. Vis. Sci. 2018, 59, 1959. [Google Scholar]
  64. Kumar, A.; Li, X. PDGF-C and PDGF-D in Ocular Diseases. Mol. Asp. Med. 2018, 62, 33–43. [Google Scholar] [CrossRef] [PubMed]
  65. Folestad, E.; Kunath, A.; Wågsäter, D. PDGF-C and PDGF-D Signaling in Vascular Diseases and Animal Models. Mol. Asp. Med. 2018, 62, 1–11. [Google Scholar] [CrossRef] [PubMed]
  66. Zhou, L.; Sun, X.; Huang, Z.; Zhou, T.; Zhu, X.; Liu, Y.; Wang, J.; Cheng, B.; Li, M.; He, C.; et al. Imatinib Ameliorated Retinal Neovascularization by Suppressing PDGFR-α and PDGFR-β. CPB 2018, 48, 263–273. [Google Scholar] [CrossRef] [PubMed]
  67. Jaffe, G.J.; Ciulla, T.A.; Ciardella, A.P.; Devin, F.; Dugel, P.U.; Eandi, C.M.; Masonson, H.; Monés, J.; Pearlman, J.A.; Quaranta-El Maftouhi, M.; et al. Dual Antagonism of PDGF and VEGF in Neovascular Age-Related Macular Degeneration: A Phase IIb, Multicenter, Randomized Controlled Trial. Ophthalmology 2017, 124, 224–234. [Google Scholar] [CrossRef] [Green Version]
  68. Falk, M.K.; Singh, A.; Faber, C.; Nissen, M.H.; Hviid, T.; Sørensen, T.L. Dysregulation of CXCR3 Expression on Peripheral Blood Leukocytes in Patients With Neovascular Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2014, 55, 4050–4056. [Google Scholar] [CrossRef] [Green Version]
  69. Palenski, T.L.; Gurel, Z.; Sorenson, C.M.; Hankenson, K.D.; Sheibani, N. Cyp1B1 Expression Promotes Angiogenesis by Suppressing NF-ΚB Activity. Am. J. Physiol. Cell Physiol. 2013, 305, C1170–C1184. [Google Scholar] [CrossRef] [Green Version]
  70. Tang, Y.; Scheef, E.A.; Wang, S.; Sorenson, C.M.; Marcus, C.B.; Jefcoate, C.R.; Sheibani, N. CYP1B1 Expression Promotes the Proangiogenic Phenotype of Endothelium through Decreased Intracellular Oxidative Stress and Thrombospondin-2 Expression. Blood 2009, 113, 744–754. [Google Scholar] [CrossRef] [Green Version]
  71. Kauppinen, A.; Paterno, J.J.; Blasiak, J.; Salminen, A.; Kaarniranta, K. Inflammation and Its Role in Age-Related Macular Degeneration. Cell. Mol. Life Sci. 2016, 73, 1765–1786. [Google Scholar] [CrossRef] [Green Version]
  72. Wang, Y.; Wang, V.M.; Chan, C.-C. The Role of Anti-Inflammatory Agents in Age-Related Macular Degeneration (AMD) Treatment. Eye 2011, 25, 127–139. [Google Scholar] [CrossRef]
  73. Hageman, G.S.; Luthert, P.J.; Victor Chong, N.H.; Johnson, L.V.; Anderson, D.H.; Mullins, R.F. An Integrated Hypothesis That Considers Drusen as Biomarkers of Immune-Mediated Processes at the RPE-Bruch’s Membrane Interface in Aging and Age-Related Macular Degeneration. Prog. Retin. Eye Res. 2001, 20, 705–732. [Google Scholar] [CrossRef]
  74. Hong, T.; Tan, A.G.; Mitchell, P.; Wang, J.J. A Review and Meta-Analysis of the Association between C-Reactive Protein and Age-Related Macular Degeneration. Surv. Ophthalmol. 2011, 56, 184–194. [Google Scholar] [CrossRef] [PubMed]
  75. Klein, R.; Myers, C.E.; Cruickshanks, K.J.; Gangnon, R.E.; Danforth, L.G.; Sivakumaran, T.A.; Iyengar, S.K.; Tsai, M.Y.; Klein, B.E.K. Markers of Inflammation, Oxidative Stress, and Endothelial Dysfunction and the 20-Year Cumulative Incidence of Early Age-Related Macular Degeneration: The Beaver Dam Eye Study. JAMA Ophthalmol. 2014, 132, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Nussenblatt, R.B.; Byrnes, G.; Sen, H.N.; Yeh, S.; Faia, L.; Meyerle, C.; Wroblewski, K.; Li, Z.; Liu, B.; Chew, E.; et al. A Randomized Pilot Study of Systemic Immunosuppression in the Treatment of Age-Related Macular Degeneration with Choroidal Neovascularization. RETINA 2010, 30, 1579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Duan, Y.; Zeng, L.; Zheng, C.; Song, B.; Li, F.; Kong, X.; Xu, K. Inflammatory Links Between High Fat Diets and Diseases. Front. Immunol. 2018, 9, 2649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Franceschi, C.; Garagnani, P.; Parini, P.; Giuliani, C.; Santoro, A. Inflammaging: A New Immune–Metabolic Viewpoint for Age-Related Diseases. Nat. Rev. Endocrinol. 2018, 14, 576–590. [Google Scholar] [CrossRef]
  79. Boulangé, C.L.; Neves, A.L.; Chilloux, J.; Nicholson, J.K.; Dumas, M.-E. Impact of the Gut Microbiota on Inflammation, Obesity, and Metabolic Disease. Genome Med. 2016, 8, 42. [Google Scholar] [CrossRef] [Green Version]
