Next Article in Journal
Establishing Treatment Effectiveness in Fabry Disease: Observation-Based Recommendations for Improvement
Previous Article in Journal
Transcriptome Analysis Reveals the Effect of Oyster Mushroom Spherical Virus Infection in Pleurotus ostreatus
Previous Article in Special Issue
Effects of Fucoidans on Activated Retinal Microglia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Impact of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623) Polymorphisms and Serum Levels on Age-Related Macular Degeneration Development and Therapeutic Responses

by
Dzastina Cebatoriene
1,
Alvita Vilkeviciute
2,*,
Greta Gedvilaite-Vaicechauskiene
2,
Monika Duseikaite
2,
Akvile Bruzaite
2,
Loresa Kriauciuniene
2,3,
Dalia Zaliuniene
3 and
Rasa Liutkeviciene
2,3
1
Medical Academy, Lithuanian University of Health Sciences, A. Mickeviciaus St. 9, LT-44307 Kaunas, Lithuania
2
Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
3
Department of Ophthalmology, Medical Academy, Lithuanian University of Health Sciences, Eiveniu St. 2, LT-50161 Kaunas, Lithuania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(17), 9750; https://doi.org/10.3390/ijms25179750
Submission received: 2 July 2024 / Revised: 3 September 2024 / Accepted: 4 September 2024 / Published: 9 September 2024

Abstract

:
Age-related macular degeneration (AMD) is a major global health problem as it is the leading cause of irreversible loss of central vision in the aging population. Anti-vascular endothelial growth factor (anti-VEGF) therapies are effective but do not respond optimally in all patients. This study investigates the genetic factors associated with susceptibility to AMD and response to treatment, focusing on key polymorphisms in the ARMS2 (rs10490924), IL1B1 (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), VEGFA (rs3024997), ARMS2, IL1B1, TNFRSF1B, TNFRSF1A, and VEGFA serum levels in AMD development and treatment efficacy. This study examined the associations of specific genetic polymorphisms and serum protein levels with exudative and early AMD and the response to anti-VEGF treatment. The AA genotype of VEGFA (rs3024997) was significantly associated with a 20-fold reduction in the odds of exudative AMD compared to the GG + GA genotypes. Conversely, the TT genotype of ARMS2 (rs10490924) was linked to a 4.2-fold increase in the odds of exudative AMD compared to GG + GT genotypes. In females, each T allele of ARMS2 increased the odds by 2.3-fold, while in males, the TT genotype was associated with a 5-fold increase. Lower serum IL1B levels were observed in the exudative AMD group compared to the controls. Early AMD patients had higher serum TNFRSF1B levels than controls, particularly those with the GG genotype of TNFRSF1B rs1061622. Exudative AMD patients with the CC genotype of TNFRSF1A rs4149576 had lower serum TNFRSF1A levels compared to the controls. Visual acuity (VA) analysis showed that non-responders had better baseline VA than responders but experienced decreased VA after treatment, whereas responders showed improvement. Central retinal thickness (CRT) reduced significantly in responders after treatment and was lower in responders compared to non-responders after treatment. The T allele of TNFRSF1B rs1061622 was associated with a better response to anti-VEGF treatment under both dominant and additive genetic models. These findings highlight significant genetic and biochemical markers associated with AMD and treatment response. This study found that the VEGFA rs3024997 AA genotype reduces the odds of exudative AMD, while the ARMS2 rs10490924 TT genotype increases it. Lower serum IL1B levels and variations in TNFRSF1B and TNFRSF1A levels were linked to AMD. The TNFRSF1B rs1061622 T allele was associated with better anti-VEGF treatment response. These markers could potentially guide risk assessment and personalized treatment for AMD.

1. Introduction

Age-related macular degeneration (AMD) is one of the most common causes of blindness in developed countries. Therefore, AMD significantly impacts quality of life, leading to difficulties in performing daily tasks, loss of independence, and increased risk of depression. The primary risk factor for AMD is age, with the severity of vision impairment ranging from mild to severe. Individuals over the age of 75 face a 25% risk of developing early AMD and an 8% risk of progressing to late AMD, with the number of cases expected to increase due to the aging population [1]. AMD leads to pathological changes in the deeper retinal layers of the macula and the surrounding blood vessels, resulting in a loss of central vision. The accumulation of deposits on the retina, called drusen, is a characteristic clinical finding in AMD and may be the first sign of the “dry” form of the disease. Dry AMD is the most common morphological type. It can progress to “wet” or neovascular AMD [1]. The neovascular, or “wet”, form of AMD is less common but accounts for 90% of cases of acute blindness caused by AMD [2]. Globally, the prevalence of advanced AMD is estimated to be 1.6% [3]. Neovascular AMD (nAMD) is characterized by the formation of neovascular choroidal membranes, exudation, and fibrosis, leading to acute vision loss [4]. Intravitreal injection of an anti-vascular endothelial growth factor is the standard therapy to maintain or improve visual acuity in most patients with nAMD. While numerous studies have shown positive outcomes with anti-VEGF therapies, there are limitations to their use. Previous studies indicate that 20% of patients continue to experience vision loss, and approximately 50% fail to achieve visual acuity 20/40 [5,6]. Despite the efficacy of anti-VEGF agents in many cases, some patients exhibit an incomplete response to treatment, characterized by persistent intraretinal or subretinal fluid and ongoing vision loss [7]. As AMD is a multifactorial condition, identifying its risk factors allows individuals to make informed lifestyle choices that may lower their risk of developing the disease. Ongoing genetic studies in AMD should prioritize determining the mechanism, pathways, and networks underlying the disease, not just the risk factors including aging, smoking, and high blood pressure, so that appropriate pathways for treatment and efficacy can be identified. Many risk factors play a role in the development of AMD but the most important role is played by genetic variants, which were identified over the years in different studies, suggesting complex oligogenic patterns of inheritance for AMD [8,9,10,11,12,13,14,15,16,17].
In the GWAS experiment [18], the identified genes were found to function in known AMD pathways and highlight the importance of additional pathways. These include complement activation, collagen synthesis, lipid metabolism and cholesterol transport, receptor-mediated endocytosis, endodermal cell differentiation, and extracellular matrix organization [18]. Researchers conducting a large genome-wide association study have identified 52 coding variants in 34 loci that occur more frequently in patients with AMD [18]. The genetic/poligenetic risk score (GRS/PRS) is important to explore in different cohorts as its calculation depends on the presence of risk variants that may be differently distributed across populations. Genetic and epidemiological research has established the undeniable role of genetic variation in the etiology of AMD, with the heritable component estimated to be between 45% and 70% [19]. This is especially true when integrating genetic information with other interacting environmental and demographic factors to better predict disease risk [20,21,22,23]. Geographic variations might explain the discrepancies and a specific genetic variant was found to be more common in Western Europe compared to other global regions, justifying its superior prevalence and effect in the Portuguese population [24]. The complex etiology of AMD depends not only on the genetic background and is greatly impacted and modified by environmental factors [20,21].
While the primary loci associated with advanced AMD are concentrated around the complement factor H gene on chromosome 1q31, recent large-scale genome-wide association studies have increasingly confirmed associations with other genes that may be risk factors [25]. Polymorphisms of the age-related maculopathy 2 gene (ARMS2, also known as LOC387715), located at the 10q26 locus, have been strongly associated with the inverse effect of hormone replacement therapy on AMD. Researchers have demonstrated that the mRNA and corresponding peptide encoded by ARMS2 are expressed in the retina, indicating that the ARMS2 transcript is responsible for the association with AMD [26]. In addition, the ARMS2 protein has been described as a component of the extracellular matrix. Its mRNA has a unique splice form in the retina that is not found in other tissues [27]. The polymorphic site A69S (rs10490924) within the ARMS2 locus has been investigated in several studies, mainly concerning AMD, and the results showed a strong correlation between poor visual acuity in advanced AMD and response to anti-VEGF intervention [28].
The human vascular endothelial growth factor A (VEGFA) gene is located on chromosome 6, with multiple common SNPs in the promoter and the 5′ and 3′ untranslated regions. It is organized into eight exons and seven introns [29]. VEGFA is a member of the VEGF-related polypeptide family and plays a key role in increasing vascular permeability, angiogenesis, endothelial cell growth, and migration [30]. Vascular leakage and inflammation caused by the excessive release of VEGFA have been found to play a crucial role in choroidal neovascularization and the development of neovascular AMD. VEGFA and its signaling pathway have been targeted at the pathogenic processes in which they are involved in the most effective therapeutic development. Polymorphisms in the VEGFA gene regulate VEGF expression and thus its angiogenic properties [31]. It is, therefore, reasonable to suggest that different expression levels of VEGF may result in different responses to anti-VEGF drugs. Finally, a study examining seven different VEGFA polymorphisms concluded that none of these significantly predicts the success of anti-VEGF treatment with Bevacizumab in patients with nAMD [32].
Through interaction with its receptors TNFR1 and TNFR2, the ligand of tumor necrosis factor α (TNFα) activates the inflammatory response, cell proliferation, and differentiation [33,34]. Several studies have shown that TNFR1 and TNFR2 receptor isoforms respond differently to TNF-α stimulation and anti-TNF-α therapy [35]. However, despite the accumulated data on the specifics of the functioning and regulation of TNFR1 and TNFR2 signaling pathways, these remain incompletely understood.
The IL-1 family of cytokines is key in triggering acute inflammatory responses [36]. IL-1β interacts with the IL-1 receptor I (IL-1RI), which consists of the IL-1R and IL-1R accessory protein subunits. The IL-1 receptor antagonist (IL-1Ra) competes with IL-1β for its binding site [36]. IL-1β is a potent inflammatory mediator with chemotactic and angiogenic properties [37,38]. It is a neurotoxic mediator in ischemic brain injury but can attenuate glutamate neurotoxicity in the retina and protect against light-induced or hereditary photoreceptor degeneration [39,40,41]. In AMD, IL-1β is secreted by retinal pigment epithelial (RPE) cells and CD68+ cells in choroidal neovascular membranes and is, therefore, a potential pro-angiogenic and neuroprotective or neurotoxic mediator in AMD [42].
Our research aims to evaluate the roles of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623), as well as the serum levels of these genetic markers, in the development of AMD and the efficacy of its treatment.

2. Results

Our current study involved 253 patients diagnosed with early AMD, 245 patients with exudative AMD, and 337 healthy controls. The control group was formed of 337 subjects that matched gender classification in the early and exudative AMD group structure; however, subjects of the control group were younger than the exudative AMD patients (p < 0.001), and further analysis was performed and adjusted by age (Table 1).

2.1. Hardy–Weinberg Equilibrium Analysis

A quality assessment based on Hardy–Weinberg equilibrium (HWE) analysis showed that the distribution of genotypes of VEGFA rs3024997, IL1B rs1143623, TNFRSF1B rs1061622, TNFRSF1A rs4149576, and ARMS2 rs10490924 did not deviate from HWE in the control group (p < 0.05).

2.2. VEGFA rs3024997, IL1B rs1143623, TNFRSF1B rs1061622, TNFRSF1A rs4149576, and ARMS2 rs10490924 Associations with Early and Exudative AMD

The frequencies of genotypes and alleles for the following SNPs were analyzed within the study groups: VEGFA rs3024997, IL1B rs1143623, TNFRSF1B rs1061622, TNFRSF1A rs4149576, and ARMS2 rs10490924.
For VEGFA rs3024997 (GG, GA, and AA), we observed a statistically significant difference between the exudative AMD and the control groups, with frequencies of 63.3%, 36.3%, and 0.4% in exudative AMD, respectively, compared to 55.5%, 38.3%, and 6.2% in the control group (p < 0.001). Furthermore, the A allele was less frequent in the exudative AMD group, accounting for 18.6% compared to 25.4% in the control group (p = 0.006). Similarly, for ARMS2 rs10490924 (GG, GT, and TT), we found a statistically significant difference between the exudative AMD and control groups, with frequencies of 31.8%, 43.3%, and 24,9%, compared to 54.3%, 38.3%, and 7.4% (p < 0.001). The T allele was more frequent in the exudative AMD group, accounting for 46.5% compared to 26.6% in the control group (p < 0.001) (Table 2).
Binary logistic regression analysis was conducted to assess the impact of selected SNPs on both early and exudative AMD. No statistically significant results were observed for the early AMD group after applying Bonferroni correction (Supplementary Table S1). However, in the exudative AMD group, VEGFA (rs3024997) exhibited a significant association. In the most robust model, the AA genotype was found to reduce the odds of exudative AMD by approximately 20-fold compared to the GG + GA genotypes (OR = 0.049, 95% CI: 0.006–0.381; p = 0.004). Similarly, ARMS2 (rs10490924) showed a significant association in the most robust model, where the TT genotype was associated with 4.2-fold increased odds of exudative AMD compared to the GG + GT genotypes (OR = 4.236, 95% CI: 2.508–7.155; p = 0.004) (Table 3).

Analysis of VEGFA rs3024997, IL1B rs1143623, TNFRSF1B rs1061622, TNFRSF1A rs4149576, and ARMS2 rs10490924 in Early and Exudative AMD in Female and Male Subgroups

We observed a statistically significant difference in the distribution of ARMS2 rs10490924 genotypes (GG, GT, and TT) between females with exudative AMD and the control groups, with frequencies of 30.3%, 46.5%, and 23.2% compared to 54.1%, 38.3%, and 7.7%, respectively (p < 0.001). Additionally, the T allele was more prevalent in the exudative AMD females’ group, accounting for 46.5% compared to 26.8% in the control group (p < 0.001) (Table 4).
Binary logistic regression analysis was performed to evaluate the impact of selected SNPs on females with early and exudative AMD. No statistically significant results were observed for early AMD females after applying Bonferroni correction (Supplementary Table S2). Meanwhile, in the exudative AMD group, ARMS2 (rs10490924) showed a significant association in the most robust genetic model, where each T allele was associated with 2.3-fold increased odds of exudative AMD in females (OR = 2.265 (1.617–3.172); p = 0.004) (Table 5).
Also, we observed a statistically significant difference in the distribution of ARMS2 rs10490924 genotypes (GG, GT, and TT) between males with exudative AMD and the control groups, with frequencies of 34.4%, 37.8%, and 27.8% compared to 54.1%, 38.3%, and 7.7%, respectively (p < 0.001). Additionally, the T allele was more prevalent in the exudative AMD male group, accounting for 46.7% compared to 26.1% in the control group (p < 0.001) (Table 6).
Binary logistic regression analysis was performed to evaluate the impact of selected SNPs on males with early and exudative AMD. No statistically significant results were observed after applying Bonferroni correction for males with early AMD (Supplementary Table S3). Meanwhile, in males with exudative AMD, ARMS2 (rs10490924) showed a significant association in the most robust model, where the TT genotype was associated with about 5-fold increased odds of exudative AMD in males compared to the GG + GT genotypes (OR = 5.049 (2.147–11.877); p < 0.001) (Table 7).

2.3. Serum IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 Associations with Early and Exudative AMD

Serum IL1B levels were measured in patients with early AMD vs. the control group (A) and in patients with exudative AMD vs. the control group (B). No statistically significant difference was found when comparing early AMD with the control group (median (IQR): 0.018 (0.008) vs. 0.018 (0.005), p = 0.890). However, statistically significantly lower serum IL1B levels were observed in the exudative AMD group compared to the controls (median (IQR): 0.017 (0.005) vs. 0.018 (0.005), respectively; p = 0.042, with a medium effect size, rrb = 0.323). The results are shown in Figure 1.
Serum TNFRSF1B levels were measured in patients with early AMD versus the control group (A) and in patients with exudative AMD versus the control group (B). A statistically significant difference was observed in the early AMD group (median (IQR): 1.359 (4.012) vs. 0.728 (2.331); p = 0.020, with a medium effect size, rrb = 371). However, no statistically significant difference was found when comparing exudative AMD with the control group (median (IQR): 0.858 (3.014) vs. 0.728 (2.331), p = 0.129). The results are shown in Figure 2.
Serum TNFRSF1A levels were measured in patients with early AMD vs. the control group (A) and in patients with exudative AMD vs. the control group (B). However, no statistically significant difference was found when comparing early AMD with the control group (median (IQR): 0.517 (0.662) vs. 0.436 (0.432), p = 0.627) or exudative AMD with the control group (median (IQR): 0.393 (0.633) vs. 0.436 (0.432), p = 0.883) (Supplementary Figure S1).
Serum ARMS2 levels were measured in patients with early AMD vs. the control group (A) and in patients with exudative AMD vs. the control group (B). However, no statistically significant difference was found when comparing early AMD with the control group (median (IQR): 0.220 (0.274) vs. 0.268 (0.224), p = 0.155) or exudative AMD with the control group (median (IQR): 0.152 (0.358) vs. 0.268 (0.224), p = 0.163) (Supplementary Figure S2).

2.4. Serum IL1B, TNFRS1B, TNFRS1A, and ARMS2 Levels and IL1B, TNFRS1B, TNFRS1A, and ARMS2 SNP Associations with AMD

Serum IL1B, TNFRS1B, TNFRS1A, and ARMS2 levels were compared among different genotypes for selected single-nucleotide polymorphisms. No statistically significant IL1B levels and IL1B rs1143623 or ARMS2 and ARMS2 rs10490924 genotype associations were revealed with early and exudative AMD occurrence (Supplementary Figures S3 and S4). However, early AMD patients with the GG genotype of TNFRS1B rs1061622 exhibited higher serum TNFRS1B levels compared to the control group (median (IQR): 3.315 (6.853) vs. 0.728 (2.460), p = 0.035, with a medium effect size, rrb = 0.412) (Figure 3B).
The analysis of TNFRS1A serum levels among different genotypes of TNFRS1A rs4149576 revealed that exudative AMD patients with the CC genotype exhibited lower serum TNFRS1A levels compared to the control group (median (IQR): 0.119 (0.241) vs. 0.503 (0.982), p = 0.033, with a large effect size, rrb = 0.833) (Figure 4A).

2.5. Response to Exudative AMD Treatment with Anti-VEGF Injections

The treatment response was evaluated for 115 patients with exudative AMD. The demographic and response to treatment parameters of the study group are summarized in Table 8. There was no difference in age or gender distribution between non-responders and responders.
We compared the median visual acuity (VA) between non-responders and responders using the Mann–Whitney U test, and the analysis showed that non-responders at the baseline had better VA than responders (0.465 (0.45) vs. 0.28 (0.26), p = 0.018, with a medium effect size, rrb = −0.336). On the other hand, while comparing the VA and CRT data before and after treatment using Wilcoxon signed-rank test, the VA was decreased after the treatment (0.465 (0.45) vs. 0.35 (0.35), p = 0.028, with a large effect size, rrb = −0.567) in non-responders and improved in responders (0.28 (0.26) vs. 375 (35), p < 0.001, with a medium effect size, rrb = −0.442) after the treatment.
The median CRT was significantly lower in responders than in non-responders after treatment (274 (95) vs. 329 (103), p = 0.032, with a medium effect size, rrb = 0.440). Also, CRT decreased after treatment statistically significantly only in responders (320.5 (113) vs. 274 (95), p < 0.001, with a strong effect size, rrb = −0.072), but not in non-responders) (Table 8).

2.6. Single-Nucleotide Polymorphism Associations with Exudative AMD Treatment Response

Binomial logistic regression analysis was performed to analyze the association between all SNPs and anti-VEGF treatment response, and only one SNP was found to be linked to treatment response with anti-VEGF. The TNFRSF1B rs1061622 T allele was found to be associated with a better response to anti-VEGF treatment under the dominant (OR  =  4.302; 95% CI: 1.181–15.674; p = 0.027) and additive (OR  =  3.999; 95% CI: 1.176–13.602; p = 0.026) genetic models (Table 9).

2.7. Serum IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 Associations with the Treatment Response to Anti-VEGF Treatment

A comparison of serum IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 was conducted among non-responders and responders but no statistically significant differences were observed comparing these groups (p > 0.05). VEGFA serum concentrations were measured and described in our previous publication [43] but did not show statistical differences between study groups.

3. Discussion

Age-related macular degeneration causes irreversible vision loss, and targeted anti-vascular endothelial growth factor therapy is now the most common and effective treatment. This paper aims to discuss whether genetic polymorphisms of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623) and their serum biomarkers could confer susceptibility to early and exudative AMD with a response to anti-VEGF treatment.
Most patients with active neovascular AMD are offered treatment with intravitreal anti-VEGF agents, such as Bevacizumab (Avastin, Genentech, South San Francisco, CA, USA), Ranibizumab (Lucentis, Genentech, South San Francisco, CA, USA), Aflibercept (Eylea, Regeneron, Tarrytown, NY, USA), Brolucizumab (Beovu, Novartis, Basel, Switzerland), and Faricimab (Vabysmo, Genentech, South San Francisco, CA, USA). Considerable diversity in treatment response among patients is evident, with some individuals responding more favorably to specific anti-VEGF agents than others. This suggests the presence of a pharmacogenetic effect [44,45]. Research findings suggest a higher prevalence of AMD among first-degree relatives of affected individuals, with an odds ratio of 2.4 [46]. Clinical genetic testing is an important part of emerging personalized medicine [47]. Improved risk stratification through clinical genetic testing can enhance patient outcomes by optimizing resource allocation and facilitating the selection of the most suitable treatment options [48,49]. Genetic variants can account for up to 70% of the clinical variability in AMD, indicating the potential for personalized medical approaches to address this disease effectively [18,48,50].
Suboptimal responses and limited effectiveness over time can significantly impact patients, leading to poor vision outcomes. Despite the positive outcomes seen in many patients, around 25–35% of individuals with nAMD experience suboptimal responses to current anti-VEGF treatments, encounter delayed treatment failure or necessitate intensive and frequent IVT therapy [51,52]. Additionally, the necessity for recurrent treatments for nAMD poses a considerable burden on healthcare systems, patients, and their caregivers. Moreover, while current anti-VEGF treatments are generally effective, they are also associated with certain adverse events that can significantly impact eyesight, albeit infrequently. For instance, endophthalmitis, a severe complication, occurs in approximately 1 in 3500 injections [53]. Another significant potential complication of anti-VEGF therapy is the risk of intraocular inflammation. In severe cases, this inflammation can lead to irreversible vision loss [54]. Furthermore, a temporary increase in intraocular pressure is frequently noticed shortly after IVT injection of all anti-VEGF agents. Additionally, a temporary increase in intraocular pressure is commonly observed shortly after intravitreal injection of all anti-VEGF agents [55].
Repeated use of anti-VEGF treatments can also lead to adverse effects. For instance, macular atrophy, an advanced phenotype of nAMD, potentially linked to long-term anti-VEGF use, may result in permanent vision loss [56]. Among the 35% who do not respond optimally to therapy, over 10% experience deterioration despite treatment, while an additional 25% show no signs of improvement [7].
Gene therapy for nAMD is challenging due to the complexity of the genes associated with the condition.