  80. Camous, X.; Pera, A.; Solana, R.; Larbi, A. NK Cells in Healthy Aging and Age-Associated Diseases. J. Biomed. Biotechnol. 2012, 2012, 195956. [Google Scholar] [CrossRef] [Green Version]
  81. Prolo, P.; Chiappelli, F.; Angeli, A.; Dovio, A.; Perotti, P.; Pautasso, M.; Sartori, M.L.; Saba, L.; Mussino, S.; Fraccalini, T.; et al. Physiologic Modulation of Natural Killer Cell Activity as an Index of Alzheimer’s Disease Progression. Bioinformation 2007, 1, 363–366. [Google Scholar] [CrossRef] [Green Version]
  82. Cui, Y.; Wan, Q. NKT Cells in Neurological Diseases. Front. Cell. Neurosci. 2019, 13, 245. [Google Scholar] [CrossRef]
  83. Baudouin, C.; Peyman, G.A.; Fredj-Reygrobellet, D.; Gordon, W.C.; Lapalus, P.; Gastaud, P.; Bazan, N.G. Immunohistological Study of Subretinal Membranes in Age-Related Macular Degeneration. Jpn. J. Ophthalmol. 1992, 36, 443–451. [Google Scholar]
  84. Hijioka, K.; Sonoda, K.-H.; Tsutsumi-Miyahara, C.; Fujimoto, T.; Oshima, Y.; Taniguchi, M.; Ishibashi, T. Investigation of the Role of CD1d-Restricted Invariant NKT Cells in Experimental Choroidal Neovascularization. Biochem. Biophys. Res. Commun. 2008, 374, 38–43. [Google Scholar] [CrossRef] [PubMed]
  85. Goverdhan, S.V.; Khakoo, S.I.; Gaston, H.; Chen, X.; Lotery, A.J. Age-Related Macular Degeneration Is Associated with the HLA-Cw*0701 Genotype and the Natural Killer Cell Receptor AA Haplotype. Investig. Ophthalmol. Vis. Sci. 2008, 49, 5077–5082. [Google Scholar] [CrossRef] [PubMed]
  86. Suzuki, A.; Yamada, R.; Kochi, Y.; Sawada, T.; Okada, Y.; Matsuda, K.; Kamatani, Y.; Mori, M.; Shimane, K.; Hirabayashi, Y.; et al. Functional SNPs in CD244 Increase the Risk of Rheumatoid Arthritis in a Japanese Population. Nat. Genet. 2008, 40, 1224–1229. [Google Scholar] [CrossRef]
  87. Lee, K.-M.; Forman, J.P.; McNerney, M.E.; Stepp, S.; Kuppireddi, S.; Guzior, D.; Latchman, Y.E.; Sayegh, M.H.; Yagita, H.; Park, C.-K.; et al. Requirement of Homotypic NK-Cell Interactions through 2B4(CD244)/CD48 in the Generation of NK Effector Functions. Blood 2006, 107, 3181–3188. [Google Scholar] [CrossRef] [Green Version]
  88. McArdel, S.L.; Terhorst, C.; Sharpe, A.H. Roles of CD48 in Regulating Immunity and Tolerance. Clin. Immunol. 2016, 164, 10–20. [Google Scholar] [CrossRef] [Green Version]
  89. Voskoboinik, I.; Whisstock, J.C.; Trapani, J.A. Perforin and Granzymes: Function, Dysfunction and Human Pathology. Nat. Rev. Immunol. 2015, 15, 388–400. [Google Scholar] [CrossRef] [PubMed]
  90. Vazirinejad, R.; Ahmadi, Z.; Arababadi, M.K.; Hassanshahi, G.; Kennedy, D. The Biological Functions, Structure and Sources of CXCL10 and Its Outstanding Part in the Pathophysiology of Multiple Sclerosis. NIM 2014, 21, 322–330. [Google Scholar] [CrossRef]
  91. Picard, C.; Fieschi, C.; Altare, F.; Al-Jumaah, S.; Al-Hajjar, S.; Feinberg, J.; Dupuis, S.; Soudais, C.; Al-Mohsen, I.Z.; Génin, E.; et al. Inherited Interleukin-12 Deficiency: IL12B Genotype and Clinical Phenotype of 13 Patients from Six Kindreds. Am. J. Hum. Genet. 2002, 70, 336–348. [Google Scholar] [CrossRef] [Green Version]
  92. Zhang, J.; Zhou, Y. Identification of Key Genes and Pathways Associated with Age-Related Macular Degeneration. J. Ophthalmol. 2020, 2020, e2714746. [Google Scholar] [CrossRef]
  93. Chen, J.; Wang, W.; Li, Q. Increased Th1/Th17 Responses Contribute to Low-Grade Inflammation in Age-Related Macular Degeneration. CPB 2017, 44, 357–367. [Google Scholar] [CrossRef] [Green Version]
  94. Yan, Y.; Chen, R.; Wang, X.; Hu, K.; Huang, L.; Lu, M.; Hu, Q. CCL19 and CCR7 Expression, Signaling Pathways, and Adjuvant Functions in Viral Infection and Prevention. Front. Cell Dev. Biol. 2019, 7, 212. [Google Scholar] [CrossRef] [PubMed]
  95. Rutar, M.; Natoli, R.; Chia, R.; Valter, K.; Provis, J.M. Chemokine-Mediated Inflammation in the Degenerating Retina Is Coordinated by Müller Cells, Activated Microglia, and Retinal Pigment Epithelium. J. Neuroinflamm. 2015, 12, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Faber, C.; Jehs, T.; Juel, H.B.; Singh, A.; Falk, M.K.; Sørensen, T.L.; Nissen, M.H. Early and Exudative Age-Related Macular Degeneration Is Associated with Increased Plasma Levels of Soluble TNF Receptor II. Acta Ophthalmol. 2015, 93, 242–247. [Google Scholar] [CrossRef]