3.1. Genetic Variants and AMD

In our study, we found potential genetic links to AMD, specifically exudative AMD.
VEGFA rs3024997 and ARMS2 rs10490924: Our analysis demonstrated a significant difference in genotype and allele frequencies of VEGFA rs3024997 and ARMS2 rs10490924 between exudative AMD patients and healthy controls. Specifically, the AA genotype of VEGFA rs3024997 was associated with a reduced risk of exudative AMD, while the TT genotype of ARMS2 rs10490924 was linked to an increased risk. These findings corroborate previous studies indicating the role of these SNPs in AMD susceptibility. A Japanese study reported that the SNP rs699946 in the VEGFA gene was associated with a better visual response after 12 months of treatment with bevacizumab [57]. An alternative study concluded that the SNP rs3025000 correlated with enhanced visual outcomes following six months of anti-VEGF treatment [58].
Polymorphisms in the VEGFA gene regulate VEGF expression and, thus, its angiogenic properties [59]. Therefore, it is possible that differing levels of VEGF expression could lead to varied responses to anti-VEGF drugs. Studies have explored genetic variations within the VEGFA and VEGFR2 genes in small cohorts to understand their influence on anti-VEGF treatment outcomes, showing mixed results. For example, one study noted a trend toward better visual outcomes after six months of ranibizumab treatment in patients with the risk genotype for the VEGFA SNP rs1413711 compared to those with the non-risk genotype [60]. A recent study investigating two VEGFA SNPs and their relationship with ranibizumab response found that rs699947 influences early functional outcomes [61]. However, other genetic variations related to blood vessel growth regulation may still be linked to treatment response [59]. Zhang and colleagues analyzed 21 studies to examine the link between the ARMS2 gene and the response to anti-VEGF treatment in advanced AMD. They found that individuals with the G allele for ARMS2 A69S had a more favorable outcome with anti-VEGF drugs, especially among East Asian patients. Further validation through large clinical trials is needed [62].
TNFRSF1A and TNFRSF1B: Our results also revealed significant associations between TNFRSF1B rs1061622 and exudative AMD, where the GG genotype was associated with higher serum levels of TNFRSF1B and increased odds of AMD. This aligns with the literature suggesting that TNFRSF1B can influence inflammatory processes and cell death, pivotal in AMD pathogenesis [63,64,65,66,67]. TNFRSF1A and TNFRSF1B are two proteins that belong to the TNF-receptor superfamily. They are mainly secreted by macrophages and can induce cell death of specific tumor cell lines. They are also potent pyrogens that can cause fever by direct action or stimulating interleukin-1 secretion. Moreover, under certain conditions, they can promote cell proliferation and differentiation [68]. The proteins encoded by these genes form a heterocomplex with TNF-receptor 1, which mediates the recruitment of two anti-apoptotic proteins. Although their function is not entirely understood, they are believed to be associated with anti-apoptotic signals. Knockout studies in mice have also suggested that these proteins protect neurons from apoptosis by stimulating antioxidative pathways. The role of TNFRSF1A in AMD remains unclear, but its association with other diseases and SNPs highlights its potential involvement in AMD [69,70,71].
IL1B: Our study found that IL1B’s association with AMD was less pronounced than in other studies. We observed significantly lower IL1B levels in the exudative AMD group compared to the controls, with no significant associations found with specific SNPs in IL1B. This contrasts with other studies linking IL1B with AMD through its inflammatory role [72,73]. IL-1β, a pro-inflammatory cytokine, plays a crucial role in immune responses, inflammation, and various disease processes. It has been implicated in retinal degenerative diseases and choroidal neovascularization (CNV). Studies have shown that inhibition of IL-1β can ameliorate these conditions [74]. Furthermore, IL-1β has also been associated with abnormal angiogenesis processes, indicating its role in promoting diverse cellular signaling pathways. The pro-inflammatory cytokine IL-1β has been shown to promote angiogenesis and can have a neurotoxic or neuroprotective effect. To determine whether IL-1β plays a role in CNV and retinal degeneration, the team conducted a study to analyze the expression of IL-1β in mice with laser-induced CNV and light-induced retinal degeneration in rats and mice [75]. IL-1β is induced in acute and chronic brain degenerative diseases and the retina. This association could result from the activation of inflammation, which further stimulates RPE cells to trigger photoreceptor degeneration and neovascularization. IL-1β was involved in the abnormal angiogenesis process. This could activate different abnormal angiogenesis processes through distinct cell signaling pathways. The variability in findings may be attributed to different abnormal angiogenesis processes through distinct cell signaling pathways, as well as differences in study populations or methodologies [39].

3.2. Serum Biomarkers and AMD

IL1B, TNFRSF1A, and TNFRSF1B: IL1B levels were significantly lower in exudative AMD patients than in the controls, though no significant differences were found between early AMD and controls. The lower levels of IL1B in exudative AMD patients might reflect a complex interplay of inflammatory processes rather than a straightforward biomarker of AMD severity. TNFRSF1B levels were significantly higher in early AMD patients than controls but not in exudative AMD patients. This suggests a potential differential role of TNFRSF1B in early versus advanced stages of AMD. This aligns with the proposed protective role of TNFRSF1B in neuronal apoptosis and its varying effects across disease stages [70,71]. In published databases, we did not uncover any studies examining the serum correlations of TNFRSF1A, TNFRSF1B, and IL1B1 with AMD.
ARMS2 and VEGFA: Despite the known genetic associations with AMD risk, we did not find significant differences in serum ARMS2 levels between AMD patients and controls. This may indicate that ARMS2’s contribution to AMD is more strongly associated with genetic factors than circulating protein levels. Battu et al. [76] found that ARMS2 serum levels were significantly elevated in the AMD group compared to the control group. The highest levels of ARMS2 and VEGF proteins were recorded for the wet AMD sub-group. The study results endorsed a significant positive correlation between the following molecules: ARMS2 and VEGF (r = 0.925, p < 0.0001), COL8A1 and VEGF (r = 0.879, p < 0.0001), and RAD51B and VEGF (r = 0.691, p < 0.0001) [76]. VEGFA serum levels, as well as VEGFA SNPs, are widely studied in AMD patients. While we did not find any statistical differences in VEGF-A serum levels between the exudative AMD patients and controls, we confirmed the results from several other studies, which included total AMD patients or only exudative AMD patients, consisting of 27 to 71 samples per group in different populations [76,77,78,79,80,81,82,83].

3.3. Response to Anti-VEGF Treatment

Our study also examined the response to anti-VEGF treatment in exudative AMD patients. Responders had improved visual acuity and reduced CRT post-treatment compared to non-responders. This is consistent with the expected clinical outcomes of effective anti-VEGF therapy. Interestingly, non-responders initially had better baseline visual acuity, which suggests that pre-treatment visual acuity might influence the response to treatment. However, the lack of significant differences in serum biomarkers between responders and non-responders indicates that these markers may not be reliable predictors of treatment efficacy.

4. Materials and Methods

4.1. Ethics

This study adhered to the guidelines of the Declaration of Helsinki and received approval from the Kaunas Regional Biomedical Research Ethics Committee of the Lithuanian University of Health Sciences (approval number BE-2-/48). Informed consent was obtained from all participants.

4.2. Study Design and Structure

The study subjects were admitted to the Ophthalmology Department at the Hospital of the Lithuanian University of Health Sciences in Kaunas from 2014 to 2023 and underwent an ophthalmological evaluation. Information about their health and other diseases was collected during examinations by general practitioners and from medical records.

4.3. SNP Selection

The single-nucleotide polymorphisms (SNPs) investigated in our study were carefully selected based on their different and various associations with disease mechanisms. We thoroughly reviewed other research papers on these polymorphisms and their links to various diseases. After extensive analysis, we identified the most relevant SNPs to study in relation to age-related macular degeneration, its stages, and potential treatments.
Many researchers have investigated AMD from the perspective of the whole genome. A potent technique for locating genetic variations that may be linked to AMD is whole exome sequencing (WES), which is performed on genomic areas of the genome that code for proteins. Typically, the goal of this research is to find both common and uncommon genetic variations that may affect the disease’s course or chance of acquiring AMD. Identifying novel risk loci, validating established genetic connections, and gaining insights into the molecular mechanisms underlying AMD are some of the major outcomes of whole exome studies in AMD, as ARMS2, VEGFA, TNFRSF [84,85,86], and IL1B1 were selected based on association with the pathogenetic mechanism of the chronic inflammatory process [87].

4.4. Study Group Formation

All study participants underwent a comprehensive ophthalmological evaluation during which data on general health and comorbidities were systematically collected. Informed consent was obtained from each participant prior to their inclusion in the study. The participants were then stratified into two groups: those diagnosed with age-related macular degeneration (AMD) and the control subjects. AMD patients were assessed by an ophthalmologist following previously established guidelines [88]. Detailed inclusion and exclusion criteria for both AMD patients and control subjects have been meticulously outlined in our earlier publication [88].

4.5. Exudative AMD Response to Anti-VEGF Injection Treatment

The efficacy of anti-VEGF treatments (Ranibizumab, Aflibercept, Bevacizumab) was evaluated in patients with exudative age-related macular degeneration (AMD) characterized by exudative or hemorrhagic macular features. Patients who had not previously received intravitreal anti-VEGF injections were followed for three to six months post-treatment. Central macular thickness (CMT) and best corrected visual acuity (BCVA) were measured before treatment and at three and six months afterward. Patients were categorized as responders or non-responders based on clinical OCT and BCVA data. Detailed methods and criteria for response evaluation, including definitions of structural changes, visual acuity deterioration, and patient categorization, were described in a previous publication [88].

4.6. DNA Extraction from Peripheral Venous Blood and Genotyping

Deoxyribonucleic acid (DNA) extraction and genotyping of selected single-nucleotide polymorphisms (SNPs)—VEGFA rs3024997, IL1B rs1143623, TNFRSF1B rs1061622, TNFRSF1A rs4149576, and ARMS2 rs10490924—were performed at the Laboratory of Ophthalmology, Neuroscience Institute, Lithuanian University of Health Sciences, using predesigned TaqManTM Genotyping assays (Thermo Fisher Scientific, Pleasanton, CA, USA) following the manufacturer’s recommendations.

4.7. Serum Protein Concentration Measurement

To prepare the serum, blood was drawn from peripheral veins and allowed to incubate at room temperature for 30 min before being centrifuged. After centrifugation, the serum was carefully separated from the cell pellet, transferred into 2 mL tubes, and stored at −80 °C until analysis. The levels of IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 in the serum of both AMD patients and control subjects were measured according to the manufacturer’s instructions.
However, the VEGFA serum concentrations were measured and described in our previous publication [43].

4.8. Statistical Analysis

The statistical analysis was conducted using SPSS/W 29.0 software (Statistical Package for the Social Sciences for Windows, Inc., Chicago, IL, USA). The normality of continuous data (age, BCVA, and CMT) was assessed using the Shapiro–Wilk test. Continuous variables that did not follow the normal distribution model were expressed as median with interquartile range (IQR) and compared using the non-parametric Mann–Whitney U test. Normally distributed data were expressed as mean with standard deviation (SD) and compared using the Student T test. The nonparametric Wilcoxon signed-rank test was used to compare differences in the BCVA and CMT before and after treatment. Statistical significance was defined as p < 0.05. Rank-biserial correlation (rrb) was used to measure the effect size of particular results.
Categorical data (sex and genotype distributions) were presented as absolute numbers with percentages in parentheses and compared between groups using the Pearson’s chi-squared test (χ2).
The influence of gene polymorphisms on early and exudative AMD was evaluated using binomial logistic regression analysis. We ensured that the key assumptions underlying our binomial regression analysis were satisfied. Specifically, the analysis was conducted on a dichotomous dependent variable, the independence of observations was maintained by design with no repeated measures or clustering, and the sample size was sufficient to yield stable and reliable estimates. Therefore, we confirm that the assumptions for the applied binomial regression analysis were adequately met. The results were presented as odds ratio (OR) with a 95% confidence interval (CI), adjusted for age in the exudative AMD groups. Genetic models (codominant, dominant, recessive, and overdominant) were employed, with the additive model assessing the effects of each minor allele on AMD. The best genetic model was selected based on the Akaike information criterion (AIC), favoring models with the lowest AIC values. Bonferroni correction was applied to account for multiple association calculations, resulting in an adjusted significance threshold for multiple comparisons of α = 0.01 (0.05/5, as five different SNPs were analyzed).

5. Conclusions

In published databases, we did not uncover any studies examining the serum correlations of TNFRSF1A, TNFRSF1B, and IL1B1 with age-related macular degeneration. In conclusion, identifying genetic variations in ARMS2, VEGFA, TNFRSF1A, TNFRSF1B, and IL1B1 provides valuable insights into the susceptibility to and treatment outcomes for age-related macular degeneration. Our analysis emphasizes the importance of personalized medicine, where genetic profiling could help choose and optimize anti-VEGF therapies. However, despite significant progress, further research through large studies is necessary to confirm these associations and fully comprehend the pharmacogenetic implications. Ultimately, integrating genetic testing into clinical practice could improve the precision and effectiveness of AMD treatment, potentially reducing the burden of this disabling condition.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25179750/s1.

Author Contributions

Conceptualization, A.V., D.C. and R.L.; methodology, A.V., D.C., A.B., M.D., R.L., L.K. and D.Z.; formal analysis, A.V., D.C., M.D. and R.L.; investigation, A.V., D.C., A.V., M.D., G.G.-V. and R.L.; resources, D.Z. and R.L.; data curation, D.C. and A.V.; writing—A.V., G.G.-V. and D.C.; writing—review and editing, A.V., D.C., R.L., L.K. and D.Z.; visualization, A.V., M.D., A.B. and D.C.; supervision, R.L.; project administration, R.L.; funding acquisition, D.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Research Council of Lithuania under the initiative of Re-searcher Group Projects, grant no. S-MIP-23-96.

Institutional Review Board Statement

This study was conducted in accordance with the Declara-tion of Helsinki and approved by the Biomedical Research Ethics Committee, Lithuanian Univer-sity of Health Sciences (no. BE-2-/48). All study participants signed the informed consent form. An ophthalmological evaluation was performed for all the study subjects admitted to the ophthalmological assessment at the Ophthalmology Department, Hospital of Lithuanian University of Health Sciences, from 2014 to 2023. Their overall health data were obtained during the general practitioner examination or collected from medical records. This research study was conducted at the Laboratory of Ophthalmology, Neuroscience Institute, LUHS.