  97. Wang, H.; Han, X.; Wittchen, E.S.; Hartnett, M.E. TNF-α Mediates Choroidal Neovascularization by Upregulating VEGF Expression in RPE through ROS-Dependent β-Catenin Activation. Mol. Vis. 2016, 22, 116–128. [Google Scholar] [PubMed]
  98. Shi, X.; Semkova, I.; Müther, P.S.; Dell, S.; Kociok, N.; Joussen, A.M. Inhibition of TNF-Alpha Reduces Laser-Induced Choroidal Neovascularization. Exp. Eye Res. 2006, 83, 1325–1334. [Google Scholar] [CrossRef] [PubMed]
  99. Fritsche, L.G.; Chen, W.; Schu, M.; Yaspan, B.L.; Yu, Y.; Thorleifsson, G.; Zack, D.J.; Arakawa, S.; Cipriani, V.; Ripke, S.; et al. Seven New Loci Associated with Age-Related Macular Degeneration. Nat. Genet. 2013, 45, 433–439. [Google Scholar] [CrossRef]
  100. Markomichelakis, N.N.; Theodossiadis, P.G.; Sfikakis, P.P. Regression of Neovascular Age-Related Macular Degeneration Following Infliximab Therapy. Am. J. Ophthalmol. 2005, 139, 537–540. [Google Scholar] [CrossRef]
  101. Fernández-Vega, B.; Fernández-Vega, Á.; Rangel, C.M.; Nicieza, J.; Villota-Deleu, E.; Vega, J.A.; Sanchez-Avila, R.M. Blockade of Tumor Necrosis Factor-Alpha: A Role for Adalimumab in Neovascular Age-Related Macular Degeneration Refractory to Anti-Angiogenesis Therapy? Case Rep. Ophthalmol. 2016, 7, 154–162. [Google Scholar] [CrossRef]
  102. Toris, C.; Gulati, V. The Biology, Pathology and Therapeutic Use of Prostaglandins in the Eye. Clin. Lipidol. 2011, 6, 577–591. [Google Scholar] [CrossRef]
  103. Kim, S.J.; Toma, H.S. Inhibition of Choroidal Neovascularization by Intravitreal Ketorolac. Arch. Ophthalmol. 2010, 128, 596–600. [Google Scholar] [CrossRef] [Green Version]
  104. Khandhadia, S.; Cipriani, V.; Yates, J.R.W.; Lotery, A.J. Age-Related Macular Degeneration and the Complement System. Immunobiology 2012, 217, 127–146. [Google Scholar] [CrossRef] [PubMed]
  105. Edwards, A.O.; Ritter, R.; Abel, K.J.; Manning, A.; Panhuysen, C.; Farrer, L.A. Complement Factor H Polymorphism and Age-Related Macular Degeneration. Science 2005, 308, 421–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Klein, R.J.; Zeiss, C.; Chew, E.Y.; Tsai, J.-Y.; Sackler, R.S.; Haynes, C.; Henning, A.K.; SanGiovanni, J.P.; Mane, S.M.; Mayne, S.T.; et al. Complement Factor H Polymorphism in Age-Related Macular Degeneration. Science 2005, 308, 385–389. [Google Scholar] [CrossRef]
  107. Chen, M.; Forrester, J.V.; Xu, H. Synthesis of Complement Factor H by Retinal Pigment Epithelial Cells Is Down-Regulated by Oxidized Photoreceptor Outer Segments. Exp. Eye Res. 2007, 84, 635–645. [Google Scholar] [CrossRef] [PubMed]
  108. Chirco, K.R.; Sohn, E.H.; Stone, E.M.; Tucker, B.A.; Mullins, R.F. Structural and Molecular Changes in the Aging Choroid: Implications for Age-Related Macular Degeneration. Eye 2017, 31, 10–25. [Google Scholar] [CrossRef]
  109. Clark, S.J.; Bishop, P.N.; Day, A.J. Complement Factor H and Age-Related Macular Degeneration: The Role of Glycosaminoglycan Recognition in Disease Pathology. Biochem. Soc. Trans. 2010, 38, 1342–1348. [Google Scholar] [CrossRef] [Green Version]
  110. Perkins, S.; Fung, K.W.; Khan, S. Molecular Interactions between Complement Factor H and Its Heparin and Heparan Sulfate Ligands. Front. Immunol. 2014, 5, 126. [Google Scholar] [CrossRef] [Green Version]
  111. Gold, B.; Merriam, J.E.; Zernant, J.; Hancox, L.S.; Taiber, A.J.; Gehrs, K.; Cramer, K.; Neel, J.; Bergeron, J.; Barile, G.R.; et al. Variation in Factor B (BF) and Complement Component 2 (C2) Genes Is Associated with Age-Related Macular Degeneration. Nat. Genet. 2006, 38, 458–462. [Google Scholar] [CrossRef] [Green Version]
  112. Seddon, J.M.; Reynolds, R.; Maller, J.; Fagerness, J.A.; Daly, M.J.; Rosner, B. Prediction Model for Prevalence and Incidence of Advanced Age-Related Macular Degeneration Based on Genetic, Demographic, and Environmental Variables. Investig. Ophthalmol. Vis. Sci. 2009, 50, 2044–2053. [Google Scholar] [CrossRef]
  113. Seth, A.; Cui, J.; To, E.; Kwee, M.; Matsubara, J. Complement-Associated Deposits in the Human Retina. Investig. Ophthalmol. Vis. Sci. 2008, 49, 743–750. [Google Scholar] [CrossRef] [Green Version]