Informed Consent Statement

Informed consent was obtained from all participants in the study.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Thomas, C.J.; Mirza, R.G.; Gill, M.K. Age-Related Macular Degeneration. Med. Clin. N. Am. 2021, 105, 473–491. [Google Scholar] [CrossRef] [PubMed]
  2. Owen, C.G.; Jarrar, Z.; Wormald, R.; Cook, D.G.; Fletcher, A.E.; Rudnicka, A.R. The Estimated Prevalence and Incidence of Late-Stage Age-Related Macular Degeneration in the UK. Br. J. Ophthalmol. 2012, 96, 752–756. [Google Scholar] [CrossRef] [PubMed]
  3. Klein, R.; Klein, B.E.K.; Linton, K.L.P. Prevalence of Age-Related Maculopathy: The Beaver Dam Eye Study. Ophthalmology 1992, 99, 933–943. [Google Scholar] [CrossRef] [PubMed]
  4. Gass, J.D. Pathogenesis of Disciform Detachment of the Neuroepithelium. Am. J. Ophthalmol. 1967, 63, 1–139. [Google Scholar]
  5. Rosenfeld, P.J.; Brown, D.M.; Heier, J.S.; Boyer, D.S.; Kaiser, P.K.; Chung, C.Y.; Kim, R.Y. Ranibizumab for Neovascular Age-Related Macular Degeneration. N. Engl. J. Med. 2006, 355, 1419–1431. [Google Scholar] [CrossRef]
  6. Heier, J.S.; Brown, D.M.; Chong, V.; Korobelnik, J.F.; Kaiser, P.K.; Nguyen, Q.D.; Kirchhof, B.; Ho, A.; Ogura, Y. Intravitreal Aflibercept (VEGF Trap-Eye) in Wet Age-Related Macular Degeneration. Ophthalmology 2012, 119, 2537–2548. [Google Scholar] [CrossRef]
  7. Martin, D.F.; Maguire, M.G.; Fine, S.L.; Ying, G.S.; Jaffe, G.J.; Grunwald, J.E.; Toth, C.; Redford, M.; Ferris, F.L., 3rd. Ranibizumab and Bevacizumab for Treatment of Neovascular Age-Related Macular Degeneration: Two-Year Results. Ophthalmology 2012, 119, 1388–1398. [Google Scholar] [CrossRef]
  8. Fisher, S.A.; Abecasis, G.R.; Yashar, B.M.; Zareparsi, S.; Swaroop, A.; Iyengar, S.K.; Klein, B.E.K.; Klein, R.; Lee, K.E.; Majewski, J.; et al. Meta-Analysis of Genome Scans of Age-Related Macular Degeneration. Hum. Mol. Genet. 2005, 14, 2257–2264. [Google Scholar] [CrossRef]
  9. Klein, M.L.; Schultz, D.W.; Edwards, A.; Matise, T.C.; Rust, K.; Berselli, C.B.; Trzupek, K.; Weleber, R.G.; Ott, J.; Acott, T.S.; et al. Age-Related Macular Degeneration: Clinical Features in a Large Family and Linkage to Chromosome 1q. Arch. Ophthalmol. 1998, 116, 1082–1088. [Google Scholar] [CrossRef]
  10. Majewski, J.; Schultz, D.W.; Weleber, R.G.; Schain, M.B.; Edwards, A.O.; Matise, T.C.; Acott, T.S.; Ott, J.; Klein, M.L. Age-Related Macular Degeneration--A Genome Scan in Extended Families. Am. J. Hum. Genet. 2003, 73, 540–550. [Google Scholar] [CrossRef]
  11. Schick, J.H.; Iyengar, S.K.; Klein, B.E.; Klein, R.; Reading, K.; Liptak, R.; Millard, C.; Lee, K.E.; Elston, R.C. A Whole-Genome Screen of a Quantitative Trait of Age-Related Maculopathy in Sibships from the Beaver Dam Eye Study. Am. J. Hum. Genet. 2003, 72, 1412–1424. [Google Scholar] [CrossRef] [PubMed]
  12. Abecasis, G.R.; Yashar, B.M.; Zhao, Y.; Zareparsi, S.; Branham, K.E.; Dewan, A.; Stanwyck, L.K.; Forman, J.J.; Scheetz, T.E.; Jacobson, S.G.; et al. Age-Related Macular Degeneration: A High-Resolution Genome Scan for Susceptibility Loci in a Population Enriched for Late-Stage Disease. Am. J. Hum. Genet. 2004, 74, 482–494. [Google Scholar] [CrossRef] [PubMed]
  13. Iyengar, S.K.; Song, D.; Klein, B.E.K.; Klein, R.; Schick, J.H.; Liptak, R.; Millard, C.; Lee, K.E.; Reading, K.; McCarty, C.A.; et al. Dissection of Genomewide-Scan Data in Extended Families Reveals a Major Locus and Oligogenic Susceptibility for Age-Related Macular Degeneration. Am. J. Hum. Genet. 2004, 74, 20–39. [Google Scholar] [CrossRef] [PubMed]
  14. Schmidt, S.; Scott, W.K.; Postel, E.A.; Agarwal, A.; Hauser, E.R.; DeAngelis, M.; Klaver, C.C.; Barbazetto, I.A.; Piermarocchi, S.; Bracken, M.B.; et al. Ordered Subset Linkage Analysis Supports a Susceptibility Locus for Age-Related Macular Degeneration on Chromosome 16p12. BMC Genet. 2004, 5, 18. [Google Scholar] [CrossRef]
  15. Kenealy, S.J.; Schmidt, S.; Agarwal, A.; Postel, E.A.; Hauser, E.R.; Pericak-Vance, M.A.; Haines, J.L.; Gilbert, J.R. Linkage Analysis for Age-Related Macular Degeneration Supports a Gene on Chromosome 10q26. Mol. Vis. 2004, 10, 57–61. [Google Scholar] [CrossRef]
  16. Jun, G.; Klein, B.E.K.; Klein, R.; Fuchs, M.; Tomany, S.C.; Lee, K.E.; Iyengar, S.K. Genome-Wide Analyses Demonstrate Novel Loci that Predispose to Drusen Formation. Investig. Ophthalmol. Vis. Sci. 2005, 46, 3081–3088. [Google Scholar] [CrossRef]
  17. Silveira, A.C.; Morrison, M.A.; Ji, F.; Xu, H.; Nichols, C.; Qin, G.; Miller, E.N.; DeAngelis, M.M. Convergence of Linkage, Gene Expression, and Association Data Demonstrates the Influence of the RAR-Related Orphan Receptor Alpha (RORA) Gene on Neovascular AMD: A Systems Biology-Based Approach. Vision Res. 2010, 50, 698–715. [Google Scholar] [CrossRef]
  18. Fritsche, L.G.; Igl, W.; Bailey, J.N.C.; Grassmann, F.; Sengupta, S.; Bragg-Gresham, J.L.; Burdon, K.P.; Hebbring, S.J.; Wen, C.; Gorski, M.; et al. A Large Genome-Wide Association Study of Age-Related Macular Degeneration Highlights Contributions of Rare and Common Variants. Nat. Genet. 2016, 48, 134–143. [Google Scholar] [CrossRef]
  19. Seddon, J.M.; Cote, J.; Page, W.F.; Aggen, S.H.; Neale, M.C. The US twin study of age-related macular degeneration: Relative roles of genetic and environmental influences. Arch. Ophthalmol. 2005, 123, 321–327. [Google Scholar] [CrossRef]
  20. Colijn, J.M.; Meester-Smoor, M.; Verzijden, T.; de Breuk, A.; Silva, R.; Merle, B.M.J.; Cougnard-Grégoire, A.; Hoyng, C.B.; Fauser, S.; Coolen, A.; et al. Genetic risk, lifestyle, and age-related macular degeneration in Europe: The EYE-RISK consortium. Ophthalmology 2021, 128, 1039–1049. [Google Scholar] [CrossRef]
  21. de Breuk, A.; Acar, I.E.; Kersten, E.; Schijvenaars, M.M.V.A.P.; Colijn, J.M.; Haer-Wigman, L.; Bakker, B.; de Jong, S.; Meester-Smoor, M.A.; Verzijden, T.; et al. Development of a genotype assay for age-related macular degeneration: The EYE-RISK consortium. Ophthalmology 2020, 128, 1604–1617. [Google Scholar] [CrossRef] [PubMed]
  22. Lambert, N.G.; ElShelmani, H.; Singh, M.K.; Mansergh, F.C.; Wride, M.A.; Padilla, M.; Keegan, D.; Hogg, R.E.; Ambati, B.K. Risk factors and biomarkers of age-related macular degeneration. Prog. Retin. Eye Res. 2016, 54, 64–102. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, J.J.; Buitendijk, G.H.; Rochtchina, E.; Lee, K.E.; Klein, B.E.; van Duijn, C.M.; Flood, V.M.; Meuer, S.M.; Attia, J.; Myers, C.; et al. Genetic susceptibility, dietary antioxidants, and long-term incidence of age-related macular degeneration in two populations. Ophthalmology 2014, 121, 667–675. [Google Scholar] [CrossRef] [PubMed]
  24. Farinha, C.; Barreto, P.; Coimbra, R.; Cachulo, M.L.; Melo, J.B.; Cunha-Vaz, J.; Lechanteur, Y.; Hoyng, C.B.; Silva, R. Common and rare genetic risk variants in age-related macular degeneration and genetic risk score in the Coimbra eye study. Acta Ophthalmol. 2023, 101, 185–199. [Google Scholar] [CrossRef]
  25. Toomey, C.B.; Johnson, L.V.; Bowes Rickman, C. Complement factor H in AMD: Bridging genetic associations and pathobiology. Progress Retin. Eye Res. 2018, 62, 38–57. [Google Scholar] [CrossRef]
  26. Kanda, A.; Chen, W.; Othman, M.; Branham, K.E.; Brooks, M.; Khanna, R.; He, S.; Lyons, R.; Abecasis, G.R.; Swaroop, A. A Variant of Mitochondrial Protein LOC387715/ARMS2, Not HTRA1, is Strongly Associated with Age-Related Macular Degeneration. Proc. Natl. Acad. Sci. USA 2007, 104, 16227–16232. [Google Scholar] [CrossRef]
  27. Wang, G.; Scott, W.K.; Whitehead, P.; Court, B.L.; Kovach, J.L.; Schwartz, S.G.; Postel, E.A.; DeAngelis, M.M.; Haines, J.L.; Pericak-Vance, M.A.; et al. A Novel ARMS2 Splice Variant is Identified in Human Retina. Exp. Eye Res. 2012, 94, 187–191. [Google Scholar] [CrossRef]
  28. Andreoli, M.T.; Morrison, M.A.; Kim, B.J.; Chen, L.; Adams, S.M.; Miller, J.W.; Kim, I.K.; Seddon, J.M.; Hageman, G.S.; DeAngelis, M.M. Comprehensive Analysis of Complement Factor H and LOC387715/ARMS2/HTRA1 Variants with Respect to Phenotype in Advanced Age-Related Macular Degeneration. Am. J. Ophthalmol. 2009, 148, 869–874. [Google Scholar] [CrossRef]
  29. Hansen, T.F.; Jakobsen, A. Clinical Implications of Genetic Variations in the VEGF System in Relation to Colorectal Cancer. Pharmacogenomics 2011, 12, 1681–1693. [Google Scholar] [CrossRef]
  30. Gupta, S.; Johnson, S.H.; Vasmatzis, G.; Aydin, H.; von Keitz, B.; Boylan, K.L.; Fan, J.; Lindgren, C.M.; Klee, E.W.; Eckloff, B.; et al. TFEB-VEGFA (6p21.1) Co-Amplified Renal Cell Carcinoma: A Distinct Entity with Potential Implications for Clinical Management. Mod. Pathol. 2017, 30, 998–1012. [Google Scholar] [CrossRef]
  31. Harper, S.J.; Bates, D.O. VEGF-A Splicing: The Key to Anti-Angiogenic Therapeutics? Nat. Rev. Cancer 2008, 8, 880–887. [Google Scholar] [CrossRef] [PubMed]
  32. Boltz, A.; Ruiß, M.; Jonas, J.B.; Shen, Y.; Schmid-Kubista, K.E.; Koch, F.; Fischer, M.; Meyer, C.H.; Holz, F.G.; Kampik, A.; et al. Role of Vascular Endothelial Growth Factor Polymorphisms in the Treatment Success in Patients with Wet Age-Related Macular Degeneration. Ophthalmology 2012, 119, 1615–1620. [Google Scholar] [CrossRef] [PubMed]
  33. Rittore, C.; Méchin, D.; Sanchez, E.; Marinèche, L.; Ea, V.; Soler, S.; Vereecke, M.; Mallavialle, A.; Richard, E.; Duroux-Richard, I.; et al. TNFR1-d2 Carrying the p.(Thr79Met) Pathogenic Variant is a Potential Novel Actor of TNFα/TNFR1 Signalling Regulation in the Pathophysiology of TRAPS. Sci. Rep. 2021, 11, 4172. [Google Scholar] [CrossRef] [PubMed]
  34. Alshevskaya, A.; Zhukova, J.; Kireev, F.; Lopatnikova, J.; Evsegneeva, I.; Demina, D.; Nepomniashchikh, V.; Gladkikh, V.; Karaulov, A.; Sennikov, S. Redistribution of TNF Receptor 1 and 2 Expression on Immune Cells in Patients with Bronchial Asthma. Cells 2022, 11, 1736. [Google Scholar] [CrossRef]
  35. Lainez, B.; Fernandez-Real, J.M.; Romero, X.; Esplugues, E.; Cañete, J.D.; Ricart, W.; Engel, P. Identification and Characterization of a Novel Spliced Variant that Encodes Human Soluble Tumor Necrosis Factor Receptor 2. Int. Immunol. 2004, 16, 169–177. [Google Scholar] [CrossRef]
  36. Dinarello, C.A. Immunological and Inflammatory Functions of the Interleukin-1 Family. Annu. Rev. Immunol. 2009, 27, 519–550. [Google Scholar] [CrossRef]
  37. Isoda, K.; Sawada, S.; Ishigami, N.; Matsuki, T.; Miyazaki, K.; Kusuhara, M.; Iwakura, Y.; Ohsuzu, F. Lack of Interleukin-1 Receptor Antagonist Modulates Plaque Composition in Apolipoprotein E-Deficient Mice. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 1068–1073. [Google Scholar] [CrossRef]
  38. BenEzra, D.; Hemo, I.; Maftzir, G. In Vivo Angiogenic Activity of Interleukins. Arch. Ophthalmol. 1990, 108, 573–576. [Google Scholar] [CrossRef]
  39. Yamasaki, Y.; Matsuura, N.; Shozuhara, H.; Onodera, H.; Itoyama, Y.; Kogure, K. Interleukin-1 as a Pathogenetic Mediator of Ischemic Brain Damage in Rats. Stroke 1995, 26, 676–680. [Google Scholar] [CrossRef]
  40. Namekata, K.; Harada, C.; Guo, X.; Kikushima, K.; Kimura, A.; Fuse, N.; Mitamura, Y.; Kohyama, K.; Matsumoto, Y.; Tanaka, K.; et al. Interleukin-1 Attenuates Normal Tension Glaucoma-Like Retinal Degeneration in EAAC1-Deficient Mice. Neurosci. Lett. 2009, 465, 160–164. [Google Scholar] [CrossRef]
  41. LaVail, M.M.; Unoki, K.; Yasumura, D.; Matthes, M.T.; Yancopoulos, G.D.; Steinberg, R.H. Multiple Growth Factors, Cytokines, and Neurotrophins Rescue Photoreceptors from the Damaging Effects of Constant Light. Proc. Natl. Acad. Sci. USA 1992, 89, 11249–11253. [Google Scholar] [CrossRef] [PubMed]
  42. Oh, H.; Takagi, H.; Takagi, C.; Suzuma, K.; Otani, A.; Ishida, K.; Matsumura, M.; Ogura, Y.; Honda, Y. The Potential Angiogenic Role of Macrophages in the Formation of Choroidal Neovascular Membranes. Investig. Ophthalmol. Vis. Sci. 1999, 40, 1891–1898. [Google Scholar]
  43. Vilkeviciute, A.; Cebatoriene, D.; Kriauciuniene, L.; Liutkeviciene, R. VEGFA Haplotype and VEGF-A and VEGF-R2 Protein Associations with Exudative Age-Related Macular Degeneration. Cells 2022, 11, 996. [Google Scholar] [CrossRef] [PubMed]
  44. Chaudhary, V.; Brent, M.; Lam, W.-C.; Devenyi, R.; Teichman, J.; Mak, M.; Barbosa, J.; Kaur, H.; Carter, R.; Farrokhyar, F.; et al. Genetic Risk Evaluation in Wet Age-Related Macular Degeneration Treatment Response. Ophthalmologica 2016, 236, 88–94. [Google Scholar] [CrossRef]
  45. Asten, F.; Rovers, M.M.; Lechanteur, Y.T.E.; Smailhodzic, D.; Muether, P.S.; Chen, J.; den Hollander, A.I.; Fauser, S.; Hoyng, C.B.; Jan van der Wilt, G.; et al. Predicting Non-Response to Ranibizumab in Patients with Neovascular Age-Related Macular Degeneration. Ophthalmic Epidemiol. 2014, 21, 347–355. [Google Scholar] [CrossRef]
  46. Seddon, J.M.; Ajani, U.A.; Mitchell, B.D. Familial Aggregation of Age-Related Maculopathy. Am. J. Ophthalmol. 1997, 123, 199–206. [Google Scholar] [CrossRef]
  47. Abul-Husn, N.S.; Owusu Obeng, A.; Sanderson, S.C.; Gottesman, O.; Scott, S.A. Implementation and Utilization of Genetic Testing in Personalized Medicine. Pharmacogenomics Pers. Med. 2014, 7, 227–240. [Google Scholar]
  48. Stradiotto, E.; Allegrini, D.; Fossati, G.; Raimondi, R.; Sorrentino, T.; Tripepi, D.; Barone, G.; Inforzato, A.; Romano, M.R. Genetic Aspects of Age-Related Macular Degeneration and Their Therapeutic Potential. Int. J. Mol. Sci. 2022, 23, 13280. [Google Scholar] [CrossRef]
  49. Warwick, A.; Lotery, A. Genetics and Genetic Testing for Age-Related Macular Degeneration. Eye 2018, 32, 849–857. [Google Scholar] [CrossRef]
  50. Cascella, R.; Strafella, C.; Caputo, V.; Errichiello, V.; Zampatti, S.; Milano, F.; Potenza, S.; Mauriello, S.; Novelli, G.; Ricci, F.; et al. Towards the Application of Precision Medicine in Age-Related Macular Degeneration. Prog. Retin. Eye Res. 2018, 63, 132–146. [Google Scholar] [CrossRef]