  114. Mullins, R.F.; Schoo, D.P.; Sohn, E.H.; Flamme-Wiese, M.J.; Workamelahu, G.; Johnston, R.M.; Wang, K.; Tucker, B.A.; Stone, E.M. The Membrane Attack Complex in Aging Human Choriocapillaris: Relationship to Macular Degeneration and Choroidal Thinning. Am. J. Pathol. 2014, 184, 3142–3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Chirco, K.R.; Tucker, B.A.; Stone, E.M.; Mullins, R.F. Selective Accumulation of the Complement Membrane Attack Complex in Aging Choriocapillaris. Exp. Eye Res. 2016, 146, 393–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zeng, S.; Whitmore, S.S.; Sohn, E.H.; Riker, M.J.; Wiley, L.A.; Scheetz, T.E.; Stone, E.M.; Tucker, B.A.; Mullins, R.F. Molecular Response of Chorioretinal Endothelial Cells to Complement Injury: Implications for Macular Degeneration. J. Pathol. 2016, 238, 446–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Doerner, S.K.; Reis, E.S.; Leung, E.S.; Ko, J.S.; Heaney, J.D.; Berger, N.A.; Lambris, J.D.; Nadeau, J.H. High-Fat Diet-Induced Complement Activation Mediates Intestinal Inflammation and Neoplasia, Independent of Obesity. Mol. Cancer Res. 2016, 14, 953–965. [Google Scholar] [CrossRef] [Green Version]
  118. Toomey, C.B.; Kelly, U.; Saban, D.R.; Bowes Rickman, C. Regulation of Age-Related Macular Degeneration-like Pathology by Complement Factor H. Proc. Natl. Acad. Sci. USA 2015, 112, E3040–E3049. [Google Scholar] [CrossRef] [Green Version]
  119. Liao, D.S.; Grossi, F.V.; El Mehdi, D.; Gerber, M.R.; Brown, D.M.; Heier, J.S.; Wykoff, C.C.; Singerman, L.J.; Abraham, P.; Grassmann, F.; et al. Complement C3 Inhibitor Pegcetacoplan for Geographic Atrophy Secondary to Age-Related Macular Degeneration: A Randomized Phase 2 Trial. Ophthalmology 2020, 127, 186–195. [Google Scholar] [CrossRef] [Green Version]
  120. Wu, J.; Sun, X. Complement System and Age-Related Macular Degeneration: Drugs and Challenges. Drug Des. Dev. Ther. 2019, 13, 2413–2425. [Google Scholar] [CrossRef]
  121. Klaver, C.C.; Kliffen, M.; van Duijn, C.M.; Hofman, A.; Cruts, M.; Grobbee, D.E.; van Broeckhoven, C.; de Jong, P.T. Genetic Association of Apolipoprotein E with Age-Related Macular Degeneration. Am. J. Hum. Genet. 1998, 63, 200–206. [Google Scholar] [CrossRef] [Green Version]
  122. Hu, M.L.; Quinn, J.; Xue, K. Interactions between Apolipoprotein E Metabolism and Retinal Inflammation in Age-Related Macular Degeneration. Life 2021, 11, 635. [Google Scholar] [CrossRef]
  123. Li, C.-M.; Clark, M.E.; Chimento, M.F.; Curcio, C.A. Apolipoprotein Localization in Isolated Drusen and Retinal Apolipoprotein Gene Expression. Investig. Ophthalmol. Vis. Sci. 2006, 47, 3119–3128. [Google Scholar] [CrossRef]
  124. Yang, P.; Skiba, N.P.; Tewkesbury, G.M.; Treboschi, V.M.; Baciu, P.; Jaffe, G.J. Complement-Mediated Regulation of Apolipoprotein E in Cultured Human RPE Cells. Investig. Ophthalmol. Vis. Sci. 2017, 58, 3073–3085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Newman, A.M.; Gallo, N.B.; Hancox, L.S.; Miller, N.J.; Radeke, C.M.; Maloney, M.A.; Cooper, J.B.; Hageman, G.S.; Anderson, D.H.; Johnson, L.V.; et al. Systems-Level Analysis of Age-Related Macular Degeneration Reveals Global Biomarkers and Phenotype-Specific Functional Networks. Genome Med. 2012, 4, 16. [Google Scholar] [CrossRef]
  126. Yu, Y.; Bhangale, T.R.; Fagerness, J.; Ripke, S.; Thorleifsson, G.; Tan, P.L.; Souied, E.H.; Richardson, A.J.; Merriam, J.E.; Buitendijk, G.H.S.; et al. Common Variants near FRK/COL10A1 and VEGFA Are Associated with Advanced Age-Related Macular Degeneration. Hum. Mol. Genet. 2011, 20, 3699–3709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Boutin, T.S.; Charteris, D.G.; Chandra, A.; Campbell, S.; Hayward, C.; Campbell, A.; UK Biobank Eye & Vision Consortium; Nandakumar, P.; Hinds, D.; 23andMe Research Team; et al. Insights into the Genetic Basis of Retinal Detachment. Hum. Mol. Genet. 2020, 29, 689–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Stone, E.M.; Braun, T.A.; Russell, S.R.; Kuehn, M.H.; Lotery, A.J.; Moore, P.A.; Eastman, C.G.; Casavant, T.L.; Sheffield, V.C. Missense Variations in the Fibulin 5 Gene and Age-Related Macular Degeneration. N. Engl. J. Med. 2004, 351, 346–353. [Google Scholar] [CrossRef]
  129. Rojo Arias, J.E.; Jászai, J. Gene Expression Profile of the Murine Ischemic Retina and Its Response to Aflibercept (VEGF-Trap). Sci. Rep. 2021, 11, 15313. [Google Scholar] [CrossRef]