  51. Spooner, K.; Hong, T.; Wijeyakumar, W.; Chang, A.A. Switching to Aflibercept among Patients with Treatment-Resistant Neovascular Age-Related Macular Degeneration: A Systematic Review with Meta-Analysis. Clin. Ophthalmol. 2017, 11, 161–177. [Google Scholar] [CrossRef] [PubMed]
  52. Broadhead, G.K.; Hong, T.; Chang, A.A. Treating the Untreatable Patient: Current Options for the Management of Treatment-Resistant Neovascular Age-Related Macular Degeneration. Acta Ophthalmol. 2014, 92, 713–723. [Google Scholar] [CrossRef] [PubMed]
  53. Torres-Costa, S.; Ramos, D.; Brandão, E.; Carneiro, Â.; Rosas, V.; Rocha-Sousa, A.; Falcão-Reis, F.; Falcão, M. Incidence of Endophthalmitis after Intravitreal Injection with and without Topical Antibiotic Prophylaxis. Eur. J. Ophthalmol. 2021, 31, 600–606. [Google Scholar] [CrossRef] [PubMed]
  54. Patil, N.S.; Dhoot, A.S.; Popovic, M.M.; Kertes, P.J.; Muni, R.H. Risk of Intraocular Inflammation after Injection of Anti-Vascular Endothelial Growth Factor Agents: A Meta-Analysis. Retina 2022, 42, 2134–2142. [Google Scholar] [CrossRef]
  55. Levin, A.M.; Chaya, C.J.; Kahook, M.Y.; Wirostko, B.M. Intraocular Pressure Elevation Following Intravitreal Anti-VEGF Injections: Short- and Long-Term Considerations. J. Glaucoma 2021, 30, 1019–1026. [Google Scholar] [CrossRef]
  56. Daien, V.; Nguyen, V.; Essex, R.W.; Guymer, R.; Arnold, J.J.; Munk, M.; Ceklic, L.; Gillies, M.C.; Barthelmes, D. Prevalence and Characteristics of Macular Atrophy in Eyes with Neovascular Age-Related Macular Degeneration. A Study from a Long-Term Observational Dataset: The Fight Retinal Blindness Project. Br. J. Ophthalmol. 2020, 104, 1064–1069. [Google Scholar] [CrossRef]
  57. Nakata, I.; Yamashiro, K.; Nakanishi, H.; Tsujikawa, A.; Otani, A.; Yoshimura, N. VEGF Gene Polymorphism and Response to Intravitreal Bevacizumab and Triple Therapy in Age-Related Macular Degeneration. Jpn. J. Ophthalmol. 2011, 55, 435–443. [Google Scholar] [CrossRef]
  58. Abedi, F.; Wickremasinghe, S.; Richardson, A.J.; Makalic, E.; Schmidt, D.F.; Sandhu, S.S.; Baird, P.N.; Guymer, R.H. Variants in the VEGFA Gene and Treatment Outcome after Anti-VEGF Treatment for Neovascular Age-Related Macular Degeneration. Ophthalmology 2013, 120, 115–121. [Google Scholar] [CrossRef]
  59. Hagstrom, S.A.; Ying, G.S.; Pauer, G.J.; Sturgill-Short, G.M.; Huang, J.; Maguire, M.G.; Martin, D.F. VEGFA and VEGFR2 Gene Polymorphisms and Response to Anti-Vascular Endothelial Growth Factor Therapy: Comparison of Age-Related Macular Degeneration Treatments Trials (CATT). JAMA Ophthalmol. 2014, 132, 521–527. [Google Scholar] [CrossRef]
  60. McKibbin, M.; Ali, M.; Bansal, S.; Baxter, P.D.; West, K.; Williams, G.; Cassidy, F.; Inglehearn, C.F. CFH, VEGF and HTRA1 Promoter Genotype May Influence the Response to Intravitreal Ranibizumab Therapy for Neovascular Age-Related Macular Degeneration. Br. J. Ophthalmol. 2012, 96, 208–212. [Google Scholar] [CrossRef]
  61. Lazzeri, S.; Figus, M.; Orlandi, P.; Fioravanti, A.; Di Desidero, T.; Agosta, E.; Sartini, M.S.; Posarelli, C.; Nardi, M.; Danesi, R.; et al. VEGF-A Polymorphisms Predict Short-Term Functional Response to Intravitreal Ranibizumab in Exudative Age-Related Macular Degeneration. Pharmacogenomics 2013, 14, 623–630. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, J.; Liu, Z.; Hu, S.; Qi, J. Meta-Analysis of the Pharmacogenetics of ARMS2 A69S Polymorphism and the Response to Advanced Age-Related Macular Degeneration. Ophthalmic Res. 2021, 64, 192–204. [Google Scholar] [CrossRef] [PubMed]
  63. Papadakis, K.A.; Targan, S.R. Tumor Necrosis Factor: Biology and Therapeutic Inhibitors. Gastroenterology 2000, 119, 1148–1157. [Google Scholar] [CrossRef] [PubMed]
  64. De Jager, P.L.; Jia, X.; Wang, J.; de Bakker, P.I.; Ottoboni, L.; Aggarwal, N.T.; Piccio, L.; Raychaudhuri, S.; Tran, D.; Aubin, C.; et al. Meta-Analysis of Genome Scans and Replication Identify CD6, IRF8 and TNFRSF1A as New Multiple Sclerosis Susceptibility Loci. Nat. Genet. 2009, 41, 776–782. [Google Scholar] [CrossRef]
  65. Davidson, S.I.; Liu, Y.; Danoy, P.A.; Wu, X.; Thomas, G.P.; Jiang, L.; Sun, L.; Wang, N.; Han, J.; Han, H.; et al. Association of STAT3 and TNFRSF1A with Ankylosing Spondylitis in Han Chinese. Ann. Rheum. Dis. 2011, 70, 289–292. [Google Scholar] [CrossRef]
  66. International Multiple Sclerosis Genetics Consortium. The Genetic Association of Variants in CD6, TNFRSF1A and IRF8 to Multiple Sclerosis: A Multicenter Case-Control Study. PLoS ONE 2011, 6, e18813. [Google Scholar]
  67. Karaderi, T.; Pointon, J.J.; Wordsworth, T.W.; Harvey, D.; Appleton, L.H.; Cohen, C.J.; Farrar, C.; Harin, A.; Brown, M.A.; Wordsworth, B.P. Evidence of Genetic Association between TNFRSF1A Encoding the p55 Tumour Necrosis Factor Receptor, and Ankylosing Spondylitis in UK Caucasians. Clin. Exp. Rheumatol. 2012, 30, 110–113. [Google Scholar]
  68. Matsukura, H.; Ikeda, S.; Yoshimura, N.; Takazoe, M.; Muramatsu, M. Genetic Polymorphisms of Tumour Necrosis Factor Receptor Superfamily 1A and 1B Affect Responses to Infliximab in Japanese Patients with Crohn’s Disease. Aliment. Pharmacol. Ther. 2008, 27, 765–770. [Google Scholar] [CrossRef]
  69. Medrano, L.M.; Taxonera, C.; Márquez, A.; Barreiro-de Acosta, M.; Gómez-García, M.; González-Artacho, C.; Pérez-Calle, J.L.; Bermejo, F.; Lopez-Sanromán, A.; Arranz, M.M.; et al. Role of TNFRSF1B Polymorphisms in the Response of Crohn’s Disease Patients to Infliximab. Hum. Immunol. 2014, 75, 71–75. [Google Scholar] [CrossRef]
  70. Tung, C.H.; Lu, M.C. Association between Ankylosing Spondylitis and Polymorphism of Tumour Necrosis Factor Receptor II in Taiwanese Patients. Scand. J. Rheumatol. 2009, 38, 395. [Google Scholar] [CrossRef]
  71. Corona-Sanchez, E.G.; Munoz-Valle, J.F.; Gonzalez-Lopez, L.; Sanchez-Hernandez, J.D.; Vazquez-Del Mercado, M.; Ontiveros-Mercado, H.; Huerta, M.; Trujillo, X.; Rocha-Muñoz, A.D.; Celis, A.; et al. −383 A/C Tumor Necrosis Factor Receptor 1 Polymorphism and Ankylosing Spondylitis in Mexicans: A Preliminary Study. Rheumatol. Int. 2012, 32, 2565. [Google Scholar] [CrossRef] [PubMed]
  72. Lopez-Castejon, G.; Brough, D. Understanding the Mechanism of IL-1β Secretion. Cytokine Growth Factor Rev. 2011, 22, 189–195. [Google Scholar] [CrossRef] [PubMed]
  73. Zhao, M.; Bai, Y.; Xie, W.; Shi, X.; Li, F.; Yang, F.; Sun, Y.; Huang, L.; Li, X. Interleukin-1β Level is Increased in Vitreous of Patients with Neovascular Age-Related Macular Degeneration (nAMD) and Polypoidal Choroidal Vasculopathy (PCV). PLoS ONE 2015, 10, e0125150. [Google Scholar] [CrossRef] [PubMed]
  74. Zhou, T.E.; Rivera, J.C.; Bhosle, V.K.; Lahaie, I.; Shao, Z.; Tahiri, H.; Zhu, T.; Polosa, A.; Dorfman, A.; Beaudry-Richard, A.; et al. Choroidal Involution is Associated with Progressive Degeneration of the Outer Retinal Function in a Model of Retinopathy of Prematurity: An Early Role for Il-1β. Am. J. Pathol. 2016, 186, 3100–3116. [Google Scholar] [CrossRef]
  75. Olson, J.L.; Courtney, R.J.; Rouhani, B.; Mandava, N.; Dinarello, C.A. Intravitreal Anakinra Inhibits Choroidal Neovascular Membrane Growth in a Rat Model. Ocul. Immunol. Inflamm. 2009, 17, 195–200. [Google Scholar] [CrossRef]
  76. Battu, P.; Sharma, K.; Rain, M.; Singh, R.; Anand, A. Serum Levels of ARMS2, COL8A1, RAD51B, and VEGF and Their Correlations in Age-Related Macular Degeneration. Curr. Neurovasc. Res. 2021, 18, 181–188. [Google Scholar]
  77. Haas, P.; Steindl, K.; Aggermann, T.; Schmid-Kubista, K.; Krugluger, W.; Hageman, G.S.; Binder, S. Serum VEGF and CFH in Exudative Age-Related Macular Degeneration. Curr. Eye Res. 2011, 36, 143–148. [Google Scholar] [CrossRef]
  78. Gu, X.; Yu, X.; Dai, H. Intravitreal Injection of Ranibizumab for Treatment of Age-Related Macular Degeneration: Effects on Serum VEGF Concentration. Curr. Eye Res. 2014, 39, 518–521. [Google Scholar] [CrossRef]
  79. Vural, E.; Hazar, L.; Karakukçu, C.; Arslan, M.E.; Sirem, M.R.; Sirakaya, E.; Ozsayglll, C.; Çiçek, A. Apelin-13: A Promising Biomarker for Age-Related Macular Degeneration? Ophthalmologica 2021, 244, 102–109. [Google Scholar] [CrossRef]
  80. Liukkonen, M.P.K.; Paterno, J.J.; Kivinen, N.; Siintamo, L.; Koskela, A.K.J.; Kaarniranta, K. Epithelial-Mesenchymal Transition-Related Serum Markers ET-1, IL-8 and TGF-Β2 Are Elevated in a Finnish Wet Age-Related Macular Degeneration Cohort. Acta Ophthalmol. 2021, 100, e1153–e1162. [Google Scholar] [CrossRef]
  81. Gonçalves, F.T.I.; Cezario, S.M.; Calastri, M.C.J.; Oliveira, C.I.F.; Souza, D.R.S.; Pinhel, M.A.d.S.; Cotrim, C.C.; Jorge, R.; Siqueira, R.C. Influence of VEGF-C936T Genetic Variant on Age-Related Macular Degeneration. Arq. Bras. Oftalmol. 2015, 78, 290–294. [Google Scholar] [CrossRef] [PubMed]
  82. Carneiro, Â.M.; Costa, R.; Falcão, M.S.; Barthelmes, D.; Mendonça, L.S.; Fonseca, S.L.; Gonçalves, R.; Gonçalves, C.; Falcão-Reis, F.M.; Soares, R. Vascular Endothelial Growth Factor Plasma Levels Before and After Treatment of Neovascular Age-Related Macular Degeneration with Bevacizumab or Ranibizumab. Acta Ophthalmol. 2012, 90, e25–e30. [Google Scholar] [CrossRef] [PubMed]
  83. Zehetner, C.; Kralinger, M.T.; Modi, Y.S.; Waltl, I.; Ulmer, H.; Kirchmair, R.; Bechrakis, N.E.; Kieselbach, G.F. Systemic Levels of Vascular Endothelial Growth Factor Before and After Intravitreal Injection of Aflibercept or Ranibizumab in Patients with Age-Related Macular Degeneration: A Randomised, Prospective Trial. Acta Ophthalmol. 2015, 93, e154–e159. [Google Scholar] [CrossRef] [PubMed]
  84. Deangelis, M.M.; Ji, F.; Adams, S.; Morrison, M.A.; Harring, A.J.; Sweeney, M.O.; Capone, A., Jr.; Miller, J.W.; Dryja, T.P.; Ott, J.; et al. Alleles in the HtrA serine peptidase 1 gene alter the risk of neovascular age-related macular degeneration. Ophthalmology 2008, 115, 1209–1215.e7. [Google Scholar] [CrossRef]
  85. Winkler, T.W.; Grassmann, F.; Brandl, C.; Kiel, C.; Günther, F.; Strunz, T.; Weidner, L.; Zimmermann, M.E.; Korb, C.A.; Poplawski, A.; et al. Genome-wide association meta-analysis for early age-related macular degeneration highlights novel loci and insights for advanced disease. BMC Med. Genom. 2020, 13, 120. [Google Scholar] [CrossRef]
  86. 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]
  87. Alcaraz-Quiles, J.; Titos, E.; Casulleras, M.; Pavesi, M.; López-Vicario, C.; Rius, B.; Lopategi, A.; de Gottardi, A.; Graziadei, I.; Gronbaek, H.; et al. Polymorphisms in the IL-1 gene cluster influence systemic inflammation in patients at risk for acute-on-chronic liver failure. Hepatology 2017, 65, 202–216. [Google Scholar] [CrossRef] [PubMed]
  88. Cebatoriene, D.; Vilkeviciute, A.; Gedvilaite, G.; Bruzaite, A.; Kriauciuniene, L.; Zaliuniene, D.; Liutkeviciene, R. CFH (rs1061170, rs1410996), KDR (rs2071559, rs1870377) and KDR and CFH Serum Levels in AMD Development and Treatment Efficacy. Biomedicines 2024, 12, 948. [Google Scholar] [CrossRef]
Figure 1. Serum IL1B levels were measured in patients with early AMD vs. control group (A) and exudative AMD vs. control groups (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Figure 1. Serum IL1B levels were measured in patients with early AMD vs. control group (A) and exudative AMD vs. control groups (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Ijms 25 09750 g001
Figure 2. Serum TNFRSF1B levels were measured in patients with early AMD vs. control group (A) and exudative AMD vs. control group (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Figure 2. Serum TNFRSF1B levels were measured in patients with early AMD vs. control group (A) and exudative AMD vs. control group (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Ijms 25 09750 g002
Figure 3. Serum TNFRSF1B levels were measured in patients with exudative AMD vs. control group and compared between TNFRSF1B rs1061622 genotypes (A) and between early AMD vs. control group (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Figure 3. Serum TNFRSF1B levels were measured in patients with exudative AMD vs. control group and compared between TNFRSF1B rs1061622 genotypes (A) and between early AMD vs. control group (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Ijms 25 09750 g003
Figure 4. Serum TNFRSF1A levels were measured in patients with exudative AMD vs. control group and compared between TNFRSF1A rs4149576 genotypes (A) and between early AMD vs. control group (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Figure 4. Serum TNFRSF1A levels were measured in patients with exudative AMD vs. control group and compared between TNFRSF1A rs4149576 genotypes (A) and between early AMD vs. control group (B). p-values marked with bold indicate statistically significant p-values, significant when p < 0.05; Mann–Whitney U test was used.
Ijms 25 09750 g004
Table 1. Demographic data of the study.
Table 1. Demographic data of the study.
CharacteristicEarly AMD
n = 253
Exudative AMD
n = 245
Control
n = 337
p-Value
Gender 2
Males, n (%)
Females, n (%)
80 (31.6)
173 (68.4)
90 (36.7)
155 (63.3)
115 (34.1)
222 (65.9)
0.522 *
0.515 **
Age years; median (IQR) 173 (12)77 (10)72 (11)0.117 *
<0.001 **
p—significance level, significant when p < 0.05; IQR—interquartile range. 1 Mann–Whitney U test was used to compare age between groups. 2 Pearson’s chi-squared test was used to compare the gender distribution between groups. * Early AMD vs. control group; ** Exudative AMD vs. control group.
Table 2. Distributions of VEGFA, IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 SNP genotypes and alleles in early and exudative AMD and control groups.
Table 2. Distributions of VEGFA, IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 SNP genotypes and alleles in early and exudative AMD and control groups.
Gene/MarkerGenotype/
Allele
Groupp-Value *p-Value **
Early AMD
(n = 253)
n (%)
Exudative AMD
(n = 245)
n (%)
Control
(n = 337)
n (%)
VEGFA rs3024997GG
GA
AA