  130. Adams, J.N.; Raffield, L.M.; Martelle, S.E.; Freedman, B.I.; Langefeld, C.D.; Carr, J.J.; Cox, A.J.; Bowden, D.W. Genetic Analysis of Advanced Glycation End Products in the DHS MIND Study. Gene 2016, 584, 173–179. [Google Scholar] [CrossRef] [Green Version]
  131. Ishibashi, T.; Murata, T.; Hangai, M.; Nagai, R.; Horiuchi, S.; Lopez, P.F.; Hinton, D.R.; Ryan, S.J. Advanced Glycation End Products in Age-Related Macular Degeneration. Arch. Ophthalmol. 1998, 116, 1629–1632. [Google Scholar] [CrossRef]
  132. Fernandez-Godino, R.; Pierce, E.A.; Garland, D.L. Extracellular Matrix Alterations and Deposit Formation in AMD. Adv. Exp. Med. Biol. 2016, 854, 53–58. [Google Scholar] [CrossRef]
  133. Butler, J.M.; Supharattanasitthi, W.; Yang, Y.C.; Paraoan, L. RNA-Seq Analysis of Ageing Human Retinal Pigment Epithelium: Unexpected up-Regulation of Visual Cycle Gene Transcription. J. Cell. Mol. Med. 2021, 25, 5572–5585. [Google Scholar] [CrossRef]
  134. Gollapalli, D.R.; Rando, R.R. The Specific Binding of Retinoic Acid to RPE65 and Approaches to the Treatment of Macular Degeneration. Proc. Natl. Acad. Sci. USA 2004, 101, 10030–10035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Zhang, J.; Bai, Y.; Huang, L.; Qi, Y.; Zhang, Q.; Li, S.; Wu, Y.; Li, X. Protective Effect of Autophagy on Human Retinal Pigment Epithelial Cells against Lipofuscin Fluorophore A2E: Implications for Age-Related Macular Degeneration. Cell Death Dis. 2015, 6, e1972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Speakman, J.R. Use of High-Fat Diets to Study Rodent Obesity as a Model of Human Obesity. Int. J. Obes. 2019, 43, 1491–1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Hintze, K.J.; Benninghoff, A.D.; Cho, C.E.; Ward, R.E. Modeling the Western Diet for Preclinical Investigations. Adv. Nutr. 2018, 9, 263–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Hintze, K.J.; Benninghoff, A.D.; Ward, R.E. Formulation of the Total Western Diet (TWD) as a Basal Diet for Rodent Cancer Studies. J. Agric. Food Chem. 2012, 60, 6736–6742. [Google Scholar] [CrossRef]
  139. Keeling, E.; Lynn, S.A.; Koh, Y.M.; Scott, J.A.; Kendall, A.; Gatherer, M.; Page, A.; Cagampang, F.R.; Lotery, A.J.; Ratnayaka, J.A. A High Fat “Western-Style” Diet Induces AMD-Like Features in Wildtype Mice. Mol. Nutr. Food Res. 2022, 66, e2100823. [Google Scholar] [CrossRef]
  140. Dighe, S.; Zhao, J.; Steffen, L.; Mares, J.A.; Meuer, S.M.; Klein, B.E.K.; Klein, R.; Millen, A.E. Diet Patterns and the Incidence of Age-Related Macular Degeneration in the Atherosclerosis Risk in Communities (ARIC) Study. Br. J. Ophthalmol. 2020, 104, 1070–1076. [Google Scholar] [CrossRef]
  141. Wang, L.; Clark, M.E.; Crossman, D.K.; Kojima, K.; Messinger, J.D.; Mobley, J.A.; Curcio, C.A. Abundant Lipid and Protein Components of Drusen. PLoS ONE 2010, 5, e10329. [Google Scholar] [CrossRef]
  142. Sasaki, M.; Kawasaki, R.; Rogers, S.; Man, R.E.K.; Itakura, K.; Xie, J.; Flood, V.; Tsubota, K.; Lamoureux, E.; Wang, J.J. The Associations of Dietary Intake of Polyunsaturated Fatty Acids With Diabetic Retinopathy in Well-Controlled Diabetes. Investig. Ophthalmol. Vis. Sci. 2015, 56, 7473–7479. [Google Scholar] [CrossRef]
  143. Hu, P.; Uddin, M.N.; Neuringer, M.; Renner, L.; Stoddard, J.; Boulton, M.E.; McGill, T.J.; Chan-Ling, T.; Grant, M. Evidence of Diabetic Retinopathy in a Western (High Fat) Diet-Induced Non-Human Primate Model of Type 2 Diabetes (T2D). Investig. Ophthalmol. Vis. Sci. 2018, 59, 3589. [Google Scholar]
  144. Reynolds, R.; Rosner, B.; Seddon, J.M. Serum Lipid Biomarkers and Hepatic Lipase Gene Associations with Age-Related Macular Degeneration. Ophthalmology 2010, 117, 1989–1995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Adams, M.K.M.; Simpson, J.A.; Richardson, A.J.; English, D.R.; Aung, K.Z.; Makeyeva, G.A.; Guymer, R.H.; Giles, G.G.; Hopper, J.; Robman, L.D.; et al. Apolipoprotein E Gene Associations in Age-Related Macular Degeneration: The Melbourne Collaborative Cohort Study. Am. J. Epidemiol. 2012, 175, 511–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Hodge, W.G.; Schachter, H.M.; Barnes, D.; Pan, Y.; Lowcock, E.C.; Zhang, L.; Sampson, M.; Morrison, A.; Tran, K.; Miguelez, M.; et al. Efficacy of Omega-3 Fatty Acids in Preventing Age-Related Macular Degeneration: A Systematic Review. Ophthalmology 2006, 113, 1165–1172; quiz 1172–1173, 1178. [Google Scholar] [CrossRef] [PubMed]
  147. Chong, E.W.-T.; Robman, L.D.; Simpson, J.A.; Hodge, A.M.; Aung, K.Z.; Dolphin, T.K.; English, D.R.; Giles, G.G.; Guymer, R.H. Fat Consumption and Its Association with Age-Related Macular Degeneration. Arch. Ophthalmol. 2009, 127, 674–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Agrón, E.; Mares, J.; Clemons, T.E.; Swaroop, A.; Chew, E.Y.; Keenan, T.D.L.; AREDS and AREDS2 Research Groups. Dietary Nutrient Intake and Progression to Late Age-Related Macular Degeneration in the Age-Related Eye Disease Studies 1 and 2. Ophthalmology 2021, 128, 425–442. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, Q.-Y.; Tie, L.-J.; Wu, S.-S.; Lv, P.-L.; Huang, H.-W.; Wang, W.-Q.; Wang, H.; Ma, L. Overweight, Obesity, and Risk of Age-Related Macular Degeneration. Investig. Ophthalmol. Vis. Sci. 2016, 57, 1276–1283. [Google Scholar] [CrossRef] [Green Version]
  150. Age-Related Eye Disease Study Research Group. A Randomized, Placebo-Controlled, Clinical Trial of High-Dose Supplementation with Vitamins C and E, Beta Carotene, and Zinc for Age-Related Macular Degeneration and Vision Loss: AREDS Report No. 8. Arch. Ophthalmol. 2001, 119, 1417–1436. [Google Scholar] [CrossRef] [Green Version]
  151. Age-Related Eye Disease Study 2 Research Group. Lutein + Zeaxanthin and Omega-3 Fatty Acids for Age-Related Macular Degeneration: The Age-Related Eye Disease Study 2 (AREDS2) Randomized Clinical Trial. JAMA 2013, 309, 2005–2015. [Google Scholar] [CrossRef]
Figure 1. Heatmap with hierarchal clustering demonstrating DEGs with |LogFC| > 1.5 and adjusted p-value < 0.01 between germ-free mice on normal diet (GF-ND, n = 4) and germ-free mice on high-fat diet (GF-HFD, n = 4). Z-score was calculated using LogFC values, with red and blue colors indicating upregulated and downregulated genes, respectively.
Figure 1. Heatmap with hierarchal clustering demonstrating DEGs with |LogFC| > 1.5 and adjusted p-value < 0.01 between germ-free mice on normal diet (GF-ND, n = 4) and germ-free mice on high-fat diet (GF-HFD, n = 4). Z-score was calculated using LogFC values, with red and blue colors indicating upregulated and downregulated genes, respectively.
Cells 11 02076 g001
Figure 2. Enrichment analysis of DEGs between GF-HFD mice (n = 4) and GF-ND mice (n = 4) using Lynx. Gene ontology analysis is shown for top 10 biological processes upregulated in GF-HFD mice compared to GF-ND mice, highlighting pathways including the angiogenic, inflammatory, and immune responses. The corresponding table demonstrates detailed statistics and genes involved in these processes.
Figure 2. Enrichment analysis of DEGs between GF-HFD mice (n = 4) and GF-ND mice (n = 4) using Lynx. Gene ontology analysis is shown for top 10 biological processes upregulated in GF-HFD mice compared to GF-ND mice, highlighting pathways including the angiogenic, inflammatory, and immune responses. The corresponding table demonstrates detailed statistics and genes involved in these processes.
Cells 11 02076 g002
Figure 3. Enrichment analysis of DEGs between GF-HFD mice (n = 4) and GF-ND mice (n = 4) using Lynx. Gene ontology analysis is shown for top 10 molecular functions upregulated in GF-HFD mice compared to GF-ND mice. Functions such as extracellular matrix (ECM) binding, ECM structural constituents, and heparin binding are highlighted. The corresponding table demonstrates detailed statistics and genes involved in these functions.
Figure 3. Enrichment analysis of DEGs between GF-HFD mice (n = 4) and GF-ND mice (n = 4) using Lynx. Gene ontology analysis is shown for top 10 molecular functions upregulated in GF-HFD mice compared to GF-ND mice. Functions such as extracellular matrix (ECM) binding, ECM structural constituents, and heparin binding are highlighted. The corresponding table demonstrates detailed statistics and genes involved in these functions.
Cells 11 02076 g003
Table 1. Top 30 differentially expressed genes upregulated by high-fat diet (HFD).
Table 1. Top 30 differentially expressed genes upregulated by high-fat diet (HFD).