G
A
150 (59.3)
86 (34)
17 (6.7)

386 (76.3)
120 (23.7)
155 (63.3)
89 (36.3)
1 (0.4)

399 (81.4)
91 (18.6)
187 (55.5)
129 (38.3)
21 (6.2)

503 (74.6)
171 (25.4)

0.563


0.513

<0.001


0.006
IL1B rs1143623CC
CG
GG

C
G
129 (51)
103 (40.7)
21 (8.3)

361 (71.3)
145 (28.7)
134 (54.7)
92 (37.6)
19 (7.8)

360 (73.5)
130 (26.5)
173 (51.3)
143 (42.4)
21 (6.2)

489 (72.6)
185 (27.4)

0.614


0.647

0.445


0.727
TNFRSF1B rs1061622GG
GT
TT

G
T
161 (63.6)
79 (31.2)
13 (5.1)

401 (79.2)
105 (20.8)
157 (64.1)
74 (30.2)
14 (5.7)

388 (79.2)
102 (20.8)
220 (65.3)
107 (31.8)
10 (3)

547 (81.2)
127 (18.8)

0.402


0.414

0.225


0.403
TNFRSF1A rs4149576TT
TC
CC

T
C
61 (24.1)
123 (48.6)
69 (27.3)

245 (48.4)
261 (51.6)
60 (24.5)
124 (50.6)
61 (24.9)

244 (49.8)
246 (50.2)
89 (26.4)
168 (49.9)
80 (23.7)

346 (51.3)
328 (48.7)

0.589


0.321

0.861


0.604
ARMS2 rs10490924GG
GT
TT

G
T
116 (45.8)
108 (42.7)
29 (11.5)

340 (67.2)
166 (32.8)
78 (31.8)
106 (43.3)
61 (24.9)

262 (53.5)
228 (46.5)
183 (54.3)
129 (38.3)
25 (7.4)

495 (73.4)
179 (26.6)
0.070



0.019
<0.001



<0.001
p—significance level. When Pearson’s chi-squared test was used, Bonferroni-corrected significance level p = 0.05/5; p-values marked with bold indicate statistically significant p-values. * Early AMD vs. control group; ** exudative AMD vs. control group.
Table 3. Associations between VEGFA (rs3024997), IL1B (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), ARMS2 (rs10490924), and exudative AMD.
Table 3. Associations between VEGFA (rs3024997), IL1B (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), ARMS2 (rs10490924), and exudative AMD.
VEGFA (rs3024997)
ModelGenotype/AlleleOR * (95% CI)p-ValueAIC
CodominantGA vs. GG
AA vs. GG
0.887 (0.617–1.275)
0.047 (0.006–0.366)
0.518
0.003
718.602
DominantGA + AA vs. GG0.755 (0.530–1.076)0.120733.422
RecessiveAA vs. GG + GA0.049 (0.006–0.381)0.004717.021
OverdominantGA vs. GG + AA0.987 (0.689–1.413)0.943735.844
AdditiveA0.657 (0.480–0.899)0.009728.805
IL1B (rs1143623)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantCG vs. CC
GG vs. CC
0.853 (0.593–1.227)
1.195 (0.599–2.384)
0.391
0.613
736.601
DominantCG + GG vs. CC0.898 (0.634–1.270)0.541735.476
RecessiveGG vs. CC + CG1.279 (0.653–2.508)0.473735.336
OverdominantCG vs. CC + GG0.835 (0.586–1.190)0.319734.856
AdditiveG0.973 (0.737–1.284)0.845735.811
TNFRSF1B (rs1061622)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantGT vs. GG
TT vs. GG
0.881 (0.603–1.288)
2.049 (0.847–4.957)
0.514
0.112
734.484
DominantGT + TT vs. GG0.974 (0.678–1.399)0.888735.829
RecessiveTT vs. GG + GT2.133 (0.890–5.116)0.090732.910
OverdominantGT vs. GG + TT0.844 (0.580–1.229)0.376735.063
AdditiveT1.078 (0.795–1.462)0.627735.614
TNFRSF1A (rs4149576)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantTC vs. TT
CC vs. TT
1.140 (0.748–1.737)
1.299 (0.796–2.119)
0.542
0.296
736.752
DominantTC + CC vs. TT1.190 (0.800–1.769)0.391735.109
RecessiveCC vs. TT + TC1.191 (0.796–1.783)0.394735.125
OverdominantTC vs. TT + CC1.004 (0.711–1.420)0.980735.849
AdditiveC1.140 (0.892–1.456)0.296734.752
ARMS2 (rs10490924)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantGT vs. GG
TT vs. GG
1.768 (1.201–2.603)
5.611 (3.205–9.822)
0.004
<0.001
697.230
DominantGT + TT vs. GG2.377 (1.659–3.404)<0.001712.955
RecessiveTT vs. GG + GT4.236 (2.508–7.155)<0.001703.644
OverdominantGT vs. GG + TT1.129 (0.794–1.606)0.499735.393
AdditiveT2.200 (1.702–2.846)<0.001697.423
* OR adjusted for age in exudative AMD group; OR—odds ratio; CI—confidence interval; p—significance level, Bonferroni-corrected significance level p = 0.05/5; p-values marked with bold indicate statistically significant p-values; AIC—Akaike information criteria.
Table 4. Distributions of VEGFA, IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 SNP genotypes and alleles in early and exudative AMD and control females.
Table 4. Distributions of VEGFA, IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 SNP genotypes and alleles in early and exudative AMD and control females.
Gene/MarkerGenotype/
Allele
Groupp-Value *p-Value **
Early AMD
(n = 173)
n (%)
Exudative AMD
(n = 155)
n (%)
Control
(n = 222)
n (%)
VEGFA rs3024997GG
GA
AA

G
A
103 (59.5)
60 (34.7)
10 (5.8)

266 (76.9)
80 (23.1)
93 (60)
61 (39.4)
1 (0.6)

247 (79.7)
63 (20.3)
117 (52.7)
92 (41.4)
13 (5.9)

326 (73.4)
118 (26.6)

0.373


0.266

0.022


0.047
IL1B rs1143623CC
CG
GG

C
G
93 (53.8)
63 (36.4)
17 (9.8)

249 (72)
97 (28)
84 (54.2)
56 (36.1)
15 (9.7)

224 (72.3)
86 (27.7)
110 (49.5)
99 (44.6)
13 (5.9)

319 (71.8)
125 (28.2)

0.140


0.970

0.152


0.901
TNFRSF1B rs1061622GG
GT
TT

G
T
111 (64.2)
55 (31.8)
7 (4)

277 (80.1)
69 (19.9)
97 (62.6)
48 (31)
10 (6.5)

242 (78.1)
68 (21.9)
146 (65.8)
68 (30.6)
8 (3.6)

360 (81.1)
84 (18.9)

0.937


0.718

0.428


0.309
TNFRSF1A rs4149576TT
TC
CC

T
C
47 (27.2)
78 (45.1)
48 (27.7)

172 (49.7)
174 (50.3)
33 (21.3)
87 (56.1)
35 (22.6)

153 (49.4)
157 (50.6)
58 (26.1)
116 (52.3)
48 (21.6)

232 (52.3)
212 (47.7)

0.279


0.478

0.555


0.433
ARMS2 rs10490924GG
GT
TT

G
T
81 (46.8)
75 (43.4)
17 (9.8)

237 (68.5)
109 (31.5)
47 (30.3)
72 (46.5)
36 (23.2)

166 (53.5)
144 (46.5)
120 (54.1)
85 (38.3)
17 (7.7)

325 (73.2)
119 (26.8)

0.342


0.147
<0.001



<0.001
p—significance level. When Pearson’s chi-squared test was used, Bonferroni-corrected significance level p = 0.05/5; p-values marked with bold indicate statistically significant p-values. * Early AMD vs. control group; ** exudative AMD vs. control group.
Table 5. Associations between VEGFA (rs3024997), IL1B (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), ARMS2 (rs10490924), and exudative AMD in females.
Table 5. Associations between VEGFA (rs3024997), IL1B (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), ARMS2 (rs10490924), and exudative AMD in females.
VEGFA (rs3024997)
ModelGenotype/AlleleOR * (95% CI)p-ValueAIC
CodominantGA vs. GG
AA vs. GG
0.890 (0.561–1.413)
0.074 (0.009–0.624)
0.622
0.017
441.925
DominantGA + AA vs. GG0.777 (0.494–1.221)0.273448.552
RecessiveAA vs. GG + GA0.078 (0.009–0.648)0.018440.168
OverdominantGA vs. GG + AA0.987 (0.626–1.557)0.955449.752
AdditiveA0.680 (0.454–1.018)0.061446.194
IL1B (rs1143623)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantCG vs. CC
GG vs. CC
0.735 (0.458–1.178)
1.567 (0.660–3.718)
0.201
0.308
448.241
DominantCG + GG vs. CC0.831 (0.531–1.299)0.416449.094
RecessiveGG vs. CC + CG1.793 (0.774–4.152)0.173447.886
OverdominantCG vs. CC + GG0.693 (0.438–1.097)0.117447.285
AdditiveG0.988 (0.694–1.408)0.947449.751
TNFRSF1B (rs1061622)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantGT vs. GG
TT vs. GG
0.897 (0.548–1.467)
1.919 (0.670–5.494)
0.664
0.225
449.878
DominantGT + TT vs. GG0.996 (0.625–1.588)0.986449.755
RecessiveTT vs. GG + GT1.986 (0.702–5.618)0.196448.067
OverdominantGT vs. GG + TT0.857 (0.525–1.393)0.533449.364
AdditiveT1.096 (0.747–1.608)0.638449.536
TNFRSF1A (rs4149576)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantTC vs. TT
CC vs. TT
1.472 (0.845–2.564)
1.600 (0.824–3.106)
0.173
0.165
449.329
DominantTC + CC vs. TT1.508 (0.887–2.562)0.129447.413
RecessiveCC vs. TT + TC1.224 (0.716–2.093)0.461449.213
OverdominantTC vs. TT + CC1.173 (0.749–1.837)0.484449.266
AdditiveC1.267 (0.911–1.762)0.160447.765
ARMS2 (rs10490924)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantGT vs. GG
TT vs. GG
1.979 (1.201–3.261)
5.628 (2.720–11.646)
0.007
<0.001
427.191
DominantGT + TT vs. GG2.550 (1.597–4.071)<0.001433.851
RecessiveTT vs. GG + GT3.974 (2.020–7.817)<0.001432.468
OverdominantGT vs. GG + TT1.262 (0.803–1.984)0.312448.735
AdditiveT2.265 (1.617–3.172)<0.001425.703
* OR adjusted for age in exudative AMD group; OR—odds ratio; CI—confidence interval; p—significance level, Bonferroni-corrected significance level p = 0.05/5; p-values marked with bold indicate statistically significant p-values; AIC—Akaike information criteria.
Table 6. Distributions of VEGFA, IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 SNP genotypes and alleles in early and exudative AMD and control males.
Table 6. Distributions of VEGFA, IL1B, TNFRSF1B, TNFRSF1A, and ARMS2 SNP genotypes and alleles in early and exudative AMD and control males.
Gene/MarkerGenotype/
Allele
Groupp-Value *p-Value **
Early AMD
(n = 80)
n (%)
Exudative AMD
(n = 90)
n (%)
Control
(n = 115)
n (%)
VEGFA rs3024997GG
GA
AA

G
A
47 (58.8)
26 (32.5)
7 (8.8)

120 (75)
40 (25)
62 (68.9)
28 (31.1)
0 (0)

152 (84.4)
28 (15.6)
70 (60.9)
37 (32.2)
8 (7)

177 (77)
53 (23)

0.890


0.655

0.034


0.058
IL1B rs1143623CC
CG
GG

C
G
36 (45)
40 (50)
4 (5)

112 (70)
48 (30)
50 (55.6)
36 (40)
4 (4.4)

136 (75.6)
44 (24.4)
63 (54.8)
44 (38.3)
8 (7)

170 (73.9)
60 (26.1)

0.260


0.395

0.745


0.704
TNFRSF1B rs1061622GG
GT
TT

G
T
50 (62.5)
24 (30)
6 (7.5)

124 (77.5)
36 (22.5)
60 (66.7)
26 (28.9)
4 (4.4)

146 (81.1)
34 (18.9)
74 (64.3)
39 (33.9)
2 (1.7)

187 (81.3)
43 (18.7)

0.131


0.357

0.426


0.960
TNFRSF1A rs4149576TT
TC
CC

T
C
14 (17.5)
45 (56.3)
21 (26.3)

73 (45.6)
87 (54.4)
27 (30)
37 (41.1)
26 (28.9)

91 (50.6)
89 (49.4)
31 (27)
52 (45.2)
32 (27.8)

114 (49.6)
116 (50.4)

0.220


0.443

0.826


0.842
ARMS2 rs10490924GG
GT
TT

G
T
35 (43.8)
33 (41.3)
12 (15)

103 (64.4)
57 (35.6)
31 (34.4)
34 (37.8)
25 (27.8)

96 (53.3)
84 (46.7)
63 (54.8)
44 (38.3)
8 (7.0)

170 (73.9)
60 (26.1)
0.121



0.043
<0.001



<0.001
p—significance level. When Pearson’s chi-squared test was used, Bonferroni-corrected significance level p = 0.05/5; p-values marked with bold indicate statistically significant p-values. * Early AMD vs. control group; ** exudative AMD vs. control group.
Table 7. Associations between VEGFA (rs3024997), IL1B (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), ARMS2 (rs10490924), and exudative AMD in males.
Table 7. Associations between VEGFA (rs3024997), IL1B (rs1143623), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), ARMS2 (rs10490924), and exudative AMD in males.
VEGFA (rs3024997)
ModelGenotype/AlleleOR * (95% CI)p-ValueAIC
CodominantGA vs. GG
AA vs. GG
0.873 (0.479–1.594)
-
0.659
-
273.752
DominantGA + AA vs. GG0.711 (0.396–1.276)0.253280.473
RecessiveAA vs. GG + GA--271.947
OverdominantGA vs. GG + AA0.974 (0.536–1.768)0.930281.782
AdditiveA0.616 (0.369–1.028)0.064278.221
IL1B (rs1143623)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantCG vs. CC
GG vs. CC
1.049 (0.588–1.870)
0.642 (0.182–2.258)
0.872
0.489
283.201
DominantCG + GG vs. CC0.986 (0.565–1.721)0.960281.787
RecessiveGG vs. CC + CG0.629 (0.183–2.163)0.462281.227
OverdominantCG vs. CC + GG1.093 (0.619–1.928)0.759281.696
AdditiveG0.926 (0.585–1.465)0.742281.681
TNFRSF1B (rs1061622)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantGT vs. GG
TT vs. GG
0.822 (0.450–1.504)
2.526 (0.445–14.345)
0.525
0.296
282.029
DominantGT + TT vs. GG0.904 (0.505–1.619)0.735281.675
RecessiveTT vs. GG + GT2.691 (0.479–15.120)0.261280.435
OverdominantGT vs. GG + TT0.791 (0.434–1.440)0.443281.197
AdditiveT1.017 (0.610–1.695)0.948281.786
TNFRSF1A (rs4149576)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantTC vs. TT
CC vs. TT
0.811 (0.416–1.583)
0.971 (0.465–2.027)
0.540
0.938
283.321
DominantTC + CC vs. TT0.871 (0.472–1.607)0.658281.595
RecessiveCC vs. TT + TC1.101 (0.593–2.043)0.760281.697
OverdominantTC vs. TT + CC0.823 (0.469–1.444)0.497281.327
AdditiveC0.984 (0.680–1.424)0.933281.783
ARMS2 (rs10490924)
ModelGenotype/AlleleOR * (95% CI)p-valueAIC
CodominantGT vs. GG
TT vs. GG
1.537 (0.824–2.867)
6.183 (2.495–15.322)
0.177
<0.001
265.951
DominantGT + TT vs. GG2.251 (1.270–3.989)0.005273.879
RecessiveTT vs. GG + GT5.049 (2.147–11.877)<0.001265.781
OverdominantGT vs. GG + TT0.960 (0.542–1.699)0.887281.770
AdditiveT2.205 (1.469–3.310)<0.001266.193
* OR adjusted for age in exudative AMD group; OR—odds ratio; CI—confidence interval; p—significance level, Bonferroni-corrected significance level p = 0.05/5; p-values marked with bold indicate statistically significant p-values; AIC—Akaike information criteria.
Table 8. Demographic and clinical parameters.
Table 8. Demographic and clinical parameters.
CharacteristicNon-Responders
n = 20
Responders
n = 95
p-Value
Gender
Males, n (%)
Females, n (%)

9 (30)
14 (70)

29 (30.5)
66 (689.5)
0.963 *
Age years; mean (SD)75.4 (7.366)77.54 (7.784)0.263 **
Response parameter
VA, median (IQR)
Baseline
Treated

0.465 (0.45) 1
0.35 (0.35) 1

0.28 (0.26) 2
0.375 (0.35) 2

0.018 ***
0.408 ***
CRT (μm), median (IQR)
Baseline
Treated

272.5 (95.25) 3
329 (103) 3

320.5 (113) 4
274 (95) 4

0.068 ***
0.032 ***
p—significance level, significant when p < 0.05; p-values marked with bold indicate statistically significant p-values; IQR—interquartile range; SD—standard deviation; VA—visual acuity; CRT—central macular thickness. * Pearson’s chi-squared test. ** Student’s T test. *** Mann–Whitney U test. 1 p = 0.028, Wilcoxon signed-rank test; 2 p < 0.001, Wilcoxon signed-rank test; 3 p = 0.441, Wilcoxon signed-rank test; 4 p < 0.001, Wilcoxon signed-rank test.
Table 9. Associations between TNFRSF1B rs1061622 and response to treatment.
Table 9. Associations between TNFRSF1B rs1061622 and response to treatment.
Genetic ModelGenotype/AlleleNon-Responders
n = 20
n (%)
Responders
n = 95
n (%)
OR (95% CI)p-ValueAIC
DominantGT + TT3 (15)41 (43.2)4.302 (1.181; 15.674)0.027102.065
Additive T3 (7.5)48 (25.3)3.999 (1.176; 13.602)0.026101.291
OR—odds ratio; CI—confidence interval; p—significance level, significant when p < 0.05; p-values marked with bold indicate statistically significant p-values; AIC—Akaike information criteria.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cebatoriene, D.; Vilkeviciute, A.; Gedvilaite-Vaicechauskiene, G.; Duseikaite, M.; Bruzaite, A.; Kriauciuniene, L.; Zaliuniene, D.; Liutkeviciene, R. The Impact of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623) Polymorphisms and Serum Levels on Age-Related Macular Degeneration Development and Therapeutic Responses. Int. J. Mol. Sci. 2024, 25, 9750. https://doi.org/10.3390/ijms25179750

AMA Style

Cebatoriene D, Vilkeviciute A, Gedvilaite-Vaicechauskiene G, Duseikaite M, Bruzaite A, Kriauciuniene L, Zaliuniene D, Liutkeviciene R. The Impact of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623) Polymorphisms and Serum Levels on Age-Related Macular Degeneration Development and Therapeutic Responses. International Journal of Molecular Sciences. 2024; 25(17):9750. https://doi.org/10.3390/ijms25179750

Chicago/Turabian Style

Cebatoriene, Dzastina, Alvita Vilkeviciute, Greta Gedvilaite-Vaicechauskiene, Monika Duseikaite, Akvile Bruzaite, Loresa Kriauciuniene, Dalia Zaliuniene, and Rasa Liutkeviciene. 2024. "The Impact of ARMS2 (rs10490924), VEGFA (rs3024997), TNFRSF1B (rs1061622), TNFRSF1A (rs4149576), and IL1B1 (rs1143623) Polymorphisms and Serum Levels on Age-Related Macular Degeneration Development and Therapeutic Responses" International Journal of Molecular Sciences 25, no. 17: 9750. https://doi.org/10.3390/ijms25179750

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