GeneLogFCAdjusted p-ValueProtein
Cd244a6.271.62 × 10−3Natural Killer Cell Receptor 2B4
Ripply15.529.98 × 10−3Ripply Transcriptional Repressor 1
Lilrb4a5.284.81 × 10−2Leukocyte Immunoglobulin-Like Receptor Subfamily B member 4
Fcer1a5.262.85 × 10−2Fc Epsilon Receptor Ia
Dnajc225.241.44 × 10−2DnaJ Heat Shock Protein Family (Hsp40) Member C22
Alkal25.113.11 × 10−2ALK And LTK Ligand 2
Ncr15.041.16 × 10−2Natural Cytotoxicity Triggering Receptor 1
Ccl194.947.60 × 10−3C-C Motif Chemokine Ligand 19
Slc38a114.813.85 × 10−2Solute Carrier Family 38 Member 11
Ces2e4.714.62 × 10−2Pyrethroid Hydrolase Ces2e
Lpar34.661.73 × 10−2Lysophosphatidic Acid Receptor 3
Sele4.624.49 × 10−2Selectin E
Sirpb1a4.619.02 × 10−3Signal-Regulatory Protein Beta 1A
Efhb4.571.33 × 10−2EF-Hand Domain-Containing Family Member B
Pgpep1l4.562.15 × 10−2Pyroglutamyl-Peptidase 1-Like Protein
Tnfrsf13b4.542.94 × 10−2Tumor Necrosis Factor Receptor Superfamily Member 13B
Il12b4.492.65 × 10−2Interleukin-12 Subunit Beta
Tmem2324.462.53 × 10−2Transmembrane Protein 232
Trbc14.411.51 × 10−2T Cell Receptor Beta Constant 1
Slc4a14.343.36 × 10−2Solute Carrier Family 4 Member 1
Olfr5744.341.60 × 10−2Olfactory Receptor Family 51 Subfamily T Member 1
Xlr4.285.51 × 10−3X-Linked Lymphocyte-Regulated Protein PM1
Gpr1414.254.73 × 10−2G Protein-Coupled Receptor 141
Cnr24.253.48 × 10−2Cannabinoid Receptor 2
Mael4.234.08 × 10−2Maelstrom Spermatogenic Transposon Silencer
Lao14.224.06 × 10−2Amine Oxidase
Mcoln24.161.72 × 10−2Mucolipin TRP Cation Channel 2
Ccl224.163.77 × 10−2C-C Motif Chemokine Ligand 22
Rnase14.112.66 × 10−2Ribonuclease A Family Member 1, Pancreatic
Ptgs2os4.113.37 × 10−2Prostaglandin-Endoperoxide Synthase 2, Opposite Strand
Table 2. Top 10 biological processes upregulated by high-fat diet (HFD).
Table 2. Top 10 biological processes upregulated by high-fat diet (HFD).
Biological Processesp-ValueAdjusted p-ValueGene RatioGenes
Melanocyte differentiation1.79 × 10−79.49 × 10−50.35Edn3, Ednrb, Mitf, Mlph, Rab27a, Slc24a5, Sox10, and Tyrp1
Angiogenesis2.08 × 10−135.51 × 10−100.13Ackr3, Angpt1, Angpt2, Angptl2, Apold1, Calcrl, Cfh, Clic4, Col18a1, Col8a1, Col8a2, Cxcr3, Cyp1b1, Ecscr, Ephb4, Esm1, Fap, Fzd8, Htatip2, Mcam, Nrp2, Pik3r6, Plxnd1, Ptgs2, Ptprb, Rapgef3, Rhoj, Rspo3, Tbx4, Tek, Tgfbr3, Tie1, Tnfaip2, and Vegfc
Cell surface receptor signaling pathway1.50 × 10−91.50 × 10−60.13Adgra3, Adgrf5, Adgrg6, Calcrl, Cd22, Cd86, Cxcr3, Cysltr1, Edn3, Fcer1a, Fzd2, Fzd4, Fzd7, Fzd8, Gpr157, Il12b, Itgal, Itpkb, Npr1, Osmr, Ostn, Pth1r, Tnfrsf1b, and Tshr
Positive regulation of angiogenesis5.68 × 10−72.15 × 10−40.12Angpt2, Brca1, Chil1, Cxcr3, Cybb, Cyp1b1, Cysltr1, Ets1, Itgb3, Itgb8, Pik3r6, Ptgis, Rapgef3, Tek, Tgfbr2, Thbs1, Tie1, and Vegfc
Inflammatory response1.14 × 10−91.50 × 10−60.10Agtr1a, Axl, Bmp2, Bmp6, Ccl19, Ccl22, Ccl4, Cfh, Chil1, Cnr2, Csf1r, Cxcl10, Cxcr3, Cyba, Cybb, Cysltr1, Gbp5, Il25, Lilrb4a, Lipa, Ly86, P2rx7, Pla2g2e, Prkcq, Ptgs1, Ptgs2, Rarres2, Sele, Selp, Slc11a1, Thbs1, Themis2, Tlr13, and Tnfrsf1b
Response to bacterium1.29 × 10−63.56 × 10−40.09Adamts9, Bank1, Bmp2, Cxcl10, Fkbp5, Gbp5, Gpc3, Gzma, Ifi211, Ifit3, Iigp1, Lrat, Ms4a1, Myo1f, Naaladl2, Nexn, Ociad2, P2rx7, Rnase1, Serpina3f, Serpinb9, Slc11a1, Tgtp1, and Trf
Immune response8.75 × 10−72.90 × 10−40.08Ackr3, Azgp1, B2m, Bmp6, Ccl19, Ccl22, Ccl4, Cd28, Cd86, Cfh, Colec12, Ctsk, Ctss, Cxcl10, Cxcl12, Cxcr3, Endou, Enpp2, H2-Ab1, H2-Eb1, H2-M3, Itgb8, Ly86, Serpinb9, Tgfbr3, Tgtp1, Tnfrsf1b, Tnfsf10, Tnfsf14, Vav1
Cell adhesion1.48 × 10−89.80 × 10−60.08Ackr3, Azgp1, Cd22, Cd33, Cd84, Cldn1, Cldn2, Cntnap4, Col12a1, Col18a1, Col8a1, Col8a2, Cyp1b1, Dpp4, Ephb4, Fap, Fblim1, Fbln5, Gpnmb, Hpse, Icam2, Itga9, Itgal, Itgb3, Itgb8, Jcad, Kitl, Lgals3bp, Ly9, Mcam, Mybpc2, Nid2, Pcdh12, Plpp3, Sele, Sell, Selp, Siglecf, Spp1, Svep1, Thbs1, Vcam1, and Vwf
Positive regulation of cell population proliferation1.98 × 10−79.49 × 10−50.07Adora2b, Agtr1a, Aldh1a2, Bambi, Calcrl, Cd38, Cdk2, Clec7a, Col18a1, Csf1r, Cxcl10, Cxcl12, Cxcr3, Dpp4, Edn3, Ednra, Ednrb, Enpp2, Esm1, Ets1, Fgf7, Gab2, Gcnt2, Gli1, Kitl, Lrp5, Nog, Ntn1, Osmr, Pax3, Pdgfc, Pdgfd, Ptgs2, Pth1r, S100b, Stox1, Tgfbr3, Thbs1, Tshr, and Vegfc
Multicellular organism development1.21 × 10−63.56 × 10−40.06Ackr3, Angpt1, Angpt2, Ano1, Axl, B2m, Bmp2, Bmp6, Cdh19, Csf1r, Ecscr, Eda2r, Ephb4, Eya1, Eya2, Fhl1, Foxd1, Foxd3, Fst, Fzd2, Fzd4, Fzd7, Fzd8, Gli1, Gpr157, Gsx2, Htatip2, Krt8, Lbx1, Lrp5, Mael, Mertk, Met, Mgp, Mitf, Nog, Nrp2, Ostn, Pax3, Pdgfc, Pdgfd, Pitx2, Plpp3, Plxnd1, Ripply1, Sema3b, Sema3c, Sema6d, Serpine2, Sfrp5, Shisa2, Smoc1, Sox6, Stpg4, Tbx4, Tek, Tie1, Tmem88, Tnfaip2, Vegfc, and Wipf3
Table 3. Top 10 molecular pathways upregulated by high-fat diet (HFD).
Table 3. Top 10 molecular pathways upregulated by high-fat diet (HFD).
Molecular Pathwaysp-ValueAdjusted p-ValueGene RatioGenes
Hemoglobin binding2.43 × 10−53.96 × 10−30.57Hbb-bs, Hbb-bt, Lrp2, Slc4a1
Sialic acid binding2.30 × 10−53.96 × 10−30.38Cd22, Cd33, Sele, Selp, Siglecf
Extracellular matrix binding2.09 × 10−53.96 × 10−30.23Adamts15, Clec14a, Dcn, Itgb3, Smoc1, Spp1, and Thbs1
Fibronectin binding2.64 × 10−53.96 × 10−30.23Ctsk, Ctss, Fbln1, Igfbp3, Igfbp5, Itgb3, Thbs1
Integrin binding2.81 × 10−72.12 × 10−40.13Cxcl12, Esm1, Fap, Fbln1, Fbln5, Fbn1, Gpnmb, Icam2, Itgb3, Itgb8, Lcp1, Lilrb4a, Plpp3, Spp1, Thbs1, Vcam1, and Vwf
Extracellular matrix structural constituent5.40 × 10−54.08 × 10−30.11Col10a1, Col18a1, Col8a1, Col8a2, Col9a3, Fbln1, Fbn1, Fbn2, Matn2, Nid2, Ntn1, Thbs1, Vwf
Signaling receptor activity4.79 × 10−61.21 × 10−30.10Cd48, Colec12, Cxcr3, Eda2r, Fzd4, Itgb8, Klrk1, Lrp2, Mrc2, P2rx7, Paqr6, Stra6, Tek, Tgfbr2, Tlr13, Tnfrsf19, Trem2, and Tshr
Carbohydrate binding2.04 × 10−67.73 × 10−40.10Agl, C4b, Cd22, Cd33, Clec12a, Clec14a, Clec1a, Clec4a2, Clec4d, Clec4n, Colec12, Galm, Galnt6, Klrk1, Man2a1, Mrc2, Sele, Sell, Selp, Siglecf
Heparin binding4.12 × 10−53.96 × 10−30.10Adamts1, Adamts15, Apoe, Cfh, Cxcl10, Fbn1, Fgf7, Gpnmb, Nrp2, Rspo3, Selp, Serpine2, Smoc1, Tgfbr3, and Thbs1
Protein homodimerization activity2.24 × 10−53.96 × 10−30.06Ano1, Ano6, Apoe, Atp2a1, B2m, Cat, Csf1r, Dpp4, Dpyd, Fap, Fbln5, Fzd4, Galm, Gbp3, Gbp5, Gzma, H2-M3, Hnf4a, Impa2, Man2a1, Mgll, Nog, Npr3, Pdgfc, Pitx2, Pon1, Pon3, Ptgs2, Pth1r, Rdh5, S100b, Slc11a1, Slc4a1, St6gal1, Tpd52l1, Trim21, Trim30d, Tyr, Tyrobp, and Tyrp1
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Xiao, J.; Xie, B.; Dao, D.; Spedale, M.; D’Souza, M.; Theriault, B.; Hariprasad, S.M.; Sulakhe, D.; Chang, E.B.; Skondra, D. High-Fat Diet Alters the Retinal Pigment Epithelium and Choroidal Transcriptome in the Absence of Gut Microbiota. Cells 2022, 11, 2076. https://doi.org/10.3390/cells11132076

AMA Style

Xiao J, Xie B, Dao D, Spedale M, D’Souza M, Theriault B, Hariprasad SM, Sulakhe D, Chang EB, Skondra D. High-Fat Diet Alters the Retinal Pigment Epithelium and Choroidal Transcriptome in the Absence of Gut Microbiota. Cells. 2022; 11(13):2076. https://doi.org/10.3390/cells11132076

Chicago/Turabian Style

Xiao, Jason, Bingqing Xie, David Dao, Melanie Spedale, Mark D’Souza, Betty Theriault, Seenu M. Hariprasad, Dinanath Sulakhe, Eugene B. Chang, and Dimitra Skondra. 2022. "High-Fat Diet Alters the Retinal Pigment Epithelium and Choroidal Transcriptome in the Absence of Gut Microbiota" Cells 11, no. 13: 2076. https://doi.org/10.3390/cells11132076

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop