IMR Press / FBL / Volume 27 / Issue 7 / DOI: 10.31083/j.fbl2707207
Open Access Review
Potential Impact of N6-Methyladenosine RNA Methylation on Vision Function and the Pathological Processes of Ocular Diseases: New Discoveries and Future Perspectives
Xiaohua Li1,2,3,4,*,†Binyun Ma5,†Mengyu Liao6,7,8Lingke Li1,2,3,4Xiaodan Zhang3Mei Du6,7,8Jinguo Yu6,7,8Shikun He9,*Hua Yan6,7,8,*
Show Less
1 Department of Ophthalmology, Henan Provincial People’s Hospital, 450003 Zhengzhou, Henan, China
2 Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, 450003 Zhengzhou, Henan, China
3 Department of Ophthalmology, People’s Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
4 Department of Ophthalmology, People’s Hospital of Henan University, 450003 Zhengzhou, Henan, China
5 Department of Medicine/Hematology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
6 Department of Ophthalmology, Tianjin Medical University General Hospital, 300052 Tianjin, China
7 Laboratory of Molecular Ophthalmology, Tianjin Medical University, 300052 Tianjin, China
8 Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, 300052 Tianjin, China
9 Department of Pathology and Ophthalmology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
*Correspondence: zyyyanhua@tmu.edu.cn (Hua Yan); xhl_6116@163.com (Xiaohua Li); shikunhe@usc.edu (Shikun He)
These authors contributed equally.
Academic Editor: Graham Pawelec
Front. Biosci. (Landmark Ed) 2022, 27(7), 207; https://doi.org/10.31083/j.fbl2707207
Submitted: 14 April 2022 | Revised: 23 May 2022 | Accepted: 24 May 2022 | Published: 28 June 2022
(This article belongs to the Special Issue Recent Advances in Eye and Vision Research)
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

N6-methyladenosine (m6A) methylation/modification plays a critical role in various biological processes through post-transcriptional ribonucleic acid (RNA) modification, which involves RNA processing, nuclear export, translation and decay. Functionally, m6A modification may be involved in ocular cell growth and differentiation, stem cell identity, development, haemostasis and innate versus adaptive immunity. Aberrations in m6A methylation may mediate numerous pathological conditions in the eye, including microorganism infection, inflammation, autoimmune disease, senescence, degeneration, epithelial–mesenchymal transition, fibrosis, angiogenesis, tumorigenesis and complex eye diseases. In this review, we have discussed the relevance of m6A modification to precision medicine, stem cell directional differentiation, biomarkers of eye diseases and m6A methylation activators and inhibitors. In addition, we summarised the challenges and future research directions in the field related to visual function and eye diseases.

Keywords
eye
m6A
pathogenesis
vision function
1. Introduction

The principle of molecular biology follows the traditional theory that genetic information flows from deoxyribonucleic acid (DNA) and ribonucleic acid (RNA) to protein. However, it is difficult to explain the ever-changing cellular phenomenon using only the central dogma. In the last decade, it has been found that epigenetic factors control the expression and function of genes and hence determine the state of cells. Epigenetics has been a prime research focus in many disciplines, including medicine, zoology, botany and reproductive development. Several aspects regarding the pathogenesis, diagnosis and treatment of diseases have been discovered owing to the remarkable achievements and breakthroughs in the field of epigenetics. In addition to DNA methylation, histone modification and non-coding RNAs, one of the novel advances in epigenetic studies is RNA methylation, also known as RNA epigenetics or epitranscriptomics [1, 2]. RNA modifications are the focus of many epigenetic studies in the field of biomedical research and are key to the epigenetic regulation of gene expression. The collective novel evidence implies that post-transcriptional modifications of RNA are crucial for the destiny and function of messenger RNAs (mRNAs). The discovery of RNA modification transcends the view that mRNA is only a message carrier. Therefore, the traditional concept of the functions of mRNA and other non-coding RNAs has changed. Moreover, researchers working on the eye and vision should pay close attention to new studies to keep up with the rapid development of research in the field.

Several RNAs undergo varied modifications, including N6-methyladenosine (m6A), N6,2’-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C), pseudouridine (Ψ), 2’-O-methylation (2’-O-Me), 7-methylguanine (m7G), N4-acetylcytidine (ac4C), uridine (U) and inosine (I) (Fig. 1) [3, 4]. To date, more than 170 RNA modifications have been reported [5], suggesting that RNA modification occurs at multiple locations in RNA molecules. The diversity of RNA modification suggests functional differences. Although m6A modification is the focus of this review, a brief introduction of other most recognised RNA modifications has been provided below.

Fig. 1.

Ribose phosphate backbone of the RNA strand and principal RNA methylation modifications. (A) Primary structure of the RNA strand. Adenine (A), cytosine (C), guanine (G) and uracil (U) are connected by 3-5ester bonds between ribose and phosphate. (B) The chemical structure of various methylated nucleosides. (C) Distribution of methylated nucleosides in the mRNA cap structure, 5- or 3-untranslated region (UTR) or the coding region of mRNA.

1.1 N6,2’-O-dimethyladenosine Modification

N6,2’-O-dimethyladenosine (m6Am) is a reversible modification that is widespread in different RNA molecules. It can be detected in 30% of mRNAs at the 5’ end [6]. Liu et al. [7] reported that m6Am levels inversely correlated with the corresponding protein expression; however, m6Am mapping via transcriptome-wide analysis showed that m6Am levels were relatively low in human tissues. A remarkable finding of the study was the recognition of phosphorylated CTD-interacting factor 1 (PCIF1) as a mammalian mRNA m6Am methyltransferase and the role of fat mass and obesity-associated (FTO) gene in removing m6Am deposition from mRNA molecules [8, 9]. Aberrant m6Am modification may be associated with the genesis of certain cancers [10] and stress response. Engel M et al. [11] discovered that m6A/m is altered after stress stimulation and glucocorticoid treatment, and knockdown of either methyltransferase-like 3 (METTL3) or FTO interferes with m6Am gene expression. In addition, glucocorticoid-mediated depressive diseases are closely associated with aberrant m6Am modification, suggesting that the alteration of m6Am modification may contribute to stress-related psychiatric disorders. However, several aspects of m6Am modification, its biological functions and its role in the pathogenesis of human diseases warrant further investigation.

1.2 N1-methyladenosine Modification

N1-methyladenosine (m1A) is one of the common RNA modifications, which is mediated by methyltransferases (TRMT10C, TRMT61B, TRMT6 and TRMT61A), demethylases (ALKBH1 and ALKBH3) and binding proteins (YTHDF1, YTHDF2, YTHDF3 and YTHDC1) [12]. Dominissini D [3] and Li X [13] mapped the distribution of m1A and demonstrated that m1A modification was enriched near the start codon, thereby highlighting its role in regulating mRNA stability and translation in addition to regulating the functions of transfer RNA (tRNA) and ribosomal RNA (rRNA). Although the physiological function and role of m1A in human diseases remain unclear, it has been reported that m1A regulates the response to physiological stress and may lead to three types of metabolic features, namely, metabolism-excluded, metabolism-high and metabolism-intermediate phenotypes [14]. Recent studies have revealed that m1A modification is related to tumorigenesis and cancer recurrence [15, 16] and regulates metabolic heterogeneity in hepatocellular carcinoma [14]. As a new type of RNA methylation, m1A modification, its function and its relevance to human diseases should be investigated further.

1.3 5-methylcytosine Modification

5-methylcytosine (m5C) methylation is one of the major post-transcriptional RNA modifications. It occurs in almost all types of RNA and is regulated by methyltransferases (NSUN, DNMT and TRDMT family members), demethylases (TET family and ALKBH1) and binding proteins (ALYREF and YBX1). It plays an active role in regulating nuclear mRNA export, RNA alternative splicing, protein translation and RNA–protein interactions; increasing RNA stability and maintaining the normal RNA structure [17]. Amort et al. [18] reported the distribution of RNA m5C in different tissues and cells. They selected mouse embryonic stem cells (ESCs) and brain tissue as experimental samples and showed that the distribution frequency of total RNA 5mC was higher in ESCs than in the brain tissue. In addition, analysis of the association between m5C locus and different genomic regions (such as coding DNA sequence [CDS], 5’ untranslated region [5’-UTR] and 3’-UTR) indicated that m5C sites in total RNA were more likely to be distributed at transcription initiation sites. The distribution of m5C in the 3’-UTR region of brain tissue and ESCs varied greatly, suggesting that RNA m5C modification in the 3’-UTR region is closely related to cell function and types. Furthermore, Wang et al. [19] mapped m5C modification and found that m5C is located at the translation initiation site of mRNA. The analysis and comparison of different tissues of humans and mice showed that the distribution characteristics of m5C in mRNA were very conserved in mammals, and genes modified in different tissues were specific. In addition, the study revealed that dynamic m5C-modified genes were significantly enriched in sperm development-related functions during penile development in mice, suggesting that m5C modifications are involved in the regulation of reproductive development. m5C is also involved in the regulation of cell differentiation, proliferation, migration and cell cycle. Moreover, abnormal levels of m5C are closely related to nervous system defects, cardiovascular system diseases, angiogenesis and tumorigenesis [20].

1.4 Pseudouridine Modification

Pseudouridine (Ψ), the five-carbon glycoside isomer of uridine, is one of the abundant post-transcriptional RNA modifications. It was initially detected in tRNA and rRNA in yeast. At present, Ψ modification is known to occur in different types of RNA, including tRNA, rRNA, snRNA, scaRNA, microRNA (miRNA) and long non-coding RNA (lncRNA). Two mechanisms have been reported for the occurrence of Ψ modification, namely, RNA-dependent and RNA-independent mechanisms. RNA-dependent catalysers include box H/ACA small nucleolar RNA (snoRNA) (NAP57 or DKC1), non-histone protein 2 (NHP2), nucleolar protein 10 (NOP10) and glycine–arginine-rich protein 1 (GAR1); of which, DKC1 is the major enzyme controlling methylation. Pseudouridine synthases (PUSs) are responsible for the occurrence of Ψ modification in RNA through the RNA-independent mechanism [20]. A study, for the first time, reported the regulatory role of PUSs in miRNA processing, thereby enriching the functions of Ψ. Another remarkable finding was the discovery of the dual function of human cellular PUS10. PUS10 not only regulates miRNA processing in the nucleus but also catalyses Ψ modification of tRNAs in the cytoplasm [21]. Furthermore, the regulation of miRNAs by PUS10 is independent of its catalytic activity. However, the catalytic activity of PUS10 plays a role in maintaining normal cell growth. PUS10 acts as a tRNA substrate modified in the cytoplasm with cytoplasm-specific catalytic activity [21]. In addition to regulating RNA metabolism, pseudouridylation helps in maintaining the stability of the RNA structure. Pseudouridylation can regulate translatability or sense codon decoding, thus affecting protein translation [22]. More recently, pseudouridylation-related knowledge has been used for designing vaccines against HIV-1, Zika virus, influenza virus and COVID-19 [23]. Pseudouridylation also contributes to certain pathological conditions, especially tumorigenesis in acute myeloid leukaemia (AML), chronic lymphocytic leukaemia (CLL), hepatocellular carcinoma (HCC), breast cancer, glioblastoma, prostate cancer, skin cancer and pituitary cancer [20]. Furthermore, ageing may also be regulated by pseudouridylation. Lack of DKC1 activity primarily affects tissues with rapid cellular turnover through impairment of telomerase activity and blocks ribosomal translation of specific mRNAs, resulting in reduced replicative potential and premature ageing. Increased expression of vascular endothelial growth factor is associated with the loss of DKC1 [20]. Although studies have reported the biological function of Ψ modification, further investigation is necessary to reveal the detailed mechanisms.

1.5 2’-O-methylation Modification

2’-O-Me modification is characterised by methylation at the 2’ position under the control of RNA methylase. It is widely distributed in mRNA, tRNA, rRNA, miRNA and other molecules. A study by Dai et al. [24] revealed the distribution characteristics of 2’-O-Me in human cells for the first time. Approximately 70.3% of 2’-O-Me sites in protein-coding transcripts are located in CDS. The signature sequence of motifs with 2’-O-Me is AGAU, with a high distribution of A or G bases. The 2’-O-Me site is also enriched in protein-coding codons for three amino acids. Studies have shown that 2’-O-Me affects the binding of mRNA to protein, regulates rRNA translation efficiency and participates in biological processes such as tRNA recognition. Recently, Bennasser et al. [25] (University of Montpellier, France) discovered a 2’-O-Me site on HIV and reported that its activity of infecting host cells was regulated by the methyltransferase FTSJ3 (2’-O-RNA methyltransferase). FTSJ3 is also involved in innate immune regulation of HIV.

1.6 N7-methylguanosine Modification

As a new focus area of epitranscriptomic research, N7-methylguanosine (m7G) has been extensively investigated by many researchers. Previous studies have shown that m7G RNA methylation modifications exist in various molecules, including mRNA 5’ cap structure, mRNA interior, pri-miRNA, tRNA and rRNA. m7G modification can regulate mRNA transcription, biosynthesis and biological functions of miRNA, tRNA stability, nuclear processing and 18S rRNA maturation. m7G RNA modification is catalysed by the METTL1/WD repeat domain 4 (WDR4) complex [26]. Furthermore, METTL1 knockout decreases m7G tRNA modification and leads to impaired ESC self-renewal and differentiation, and its mutations are associated with developmental disorders, suggesting that m7G modification in tRNA has more important physiological functions in mammals [27]. Malbec et al. [28] found that the distribution of m7G in eukaryotic mRNA is species-conserved. They performed m7G-seq on mouse ESCs (mESCs) and brain tissue and found that the distribution of m7G genomic signatures in mice was highly consistent with that in humans. In a study, m7G was found to be significantly enriched in the 5’-UTR of human and mouse mRNAs, especially in the AG-rich region near the translation initiation site. However, after heat shock treatment, the distribution of m7G dynamically changed and was significantly enriched in the gene region (CDS) and 3’-UTR [28]. Furthermore, m7G formed under stress conditions can promote protein translation. METTL1 (m7G methylase) may be involved in regulating m7G formation in eukaryotic mRNA under stress conditions [28]. One of the specific examples of stress response regulated by m7G was reported by Wang et al. [29] who used m7G-MeRIP-seq and lncRNA-seq technology to analyse the m7G modification profile in hypoxic pulmonary hypertension. The results showed that the m7G modification pattern was significantly different between the disease and control groups. In the hypoxic pulmonary hypertension group, m7G modification affected RNA expression, and m7G lncRNAs were significantly upregulated compared with non-m7G lncRNAs. Although the clinical significance requires further validation, the study helped to reveal the potential role of m7G modification in the pathogenesis of hypoxic pulmonary hypertension [29]. Studies have also investigated the role of m7G in tumorigenesis. The development and malignancy of head and neck squamous cell carcinoma are highly related to the tRNA m7G methyltransferase METTL1 through the regulation of entire mRNA translation, PI3K/AKT/mTOR signalling pathway and immune response [26].

1.7 N4-acetylcytidine Modification

N4-acetylcytidine (ac4C) is a highly conserved RNA modification of tRNA and rRNA and has been recently studied in eukaryotic mRNA. However, the distribution and biologic functions of ac4C remain to be further elucidated. As early as 2018, Arango et al. [30] found that N-acetyltransferase 10 (NAT10) catalysed ac4C modification in mRNA and revealed the discrete acetylated regions enriched in coding sequences. In addition, ablation of NAT10 reduced ac4C detection at mapped mRNA sites and was found to be associated with target mRNA downregulation, indicating an important role of ac4C in regulating mRNA translation. In 2020, a research team from the National Institutes of Health conducted a study that provided a technical and theoretical basis for elucidating the role of ac4c modification in biology and disease using ac4C-seq, a chemogenomic approach for transcriptome-wide quantification of ac4C at single-nucleotide resolution [31]. ac4C is markedly induced in response to an increase in temperature, and acetyltransferase-deficient archaeal strains exhibit temperature-dependent growth defects.

ac4C is related to gene regulation, protein translation and stress response. The content of ac4C in the human body significantly changes under diseased conditions; therefore, ac4C may also regulate inflammatory responses, metabolic diseases and autoimmune diseases. The importance of the role of ac4C in the pathogenesis and treatment of cancer has also been reported [32]. In addition, the abnormality of ac4C modification and its catalyze enzyme NAT10 in the condition of HIV-1 infection is validated [33].

1.8 Uridine Modification

Uridine (m5U) modification was demonstrated in rRNA and tRNA a decade ago [34] but was discovered in eukaryotic mRNA in 2020 by Cheng et al. [35], who showed that m5U in mRNA can be found in various mammalian cells and tissues. However, the distribution, function and relevance of m5U modification in human diseases remain to be demonstrated.

m6A methylation is the most common and abundant RNA modification and accounts for >50% of all methylated ribonucleotides [36]. Since its discovery in the 1970s (Fig. 2) [37], m6A modification has attracted great attention in the area of post-transcriptional gene regulation. In addition to mRNA, m6A modification can be seen in miRNA, lncRNA, circular RNA (circRNA), rRNA, tRNA and snoRNA (Fig. 3A). The regulation of coding and non-coding RNAs by m6A represents a new way of understanding the regulation of genetic information [38, 39].

Fig. 2.

Historic events of the study of RNA methylation. The discovery of m5C RNA methylation initiated the study of RNA methylation, and that of the RNA methylase METTL3 began the golden period of RNA methylation research. RNA methylation became a prime focus of epigenetic research after 2011.

Fig. 3.

The role of m6A modification in maintaining the physiological function of cells and its contribution to pathological conditions and molecular composition of m6A RNA methylation. (A) m6A modification can occur in a majority of RNAs, from mRNAs to non-coding RNAs. Therefore, aberrant expression of m6A modification-related factors may result in numerous diseases. (B) m6A methylation is a reversible process controlled by m6A methylases and demethylases. m6A reader proteins bind to m6A-containing transcripts.

In terms of its distribution in genes, m6A is located in highly conserved genomic regions. Specifically, the priority enrichment area of m6A is close to the 3’-UTR and stop codon within the longer internal exons, and m6A is found at the 3’-UTR in a majority of mRNAs. Therefore, m6A controls the translational affinity of RNA-binding proteins or unique m6A-derived transcriptomes. It can affect metabolic processes of mRNA, such as splicing, nuclear export, stabilisation and translation, and RNA–protein interactions, thereby regulating gene expression (Fig. 3B). However, it remains unclear whether the genetic code is influenced by m6A modification. A recent study (2020) [40] provided new ideas for basic biological research, which subverts the inherent unidirectional flow of genetic instructions from DNA to RNA to protein. The study reported that knocking out the m6A methylation-related enzyme METTL3 or YTHDC1 in mouse ESCs increased the openness of chromatin through chromosome-associated regulatory RNA (carRNA) in an m6A-dependent manner and activated transcription. Because RNA structural changes caused by the introduction of m6A may also alter the interaction between RNAs, proteins and chromatin, this new finding may extensively influence our comprehension of human diseases and their treatment [40], suggesting that m6A methylation regulates cellular functions and systemic diseases and is essential for vision function and eye diseases. Therefore, this review highlights the role of m6A modification in the pathogenesis of ocular diseases and vision function.

Previous studies have demonstrated the expression of enzymes required for m6A modification in ocular tissues [41, 42, 43, 44, 45]. m6A modification and its biological functions require specific methyltransferases (writers), demethylases (erasers) and proteins that recognise methylated sites (readers) [46] (Fig. 3B). m6A modification plays a key role in regulating cell division, growth and differentiation; stem cell identity [47]; embryonic and reproductive development; immunity; cell metabolism and homeostasis. Abundant expression of m6A in brain cells is related to neurodevelopment, learning and memory, synaptic plasticity, stress response and heat shock stress response [48] (Fig. 3A). In addition, the importance of m6A RNA modification in the circadian clock and retina has been addressed by Zelinger and Swaroop [49]. Furthermore, m6A mRNA methylation affects testosterone synthesis by regulating LC autophagy by affecting the stability of calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) transcription and the translation efficiency of protein phosphatase 1A (PPM1A) [50], which provides novel insights into the functional mechanism of m6A RNA methylation.

The regulatory mechanisms of post-transcriptional gene expression in eukaryotes warrant further investigation. Aberrant expression of enzymes involved in m6A modification may mediate numerous ocular and systemic pathological conditions, including abnormal retinal development, age-related eye diseases, ocular angiogenesis, ocular surface infection [43, 51, 52, 53], learning and memory decline [54], tumorigenesis [42, 55, 56, 57, 58], degeneration [59, 60, 61, 62] and fibrosis [63, 64, 65] (Fig. 3A). Therefore, the concept of the epitranscriptomic era is well established now [66].

2. m6A Modification and Vision Function
2.1 Factors that Control m6A Modification

The essential factors that control m6A modification and its biological functions in cells, including ocular cells, are methyltransferases (writers, including METTL3, METTL14, METTL16, WTAP, KIAA1429, RBM15 and ZFP217), RNA demethylases (erasers, including FTO and ALKBH5) and specific proteins that recognise methylated sites (readers, including YTHDF1–3, YTHDC1–2, eIF3, IGF2BP1–3, HNRNPA2B1, FMR1 and LRPPRC) [67, 68, 69, 70] (Fig. 3B). METTL3 is a major methylase responsible for adding m6A to RNA molecules. FTO is the first known ‘eraser’ protein that removes methylase and regulates cellular homeostasis. ALKBH5 serves as a demethylase that is also related to DNA damage repair. Reader proteins can recognise m6A sites. YTHDF1 and 3 can promote the translation of m6A-modified mRNA, whereas YTHDF2 can promote RNA degradation. The writer and eraser proteins of mRNA determine the m6A modification level of the target mRNA, and the reader protein determines the translation efficiency of m6A-modified mRNA or affects mRNA stability [71]. The factors related to m6A modification mentioned above may be critical for regulating ocular cell function and behaviour and are discussed below.

2.2 m6A Distribution in Ocular Tissue

The expression of m6A writers (METTL1, METTL3, METTL14 and WTAP), erasers (FTO and ALKBH5) and readers (YTHDF1–3, YTHDC1 and 2, eIF3 and IGF2BP1–3) in ocular tissues has been reported in our previous study and other studies as well [11, 12, 13, 15, 39]. The expression of m6A modification factors (METTL3, METTL14, WTAP, FTO, ALKBH5, YTHDF1, YTHDF3 and YTHDC1) can be found in numerous ocular tissues (cornea, lens, retina, retinal pigmental epithelium [RPE], extraocular muscles and uveal melanoma cells) [41, 42, 43, 45, 51, 72, 73, 74, 75, 76]. m6A methylation usually varies among different tissues [77, 78], indicating that m6A methylation is not only variable but also dynamic, and data obtained from one specific tissue or cell cannot represent the status of m6A methylation in other tissues or cells. For example, the outcome of m6A methylation will be different in the same tissue if the experimental time point is different.

2.3 Implication of m6A Modification in Vision Function

Because of limited studies on the role of m6A in regulating vision function under normal conditions, we systemically discussed some major aspects related to its role physiologically and its implication in vision function.

2.3.1 Implication of m6A Modification in Eye Development

m6A is known to control mammalian neurogenesis and development [79]. Knockdown of METTL3 inhibits the duplication of neural stem cells and morphological maturation of neurons, and depletion of m6A in the mRNA of histone methyltransferase EZH2 downregulates EZH2 expression, leading to defects in neuronal development [80]. The eye is derived from the neuroepithelium, ectoderm and mesenchyme and is considered a part of the brain. Therefore, it would be interesting to discover whether the above mentioned mechanism underlies the development of ocular tissues as well (eyeball, appendices of the eye and visual pathway).

The retina is the most important part of the human eye, and its correct development depends on the activation of transcription factors including epigenetic factors in a timely manner, as demonstrated by Chen et al. [81], which in turn controls the expression of retinal cell structure proteins. Genetic control of retinal development has been well recognised [82]. However, basic knowledge regarding the control of retinal development (cell fate, proliferation and neurogenesis) via epigenetic factors, especially m6A methylation, remains limited. Accumulating evidence suggests that m6A modification is correlated with embryonic development in mammals [83]. Therefore, retinal development may also be subjected to the regulation of m6A methylation [53, 84]. A study showed that METTL3 and METTL14 are expressed in amacrine, bipolar and ganglion cells but not in the rod and cone photoreceptors and horizontal glial cells of the mouse retina [53]. The expression of these two methylases peaked on post-natal day 6 and the proliferation and differentiation of neuronal precursor cells and the maturation of neuronal synapses of the retina peaked on post-natal day 8, implying that m6A methylation may actively regulate retinal development [85]. In addition, the global expression of m6A RNA was significantly lower in adult mouse retina than in newborn mouse retina, which may be associated with a reduction in the expression of m6A writers such as METTL3, METTL14, and WTAP [84]. Similarly, Huang et al. [86] found that the expression of m6A modification factors such as METTL3, METTL14 and WTAP was high in the developing eye of zebrafish, indicating that these factors were involved in retinal development in zebrafish. Functional analysis verified the critical role of m6A methylases in the regulation of eye development in zebrafish. Moreover, deletion of the m6A methylation complex resulted in impairment of eye formation and retinal progenitor cell differentiation, suggesting that the opportune retinal development is m6A-dependent. These studies suggest that m6A modification occurs earlier than cell proliferation and differentiation. The eye, especially the retina, is considered a part of the neural system. Although the abovementioned studies suggest that m6A modification regulates retinal development, further investigation is warranted to examine the mechanism of action of m6A methylation factors, such as writers, erasers and readers, in regulating retinal development during the embryonic and post-natal stages.

2.3.2 Implication of m6A Modification in Normal Ocular Function

Whether the balance between extraocular muscles, intraocular pressure maintenance, lens transparency, vision acuity and the metabolism of vision cycle proteins is controlled by m6A modulation remains unclear. However, the expression of m6A modification factors is observed in the cornea, sclera, iris, lens, retina and optic nerve, and protein expression indicates its function in the tissue. Therefore, we speculate that m6A participates in the maintenance of normal vision function. A recent study showed that a majority of genes expressed in the mouse retina exhibit circadian rhythmic fluctuations [87]. Retinal cell response and hypoxia-related pathways are more active at night, whereas the expression of genes involved in photoreceptor function is higher during the day [87]. These studies suggest that m6A modification is important for maintaining normal vision, rhodopsin metabolism, vision adaptation and memory effects on colour vision especially perception and highlight the possible influence of circadian rhythm disorders on vision function.

2.3.3 Implication of m6A Modification and Cell Haemostasis in Vision Function

m6A modification is controlled by methyltransferases and demethylases, which should be balanced under normal conditions because the loss of balance may result in disease occurrence. Several ocular haemostasis mechanisms have been reported, including maintenance of the delicate balance among tear production, evaporation and drainage; corneal endothelial cell-dependent thickness maintenance; accommodation of the eye; aqueous humor production and drainage; blood–retinal barrier (BRB) intake maintenance and molecular trafficking between the retina and retinal microvessels; innate immune response and subretinal immune privilege; retinol metabolism; photoreceptor outer segment shedding and RPE phagocytosis; and nutrient and waste material transportation between the retina and choroid. Ocular haemostasis may be regulated by m6A modification factors. Although the following data were obtained from systemic studies, the principle explained by them may also apply to ocular cells. m6A methylases/demethylases are essential for producing a normal hypertrophic response in cardiomyocytes [88]. However, reduced levels of m6A methylation/increased levels of demethylases are related to cardiomyocyte dysfunction, implying that m6A modification regulates cardiac homeostasis. In addition, a study showed that METTL3 played a critical role in maintaining mouse T-cell homeostasis, and T cells failed to undergo homeostatic expansion under METTL3-deficient conditions [89]. Therefore, extremely high or low expression of either methyltransferases or demethylases may break the balance between them. As discussed in previous sections, m6A modification factors, especially the major enzymes of m6A methylation/demethylation, namely, METTL3/FTO, can be found in most ocular tissues. The balance between m6A methylation and demethylation may be critical for maintaining normal cell function, and its loss may contribute to the development of numerous diseases including eye diseases. These studies suggest a novel mechanism of m6A methylation/demethylation underlying the maintenance of cell homeostasis [89, 90], therefore, whether m6A modification participates in maintaining and regulating the homeostasis of vision function should be investigated further.

2.3.4 Implication of m6A Modification and Innate Immune Response in Vision Function

The eye has both innate and adaptive immune responses, which are critical for maintaining normal vision function [91, 92]. The ocular innate immune system comprises tears (containing lysozymes, lactoferrins, lipocalin, beta-lysine and immunoglobulins), conjunctival epithelium, corneal epithelium, the complement system, RPE [93], interferons (IFNs), toll-like receptors (TLRs) [94] and transforming growth factor-beta (TGF-β) [95]. In addition, dendritic cells are important for initiating the innate immune response, including recruitment and activation of retinal dendritic cells [96]. A study reported that the expression of MHC class II is upregulated following retinal injury [97]. Among innate immune system-related factors, IFNs play an important role as a mediator of innate immunity in ocular tissues. The expression of IFNs has been confirmed in tears [98], cornea [99], aqueous humor [100], vitreous [101] and retina [102, 103]. The association between m6A regulatory mechanisms and the innate immune system has been reported in several studies. DDX46, a member of the DEAD-box RNA helicase family, can suppress IFN production after viral infection by recruiting ALKBH5 [104]. In addition, overexpression of METTL3 inhibits IFN expression by destabilising the IFN transcript by increasing m6A methylation [105], thereby suppressing the antiviral innate immune response. However, inhibition of METTL3 increases the expression of IFN genes, suggesting that m6A methylation negatively regulates IFN response [105]. Gao et al. [106] reported that m6A modification is necessary for innate immune response because deletion of METTL3 causes an undesirable innate immune response during haematopoiesis. Although IFN and m6A modification factors are expressed in ocular tissues, further investigation is required to examine whether m6A can regulate the innate immune response in the eye.

3. Regulation of Common Ocular Pathogenic Processes via m6A Modification
3.1 m6A Modification and Microorganism Infection in the Eye

Ocular infectious diseases may be caused by viruses, bacteria, fungi or parasites. Such eye diseases may become difficult to control if the infectious agent becomes resistant to commonly used drugs or if there is no cure for the infection, such as infection with deadly bacteria or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, it is essential to recognise the complexity and diversity of ocular microorganisms. To facilitate diagnosis and understand the pathogenesis of ocular diseases related to microorganism infections, genomic approaches are highly recommended for precise detection of the diversity of ocular surface microorganisms. However, the interruption of homeostatic microbiota may contribute to some ocular infections, and the gut and oral microbiota may be closely associated with the pathogenesis of glaucoma, uveitis and AMD [107].

Research into the role of m6A in infectious diseases may help to improve our understanding of the pathogenesis of systemic and ocular infectious diseases and their treatment. Among ocular microorganism infections, the alteration of m6A modification has been reported in experimental fungal keratitis [43], which was the first evidence of the relevance of m6A modification in ocular microorganism infections. Increased m6A methylation, especially METTL3 expression, contributes to the pathogenesis of fungal keratitis [43], whereas aberrant m6A methylation is associated with the production of inflammatory cytokines. In terms of the interaction between m6A and bacterial infections, a recent study revealed that the production of inflammatory cytokines is increased in bacterial infections through demethylation of histone H3 lysine 27 trimethylation (H3K27me3) owing to the loss of YTHDF2. Specifically, YTHDF2 silencing promoted bacterial infection-induced inflammatory responses [108]. Therefore, the study revealed a new mechanism of cross-talk between m6A and H3K27me3 in bacterial infections [108]. In a study on the effect of gut microbiota on the expression of m6A modification factors, the expression of the methyltransferase METTL16 was found to be reduced in microbiota infections, and the methylation of METTL16-targeted mRNA encoding S-adenosylmethionine synthase MAT2a was also inhibited [109]. These studies reveal that m6A modification is a novel mechanism underlying the interaction between bacterial infections and the host.

The relevance of viral infection to m6A modification has attracted significant attention from biomedical researchers, and viral mRNA modified by m6A regulatory factors has been found in prototypic polyomavirus simian virus 40 (SV40) [110], influenza virus, Rous sarcoma virus, human immunodeficiency virus type 1 (HIV-1), Flaviviridae [111], dengue virus, Zika virus, West Nile virus and hepatitis C virus [112]. Herpes simplex virus 1 (HSV1) infection is the most common viral infection in the cornea, and persistent HSV1 infection in the cornea often results in serious vision loss or blindness. The recurrence of herpetic keratitis is owing to the activation of latent HSV1 in trigeminal ganglia cells [113]. The lifelong establishment of the latent infection of HSV1 in the host cell nucleus may occur after the first HSV infection, and the transcription of activation stimulated by environmental factors (such as ultraviolet [UV] light and stress) is limited by the latency-associated transcript; therefore, lytic-related genes are silent [113]. In addition, activation of the latent viral genome may be regulated by post-translational modifications besides DNA methylation [113]. Therefore, active and inactive transcription of HSV1 may be regulated by m6A modification. More recently, a study showed that SARS-CoV-2 infection triggers a global response in m6A methylome of the host, and m6A negatively regulates SARS-CoV-2 infection [114]. SARS-CoV-2 enters the human body mainly through respiratory cells; however, it may enter through the eye as well. Troisi et al. [115] showed that the positive rate for conjunctival swabs was as high as 72% in patients with viral infection; in addition, corneal epithelial defects were reported in some patients. The study indicated that keratoconjunctivitis caused by viral infection occurs if the eye is unprotected, and the infected eye may transmit the virus. Therefore, eye protection may be one of the measures for preventing the spread of a virus. These studies suggest that viral infection may alter the m6A modification of antiviral mRNAs in the host to regulate infection [112]. In addition, m6A modification of viral RNA may represent a new mechanism of host–pathogen interactions during viral infection [111]. Therefore, in addition to anti-microbial agents, m6A regulatory factors related to microorganism infections may also lead to infection control. In addition, the mechanism of action of m6A in regulating the establishment and activation of HSV1 latent infection and its recurrence in the cornea warrants further investigation.

The purpose of understanding the pathogenesis of eye diseases is to transfer the bench-top knowledge to clinical application. With the advancement of technology, many new therapies have emerged for tumour treatment; among which, oncolytic viruses (OVs) have received increasing scientific and industrial attention because of their ability to specifically replicate in tumour cells and cause tumour cell lysis without affecting normal cells. OVs are a class of natural or genetically engineered viruses that can selectively kill or lyse tumour cells but have no killing effect on normal tissues [116]. In addition, OVs can be combined with inhibitors of DNA methyltransferases and histone deacetylases as well as microRNAs [117], which may be a promising treatment strategy for some complex eye diseases including ocular microorganism infections.

3.2 m6A Modification and Inflammatory Response in the Eye

Ocular inflammatory responses caused by pathogens, injuries and toxins are characterised by the release of pro-inflammatory factors from activated inflammatory cells such as macrophages, monocytes, neutrophils and lymphocytes. The primary inflammatory factors are cytokines such as interleukin-1β(IL-1β) [118], IL-6 [119], IL-17 [120], IL-18 [121, 122] and tumour necrosis factor-alpha (TNF-α) [123], which have been identified in ocular tissues. Inflammatory responses can be seen in many ocular diseases, including corneal diseases, uveitis, glaucoma, hypermature cataract, diabetic retinopathy (DR), AMD, proliferative vitreoretinopathy (PVR) and ocular and optic trauma. Although some studies have shown that IL-18 can be used for treating macular neovascularisation (MNV), five independent studies have reported that IL-18 is not therapeutic for MNV [122].

Wei et al. [124] reported that ocular inflammatory diseases are regulated by genetic and epigenetic factors. Recent studies have demonstrated that m6A plays a crucial role in controlling inflammatory responses, and several studies have shown that TNF-α is one of the major inflammatory response factors found in numerous pathological conditions in the eye [123, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134]. In a recent study, transcriptomic analysis demonstrated that TNF levels were significantly increased after fungal infection in mice. In addition, the regulation of corneal inflammatory response mediated by TNF-α in corneal fungal infection was found to be regulated by m6A modification [43]. Therefore, the increased expression of TNF-α may represent aberrant m6A modification after fungal infection.

The findings of the above mentioned studies, which state that m6A is involved in the regulation of inflammatory responses in the eye, have been supported by a systemic study. An interesting finding was that METTL3 knockdown downregulated the expression of LPS-induced inflammatory cytokines, including IL-6, IL-8 and growth-regulated oncogene-alpha (GRO-α) in human dental pulp cells [135]; these cytokines are associated with inactivation of the NF-κB and MAPK signalling pathways, suggesting that the effects of METTL3 on LPS-induced inflammation in macrophages were dependent on NF-κB. In addition, inhibition of m6A also reduces the innate immune response mediated by myeloid differentiation factor 88 (MyD88) and the Toll/IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF) pathway [135]. Zhang et al. [136] validated the importance of METTL3 in the inflammatory response. They reported that m6A modification levels were significantly increased in chronic inflammation induced by complete Freund’s adjuvant in the spinal cord of a mouse model. In addition, METTL3 overexpression augmented local inflammation, whereas its knockdown prevented inflammation-induced pain in the mouse model. Furthermore, Wang et al. [137] validated the role of METTL3 in autoimmune diseases (rheumatoid arthritis). They reported that METTL3 expression was higher in the peripheral blood mononuclear cells (PBMCs) of patients with rheumatoid arthritis than in those of healthy controls, indicating that the autoimmune response induced by LPS may be METTL3-dependent.

In addition to METTL3, the role of the m6A eraser FTO and reader YTHDF2 in inflammation has been demonstrated in a mouse model of alcohol-induced kidney injury. Aberrant modification of peroxisome proliferator-activated receptor-alpha (PPAR-α) mRNA mediated by FTO and YTHDF2 results in renal inflammation in the kidney by activating nucleotide-binding oligomerisation domain-like receptor protein 3 (NLRP3) inflammasomes and NF-κB [138]. However, recent studies have suggested that T-cell homeostasis is controlled by m6A modification [88], and chronic intestinal inflammation in mice occurs owing to the deficiency of METTL3 because of abnormal regulatory T-cell function [139].

Altogether, these studies reveal an important role of METTL3 in regulating inflammatory responses and provide a novel therapeutic target for inflammation-related eye and systemic diseases. However, the role of m6A modification factors in specific inflammatory ocular diseases should be investigated further.

3.3 m6A Modification and Degenerative Eye Diseases

Many degenerative diseases, including ocular diseases (keratoconus, cataract, retinitis pigmentosa and AMD) and systemic diseases (Alzheimer’s disease, multiple sclerosis, arthritis, Parkinson’s disease, muscular dystrophy, Huntington’s disease and cystic fibrosis), share some common characteristics of sustained degenerative response and cell death (apoptosis, necroptosis, autophagy, ferroptosis and pyroptosis) [140]. m6A modification is closely related to the pathogenesis of ocular cell degeneration. Age-related cataract is a typical ocular disease associated with lens epithelial cell degeneration. A study reported that the expression of the key element of m6A demethylases was significantly increased in lens epithelial cells in patients with age-related cataracts [51]. Furthermore, the formation of diabetic cataracts induced by high glucose levels is also associated with aberrant m6A modification, and METTL3 mediates lens epithelial cell apoptosis in patients with diabetes [141]. RPE cell dysfunction is closely related to the pathogenesis of AMD, and RPE degeneration may lead to photoreceptor death. Amyloid-β (Aβ), the major causative factor of Alzheimer’s disease [142] and one of the components of drusen, contributes to RPE degeneration [143], and RPE degeneration induced by Aβ1-40 is correlated with the reduced expression of FTO, suggesting that enhanced expression of FTO can protect RPE cells from degeneration [72]. Systemically, increased expression of METTL3 has been observed in an experimental mouse model of Alzheimer’s disease and was associated with the reduced expression of FTO [61]. In addition to the m6A machinery, single nucleotide polymorphisms in FTO have been implicated in some neurodegenerative diseases [62], and genetic variations in the FTO gene (the FTOrs9939609 risk allele) are associated with a high risk of Alzheimer’s disease [59].

The role of NLRP-3 and IL-1β in the pathogenesis of AMD (a typical degenerative retinal disease), especially in the induction of RPE apoptosis, has been validated [144, 145]. IL-1β has been demonstrated as an important factor in cell damage associated with NLRP-3 inflammasome-mediated apoptosis, and it has been shown that METTL3, the core m6A methylation transferase, is highly upregulated in endplate chondrocytes, and the increased expression of METTL3 is stimulated by one of the important cytokines, IL-1β [146]. METTL3 inhibits the activation of the PI3K/Akt pathway on endplate chondrocytes through the downstream target genes miR126-5p and PIK3R2, eventually resulting in the degeneration of endplate chondrocytes. This phenomenon indicates that IL-1β-mediated increase in METTL3 expression may play a key role in initiating degeneration, and the pathogenesis of cell degeneration may involve the inflammation–METTL3–PI3K/Akt pathway (Fig. 4).

Fig. 4.

The proposed role of abnormal m6A modification contributes to the pathological process of cellular degeneration. m6A modification may either upregulate IL-1B or downregulate FTO to increase the expression of METTL3, which may inhibit PI3K/Akt/GSK-3β by modulating miR126-5p/PIK3R2 to subsequently induce cell degeneration.

3.4 m6A Modification and Oxidative Stress Damage in the Eye

The development of ocular diseases (corneal dysfunction, cataract, glaucoma, uveitis, AMD and DR) [147, 148, 149, 150, 151, 152] and systemic diseases, including obesity, diabetes, autoimmune diseases, neurodegeneration, ischaemia/reperfusion injury and cancer, is closely associated with oxidative stress-induced damage caused by pollutants, smoking, UV rays, radiation and toxic chemicals [153, 154, 155, 156]. One of the most common features of oxidative stress is the accumulation of reactive oxygen species (ROS). Excess ROS production induces cell damage and leads to cell death, including but not limited to corneal endothelial cells [156, 157], lens epithelial cells [158], RPE cells [159] and retinal ganglion cells [149, 160], and photoreceptor dysfunction [161, 162], which may be regulated by m6A modification [163, 164]. Li et al. [51] reported that m6A is involved in the pathogenesis of age-related cataracts through its target gene of circRNA in lens epithelial cells, and the m6A methyltransferase ALKBH5 may majorly contribute to aberrant m6A methylation in lens epithelial cells. The pathogenesis of AMD involves oxidative stress-induced damage in RPE [165, 166]. Amyloid-β (Aβ), one of the factors deposited in Bruch membranes and an inducer of oxidative stress, can induce RPE degeneration [167, 168]. As mentioned above, Aβ-induced RPE degeneration may be related to m6A modifications [72].

Although the role of m6A modification in inducing oxidative stress in ocular diseases remains unclear, several studies have shown that oxidative stress-induced damage is closely related to mRNA modification, especially m6A [137, 163, 169]. The involvement of m6A modification in redox homeostasis is evidenced by the following findings. The level of m6A methylation is increased globally in response to oxidative stress [170, 171]. Hypoxic preconditioning treatment-induced protection of myoblast heart cells (H9c2) from H2O2 is associated with a significant increase in the total level of m6A methylation and METTL3 expression [170]; however, inhibition of m6A methylation attenuates the protection [170]. Additional studies have shown that kidney injury and dysfunction induced by excess ROS production are regulated by m6A RNA methylation [171, 172].

In addition to m6A writers (METTL3), m6A demethylation factors may also regulate oxidative stress [137, 173, 174]. Recent studies have reported that increased PGC-1α mRNA stability and protein expression in renal cells are mediated by the m6A eraser FTO. Therefore, oxidative stress and cell death are induced in HEK293T renal cells owing to decreased PGC-1α expression. In addition, FTO has been demonstrated to inhibit apoptosis induced by oxidative stress by regulating the expression of BAX and Bcl-2 [175], suggesting that m6A methylation is closely associated with oxidative stress [171].

m6A modification can regulate the oxidative stress response through lncRNAs [176]. Oxygen–glucose deprivation/re-oxygenation enhances free radical production, induces METTL3-dependent lnc-D63785 m6A methylation, resulting in miR-422a accumulation, and reduces the expression of the downstream gene MEF2D-MAPKK6, leading to neuronal cell apoptosis [177]. In addition, the increased expression of m6A induced by oxidative stress is associated with an enhanced expression of p21, which may regulate apoptosis [164]. These studies show that oxidative stress may be regulated by m6A methylation, and m6A expression may also be affected by oxidative stress, indicating that m6A and oxidative stress may regulate each other.

The purpose of understanding the pathogenesis of ocular diseases is to interfere with or cure a disease. Resveratrol, which stimulates the expression of SIRT1, is a well-recognised agent with broad-spectrum anti-oxidative and apoptosis-inhibiting properties [178]. Intravitreal injection of resveratrol can prevent retinal ganglion cell death caused by high intraocular pressure [179]. In a study, dietary resveratrol inhibited ROS production, reduced the expression of m6A and downregulated the expression of cleaved caspase-3 in a mouse model of AFB1-induced liver injury [180], suggesting that resveratrol is useful for treating systemic and ocular diseases related to oxidative stress through the regulation of m6A methylation.

Although ROS has been traditionally considered harmful to the human body, this concept is increasingly challenged because the dynamic modification of redox proteins by ROS is similar to the phosphorylation and acetylation modification of other proteins in various physiological processes [181]. ROS is required for cytokine, insulin growth factor (IGF), AP-1 and NF-κB signalling as well as differentiation, autophagy, metabolic adaptation and immune cell activation [181, 182, 183]. It is one of the systems in which cells respond to various stresses under physiological (oxidative stress in the nanomolar range) or pathological conditions. Therefore, when studying the relevance of m6A in relation to oxidative stress, we have to rethink the traditional concept of using antioxidants to treat oxidative stress-related diseases because antioxidants eliminate free radicals while interfering with the redox signal network regulated by ROS in cells, which may promote the development of some diseases [184, 185, 186].

3.5 m6A Modification and Ocular Angiogenetic Diseases

Angiogenesis is a complex process mediated by the overproduction of growth factors, cytokines and ECM proteins and the loss of balance between angiogenic inducers such as vascular endothelial growth factor (VEGF) and anti-angiogenic factors such as pigment epithelium-derived factor (PEDF). In addition to PEDF, the second factor of VEGF negative regulator, vascular endothelial growth inhibitor (VEGI; known as TNFSF15), plays an important role in vascular homeostasis, which specifically targets VEGF production, VEGF receptor (including R3) activation, endothelial cell proliferation and apoptosis [187, 188, 189]. The significance of VEGFI in ocular angiogenesis has been demonstrated in our previous study and other studies as well [190, 191]. In a study, VEGFI treatment prevented the leakage of retinal blood vessels in a diabetic rat model [191]. Many ocular diseases such as corneal neovascularisation, neovascular glaucoma, neovascular AMD, DR, central retinal vein occlusion and other systemic diseases (tumours, heart diseases, skin diseases, cardiovascular diseases and stroke) are associated with angiogenesis. VEGF is known to be a critical mediator of angiogenesis. However, maximal angiogenesis cannot be induced by VEGF alone, and resistance to VEGF treatment often presents in clinical practice, implying that more currently unknown factors may participate in the underlying pathogenesis of angiogenesis. m6A modification may play a critical role in regulating angiogenesis, including ocular angiogenesis [52, 75, 192, 193, 194]. To date, m6A methyltransferases (METTL3, METTL14 and WTAP), the demethyltransferase FTO and the m6A-binding protein IGF2BP3 have attracted greater attention in different cell types and animal models of angiogenesis [52, 192, 193, 194]. In DR, a break in the outer BRB is caused by RPE cell dysfunction or apoptosis induced by high glucose levels [195, 196, 197]. A recent study demonstrated that reduced expression of METTL3 may respond to the RPE barrier breakdown, whereas overexpression of METTL3 can prevent RPE cell death [42]. Furthermore, VEGF contributes to inner BRB breaks and the progression of DR [198]. The most impressive finding in this field was the relevance of m6A modification to VEGF during the formation of new vessels [194]. VEGF mRNA expression and VEGF secretion are regulated by the m6A-binding protein IGF2BP3 and YTHDF1 and 2 [194]. IGF2BP3 silencing suppresses VEGF mRNA stability and expression [194]. Furthermore, an IGF2BP3 binding site is present in the VEGF gene, and mutations in this abrogate the binding of IGF2BP3. Moreover, VEGF expression is enhanced by overexpression of IGF2BP3 [194]. These studies suggest that angiogenesis inducers may participate in VEGF-induced angiogenesis through m6A methylases, IGF2BP3 and YTHDF1 and 2, indicating that interfering m6A modification factors may be a potential therapeutic target for inhibiting ocular and systemic angiogenesis (Fig. 5).

Fig. 5.

Proposed role of m6A modification in the pathogenesis of angiogenesis. Angiogenesis inducers upregulate the expression of m6A methylases and IGF2BP3, through which YTHDF1 and 2 increase VEGF mRNA stability and promote angiogenesis.

The relationship between methyltransferases and VEGF during angiogenesis has also been revealed. In an oxygen-induced retinopathy model (retinal neovascularisation) and alkali burn-induced corneal neovascularisation model, in which VEGF serves as a major player, neovascularisation was significantly inhibited by METTL3 knockout [52]. The importance of METTL3 in angiogenesis has been emphasised in several studies on different systems [199, 200]. METTL3 knockdown reduces the expression of pro-angiogenic factors such as hepatocyte growth factor (HGF), TGF-β, granulocyte–macrophage colony-stimulating factor (GM-CSF), basic fibroblast growth factor (bFGF) and stromal cell-derived factor-1 (SDF-1) in adipose-derived stem cells [198], which are involved in ocular angiogenesis [201, 202, 203, 204]. The expression of VEGFA and its splice variants, VEGFA-164 and VEGFA-188, is inhibited in METTL3-deficient bone marrow stromal cells [205]. In addition, METTL14, one of the m6A methylase components, plays an active role in regulating cardiovascular endothelial cell proliferation, invasion [206] and angiogenesis in haemorrhoids [207].

One of the important factors for m6A demethylation is FTO. FTO expression was found to be increased in a model of corneal neovascularisation [74]. A study reported that FTO knockdown inhibited the growth and survival of von Hippel–Lindau (tumour suppressor)-deficient cells [208]. In another study, inhibition of FTO reduced the survival of both HIF-wildtype and HIF-deficient mice with tumours, indicating that FTO is an important mediator of angiogenesis [209].

Although the role of m6A in regulating angiogenesis has been extensively investigated, no studies have reported the role of m6A in maintaining the balance between VEGF and PEDF and VEGF/VEGI under normal and pathological conditions of the eye. In addition, neither dynamic changes in m6A methylation during overall angiogenesis nor specific changes in m6A methylation during angiogenesis of specific ocular tissues have been reported. Therefore, this field should be investigated further. Therefore, understanding the role of m6A modification in the regulation of ocular and systemic angiogenesis will provide insights into the pathogenesis of ocular angiogenesis.

3.6 m6A Modification with Epithelial–Mesenchymal Transition and Fibrosis in Ocular Diseases

During epithelial–mesenchymal transition (EMT), epithelial cells transform into mesenchymal cells in response to physiological and pathological changes in the microenvironment of cells. EMT is involved in the initial steps of pathogenesis of many ocular fibrotic diseases, such as advanced dry eye, corneal fibrosis, glaucoma, filtration surgery, posterior capsule opacification after cataract surgery, PVR, proliferative DR (PDR), macular neovascularisation, ocular wound healing and many systemic fibrotic diseases (pulmonary fibrosis, cystic fibrosis, cirrhosis, retroperitoneal fibrosis, myocardial fibrosis, kidney fibrosis and idiopathic pulmonary fibrosis). However, the aetiology of EMT and fibrosis is not well understood. There are three types of EMT. Type 1 EMT refers to embryonic development. Type 2 EMT is often associated with wound healing response and fibrosis after trauma, inflammation and angiogenesis. Type 3 EMT occurs with tumour invasion and metastasis [210]. Most ocular EMTs belong to type 2. In addition to EMT, another pathological process called endothelial–mesenchymal transition (EndMT) also contributes to subretinal fibrosis, including PDR [211, 212], macular neovascularisation [213] and retinopathy of prematurity [214]. EndMT is characterised by the loss of endothelial markers, including vascular endothelial (VE)-cadherin, CD31, platelet–endothelial cell adhesion molecule (PECAM) 1, TIE-1, TIE-2 and von Willebrand Factor (vWF) [215], and the upregulation of N-cadherin, vimentin and α-SMA [215]. Under the right conditions, EMT can initiate the reverse process, MET, which also plays a key role in stem cell differentiation and de-differentiation [216] as well as wound healing. EMT can be induced by inflammatory cytokines, growth factors and ECM components through numerous signalling pathways and EMT transcription factors [217]. The regulation of EMT by epigenetic factors such as DNA methylation, histone modification and non-coding RNAs has been extensively studied [218]. However, research into the role of m6A methylation in the regulation of EMT has recently begun. EMT behaviours, such as proliferation, migration and fibroblast-like morphological changes, have been observed during corneal neovascularisation in mice [74], in uveal melanoma cells [73], after glaucoma filtration surgery [114] and in PVR [65] and are regulated by FTO and METTL3. The failure of glaucoma filtration surgery is largely owing to postoperative scar formation initiated with EMT [182]. Liu et al. [219] showed that the expression of METTl3 and Smad was increased in a rabbit model after glaucoma surgery; in addition, METTL3 promoted fibrosis, whereas its knockdown inhibited the expression of SMA and fibronectin induced by TGF-β. The regulation of EMT and fibrosis by m6A in ocular tissues has been also supported by systemic studies. m6A modification is implicated in the development of EMT induced by the major EMT and fibrosis inducer TGF-β [64, 218, 220]. In a study, the expression of METTL3 and YTHDF1 was high in liver specimens obtained from patients, which was associated with EMT and a worse prognosis [63]. Moreover, a global increase in m6A levels is seen during the reprogramming of cancer cells during EMT in response to TGF-β treatment [64]. Furthermore, a study reported that the expression of m6A methylases, such as METTL3 and WTAP, was significantly enhanced in cells treated with TGF-β. A more important phenotype noticed during EMT is characterised by cell behaviour changes induced by TGF-β after METTL3 knockdown, and the loss of E-cadherin, an important epithelial marker, is recovered after METTL3 knockdown even in the presence of TGF-β. However, some EMT features of cytoskeletal rearrangements and the upregulated expression of fibronectin and vimentin stimulated by TGF-β are inhibited after METTL3 silencing [64, 218]. Furthermore, tumour cell migration and invasion in vitro and in vivo are suppressed after the deletion of METTL3 or overexpression of m6A demethylases [65]. Mechanistically, m6A may regulate TGF-β-induced EMT in multiple ways as follows: (1) m6A regulates TGF-β expression and secretion, and TGF-β secretion is almost absent if METTL3 is deleted [218]. (2) m6A regulates TGF-β dimer formation, which is essential for the initiation of TGF-β signalling. Therefore, METTL3 silencing disturbs the autocrine signalling of TGF-β [218]. (3) Smad2/3 signalling is the key signalling pathway of TGF-β for inducing EMT, and an interaction between the METTL3–METTL14–WTAP complex and Smad2/3 signalling has also been reported [219, 221]. Inhibition of METTL3 suppresses TGF-β-induced Smad3 expression in Tenon’s capsule fibroblasts [219]. (4) In addition to the Smad signalling pathway, TGF-β can induce EMT by activating non-canonical pathways, including the PI3K/Akt/mTOR and Rho GTPase pathways [222]. METTL14 is a negative regulator of TGF-β-induced EMT, and its overexpression inhibits the EMT of kidney cells by inactivating the PI3K/Akt pathway and the expression of TGF-β [223]. (5) m6A regulates the key transcription factor snail, which is responsible for TGF-β1-induced EMT, and the expression of snail protein stimulated by TGF-β is greatly inhibited by METTL3 deletion [220]. (6) METTL3 regulates JUNB, an important transcriptional regulator of TGF-β-induced EMT, by increasing JUNB mRNA levels and stability [31]. (7) m6A may regulate TGF-β-induced EMT through FTO. Increased expression of alpha-smooth muscle actin (α-SMA) is considered a marker of EMT. In a study, α-SMA was upregulated in renal tubular cells after TGF-β treatment, which was associated with significantly increased expression of FTO. In addition, both α-SMA and FTO were found to be abundant in kidney fibrosis tissue specimens, and FTO silencing suppressed TGF-β stimulation of α-SMA expression in kidney cells [224].

Altogether, these studies suggest that TGF-β-induced EMT may be tightly regulated by m6A methylation, and the TGF-β/METTL3/snail axis may control EMT [218]. Other m6A modification factors may participate in EMT processing. HNRNPA2B1 is considered one of the mediators that regulate the development of EMT through the ERK/snail signalling pathway by increasing the expression of N-cadherin and vimentin while decreasing the expression of E-cadherin [225]. To date, no studies have reported an RNA modification factor that can reverse EMT, and studies reporting on the effects of m6A on EMT or MET induced by other growth factors (specifically PDGF and CTGF), cytokines (TNF and IL-1) and ECM components are lacking. Therefore, understanding the role of m6A in EMT during the development of fibrosis in ocular diseases will provide novel insights into the pathogenesis of fibrosis and may help in reversing EMT through the regulation of m6A. In addition, such studies may help to design novel therapeutic approaches for ocular fibrotic diseases.

3.7 m6A Modification and Senescence in Ocular Cells

Senescence is a complex process that involves various cellular and tissue changes throughout the body [226, 227]. All cells and tissues undergo senescence, which is characterized by reduced response to stress, declined homeostasis, and age-related diseases. Senescence is an important predisposing factor in several ocular diseases. The effect of senescence in ocular disease is often seen in corneal endothelial cell dysfunction [228], glaucoma [229], age-related cataracts [230, 231], and age-related macular degeneration (AMD) [232, 233, 234]. Recent discoveries have shown that the m6A RNA methylation modification plays an important role in age-related disorders [235, 236]. A study of the effects of m6A modification on the pathogenesis of age-related cataracts by m6A-IP-seq and RNA-seq analysis showed that major enriched and meaningful host circRNA genes are involved in the processes of autophagy, DNA repair, and aging in age-related cataracts [51, 237, 238, 239]. In addition, Aβ induces senescence [240], and the role of m6A demethylase FTO in Aβ-induced RPE degeneration has been suggested [72]. Therefore, m6A modifications seem to be of importance in senescence in eye diseases.

There is no tissue-specific biomarker of senescence. Therefore, the following parameters may be used as indicators of senescence in ocular cells to study its relevance to m6A: loss of genomic stability, telomere shortening, aberrant epigenetic factors [241], mitochondrial dysfunction [242], increased senescence-associated beta-galactosidase [243] and senescence-associated secretory phenotype (SASP) [244], accumulation of ROS in cells, and activation of P53 [245] and p16 [246].

It would be interesting to determine whether m6A modification factors could be epigenetic biomarkers of senescence. Importantly, the global reduction in m6A methylation and METTL3 levels have been shown to be associated with the process of senescence [236, 247, 248]. The level of m6A methylation in human peripheral blood mononuclear cell (PBMCs) and human diploid fibroblasts was significantly reduced in older individuals compared with that in younger individuals [247]. Importantly, mRNA expression of the m6A methylated gene argonaute 2 (AGO2) in the older PBMC subjects was decreased, and senescence-associated β-galactosidases (SA-β-Gal) increased after knockdown of METTL3 or METTL14, indicating that suppression of m6A methylation may destabilize AGO2 mRNA and decrease AGO2 miRNA abundance, leading to cell senescence [247]. The notion of reduced m6A methylation being involved in the senescence process was further supported by a study by Wu et al. [236] which showed that METTL3 was decreased in prematurely senescent human mesenchymal stem cell models of progeroid syndromes, and deletion of METTL3 promoted cell senescence. They also found that the senescent phenotypes were preserved with the overexpression of METTL3, and that the expression of senescence inhibition gene MIS12 is under the control of the m6A reader IGF2BP2, and the loss of m6A modifications decreased the expression of MIS12 mRNA and accelerated cellular senescence [236].

RNA modifications associated with cell senescence are not limited to m6A. The synergistic effect of RNA 5-methylcytosine (m5C) and m6A on senescence regulation was recently reported. Moreover, NSUN2 (an RNA methyltransferase) can accelerate oxidative stress-induced cellular senescence by upregulation of P21 through m5C and METTL3/METTL14-mediated m6A methylation [163].

Furthermore, the possible signalling mechanisms through which m6A mediates cell senescence induction have not been characterized. Therefore, precise understanding of the mechanism underpinning m6A and senescence warrants further research. The most recent hypothesis states that Ras activation induces ROS overproduction which inhibits the expression of m6A reading protein YTHDF2 and subsequently increases the activation of MAP2K4 and MAP4K4, leading to SASP expression. Importantly, it was found that the senescence process mediated by the ROS-YTHDF2-MAPK-NF-κB pathway could be suppressed by the application of melatonin in ovarian cells [248].

The key elements of m6A modification in regulating the senescence process in ocular tissues need to be determined. Senescence is not a static endpoint but rather reflects diverse cellular abnormalities with a series of progressive cellular and behavioral changes [249]. Importantly, there is no m6A dynamic analysis in vivo from human tissues during ocular tissue senescence. Furthermore, no large-scale longitudinal cohort studies have been conducted to determine the specific transcripts that are modified by m6A in young and old people. Therefore, in this review, we discuss the key themes of m6A modification, with a focus on their role in vision function and eye diseases.

3.8 m6A Modifications and Immune Response in the Eye

There are two types of immune responses, innate and adaptive immune responses, which play an important role in maintaining ocular cell homeostasis by eliminating damaged cells and protecting the ocular tissue from pathogen infection. The innate immune response acts rapidly and can identify molecules from numerous pathogens. In contrast, adaptive immune response is slower but more specific. Aberrant immune responses are linked to many ocular diseases, including immunodeficiency due to diabetes [250, 251], HIV infection/ocular involvement, and overreaction of the immune system to the native existing cell, such as in autoimmune diseases like Sjögren’s syndrome [252], keratoconus [253], multiple sclerosis [254], rheumatoid arthritis, Graves’ disease [255], uveitis [256], and autoimmune retinopathy [257]. Of the ocular autoimmune diseases, Graves’ ophthalmopathy (GO) is a typical autoimmune disorder that causes ophthalmopathy due to hyperthyroidism and excess autoantibodies production [255, 258]. In a study of the role of m6A RNA modification in the pathogenesis of GO, we demonstrated that the expression of genes related to immune response and inflammatory processes such as lymphocyte activation and inflammatory pathways, which are associated with aberrant m6A modification factors, including m6A methylase WTAP, m6A demethylase ALKBH5, and m6A readers YTHDF2, YTHDF3, and YTHDC2, are significantly high in surgically excised extraocular muscles from patients with GO compared with that of normal subjects [45]. The results imply that m6A modification participates in the regulation of the pathogenesis of the autoimmune response in GO.

Many ocular immune-related diseases occur secondary to systemic disease. Therefore, the ocular immune diseases caused by the aberrant immune response systemically, cannot be ignored. Both systemic and ocular innate immune responses are characterized by the expression of interferon during viral infection. In addition to METTL3, the m6A reader YTHDF3 regulates innate immunity by binding to the transcription corepressor forkhead box protein O3 (FOXO3) mRNA [259]. Importantly, the innate immune response during hematopoietic development is closely related to m6A methylation, and deletion of METTL3 results in an abnormal innate immune response, which manifests as endogenous double-stranded RNA production, and activation of pattern recognition receptor pathways.

The role of m6A modification in the regulation of adaptive immunity has recently been recognized. The most impressive discovery is a new function of the m6A modification in tumor immune response [260], which shows that antigen presentation from dendritic cells to CD8+ T-cells is regulated by m6A, specifically by the m6A reader protein YTHDF1, which enhances the translation of lysosomal cathepsins, leading to reduced tumor antigen presentation. Tumor growth was significantly inhibited in mice with silenced YTHDF1 compared with wild-type mice because more CD8+ T-cells and natural killer cells were recruited in YTHDF1-deficient mice [260], suggesting that control of YTHDF1 expression may be a new approach for the treatment of tumors.

The regulation of the immune response mediated by m6A occurs through multiple mechanisms. One of the critical functions of m6A modification is the control of T-cell homeostasis. In the last few years, several studies have validated that the suppressor of cytokine signaling (SOCS) family proteins SOCS1 and SOCS3, and cytokine-inducible SH2 containing protein (CISH) SOCS genes control IL-7 signaling and play critical roles in adaptive immunity [89]. SOCS gene activation induces the proliferation and differentiation of naïve T-cells, which are tightly regulated by m6A [89]. Modification of m6A in IL-7-induced naïve T-cells boosts the inactivation of SOCS (JAK-STAT) [261, 262]. Deletion of METTL14 induces colitis in mice, suggesting that m6A methylation is critical for maintaining immune homeostasis stability of T regulatory cells [139] and alleviating various autoimmune diseases [262, 263, 264]. Nevertheless, whether these principles can be applied to ocular immune homeostasis is unclear.

Finally, the clinical value of m6A in the regulation of immune response in the prognosis and treatment of tumors has also been recognized [265, 266]. It was found that m6A regulators are related to malignancy and antitumor immune response in cancer cells [266] and that the effectiveness of anti-PD-1 therapy could be highly enhanced by knocking down METTL3 or METTL14 due to increased infiltration of CD8+ T-cells, production of IFN, monokine, chemokine (C-X-C motif) (CXCL9), and interferon-gamma induced protein CXCL10 in tumors. METTL3 and METTL14 may therefore be used as potential therapeutic targets in anti-cancer immunotherapy, including ocular melanoma [42, 73, 265].

Collectively, m6A modification is involved in numerous ocular pathologic processes (Fig. 6), and its understanding may be valuable in improving our understanding of the pathogenesis of ocular diseases.

Fig. 6.

Roles of m6A modification in the pathogenesis of eye diseases. m6A may play a significant role in various pathological processes of eye disease such as microorganism infection, inflammation, autoimmune response, senescence, angiogenesis, degeneration, EMT, fibrosis, and ocular tumorigenesis.

4. Other Important Aspects of m6A Modification in Vision Function and Eye Diseases
4.1 Implication of m6A Modification in Ocular Cell Reprogramming

Through reprogramming, we can obtain pluripotent stem cells with re-established epigenetic marks [267]. The discovery of cell reprogramming has changed the traditional view of how cells function and opens up exciting possibilities for the treatment of eye diseases [268]. Different types of cells can be genetically and epigenetically reprogramed. For example, Nrl knockdown prevents retinal degeneration [269], and reprogramming of photoreceptors can be used for vision rescue in mice [268]. Therefore, cell reprogramming has emerged as a promising strategy for the treatment of ocular diseases. However, several problems concerning cell reprogramming persist, including low efficiency, tumorigenic potential, and instability of the reprogrammed cell, Therefore, the molecular mechanisms underlying cellular reprogramming are still needed to be further investigated. The study of m6A methylation may improve our understanding of the control of cell reprogramming.

Recent studies suggest that reprogramming from one cell type to another may occur through the modulation of m6A modification. Chen et al. [270] mapped m6A modification in four cell types with different degrees of pluripotency and found that reprogramming of mouse embryonic fibroblasts to pluripotent stem cells was boosted under increased m6A levels, whereas it was hindered under reduced m6A levels. Metabolic reprogramming in hepatocellular carcinoma cells was also mediated by m6A modification [171], suggesting that m6A modification regulation may restore normal cell functions under various pathological conditions.

4.2 Implication of m6A Modification and Stem Cell Differentiation in Vision Function and Eye Diseases

Stem cells, including ocular stem cells, are pluripotent cells that can differentiate into different types of cells. Normal cell differentiation is an important process in biological development. Ocular stem cells are one of the earliest stem cells used in regenerative medicine [271]. Ocular stem cells comprising the iris, sclera, photoreceptors, corneal limbus, cornea and conjunctiva, iris pigment epithelium, ciliary body epithelium, trabecular meshwork, retina, RPE, and orbital stem cells can be potentially used to treat eye diseases [272], including corneal disease, cataract, glaucoma, retinitis pigmentosa, and age-related macular degeneration AMD. However, understanding the precise mechanisms controlling single stem cell differentiation warrants further research. For example, the transcription factors that control ocular stem cell differentiation still need to be ascertained. m6A methylation plays a fundamental role in stem cell self-renewal and differentiation [68, 270]. It has been shown that the expression of pluripotency genes SOX2, NANOG, and DPPA3, was compromised when METTL3 and METTL14 were silenced. This phenomenon was associated with an increased expression of FGF5, CDX2, and SOX17, genes related to development in mouse embryonic stem cells [273]. Furthermore, METTL3 depletion prevented the self-renewal of porcine-induced pluripotent stem cells [83, 274]. The stability and expression levels of transcripts encoding developmental regulatory factors are negatively correlated with m6A because of the inhibition of the methyltransferases METTL3 and METTL14 compromise the expression of pluripotency genes such as SOX2 and NANOG [273]. The importance of methyltransferases and the m6A modification is supported by a recent report that showed that the self-renewal of porcine iPSC was inhibited by METTL3 knockdown [275].

Stem cells have been used for cell therapies, the development of new drugs, and for understanding the regulation of stem cell differentiation, including that of ocular stem cells is critic important [276, 277, 278, 279]. However, directional control of the differentiation of stem cells into specific organs or tissues, including ocular tissues, is still under investigation [280]. In addition to physical, chemical, mechanical, and biological (DNA methylation, histone modification, and non-coding RNA) approaches, regulation of m6A modification may be promising for the control of stem cell directional differentiation. Mapping of m6A modifications during embryo development in zebrafish revealed that the key gene notch1a controls the direction of endothelial–hematopoietic transformation through the cooperation of YTHDF2 with the activation of the Notch signaling pathway [281]. These results indicate the role of mRNA m6A modification in regulating the fate determination of stem cells and explain the key role of m6A in directional stem cell differentiation [281]. The expression of m6A modification factors has been illustrated in ocular cells, as mentioned previously. It has also been demonstrated that transcriptional factors, such as NANOG, PAX6, and SOX2, can control stem cell differentiation in numerous ocular stem cells [271, 282].

In conclusion, m6A RNA methylation provides an additional regulatory mechanism in regulating stem cell differentiation and pluripotency.

4.3 Implication of m6A as a New Biomarker of Ocular Diseases

Biomarkers are crucial in the study of the pathogenesis and the early detection, diagnosis, and prognosis of diseases. Moreover, biomarkers are closely observed for monitoring treatments [283, 284]. However, few biomarkers are translated into clinical practice. Biologic stability and specificity are the major issues in using some biomarkers in clinical practice. The use of m6A as a biomarker could be challenging as m6A modification factors are likely to vary in different cells, and therefore, may be tissue-specific [11, 39, 57, 285]. As RNA and m6A modification factors are universally expressed in all eukaryotic cells, including ocular cells, plasma, serum, aqueous humor, vitreous, and any solid ocular tissue can be used to identify m6A biomarkers for the screening, diagnosis, and therapy of ocular diseases.

In the last few years, the value of RNA modification, especially m6A, for its potential roles in tumors [286], neovascular disease [74], degenerative disease [287], diabetes [288], and fibrotic diseases [220, 289] has been recognized. Potential biomarkers for ocular disease, particularly for uveal melanoma, have also been suggested [42, 73, 290]. In particular, the aberrant expression of METTL3 in melanoma cell lines and tissues may be considered a new biomarker for tumor diagnosis and treatment. Notably, reduced m6A is suggested as a possible biomarker of type 2 diabetes [291]. ZFAS1 expression regulated by METTl14 has been shown to be significantly increased in patients with atherosclerosis compared with those without atherosclerosis, suggesting that m6A modification factors may be a novel biomarker for monitoring atherosclerosis susceptibility [292, 293, 294]. Moreover, the prognostic value of m6A methylation has also been reported in patients with uveal melanoma [290], bladder cancer [295], and colon cancer [296]. In addition, METTL3 is considered a promising biomarker for determining the progression of gastrointestinal cancer and therapeutic response prediction [297]. Thus, collectively, the alteration of m6A modification may serve as a potential diagnostic biomarker for human diseases, including ocular diseases. However, the m6A signature in normal ocular tissues needs to be determined before m6A modification factors could be used as a biomarker for the screening of ocular disease.

4.4 Implication of m6A Modification and Autophagy on Vision Function

Autophagy clears degenerated cells and reuses cellular components to maintain cell hemostasis. Without exception, autophagy participates in maintaining homeostasis of the cornea, aqueous humor, RPE, neural retina, and lens. Abnormalities in autophagy function contribute to retinal degeneration [298, 299], retinal injury [300], light-induced stress [301, 302], hypoglycemia [303, 304], lipofuscin accumulation in the RPE, and the pathogenesis of AMD [305]. Moreover, dysfunction of autophagy may be linked to photoreceptor degeneration [183, 220]. Furthermore, inhibition of autophagy induces retinal ganglion cell death [271, 306]. The implication of m6A modification and autophagy in ocular disease also was reported recently, a study investigating the relationship between m6A and the pathogenesis of age-related cataracts using gene ontology analysis revealed that autophagy-related genes are associated with altered m6A-circRNAs and methyltransferases in lens epithelium cells [51]. Systemic studies have shown that m6A plays a critical role in regulating macro autophagy/autophagy by targeting ATG5 and ATG7 [307], which are key proteins in autophagic vesicles. Interestingly, knockdown of FTO inhibited autophagosome formation and autophagy, suggesting that the expression of autophagy-related genes and autophagy is regulated by m6A modification, and m6A modification is required for autophagy [308]. Data from ocular and systemic studies on the relevance of m6A to autophagy imply the importance of autophagy in ocular homeostasis and diseases. Nevertheless, several fundament questions need to be answered before linking m6A and autophagy to vision function and eye diseases. For example, whether autophagy in ocular tissues is consistent with the expression of m6A modification factors and what is the role of the m6A and autophagy in the maintenance of the structure and/or normal physiological function of the eye (from the cornea to the vision pathway), need to be determined.

4.5 Implication of m6A Modification on Exosomes Mediating Ocular Function

Exosomes provide intercellular communication and transmission of macromolecules between cells and they are enriched in proteins, lipids, mRNA, miRNA, and DNA. Therefore, they may contribute to the maintenance of normal cell functions, including cellular proliferation and differentiation, extracellular matrix (ECM) production, wound healing, immune response, and metabolic waste removal [309, 310]. In addition, there is a growing interest in using exosomes as biomarkers and therapy carriers [309]. Exosomes have been found in most ocular tissues, including corneal epithelial cells, RPE, trabecular meshwork, iris, and ciliary body epithelial cells, tear fluid, vitreous humor, human iPSC-RPE, retinal astrocyte, and uvea [311]. RNA is shuttled from one cell to another via exosomes, which is known as “exosomal shuttle RNA”. Interestingly, depletion of METTL3 reduced cellular and extracellular levels of miRNAs containing m6A consensus sequences [312]. To date, no study has demonstrated the direct regulation of the mRNA in exosomes by m6A. Nevertheless, it is known that exosomes are enriched in mRNA and miRNA, and it is likely that RNA is subject to m6A regulation and could potentially affect gene expression and protein translation. The roles of m6A modification factors derived from exosomes of ocular tissue in the maintenance of corneal and lens transparency, normal intraocular pressure, retinal blood barrier integrity, and ocular immune tolerance, the prevention of ocular autoimmune diseases and the retinal protective effects are largely unknown. Therefore, the precise role of exosomes mediated by m6A in maintaining normal cellular function and its effects on the pathogenesis of diseases, including ocular disease, should be determined.

4.6 Signalling Pathways Involved in the Manifestation of the Effects of m6A Modification in Ocular Cells

Cell signalling is complex and essential for ocular cell differentiation, proliferation, migration, survival, cell polarity, stem cell renewal, apoptosis, and the development of many pathological conditions. Common cell signalling pathways include TGF-β signalling, mitogen-activated protein kinase (MAPK), Ras homolog (Rho) kinase, brain-derived neurotrophic factor (BDNF), c-Jun N-terminal kinase (JNK), phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt), phosphatase and tensin homolog (PTEN), B-cell lymphoma 2 (Bcl-2), caspase, calcium-calpain signalling, WNT/β-catenin signalling [313, 314], Hippo-YAP signalling [315], and the Hedgehog signalling pathway [316]. The PI3K/Akt pathway has been found to be the most common ocular cell signalling pathway associated with m6A modification [42, 43, 75]. It was found that the activation of the PI3K/Akt signalling pathway, especially phosphorylation of PI3K/Akt, significantly increased after corneal infection, which was also enriched in differential m6A-containing mRNAs. This suggests that m6A may regulate the PI3K/Akt signalling pathway during fungal infection [43]. In addition, overexpression of the m6A methylase METTL3 could enhance phosphorylated Akt levels through the miR-25-3p/PTEN axis and lead to the rescue of RPE cells from high glucose insults [75]. Similarly, PI3K/Akt was found to be one of the major molecular signalling pathways involved in the formation of nuclear cataracts in patients with high myopia, as well as the pathogenesis of retinoblastoma [317]. Furthermore, the mesenchymal–epithelial transition (MET)/Akt signalling pathway has been suggested to participate in the pathogenesis of uveal melanoma mediated by m6A [73]. The Hippo-YAP signalling pathway is differentially expressed in uveal melanoma cells, which are regulated by m6A [42]. Another common signalling pathway closely related to m6A-mediated cellular function is focal adhesion kinase (FAK). The m6A reader protein YTHDF2 and the eraser protein FTO target FAK in the regulation of corneal angiogenesis. Furthermore, silencing of YTHDF2 increases FAK expression, while knockdown of FTO reduces FAK expression [74].

Besides the PI3K/Akt signalling pathway involved in m6A modification in ocular cells, MAPK signalling is also an important signalling pathway in the lens epithelium of age-related cataracts modulated by m6A methylation [51]. Importantly, PI3K and FAK have a cross-talk with MAPK [318], suggesting that activation of PI3K and FAK signalling may also activate MAPK [319].

Protein kinase A (PKA), commonly known as cAMP-dependent A kinase, is responsible for cAMP-mediated stimulation. It was found that FTO can downregulate PKA expression by demethylating PKA mRNA in RPE cells. Importantly, RPE cell death is increased by activation of the PKA/cyclin AMP-responsive element-binding (CREB) signalling pathway on FTO inhibition [72].

The NF-κB singling pathway may mediate many physiologic and pathologic events. A previous study demonstrated that NF-κB is listed among the top five of the 10 signalling pathways identified in the specimens of extraocular muscles of GO during the study of m6A methylation in the pathogenesis of GO. Notably, there is a close relationship between YTHDF expression and the NF-κB signalling pathway in controlling inflammation in the pathogenesis of GO [45]. The expression of NF-κB is regulated by the m6A methylation transferase METTL3 through its upstream gene MYD88, which controls cell differentiation [320], suggesting that RNA methylation, especially m6A modification plays an essential role in regulating signalling in many cells, including ocular cells.

4.7 Implication of m6A Modification in Improving Precision Medicine Use in Ocular Tumors

It is currently challenging to treat ocular tumors, such as RB and melanoma, using a non-surgical approach. The genetic background may be quite different in people with the same tumor, therefore, different people may respond differently to the same treatment. After decades of research, the application of precision medicine wherein treatment is tailored based on the understanding of the genetic variability of patients’ tumors, has gained impetus. Precision medicine aims to target disease with patient-specific treatment. Interestingly, m6A modification may facilitate precision medicine. For example, the status of m6A methylation in uveal melanoma differed between patients with uveal melanoma in two separate reports [42, 73], suggesting that m6A modification is individual-specific. Thus, m6A RNA methylation may be dynamic and relevant to precision medicine. m6A methylation signatures may thus improve our understanding of the mechanism of many complex eye diseases. According to the Precision Medicine Initiative (https://obamawhitehouse.archives.gov/precision-medicine), precision medicine emphasizes the variability of the genetic background, environment, and lifestyle. However, precision medicine has not been widely applied in clinical practice, especially in relation to ocular tumor therapy.

Many studies have shown that m6A modifications are characteristic of tumor development. Remarkably, the m6A modification patterns in the pathogenesis of tumors, such as uveal melanoma [42, 73, 321], retinoblastoma [317], and glioblastoma [322, 323], female reproductive system cancers [324], acute myeloid leukemia [325], breast cancer [326], prostate cancer [327], liver cancer [57, 328], lung cancer [329], pancreatic cancer [330], colorectal cancer [331], kidney cancer [332], and gastric cancer [333] are highly variable. The role of m6A modifications in cancer genesis is controversial. The effects of m6A modifications on cellular function vary frequently. Some methylated genes promote tumor development, while the demethylation of other genes enhances tumor growth. For example, m6A methylation can promote or inhibit uveal melanoma [42, 73]. Elevated levels of METTL3 or METTL14 (suggesting increases in RNA methylation) facilitate the translation of the oncogene c-MYC and BCL2 in AML. In contrast, demethylation of tumor suppression genes, ankyrin repeats, and SOCS box protein 2 and retinoic acid receptor alpha (RARα) by m6A eraser FTO leads to the downregulation of tumor suppression genes [58]. METTL3 was considered an oncogenic factor of uveal melanoma [40, 73] and retinoblastoma [317]; however, METTL3 has also been reported as a tumor suppressor in ocular melanoma [42] and other cancers [39], indicating that higher expression of METTL3 is predictive of better survival in patients with renal cell carcinoma and that knockdown of METTL3 inhibits tumor cell EMT [334]. Therefore, the role of METTL3 may be altered in different tissues and cells.

The role of m6A methylation in tumorigenesis is variable in different tumors, and the interplay between methylation and demethylation in specific tumors and eye diseases and specific target genes seems to be critical for tumorigenesis and tumor treatment [42, 58]. However, studies that have used the parameters of m6A modification factors as a specific target (implying precision medicine) for cancer, including ocular tumor and complex eye disease treatments, are limited. A biopsy procedure might be required to determine which m6A modification factor is unique in a specific cancers and individuals, followed by a test to obtain the profiling of m6A regulatory factors in the ocular tumor. Through this, ophthalmologists may be able to develop different treatments tailored to different individuals.

4.8 Potential Application of m6A Methylation Activators and Inhibitors in the Treatment of Ocular Disease

Owing to the extensive involvement of m6A modification in the pathogenesis of diseases, the targeting of m6A regulatory factors may be a treatment strategy for human diseases by promoting or inhibiting m6A methylation.

4.8.1 m6A Methylation Activators

Small molecular compounds such as m6A activators may be useful in the treatment of certain diseases associated with the decreased expression of m6A methylase [335]. A small molecular compound was found to bind the active site of the METTL3–METTL14–WTAP complex and promote the activation of m6A RNA methylation [335], suggesting a new treatment for the regulation of m6A modification externally and promoting the development of more such small molecules for possible applications in ocular disease. In conclusion, m6A activators may be considered potential therapies for the treatment of some ocular diseases, such as melanoma, in which m6A methylation is reduced [42].

4.8.2 m6A Methylation Inhibitors

The aberrant expression of m6A modification factors has been shown to be related to many ocular diseases [42, 45, 51, 72, 73, 74, 75]. The altered expression of m6A modification factors in ocular diseases has also been demonstrated. Therefore, inhibition of m6A modification factors may be promising for the treatment of ocular diseases, especially complex diseases (macular neovascularization, uveitis, DR, glaucoma, and myopia) and diseases resistant to regular therapy. For example, the success of anti-VEGF treatment for macular neovascularization is variable; 15–45% of patients do not respond well to anti-VEGF treatment [336, 337]. In a review paper, Garbo et al. [338] summarized the most recent updates on m6A related modulators and their potential treatment in human diseases, including 15 m6A modulators targeting METTL3, FTO, ALKH5 [338]. The review discussed the potential of FTO inhibitors, including dihydroxyfuran sulfonamides, inhibitors of hypoxia-inducible factor prolyl-hydroxylases (PHDs), chlororesorcinol analogs, meclofenamic acid (MA) and entacapone, in inhibiting FTO. Furthermore, imidazobenzoxazin-5-thione MV1035 as a specific ALKBH5 inhibitor and its ability to modulate the immune response in tumors were discussed [338]. There are several approaches to inhibit or regulate m6A modification factors. These include the new use of old drugs. We now realize that drugs, such as meclofenamic acid, can regulate m6A modification factors. Meclofenamic acid is a non-steroidal anti-inflammatory drug that can inhibit FTO function. The use of meclofenamic acid in ocular diseases has not yet been reported. Thus, research is being conducted to identify specific inhibitors of FTO for application in clinical settings [339]. Furthermore, the use of general methyltransferase inhibitors is also being explored. Currently, no m6A writer, eraser, and reader-specific inhibitors are available commercially, except for the general nucleoside analog Sinefungin-SAH [340, 341], which is a nucleoside S-adenosyl-1-methionine analogous to methyltransferase inhibitors for protein, DNA, and RNA methyltransferases [342] and lacks specificity. Next, the application of small molecular compounds with a relatively specific function for easy penetration into tissue has drawn great attention from researchers in the field. Two studies have been conducted using small molecular compounds to treat acute myeloid leukemia in vitro and in experimental animals. One study used the m6A writer METTL3 [343], while the other study used an FTO inhibitor [344, 345]. Both studies initially aimed at assessing the effects of METTL3 and FTO inhibition on the treatment of acute myeloid leukemia. The FTO inhibitor FB23-2 inhibited cell proliferation and induced cell apoptosis in myeloid leukemia cell lines and primary cells in xeno-transplanted mice by specifically binding to FTO molecules [344]. Later, the other two small molecular compounds were named CS1 and CS2 (or Brequinar), both of which could selectively inhibit FTO. Importantly, they have much higher efficiency and are stronger than the previously reported FTO inhibitors [309]. The compounds not only significantly inhibited tumor growth but also improved the survival of experimental animals. Mechanically, these compounds can enhance the sensitivity of cancer cells to T-cell cytotoxicity and immune evasion. Other FTO and ALKBH5 inhibitors, 2-oxoglutarate (2OG), N-oxalylglycine (NOG), pyridine-2, 4-dicarboxylate, Rhein, MA, IOX3, and R-2-hydroxyglutarate (R-2HG), have been shown to inhibit tumor cell growth and induce apoptosis [346, 347]. Recently, the METTL3 inhibitor STM2457 was reported to suppress myeloid leukemia cell growth in vitro and in animals through the selective inhibition of METTL3 and induction of tumor cell apoptosis [348]. Bedi et al. [349] also identified two derivatives of S-adenosyl-L-methionine (SAM) that show a high biding efficiency to METTL3 and may be considered as a candidate for a small molecular compound for the inhibition of METTL3. Moroz-Omori et al. [350] showed that an inhibitor of METTL3, UZH1a, which is selective and cell permeable, can function for up to six days and shows no negative effects on other RNA modifications (m1A, m6Am, m7G). Other consideration of the control of the expression of m6A modification factors is the application of CRISPR/Cas9, the CRISPR/Cas9 techniques can not only enable precise and target-specific modifications of DNA but also modify RNA to edit m6A at a specific site on mRNA [209, 351, 352, 353]. A study by Barbieri showed that the growth of acute myeloid leukemia cells was significantly inhibited by the deletion of METTL3 using the CRISPR technique [209]. Furthermore, specific deletion of ALKBH5 using the CRISPR technique led to the demethylation of oncoproteins of epidermal growth factor receptor and MYC, and inhibited proliferation of tumor cells [353]. Therefore, targeting m6A modification factors such as writers, erasers, and readers as a potential treatment for tumors is gaining attention in m6A research [346, 354].

Besides the progress in the study of the inhibitors of m6A modification factors, a number of manufacturers are also developing drugs targeting m6A modification factors, such as METTL3 inhibitors, which are also inching closer toward clinical trials [355]. The majority of these compounds, for the manipulation of m6A modification, focus on tumor treatment, which may be beneficial for patients who do not respond well to traditional cancer therapy or develop drug resistance [354, 355]. These developments in tumor treatment will help drive the development of potential therapies for ocular and systemic diseases, Notably, we are still at an early stage of the use of m6A-related drug in the treatment of human diseases.

5. Conclusions and Challenges

In addition to m6A modification, the importance of other RNA methylations such as N1-methyladenosine (m1A), 5-methylcytosine (m5C), pseudouridine (Ψ), 2’-Omethylation (2’O-Me), inosine (I), uridine (U),7-methylguanine (m7G), and N4-acetylcytidine (ac4) is gradually recognized in biomedical research. These modifications occur through specific modification enzymes (methylase, demethylase, and binding proteins), which may function in multiple locations synergistically or alone. Therefore, gene expression and behavior in ocular cells may be determined by the interaction of multiple factors. Regardless, understanding RNA methylation modifications in ocular cells may provide new insights into the molecular mechanisms of ocular function and eye diseases.

The regulation of RNA methylation, specifically m6A methylation, may provide novel therapeutic strategies for the treatment of eye diseases, particularly for diseases that produce resistance to traditional therapies, as well as metabolic, degenerative, inflammatory, and vascular eye diseases. Determination of RNA methylation patterns may be facilitated by the application of single-cell sequencing and third-generation sequencing and will enable and improve prognostic analysis, early screening, diagnosis, and targeted therapy for eye diseases.

Nevertheless, the importance of m6A modification in biomedical research has been recognized relatively recently and its potential clinical applications in the treatment of eye diseases is a relatively new concept. Some of the challenges and questions we face going forward are as follows:

(1) Do m6A writers, erasers, and readers participate in the regulation of other cellular functions besides RNA transcription, processing, translation, and metabolism in ocular cells? Do different m6A modification factors play the same function in different ocular cells? Are they tissue- or cell-specific?

(2) The role of RNA methylation in vision function and the pathogenesis of eye diseases is complex and variable. The most challenging question in the study of m6A modification in vision function and eye diseases is to monitor dynamic (real-time) m6A changes under physiological and pathological conditions. For example, what is the role of m6A methylation in vision and eye embryonic development, maintenance of corneal transparency, maintenance of aqueous humor homeostasis, lens epithelial cell metabolism, blood–retinal barrier intake, subretinal vascularization, immune privilege, RPE, photoreceptor cell, glial cell function, extraocular muscles, maintenance of intraocular pressure, lens transparency, vision acuity, and the metabolism of vision cycle proteins? What is the pattern of m6A methylation after birth and what changes occur during a person’s life in specific ocular tissues and cells, and what is the relevance to vision function? Importantly, what are the dynamic changes in m6A in the pathogenesis of all eye diseases?

(3) The discovery of glycoRNAs on the surface of multiple cell types in different organisms, such as humans, mice, hamsters, and zebrafish, was a revolutionary finding in biological research [356]. The RNA molecules modified by glycosylation may exert some important cellular functions and insight into the mechanism of human diseases. If RNA methylation occurs in glycoRNAs, what is its significance in the regulation of cellular function and its relationship to ocular disease?

(4) What is the relationship between RNA methylation and extrachromosomal circular DNA (eccDNA)? EccDNA is widely present in various eukaryotes, including humans, carries complete genes and special regulatory elements and can replicate independently [357, 358, 359, 360]. The genes carried by eccDNA can actively transcribe into mRNA and play an important role in regulating cell function and in numerous human diseases as independent DNA molecules. Therefore, it would be interesting to determine if RNA from eccDNA is modified by m6A modification factors. The most recent study led by Zhang showed that mRNA m6A modification can also contribute to human disease heritability [361]. This concept in vision function and eye research remains to be elucidated.

Abbreviations

m6A, N6-methyladenine; RPE, retinal pigment epithelium; AMD, age-related macular degeneration; TGF-β, transforming growth factor-beta; ECM, extracellular matrix; MET, mesenchymal–epithelial transition; GO, Graves’ ophthalmopathy; HCMV, human cytomegalovirus; MNV, macular neovascularization; DR, diabetic retinopathy; PVR, proliferative vitreoretinopathy; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor; PEDF, pigment epithelium-derived factor; HGF, hepatocyte growth factor; HIF, hypoxia-inducible factor; EMT, epithelial–mesenchymal transition; PBMC, peripheral blood mononuclear cells; BRB, blood–retinal barrier.

Author Contributions

XL, SH and HY carried out the conception of this review; XL, SH and HY designed this review; XL, BM and SH drafted the manuscript and prepared the Figs; ML, LL, XZ, MD and JY edited the manuscript and collected the related references; XL, SH, HY and BM revised the manuscript and improved the language.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

Not applicable.

Funding

This work was supported by National Natural Science Foundation of China (81873681, 81770952), the Basic Research Program of Henan Eye Hospital (20JCQN005) and the “23456” project of Henan Provincial People’s Hospital (ZC20200296).

Conflict of Interest

The authors declare no conflict of interest. SH is serving as one of the Editorial Board members of this journal. We declare that SH had no involvement in the peer review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to GP.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
Nilsen TW. Internal mRNA Methylation Finally Finds Functions. Science. 2014; 343: 1207–1208.
[2]
Roundtree IA, He C. RNA epigenetics — chemical messages for posttranscriptional gene regulation. Current Opinion in Chemical Biology. 2016; 30: 46–51.
[3]
Dominissini D, Nachtergaele S, Moshitch-Moshkovitz S, Peer E, Kol N, Ben-Haim MS, et al. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature. 2016; 530: 441–446.
[4]
Li J, Yang X, Qi Z, Sang Y, Liu Y, Xu B, et al. The role of mRNA m6a methylation in the nervous system. Cell & Bioscience. 2019; 9: 66.
[5]
Boccaletto P, Machnicka MA, Purta E, Piątkowski P, Bagiński B, Wirecki TK, et al. MODOMICS: a database of RNA modification pathways. 2017 update. Nucleic Acids Research. 2018; 46: D303–D307.
[6]
Wei C, Gershowitz A, Moss B. N6, O2′-dimethyladenosine a novel methylated ribonucleoside next to the 5′ terminal of animal cell and virus mRNAs. Nature. 1975; 257: 251–253.
[7]
Liu J, Li K, Cai J, Zhang M, Zhang X, Xiong X, et al. Landscape and Regulation of m6A and m6Am Methylome across Human and Mouse Tissues. Molecular Cell. 2020; 77: 426–440.e6
[8]
Mauer J, Luo X, Blanjoie A, Jiao X, Grozhik AV, Patil DP, et al. Reversible methylation of mA in the 5’ cap controls mRNA stability. Nature. 2017; 541: 371–375.
[9]
Jiang J, Song B, Chen K, Lu Z, Rong R, Zhong Y, et al. M6AmPred: Identifying RNA N6, 2′-O-dimethyladenosine (m6a_m) sites based on sequence-derived information. Methods. 2021. (in press)
[10]
Xie S, Chen W, Chen K, Chang Y, Yang F, Lin A, et al. Emerging roles of RNA methylation in gastrointestinal cancers. Cancer Cell International. 2020; 20: 585.
[11]
Engel M, Eggert C, Kaplick PM, Eder M, Röh S, Tietze L, et al. The Role of m6a/m-RNA Methylation in Stress Response Regulation. Neuron. 2018; 99: 389–403.e9.
[12]
Xiong X, Li X, Yi C. N1-methyladenosine methylome in messenger RNA and non-coding RNA. Current Opinion in Chemical Biology. 2018; 45: 179–186.
[13]
Li X, Xiong X, Wang K, Wang L, Shu X, Ma S, et al. Transcriptome-wide mapping reveals reversible and dynamic N1-methyladenosine methylome. Nature Chemical Biology. 2016; 12: 311–316.
[14]
Tong C, Wang W, He C. M1a methylation modification patterns and metabolic characteristics in hepatocellular carcinoma. BMC Gastroenterology. 2022; 22: 93.
[15]
Zhang C, Jia G. Reversible RNA Modification N1-methyladenosine (m1A) in mRNA and tRNA. Genomics, Proteomics & Bioinformatics. 2018; 16: 155–161.
[16]
Han X, Wang M, Zhao Y, Yang Y, Yang Y. RNA methylations in human cancers. Seminars in Cancer Biology. 2021; 75: 97–115.
[17]
Chen YS, Yang WL, Zhao YL, Yang YG. Dynamic transcriptomic m5 C and its regulatory role in RNA processing. Wiley Interdisciplinary Reviews: RNA. 2021; 12: e1639.
[18]
Amort T, Rieder D, Wille A, Khokhlova-Cubberley D, Riml C, Trixl L, et al. Distinct 5-methylcytosine profiles in poly(A) RNA from mouse embryonic stem cells and brain. Genome Biology. 2017; 18: 1.
[19]
Yang X, Yang Y, Sun B, Chen Y, Xu J, Lai W, et al. 5-methylcytosine promotes mRNA export — NSUN2 as the methyltransferase and ALYREF as an m5C reader. Cell Research. 2017; 27: 606–625.
[20]
Nombela P, Miguel-López B, Blanco S. The role of m6a, m5C and Ψ RNA modifications in cancer: Novel therapeutic opportunities. Molecular Cancer. 2021; 20: 18.
[21]
Song J, Zhuang Y, Zhu C, Meng H, Lu B, Xie B, et al. Differential roles of human PUS10 in miRNA processing and tRNA pseudouridylation. Nature Chemical Biology. 2020; 16: 160–169.
[22]
Eyler DE, Franco MK, Batool Z, Wu MZ, Dubuke ML, Dobosz-Bartoszek M, et al. Pseudouridinylation of mRNA coding sequences alters translation. Proceedings of the National Academy of Sciences. 2019; 116: 23068–23074.
[23]
Morais P, Adachi H, Yu YT. The Critical Contribution of Pseudouridine to mRNA COVID-19 Vaccines. Frontiers in Cell and Developmental Biology. 2021; 9: 789427.
[24]
Dai Q, Moshitch-Moshkovitz S, Han D, Kol N, Amariglio N, Rechavi G, et al. Nm-seq maps 2′-O-methylation sites in human mRNA with base precision. Nature Methods. 2017; 14: 695–698.
[25]
Ringeard M, Marchand V, Decroly E, Motorin Y, Bennasser Y. FTSJ3 is an RNA 2′-O-methyltransferase recruited by HIV to avoid innate immune sensing. Nature. 2019; 565: 500–504.
[26]
Chen J, Li K, Chen J, Wang X, Ling R, Cheng M, et al. Aberrant translation regulated by METTL1/WDR4‐mediated tRNA N7‐methylguanosine modification drives head and neck squamous cell carcinoma progression. Cancer Communications. 2022; 42: 223–244.
[27]
Han H, Yang C, Ma J, Zhang S, Zheng S, Ling R, et al. N7-methylguanosine tRNA modification promotes esophageal squamous cell carcinoma tumorigenesis via the RPTOR/ULK1/autophagy axis. Nature Communications. 2022; 13: 1478.
[28]
Malbec L, Zhang T, Chen Y, Zhang Y, Sun B, Shi B, et al. Dynamic methylome of internal mRNA N7-methylguanosine and its regulatory role in translation. Cell Research. 2019; 29: 927–941.
[29]
Wang H, Chen RB, Zhang SN, Zhang RF. N7-methylguanosine modification of lncRNAs in a rat model of hypoxic pulmonary hypertension: a comprehensive analysis. BMC Genomics. 2022; 23: 33.
[30]
Arango D, Sturgill D, Alhusaini N, Dillman AA, Sweet TJ, Hanson G, et al. Acetylation of Cytidine in mRNA Promotes Translation Efficiency. Cell. 2018; 175: 1872–1886.e24.
[31]
Sas-Chen A, Thomas JM, Matzov D, Taoka M, Nance KD, Nir R, et al. Dynamic RNA acetylation revealed by quantitative cross-evolutionary mapping. Nature. 2020; 583: 638–643.
[32]
Jin G, Xu M, Zou M, Duan S. The Processing, Gene Regulation, Biological Functions, and Clinical Relevance of N4-Acetylcytidine on RNA: a Systematic Review. Molecular Therapy - Nucleic Acids. 2020; 20: 13–24.
[33]
Tsai K, Jaguva Vasudevan AA, Martinez Campos C, Emery A, Swanstrom R, Cullen BR. Acetylation of Cytidine Residues Boosts HIV-1 Gene Expression by Increasing Viral RNA Stability. Cell Host & Microbe. 2020; 28: 306–312.e6.
[34]
Motorin Y, Helm M. RNA nucleotide methylation. WIREs RNA. 2011; 2: 611–631.
[35]
Cheng Q, Xiong J, Ma C, Dai Y, Ding J, Liu F, et al. Chemical tagging for sensitive determination of uridine modifications in RNA. Chemical Science. 2020; 11: 1878–1891.
[36]
Wei C, Gershowitz A, Moss B. Methylated nucleotides block 5′ terminus of HeLa cell messenger RNA. Cell. 1975; 4: 379–386.
[37]
Desrosiers R, Friderici K, Rottman F. Identification of Methylated Nucleosides in Messenger RNA from Novikoff Hepatoma Cells. Proceedings of the National Academy of Sciences. 1974; 71: 3971–3975.
[38]
Coker H, Wei G, Brockdorff N. M6a modification of non-coding RNA and the control of mammalian gene expression. Biochimica Et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 2019; 1862: 310–318.
[39]
Ma S, Chen C, Ji X, Liu J, Zhou Q, Wang G, et al. The interplay between m6a RNA methylation and noncoding RNA in cancer. Journal of Hematology & Oncology. 2019; 12: 121.
[40]
Liu J, Dou X, Chen C, Chen C, Liu C, Xu MM, et al. N6-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science. 2020; 367: 580–586.
[41]
Weng Y, Wang X, An R, Cassin J, Vissers C, Liu Y, et al. Epitranscriptomic m6a Regulation of Axon Regeneration in the Adult Mammalian Nervous System. Neuron. 2018; 97: 313–325.e6.
[42]
Jia R, Chai P, Wang S, Sun B, Xu Y, Yang Y, et al. M6a modification suppresses ocular melanoma through modulating HINT2 mRNA translation. Molecular Cancer. 2019; 18: 161.
[43]
Hu J, Lin Y. Fusarium infection alters the m6a-modified transcript landscape in the cornea. Experimental Eye Research. 2020; 200: 108216.
[44]
Zhang H, Shi X, Huang T, Zhao X, Chen W, Gu N, et al. Dynamic landscape and evolution of m6a methylation in human. Nucleic Acids Research. 2020; 48: 6251–6264.
[45]
Zhu L, Li S, He S, Tong Q, Wang L, Li X, et al. The critical role of m6a methylation in the pathogenesis of Graves’ ophthalmopathy. Eye and Vision. 2020; 7: 55.
[46]
Yang Y, Hsu PJ, Chen Y, Yang Y. Dynamic transcriptomic m6a decoration: writers, erasers, readers and functions in RNA metabolism. Cell Research. 2018; 28: 616–624.
[47]
Cheng Y, Luo H, Izzo F, Pickering BF, Nguyen D, Myers R, et al. M6a RNA Methylation Maintains Hematopoietic Stem Cell Identity and Symmetric Commitment. Cell Reports. 2019; 28: 1703–1716.e6.
[48]
Mathoux J, Henshall DC, Brennan GP. Regulatory Mechanisms of the RNA Modification m6A and Significance in Brain Function in Health and Disease. Frontiers in Cellular Neuroscience. 2021; 15: 671932.
[49]
Zelinger L, Swaroop A. RNA Biology in Retinal Development and Disease. Trends in Genetics. 2018; 34: 341–351.
[50]
Chen Y, Wang J, Xu D, Xiang Z, Ding J, Yang X, et al. m6A mRNA methylation regulates testosterone synthesis through modulating autophagy in Leydig cells. Autophagy. 2021; 17: 457–475.
[51]
Li P, Yu H, Zhang G, Kang L, Qin B, Cao Y, et al. Identification and Characterization of N6-Methyladenosine CircRNAs and Methyltransferases in the Lens Epithelium Cells From Age-Related Cataract. Investigative Ophthalmology & Visual Science. 2020; 61: 13.
[52]
Yao M, Jiang Q, Ma Y, Liu C, Zhu C, Sun Y, et al. Role of METTL3-Dependent N6-Methyladenosine mRNA Modification in the Promotion of Angiogenesis. Molecular Therapy. 2020; 28: 2191–2202.
[53]
Zhong Q, Guo J, Chen S. Spatio-Temporal Expression Profile of Mettl3/Mettl14 During Mouse Retina Development. International Journal of Morphology. 2020; 38: 1668–1675.
[54]
Leonetti AM, Chu MY, Ramnaraign FO, Holm S, Walters BJ. An Emerging Role of m6A in Memory: A Case for Translational Priming. International Journal of Molecular Sciences. 2020; 21: 7447.
[55]
Cui Q, Shi H, Ye P, Li L, Qu Q, Sun G, et al. M 6 a RNA Methylation Regulates the Self-Renewal and Tumorigenesis of Glioblastoma Stem Cells. Cell Reports. 2017; 18: 2622–2634.
[56]
Chen M, Wei L, Law C, Tsang FH, Shen J, Cheng CL, et al. RNA N6-methyladenosine methyltransferase-like 3 promotes liver cancer progression through YTHDF2-dependent posttranscriptional silencing of SOCS2. Hepatology. 2018; 67: 2254–2270.
[57]
Chen M, Wong C. The emerging roles of N6-methyladenosine (m6A) deregulation in liver carcinogenesis. Molecular Cancer. 2020; 19: 44.
[58]
Han SH, Choe J. Diverse molecular functions of m6a mRNA modification in cancer. Experimental & Molecular Medicine. 2020; 52: 738–749.
[59]
Ho AJ, Stein JL, Hua X, Lee S, Hibar DP, Leow AD, et al. A commonly carried allele of the obesity-related FTO gene is associated with reduced brain volume in the healthy elderly. Proceedings of the National Academy of Sciences of the United States of America. 2010; 107: 8404–8409.
[60]
Dermentzaki G, Lotti F. New Insights on the Role of N6-Methyladenosine RNA Methylation in the Physiology and Pathology of the Nervous System. Frontiers in Molecular Biosciences. 2020; 7: 555372.
[61]
Han M, Liu Z, Xu Y, Liu X, Wang D, Li F, et al. Abnormality of m6A mRNA Methylation Is Involved in Alzheimer’s Disease. Frontiers in Neuroscience. 2020; 14: 98.
[62]
Shafik AM, Allen EG, Jin P. Dynamic N6-methyladenosine RNA methylation in brain and diseases. Epigenomics. 2020; 12: 371–380.
[63]
Lin X, Chai G, Wu Y, Li J, Chen F, Liu J, et al. RNA m6a methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail. Nature Communications. 2019; 10: 2065.
[64]
Wanna-udom S, Terashima M, Lyu H, Ishimura A, Takino T, Sakari M, et al. The m6a methyltransferase METTL3 contributes to Transforming Growth Factor-beta-induced epithelial-mesenchymal transition of lung cancer cells through the regulation of JUNB. Biochemical and Biophysical Research Communications. 2020; 524: 150–155.
[65]
Ma X, Long C, Wang F, Lou B, Yuan M, Duan F, et al. METTL3 attenuates proliferative vitreoretinopathy and epithelial‐mesenchymal transition of retinal pigment epithelial cells via wnt/β‐catenin pathway. Journal of Cellular and Molecular Medicine. 2021; 25: 4220–4234.
[66]
Stellos K. The rise of epitranscriptomic era: implications for cardiovascular disease. Cardiovascular Research. 2017; 113: e2–e3.
[67]
Wang X, Lu Z, Gomez A, Hon GC, Yue Y, Han D, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014; 505: 117–120.
[68]
Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AA, Kol N, Salmon-Divon M, et al. Stem cells. m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science. 2015; 347: 1002–1006.
[69]
Meyer KD, Jaffrey SR. Rethinking m6A Readers, Writers, and Erasers. Annual Review of Cell and Developmental Biology. 2017; 33: 319–342.
[70]
Zaccara S, Ries RJ, Jaffrey SR. Reading, writing and erasing mRNA methylation. Nature Reviews Molecular Cell Biology. 2019; 20: 608–624.
[71]
Wang C, Shie S, Hsieh I, Tsai M, Wen M. FTO modulates circadian rhythms and inhibits the CLOCK-BMAL1-induced transcription. Biochemical and Biophysical Research Communications. 2015; 464: 826–832.
[72]
Hu Y, Chen J, Sun J, Wang Y, Huang P, Feng J, et al. FTO alleviates Aβ1-40 induced retinal pigment epithelium degeneration via PKA/CREB signaling pathway. 2020. (in press)
[73]
Luo G, Xu W, Zhao Y, Jin S, Wang S, Liu Q, et al. RNA m6A methylation regulates uveal melanoma cell proliferation, migration, and invasion by targeting c-Met. Journal of Cellular Physiology. 2020; 235: 7107–7119.
[74]
Shan K, Zhou R, Xiang J, Sun Y, Liu C, Lv M, et al. FTO regulates ocular angiogenesis via m6a-YTHDF2-dependent mechanism. Experimental Eye Research. 2020; 197: 108107.
[75]
Zha X, Xi X, Fan X, Ma M, Zhang Y, Yang Y. Overexpression of METTL3 attenuates high-glucose induced RPE cell pyroptosis by regulating miR-25-3p/PTEN/Akt signaling cascade through DGCR8. Aging. 2020; 12: 8137–8150.
[76]
Qu X, Zhu K, Li Z, Zhang D, Hou L. The Alteration of M6A-Tagged Transcript Profiles in the Retina of Rats After Traumatic Optic Neuropathy. Frontiers in Genetics. 2021; 12: 628841.
[77]
Meyer KD, Jaffrey SR. The dynamic epitranscriptome: N6-methyladenosine and gene expression control. Nature Reviews Molecular Cell Biology. 2014; 15: 313–326.
[78]
Chang M, Lv H, Zhang W, Ma C, He X, Zhao S, et al. Region-specific RNA m6A methylation represents a new layer of control in the gene regulatory network in the mouse brain. Open Biology. 2017; 7: 170166.
[79]
Yoon K, Ringeling FR, Vissers C, Jacob F, Pokrass M, Jimenez-Cyrus D, et al. Temporal Control of Mammalian Cortical Neurogenesis by m6a Methylation. Cell. 2017; 171: 877–889.e17.
[80]
Chen J, Zhang Y, Huang C, Shen H, Sun B, Cheng X, et al. M6a Regulates Neurogenesis and Neuronal Development by Modulating Histone Methyltransferase Ezh2. Genomics, Proteomics & Bioinformatics. 2019; 17: 154–168.
[81]
Rao RC, Tchedre KT, Malik MTA, Coleman N, Fang Y, Marquez VE, et al. Dynamic Patterns of Histone Lysine Methylation in the Developing Retina. Investigative Opthalmology & Visual Science. 2010; 51: 6784.
[82]
Reese BE. Development of the retina and optic pathway. Vision Research. 2011; 51: 613–632.
[83]
Zhang M, Zhai Y, Zhang S, Dai X, Li Z. Roles of N6-Methyladenosine (m6A) in Stem Cell Fate Decisions and Early Embryonic Development in Mammals. Frontiers in Cell and Developmental Biology. 2020; 8: 782.
[84]
Yan D, Luo G, Chen X, Xu W. A Genome-wide Map of RNA m6A Modification in mouse retina. Investigative Ophthalmology & Visual Science. 2021; 62: 1572–1572.
[85]
Dorrell MI, Aguilar E, Weber C, Friedlander M. Global Gene Expression Analysis of the Developing Postnatal Mouse Retina. Investigative Opthalmology & Visual Science. 2004; 45: 1009–1019.
[86]
Huang L, Liang H, Wang S, Chen, S. m6A writer complex promotes timely differentiation and survival of retinal progenitor cells in zebrafish. Biochemical and biophysical research communications. 2021; 567: 171–176.
[87]
Wang Z, Ji S, Huang Y, Liao K, Cui Z, Chu F, et al. The daily gene transcription cycle in mouse retina. Experimental Eye Research. 2021; 207: 108565.
[88]
Qin Y, Li L, Luo E, Hou J, Yan G, Wang D, et al. Role of m6A RNA methylation in cardiovascular disease (Review). International Journal of Molecular Medicine. 2020; 46: 1958–1972.
[89]
Li H, Tong J, Zhu S, Batista PJ, Duffy EE, Zhao J, et al. M6a mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature. 2017; 548: 338–342.
[90]
Pagiatakis C, Condorelli G. The RNA Methylome Blackboard. Circulation. 2019; 139: 546–548.
[91]
Perez VL, Saeed AM, Tan Y, Urbieta M, Cruz-Guilloty F. The eye: a window to the soul of the immune system. Journal of Autoimmunity. 2013; 45: 7–14.
[92]
Perez VL, Caspi RR. Immune mechanisms in inflammatory and degenerative eye disease. Trends in Immunology. 2015; 36: 354–363.
[93]
Akhtar‐Schäfer I, Wang L, Krohne TU, Xu H, Langmann T. Modulation of three key innate immune pathways for the most common retinal degenerative diseases. EMBO Molecular Medicine. 2018; 10: e8259.
[94]
Ueta M, Kinoshita S. Innate immunity of the ocular surface. Brain Research Bulletin. 2010; 81: 219–228.
[95]
Vega JL, Keino H, Masli S. Surgical denervation of ocular sympathetic afferents decreases local transforming growth factor-beta and abolishes immune privilege. The American Journal of Pathology. 2009; 175: 1218–1225.
[96]
Heuss ND, Pierson MJ, Montaniel KRC, McPherson SW, Lehmann U, Hussong SA, et al. Retinal dendritic cell recruitment, but not function, was inhibited in MyD88 and TRIF deficient mice. Journal of Neuroinflammation. 2014; 11: 143.
[97]
Lehmann U, Heuss ND, McPherson SW, Roehrich H, Gregerson DS. Dendritic cells are early responders to retinal injury. Neurobiology of Disease. 2010; 40: 177–184.
[98]
Pflugfelder SC, De Paiva CS, Moore QL, Volpe EA, Li D, Gumus K, et al. Aqueous Tear Deficiency Increases Conjunctival Interferon-γ (IFN-γ) Expression and Goblet Cell Loss. Investigative Opthalmology & Visual Science. 2015; 56: 7545.
[99]
Conrady CD, Jones H, Zheng M, Carr DJJ. A functional type i interferon pathway drives resistance to cornea herpes simplex virus type 1 infection by recruitment of leukocytes. Journal of Biomedical Research. 2011; 25: 111–119.
[100]
Zhang Y, Liang Q, Liu Y, Pan Z, Baudouin C, Labbé A, et al. Expression of cytokines in aqueous humor from fungal keratitis patients. BMC Ophthalmology. 2018; 18: 105.
[101]
Ozek D, Yildirim F, Özay Y, Yıldırım Z. The effect of diabetes on vitreous levels of adiponectin and inflammatory cytokines in experimental rat model. Advances in Clinical and Experimental Medicine. 2020; 29: 449–452.
[102]
Hooks JJ, Nagineni CN, Hooper LC, Hayashi K, Detrick B. IFN-beta provides immuno-protection in the retina by inhibiting ICAM-1 and CXCL9 in retinal pigment epithelial cells. Journal of Immunology. 2008; 180: 3789–3796.
[103]
Li T, Zhuang Y, Yang W, Xie Y, Shang W, Su S, et al. Silencing of METTL3 attenuates cardiac fibrosis induced by myocardial infarction via inhibiting the activation of cardiac fibroblasts. The THE FASEB JOURNALournal. 2021; 35: e21162.
[104]
Zheng Q, Hou J, Zhou Y, Li Z, Cao X. The RNA helicase DDX46 inhibits innate immunity by entrapping m6a-demethylated antiviral transcripts in the nucleus. Nature Immunology. 2017; 18: 1094–1103.
[105]
Winkler R, Gillis E, Lasman L, Safra M, Geula S, Soyris C, et al. M6a modification controls the innate immune response to infection by targeting type i interferons. Nature Immunology. 2019; 20: 173–182.
[106]
Gao Y, Vasic R, Song Y, Teng R, Liu C, Gbyli R, et al. M6a Modification Prevents Formation of Endogenous Double-Stranded RNAs and Deleterious Innate Immune Responses during Hematopoietic Development. Immunity. 2020; 52: 1007–1021.e8.
[107]
Petrillo F, Pignataro D, Lavano MA, Santella B, Folliero V, Zannella C, et al. Current evidence on the ocular surface microbiota and related diseases. Microorganisms. 2020; 8: 1033.
[108]
Wu C, Chen W, He J, Jin S, Liu Y, Yi Y, et al. Interplay of m6A and H3K27 trimethylation restrains inflammation during bacterial infection. Science Advances. 2020; 6: eaba0647.
[109]
Jabs S, Biton A, Bécavin C, Nahori M, Ghozlane A, Pagliuso A, et al. Impact of the gut microbiota on the m6a epitranscriptome of mouse cecum and liver. Nature Communications. 2020; 11: 1344.
[110]
Yang C, Hu Y, Zhou B, Bao Y, Li Z, Gong C, et al. The role of m6a modification in physiology and disease. Cell Death & Disease. 2020; 11: 960.
[111]
Brocard M, Ruggieri A, Locker N. M6a RNA methylation, a new hallmark in virus-host interactions. Journal of General Virology. 2017; 98: 2207–2214.
[112]
Gokhale NS, McIntyre ABR, Mattocks MD, Holley CL, Lazear HM, Mason CE, et al. Altered m6a Modification of Specific Cellular Transcripts Affects Flaviviridae Infection. Molecular Cell. 2020; 77: 542–555.e8.
[113]
Bloom DC, Giordani NV, Kwiatkowski DL. Epigenetic regulation of latent HSV-1 gene expression. Biochimica Et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 2010; 1799: 246–256.
[114]
Liu J, Xu Y, Li K, Ye Q, Zhou H, Sun H, et al. The m6a methylome of SARS-CoV-2 in host cells. Cell Research. 2021; 31: 404–414.
[115]
Troisi M, Zannella C, Troisi S, De Bernardo M, Galdiero M, Franci G, et al. Ocular Surface Infection by SARS-CoV-2 in COVID-19 Pneumonia Patients Admitted to Sub-Intensive Unit: Preliminary Results. Microorganisms. 2022; 10: 347.
[116]
Kaufman HL, Maciorowski D. Advancing oncolytic virus therapy by understanding the biology. Nature Reviews Clinical Oncology. 2021; 18: 197–198.
[117]
Chianese A, Santella B, Ambrosino A, Stelitano D, Rinaldi L, Galdiero M, et al. Oncolytic viruses in combination therapeutic approaches with epigenetic modulators: Past, present, and future perspectives. Cancers. 2021; 13: 2761.
[118]
Todd L, Palazzo I, Suarez L, Liu X, Volkov L, Hoang TV, et al. Reactive microglia and IL1β/IL-1R1-signaling mediate neuroprotection in excitotoxin-damaged mouse retina. Journal of Neuroinflammation. 2019; 16: 118.
[119]
Mesquida M, Leszczynska A, Llorenç V, Adán A. Interleukin-6 blockade in ocular inflammatory diseases. Clinical and Experimental Immunology. 2014; 176: 301–309.
[120]
Shin JI, Bayry J. A role for IL-17 in age-related macular degeneration. Nature Reviews Immunology. 2013; 13: 701–701.
[121]
Doyle SL, Campbell M, Ozaki E, Salomon RG, Mori A, Kenna PF, et al. NLRP3 has a protective role in age-related macular degeneration through the induction of IL-18 by drusen components. Nature Medicine. 2012; 18: 791–798.
[122]
Hirano Y, Yasuma T, Mizutani T, Fowler BJ, Tarallo V, Yasuma R, et al. IL-18 is not therapeutic for neovascular age-related macular degeneration. Nature Medicine. 2014; 20: 1372–1375.
[123]
Xu H, Song Z, Fu S, Zhu M, Le Y. RPE barrier breakdown in diabetic retinopathy: seeing is believing. Journal of Ocular Biology, Diseases, and Informatics. 2011; 4: 83–92.
[124]
Wei L, Liu B, Tuo J, Shen D, Chen P, Li Z, et al. Hypomethylation of the IL17RC Promoter Associates with Age-Related Macular Degeneration. Cell Reports. 2012; 2: 1151–1158.
[125]
Jin M, He S, Wörpel V, Ryan SJ, Hinton DR. Promotion of adhesion and migration of RPE cells to provisional extracellular matrices by TNF-alpha. Investigative Ophthalmology & Visual Science. 2000; 41: 4324–4332.
[126]
Limb GA, Hollifield RD, Webster L, Charteris DG, Chignell AH. Soluble TNF receptors in vitreoretinal proliferative disease. Investigative Ophthalmology & Visual Science. 2001; 42: 1586–1591.
[127]
Derevjanik NL, Vinores SA, Xiao WH, Mori K, Turon T, Hudish T, et al. Quantitative assessment of the integrity of the blood-retinal barrier in mice. Investigative Ophthalmology & Visual Science. 2002; 43: 2462–2467.
[128]
Prada J. Tumour necrosis factor alpha and interleukin 6 gene expression in keratocytes from patients with rheumatoid corneal ulcerations. British Journal of Ophthalmology. 2003; 87: 548–550.
[129]
Grant MB, Afzal A, Spoerri P, Pan H, Shaw LC, Mames RN. The role of growth factors in the pathogenesis of diabetic retinopathy. Expert Opinion on Investigational Drugs. 2004; 13: 1275–1293.
[130]
Theodossiadis PG, Markomichelakis NN, Sfikakis PP. Tumor Necrosis Factor Antagonists. Retina. 2007; 27: 399–413.
[131]
Berger S, Savitz SI, Nijhawan S, Singh M, David J, Rosenbaum PS, et al. Deleterious role of TNF-alpha in retinal ischemia-reperfusion injury. Investigative Ophthalmology & Visual Science. 2008; 49: 3605–3610.
[132]
Rodrigues EB, Farah ME, Maia M, Penha FM, Regatieri C, Melo GB, et al. Therapeutic monoclonal antibodies in ophthalmology. Progress in Retinal and Eye Research. 2009; 28: 117–144.
[133]
Mirshahi A, Hoehn R, Lorenz K, Kramann C, Baatz H. Anti-tumor necrosis factor alpha for retinal diseases: current knowledge and future concepts. Journal of Ophthalmic and Vision Research. 2012; 7: 39–44.
[134]
Nicolela Susanna F, Pavesio C. A review of ocular adverse events of biological anti-TNF drugs. Journal of Ophthalmic Inflammation and Infection. 2020; 10: 11.
[135]
Feng Z, Li Q, Meng R, Yi B, Xu Q. METTL3 regulates alternative splicing of MyD88 upon the lipopolysaccharide-induced inflammatory response in human dental pulp cells. Journal of Cellular and Molecular Medicine. 2018; 22: 2558–2568.
[136]
Zhang C, Wang Y, Peng Y, Xu H, Zhou X. METTL3 regulates inflammatory pain by modulating m6A-dependent pri-miR-365-3p processing. The THE FASEB JOURNALournal. 2020; 34: 122–132.
[137]
Wang J, Yan S, Lu H, Wang S, Xu D. METTL3 Attenuates LPS-Induced Inflammatory Response in Macrophages via NF-κB Signaling Pathway. Mediators of Inflammation. 2019; 2019: 3120391.
[138]
Yu J, Hu X, Chen H, Yang Q, Li H, Dong Y, et al. DNA methylation of FTO promotes renal inflammation by enhancing m6a of PPAR-α in alcohol-induced kidney injury. Pharmacological Research. 2021; 163: 105286.
[139]
Lu TX, Zheng Z, Zhang L, Sun H, Bissonnette M, Huang H, et al. A New Model of Spontaneous Colitis in Mice Induced by Deletion of an RNA m6a Methyltransferase Component METTL14 in T Cells. Cellular and Molecular Gastroenterology and Hepatology. 2020; 10: 747–761.
[140]
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death & Differentiation. 2018; 25: 486–541.
[141]
Yang J, Liu J, Zhao S, Tian F. N6-Methyladenosine METTL3 Modulates the Proliferation and Apoptosis of Lens Epithelial Cells in Diabetic Cataract. Molecular Therapy - Nucleic Acids. 2020; 20: 111–116.
[142]
Penke B, Szűcs M, Bogár F. Oligomerization and Conformational Change Turn Monomeric β-Amyloid and Tau Proteins Toxic: Their Role in Alzheimer’s Pathogenesis. Molecules. 2020; 25: 1659.
[143]
Lashkari K, Teague GC, Beattie U, Betts J, Kumar S, McLaughlin MM, et al. Plasma biomarkers of the amyloid pathway are associated with geographic atrophy secondary to age-related macular degeneration. PLoS ONE. 2020; 15: e0236283.
[144]
Tseng WA, Thein T, Kinnunen K, Lashkari K, Gregory MS, D’Amore PA, et al. NLRP3 Inflammasome Activation in Retinal Pigment Epithelial Cells by Lysosomal Destabilization: Implications for Age-Related Macular Degeneration. Investigative Opthalmology & Visual Science. 2013; 54: 110–120.
[145]
Wooff Y, Man SM, Aggio-Bruce R, Natoli R, Fernando N. IL-1 Family Members Mediate Cell Death, Inflammation and Angiogenesis in Retinal Degenerative Diseases. Frontiers in Immunology. 2019; 10: 1618.
[146]
Xiao L, Zhao Q, Hu B, Wang J, Liu C, Xu H. METTL3 promotes IL‐1β–induced degeneration of endplate chondrocytes by driving m6a‐dependent maturation of miR‐126‐5p. Journal of Cellular and Molecular Medicine. 2020; 24: 14013–14025.
[147]
Osborne NN. Mitochondria: their role in ganglion cell death and survival in primary open angle glaucoma. Experimental Eye Research. 2010; 90: 750–757.
[148]
Kruk J, Kubasik-Kladna K, Aboul-Enein HY. The Role Oxidative Stress in the Pathogenesis of Eye Diseases: Current Status and a Dual Role of Physical Activity. Mini-Reviews in Medicinal Chemistry. 2015; 16: 241–257.
[149]
Osborne NN, Núñez-Álvarez C, Joglar B, del Olmo-Aguado S. Glaucoma: Focus on mitochondria in relation to pathogenesis and neuroprotection. European Journal of Pharmacology. 2016; 787: 127–133.
[150]
Calderon GD, Juarez OH, Hernandez GE, Punzo SM, De la Cruz ZD. Oxidative stress and diabetic retinopathy: development and treatment. Eye. 2017; 31: 1122–1130.
[151]
Masuda T, Shimazawa M, Hara H. Retinal Diseases Associated with Oxidative Stress and the Effects of a Free Radical Scavenger (Edaravone). Oxidative Medicine and Cellular Longevity. 2017; 2017: 9208489.
[152]
Ruan Y, Jiang S, Gericke A. Age-Related Macular Degeneration: Role of Oxidative Stress and Blood Vessels. International Journal of Molecular Sciences. 2021; 22: 1296.
[153]
Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, et al. Oxidative Stress: Harms and Benefits for Human Health. Oxidative Medicine and Cellular Longevity. 2017; 2017: 8416763.
[154]
Sharifi-Rad M, Anil Kumar NV, Zucca P, Varoni EM, Dini L, Panzarini E, et al. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Frontiers in Physiology. 2020; 11: 694.
[155]
Chen A, Huang X, Xue Z, Cao D, Huang K, Chen J, et al. The Role of p21 in Apoptosis, Proliferation, Cell Cycle Arrest, and Antioxidant Activity in UVB-Irradiated Human HaCaT Keratinocytes. Medical Science Monitor Basic Research. 2015; 21: 86–95.
[156]
Azizi B, Ziaei A, Fuchsluger T, Schmedt T, Chen Y, Jurkunas UV. P53-Regulated Increase in Oxidative-Stress–Induced Apoptosis in Fuchs Endothelial Corneal Dystrophy: a Native Tissue Model. Investigative Opthalmology & Visual Science. 2011; 52: 9291–9297.
[157]
Núñez-Álvarez C, Osborne NN. Enhancement of corneal epithelium cell survival, proliferation and migration by red light: Relevance to corneal wound healing. Experimental Eye Research. 2019; 180: 231–241.
[158]
Ma T, Lan D, He S, Ye Z, Li P, Zhai W, et al. Nrf2 protects human lens epithelial cells against H 2 O 2 -induced oxidative and ER stress: the ATF4 may be involved. Experimental Eye Research. 2018; 169: 28–37.
[159]
Datta S, Cano M, Ebrahimi K, Wang L, Handa JT. The impact of oxidative stress and inflammation on RPE degeneration in non-neovascular AMD. Progress in Retinal and Eye Research. 2017; 60: 201–218.
[160]
Osborne NN, Núñez-Álvarez C, del Olmo-Aguado S. The effect of visual blue light on mitochondrial function associated with retinal ganglions cells. Experimental Eye Research. 2014; 128: 8–14.
[161]
Brown EE, DeWeerd AJ, Ildefonso CJ, Lewin AS, Ash JD. Mitochondrial oxidative stress in the retinal pigment epithelium (RPE) led to metabolic dysfunction in both the RPE and retinal photoreceptors. Redox Biology. 2019; 24: 101201.
[162]
Overbey EG, da Silveira WA, Stanbouly S, Nishiyama NC, Roque-Torres GD, Pecaut MJ, et al. Spaceflight influences gene expression, photoreceptor integrity, and oxidative stress-related damage in the murine retina. Scientific Reports. 2019; 9: 13304.
[163]
Li Q, Li X, Tang H, Jiang B, Dou Y, Gorospe M, et al. NSUN2-Mediated m5C Methylation and METTL3/METTL14-Mediated m6a Methylation Cooperatively Enhance p21 Translation. Journal of Cellular Biochemistry. 2017; 118: 2587–2598.
[164]
Yao W, Han X, Ge M, Chen C, Xiao X, Li H, et al. N6-methyladenosine (m6A) methylation in ischemia–reperfusion injury. Cell Death & Disease. 2020; 11: 478.
[165]
He S, Yaung J, Kim YH, Barron E, Ryan SJ, Hinton DR. Endoplasmic reticulum stress induced by oxidative stress in retinal pigment epithelial cells. Graefe’S Archive for Clinical and Experimental Ophthalmology. 2008; 246: 677–683.
[166]
Dou G, Sreekumar PG, Spee C, He S, Ryan SJ, Kannan R, et al. Deficiency of αB crystallin augments ER stress-induced apoptosis by enhancing mitochondrial dysfunction. Free Radical Biology and Medicine. 2012; 53: 1111–1122.
[167]
Hoh Kam J, Lenassi E, Jeffery G. Viewing ageing eyes: diverse sites of amyloid Beta accumulation in the ageing mouse retina and the up-regulation of macrophages. PLoS ONE. 2010; 5: e13127.
[168]
Dong ZZ, Li J, Gan YF, Sun XR, Leng YX, Ge J. Amyloid beta deposition related retinal pigment epithelium cell impairment and subretinal microglia activation in aged APPswePS1 transgenic mice. International Journal of Ophthalmology. 2018; 11: 747–755.
[169]
Fiorentino M, Grandaliano G, Gesualdo L, Castellano G. Acute Kidney Injury to Chronic Kidney Disease Transition. Contributions to Nephrology. 2018; 193: 45–54.
[170]
Su Y, Xu R, Zhang R, Qu Y, Zuo W, Ji Z, et al. N6-methyladenosine methyltransferase plays a role in hypoxic preconditioning partially through the interaction with lncRNA H19. Acta Biochimica Et Biophysica Sinica. 2020; 52: 1306–1315.
[171]
Yang B, Chen Q. Cross-Talk between Oxidative Stress and m6a RNA Methylation in Cancer. Oxidative Medicine and Cellular Longevity. 2021; 2021: 6545728.
[172]
Xu Y, Yuan XD, Wu JJ, Chen RY, Xia L, Zhang M, et al. The N6‐methyladenosine mRNA methylase METTL14 promotes renal ischemic reperfusion injury via suppressing YAP1. Journal of Cellular Biochemistry. 2020; 121: 524–533.
[173]
Bravard A, Lefai E, Meugnier E, Pesenti S, Disse E, Vouillarmet J, et al. FTO is Increased in Muscle during Type 2 Diabetes, and its Overexpression in Myotubes Alters Insulin Signaling, Enhances Lipogenesis and ROS Production, and Induces Mitochondrial Dysfunction. Diabetes. 2011; 60: 258–268.
[174]
Wilkinson E, Cui YH, He YY. Context-Dependent Roles of RNA Modifications in Stress Responses and Diseases. International Journal of Molecular Sciences. 2021; 22: 1949.
[175]
Shen W, Li H, Su H, Chen K, Yan J. FTO overexpression inhibits apoptosis of hypoxia/reoxygenation-treated myocardial cells by regulating m6a modification of Mhrt. Molecular and Cellular Biochemistry. 2021; 476: 2171–2179.
[176]
Huisman B, Manske G, Carney S, Kalantry S. Functional Dissection of the m6a RNA Modification. Trends in Biochemical Sciences. 2017; 42: 85–86.
[177]
Xu S, Li Y, Chen J, Li D, Jiang Q, Wu T, et al. Oxygen glucose deprivation/re-oxygenation-induced neuronal cell death is associated with Lnc-D63785 m6a methylation and miR-422a accumulation. Cell Death & Disease. 2020; 11: 816.
[178]
Abu-Amero KK. Resveratrol and Ophthalmic Diseases. Nutrients. 2016; 8: 200–210.
[179]
Cao K, Ishida T, Fang Y, Shinohara K, Li X, Nagaoka N, et al. Protection of the Retinal Ganglion Cells: Intravitreal Injection of Resveratrol in Mouse Model of Ocular Hypertension. Investigative Opthalmology & Visual Science. 2020; 61: 13.
[180]
Wu J, Gan Z, Zhuo R, Zhang L, Wang T, Zhong X. Resveratrol Attenuates Aflatoxin B_1-Induced ROS Formation and Increase of m6A RNA Methylation. Animals. 2020; 10: 677.
[181]
Sena L, Chandel N. Physiological Roles of Mitochondrial Reactive Oxygen Species. Molecular Cell. 2012; 48: 158–167.
[182]
Finkel T. Oxygen radicals and signaling. Current Opinion in Cell Biology. 1998; 10: 248–253.
[183]
Rhee SG, Bae YS, Lee S, Kwon J. Hydrogen Peroxide: a Key Messenger that Modulates Protein Phosphorylation through Cysteine Oxidation. Science’s STKE. 2000; 2000: pe1.
[184]
Watson J. Oxidants, antioxidants and the current incurability of metastatic cancers. Open Biology. 2013; 3: 120144.
[185]
Holmström KM, Finkel T. Cellular mechanisms and physiological consequences of redox-dependent signalling. Nature Reviews Molecular Cell Biology. 2014; 15: 411–421.
[186]
Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nature Reviews Molecular Cell Biology. 2020; 21: 363–383.
[187]
Tan KB, Harrop J, Reddy M, Young P, Terrett J, Emery J, et al. Characterization of a novel TNF-like ligand and recently described TNF ligand and TNF receptor superfamily genes and their constitutive and inducible expression in hematopoietic and non-hematopoietic cells. Gene. 1997; 204: 35–46.
[188]
Zhai Y, Ni J, Jiang GW, Lu J, Xing L, Lincoln C, et al. VEGI, a novel cytokine of the tumor necrosis factor family, is an angiogenesis inhibitor that suppresses the growth of colon carcinomas in vivo. The FASEB Journal. 1999; 13: 181–189.
[189]
Gao W, Zhao Z, Yu G, Zhou Z, Zhou Y, Hu T, et al. VEGI attenuates the inflammatory injury and disruption of blood–brain barrier partly by suppressing the TLR4/NF-κB signaling pathway in experimental traumatic brain injury. Brain Research. 2015; 1622: 230–239.
[190]
Metheny-Barlow LJ, Li L. Vascular Endothelial Growth Inhibitor (VEGI), an Endogenous Negative Regulator of Angiogenesis. Seminars in Ophthalmology. 2006; 21: 49–58.
[191]
Jiang F, Chen Q, Huang L, Wang Y, Zhang Z, Meng X, et al. TNFSF15 Inhibits Blood Retinal Barrier Breakdown Induced by Diabetes. International Journal of Molecular Sciences. 2016; 17: 615.
[192]
Fry NJ, Law BA, Ilkayeva OR, Holley CL, Mansfield KD. N6-methyladenosine is required for the hypoxic stabilization of specific mRNAs. RNA. 2017; 23: 1444–1455.
[193]
Wang L, Xue Y, Li H, Huo R, Yan Z, Wang J, et al. Wilms’ tumour 1‐associating protein inhibits endothelial cell angiogenesis by m6a‐dependent epigenetic silencing of desmoplakin in brain arteriovenous malformation. Journal of Cellular and Molecular Medicine. 2020; 24: 4981–4991.
[194]
Yang Z, Wang T, Wu D, Min Z, Tan J, Yu B. RNA N6-methyladenosine reader IGF2BP3 regulates cell cycle and angiogenesis in colon cancer. Journal of Experimental & Clinical Cancer Research. 2020; 39: 203.
[195]
Rizzolo LJ, Peng S, Luo Y, Xiao W. Integration of tight junctions and claudins with the barrier functions of the retinal pigment epithelium. Progress in Retinal and Eye Research. 2011; 30: 296–323.
[196]
Xia T, Rizzolo LJ. Effects of diabetic retinopathy on the barrier functions of the retinal pigment epithelium. Vision Research. 2017; 139: 72–81.
[197]
Xiao Q, Zhao Y, Xu J, Li W, Chen Y, Sun H. NFE2/miR-423-5p/TFF1 axis regulates high glucose-induced apoptosis in retinal pigment epithelial cells. BMC Molecular and Cell Biology. 2019; 20: 39.
[198]
Qaum T, Xu Q, Joussen AM, Clemens MW, Qin W, Miyamoto K, et al. VEGF-initiated blood-retinal barrier breakdown in early diabetes. Investigative Ophthalmology & Visual Science. 2001; 42: 2408–2413.
[199]
Lin J, Zhu Q, Huang J, Cai R, Kuang Y. Hypoxia Promotes Vascular Smooth Muscle Cell (VSMC) Differentiation of Adipose-Derived Stem Cell (ADSC) by Regulating Mettl3 and Paracrine Factors. Stem Cells International. 2020; 2020: 2830565.
[200]
Wang G, Dai Y, Li K, Cheng M, Xiong G, Wang X, et al. Deficiency of Mettl3 in Bladder Cancer Stem Cells Inhibits Bladder Cancer Progression and Angiogenesis. Frontiers in Cell and Developmental Biology. 2021; 9: 627706.
[201]
Paques M, Massin P, Gaudric A. Growth factors and diabetic retinopathy. Diabetes & Metabolism. 1997; 23: 125–130.
[202]
Nagineni CN, Samuel W, Nagineni S, Pardhasaradhi K, Wiggert B, Detrick B, et al. Transforming growth factor-beta induces expression of vascular endothelial growth factor in human retinal pigment epithelial cells: involvement of mitogen-activated protein kinases. Journal of Cellular Physiology. 2003; 197: 453–462.
[203]
Rubio RG, Adamis AP. Ocular Angiogenesis: Vascular Endothelial Growth Factor and Other Factors. Developments in Ophthalmology. 2016; 55: 28–37.
[204]
Selvam S, Kumar T, Fruttiger M. Retinal vasculature development in health and disease. Progress in Retinal and Eye Research. 2018; 63: 1–19.
[205]
Tian C, Huang Y, Li Q, Feng Z, Xu Q. Mettl3 Regulates Osteogenic Differentiation and Alternative Splicing of Vegfa in Bone Marrow Mesenchymal Stem Cells. International Journal of Molecular Sciences. 2019; 20: 551.
[206]
Zhang BY, Han L, Tang YF, Zhang GX, Fan XL, Zhang JJ, et al. METTL14 regulates M6A methylation-modified primary miR-19a to promote cardiovascular endothelial cell proliferation and invasion. European Review for Medical and Pharmacological Sciences. 2020; 24: 7015–7023.
[207]
Liu T, Zhou H, Lu H, Luo C, Wang Q, Peng Y, et al. MiR-4729 regulates TIE1 mRNA m6a modification and angiogenesis in hemorrhoids by targeting METTL14. Annals of Translational Medicine. 2021; 9: 232–232.
[208]
Xiao Y, Thakkar KN, Zhao H, Broughton J, Li Y, Seoane JA, et al. The m6A RNA demethylase FTO is a HIF-independent synthetic lethal partner with the VHL tumor suppressor. Proceedings of the National Academy of Sciences of the United States of America. 2020; 117: 21441–21449.
[209]
Barbieri I, Kouzarides T. Role of RNA modifications in cancer. Nature Reviews Cancer. 2020; 20: 303–322.
[210]
Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. Journal of Clinical Investigation. 2009; 119: 1420–1428.
[211]
Friedlander M. Fibrosis and diseases of the eye. Journal of Clinical Investigation. 2007; 117: 576–586.
[212]
Abu El-Asrar AM, De Hertogh G, van den Eynde K, Alam K, Van Raemdonck K, Opdenakker G, et al. Myofibroblasts in proliferative diabetic retinopathy can originate from infiltrating fibrocytes and through endothelial-to-mesenchymal transition (EndoMT) Experimental Eye Research. 2015; 132: 179–189.
[213]
Shu DY, Butcher E, Saint-Geniez M. EMT and EndMT: Emerging Roles in Age-Related Macular Degeneration. International Journal of Molecular Sciences. 2020; 21: 4271.
[214]
Sun J, Chang T, Li M, Sun L, Yan X, Yang Z, et al. SNAI1, an endothelial–mesenchymal transition transcription factor, promotes the early phase of ocular neovascularization. Angiogenesis. 2018; 21: 635–652.
[215]
Medici D, Kalluri R. Endothelial–mesenchymal transition and its contribution to the emergence of stem cell phenotype. Seminars in Cancer Biology. 2012; 22: 379–384.
[216]
Chen T, You Y, Jiang H, Wang ZZ. Epithelial-mesenchymal transition (EMT): a biological process in the development, stem cell differentiation, and tumorigenesis. Journal of Cellular Physiology. 2017; 232: 3261–3272.
[217]
Zhou M, Geathers JS, Grillo SL, Weber SR, Wang W, Zhao Y, et al. Role of Epithelial-Mesenchymal Transition in Retinal Pigment Epithelium Dysfunction. Frontiers in Cell and Developmental Biology. 2020; 8: 501.
[218]
Li J, Chen F, Peng Y, Lv Z, Lin X, Chen Z, et al. N6-Methyladenosine Regulates the Expression and Secretion of TGFβ1 to Affect the Epithelial-Mesenchymal Transition of Cancer Cells. Cells. 2020; 9: 296.
[219]
Liu Y, Gu C, Li X, Wang T, Yu L. Involvement of METTL3/m6Adenosine and TGFβ/Smad3 signaling on Tenon’s fibroblasts and in a rabbit model of glaucoma surgery. Journal of Molecular Histology. 2021; 52: 1129–1144.
[220]
Lin W, Xu G. Autophagy: a Role in the Apoptosis, Survival, Inflammation, and Development of the Retina. Ophthalmic Research. 2019; 61: 65–72.
[221]
Bertero A, Brown S, Madrigal P, Osnato A, Ortmann D, Yiangou L, et al. The SMAD2/3 interactome reveals that TGFβ controls m6a mRNA methylation in pluripotency. Nature. 2018; 555: 256–259.
[222]
Liu X, Xiao M, Zhang L, Li L, Zhu G, Shen E, et al. The m6a methyltransferase METTL14 inhibits the proliferation, migration, and invasion of gastric cancer by regulating the PI3K/AKT/mTOR signaling pathway. Journal of Clinical Laboratory Analysis. 2021; 35: e23655.
[223]
Xu Z, Jia K, Wang H, Gao F, Zhao S, Li F, et al. METTL14-regulated PI3K/Akt signaling pathway via PTEN affects HDAC5-mediated epithelial–mesenchymal transition of renal tubular cells in diabetic kidney disease. Cell Death & Disease. 2021; 12: 32.
[224]
Wang C, Shie S, Tsai M, Yang C, Hung K, Wang C, et al. FTO modulates fibrogenic responses in obstructive nephropathy. Scientific Reports. 2016; 6: 18874.
[225]
Dai S, Zhang J, Huang S, Lou B, Fang B, Ye T, et al. HNRNPA2B1 regulates the epithelial–mesenchymal transition in pancreatic cancer cells through the ERK/snail signalling pathway. Cancer Cell International. 2017; 17: 12.
[226]
Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011; 479: 232–236.
[227]
Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature. 2016; 530: 184–189.
[228]
Abib FC, Barreto J. Behavior of corneal endothelial density over a lifetime. Journal of Cataract and Refractive Surgery. 2001; 27: 1574–1578.
[229]
Caprioli J. Glaucoma: a Disease of Early Cellular Senescence. Investigative Opthalmology & Visual Science. 2013; 54: ORSF60-7.
[230]
Fu Q, Qin Z, Yu J, Yu Y, Tang Q, Lyu D, et al. Effects of senescent lens epithelial cells on the severity of age-related cortical cataract in humans. Medicine. 2016; 95: e3869.
[231]
Xu J, Li D, Zheng T, Lu Y. β-amyloid expression in age-related cataract lens epithelia and the effect of β-amyloid on oxidative damage in human lens epithelial cells. Molecular Vision. 2017; 23: 1015–1028.
[232]
Blasiak J. Senescence in the pathogenesis of age-related macular degeneration. Cellular and Molecular Life Sciences. 2020; 77: 789–805.
[233]
Sreekumar PG, Hinton DR, Kannan R. The Emerging Role of Senescence in Ocular Disease. Oxidative Medicine and Cellular Longevity. 2020; 2020: 2583601.
[234]
Lee KS, Lin S, Copland DA, Dick AD, Liu J. Cellular senescence in the aging retina and developments of senotherapies for age-related macular degeneration. Journal of Neuroinflammation. 2021; 18: 32.
[235]
Cascella R, Strafella C, Caputo V, Errichiello V, Zampatti S, Milano F, et al. Towards the application of precision medicine in Age-Related Macular Degeneration. Progress in Retinal and Eye Research. 2018; 63: 132–146.
[236]
Wu Z, Shi Y, Lu M, Song M, Yu Z, Wang J, et al. METTL3 counteracts premature aging via m6a-dependent stabilization of MIS12 mRNA. Nucleic Acids Research. 2020; 48: 11083–11096.
[237]
Xu Y, Li Y, Ma L, Xin G, Wei Z, Zeng Z, et al. D-galactose induces premature senescence of lens epithelial cells by disturbing autophagy flux and mitochondrial functions. Toxicology Letters. 2018; 289: 99–106.
[238]
Wang Y, Zhang J, Wu J, Guan H. Expression of DNA repair genes in lens cortex of age-related cortical cataract. Experimental and Molecular Pathology. 2017; 102: 219–223.
[239]
Kim C, Kang D, Lee EK, Lee J. Long Noncoding RNAs and RNA-Binding Proteins in Oxidative Stress, Cellular Senescence, and Age-Related Diseases. Oxidative Medicine and Cellular Longevity. 2017; 2017: 2062384.
[240]
Wang J, Ohno-Matsui K, Morita I. Elevated amyloid β production in senescent retinal pigment epithelium, a possible mechanism of subretinal deposition of amyloid β in age-related macular degeneration. Biochemical and Biophysical Research Communications. 2012; 423: 73–78.
[241]
Contrepois K, Coudereau C, Benayoun BA, Schuler N, Roux P, Bischof O, et al. Histone variant H2a.J accumulates in senescent cells and promotes inflammatory gene expression. Nature Communications. 2017; 8: 14995.
[242]
Helman A, Klochendler A, Azazmeh N, Gabai Y, Horwitz E, Anzi S, et al. P16Ink4a-induced senescence of pancreatic beta cells enhances insulin secretion. Nature Medicine. 2016; 22: 412–420.
[243]
Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proceedings of the National Academy of Sciences. 1995; 92: 9363–9367.
[244]
Narita M, Nuñez S, Heard E, Narita M, Lin AW, Hearn SA, et al. Rb-Mediated Heterochromatin Formation and Silencing of E2F Target Genes during Cellular Senescence. Cell. 2003; 113: 703–716.
[245]
López-Luppo M, Catita J, Ramos D, Navarro M, Carretero A, Mendes-Jorge L, et al. Cellular Senescence is Associated with Human Retinal Microaneurysm Formation during Aging. Investigative Opthalmology & Visual Science. 2017; 58: 2832–2842.
[246]
Liu Y, Sanoff HK, Cho H, Burd CE, Torrice C, Ibrahim JG, et al. Expression of p16(INK4a) in peripheral blood T-cells is a biomarker of human aging. Aging Cell. 2009; 8: 439–448.
[247]
Min KW, Zealy RW, Davila S, Fomin M, Cummings JC, Makowsky D, et al. Profiling of m6A RNA modifications identified an age-associated regulation of AGO2 mRNA stability. Aging Cell. 2018; 17: e12753.
[248]
Zhu R, Ji X, Wu X, Chen J, Li X, Jiang H, et al. Melatonin antagonizes ovarian aging via YTHDF2-MAPK-NF-κB pathway. Genes & Diseases. 2020; 9: 494–509.
[249]
van Deursen JM. The role of senescent cells in ageing. Nature. 2014; 509: 439–446.
[250]
Geerlings SE, Hoepelman AIM. Immune dysfunction in patients with diabetes mellitus (DM). FEMS Immunology & Medical Microbiology. 1999; 26: 259–265.
[251]
Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 Diabetes and its Impact on the Immune System. Current Diabetes Reviews. 2020; 16: 442–449.
[252]
Akpek EK, Bunya VY, Saldanha IJ. Sjögren’s Syndrome: more than Just Dry Eye. Cornea. 2019; 38: 658–661.
[253]
Nemet AY, Vinker S, Bahar I, Kaiserman I. The Association of Keratoconus with Immune Disorders. Cornea. 2010; 29: 1261–1264.
[254]
Nerrant E, Tilikete C. Ocular Motor Manifestations of Multiple Sclerosis. Journal of Neuro-Ophthalmology. 2017; 37: 332–340.
[255]
Bartalena L. Diagnosis and management of Graves disease: a global overview. Nature Reviews Endocrinology. 2013; 9: 724–734.
[256]
O’Keefe GAD, Rao NA. Vogt-Koyanagi-Harada disease. Survey of Ophthalmology. 2017; 62: 1–25.
[257]
Grange L, Dalal M, Nussenblatt RB, Sen HN. Autoimmune Retinopathy. American Journal of Ophthalmology. 2014; 157: 266–272.e1.
[258]
Bahn RS. Graves’ Ophthalmopathy. New England Journal of Medicine. 2010; 362: 726–738.
[259]
Zhang C, Zhang M, Ge S, Huang W, Lin X, Gao J, et al. Reduced m6a modification predicts malignant phenotypes and augmented Wnt/PI3K‐Akt signaling in gastric cancer. Cancer Medicine. 2019; 8: 4766–4781.
[260]
Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, et al. Anti-tumour immunity controlled through mRNA m6a methylation and YTHDF1 in dendritic cells. Nature. 2019; 566: 270–274.
[261]
Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nature Reviews Immunology. 2007; 7: 454–465.
[262]
Palmer DC, Restifo NP. Suppressors of cytokine signaling (SOCS) in T cell differentiation, maturation, and function. Trends in Immunology. 2009; 30: 592–602.
[263]
Tong J, Cao G, Zhang T, Sefik E, Amezcua Vesely MC, Broughton JP, et al. M6a mRNA methylation sustains Treg suppressive functions. Cell Research. 2018; 28: 253–256.
[264]
Paramasivam A, Priyadharsini JV, Raghunandhakumar S. Implications of m6a modification in autoimmune disorders. Cellular & Molecular Immunology. 2020; 17: 550–551.
[265]
Wang L, Hui H, Agrawal K, Kang Y, Li N, Tang R, et al. m6 A RNA methyltransferases METTL3/14 regulate immune responses to anti-PD-1 therapy. The EMBO Journal. 2020; 39: e104514.
[266]
He X, Tan L, Ni J, Shen G. Expression pattern of m6A regulators is significantly correlated with malignancy and antitumor immune response of breast cancer. Cancer Gene Therapy. 2021; 28: 188–196.
[267]
Yagi M, Yamanaka S, Yamada Y. Epigenetic foundations of pluripotent stem cells that recapitulate in vivo pluripotency. Laboratory Investigation. 2017; 97: 1133–1141.
[268]
Mahato B, Kaya KD, Fan Y, Sumien N, Shetty RA, Zhang W, et al. Pharmacologic fibroblast reprogramming into photoreceptors restores vision. Nature. 2020; 581: 83–88.
[269]
Yu W, Mookherjee S, Chaitankar V, Hiriyanna S, Kim J, Brooks M, et al. Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal degeneration in mice. Nature Communications. 2017; 8: 14716.
[270]
Chen T, Hao Y, Zhang Y, Li M, Wang M, Han W, et al. M6a RNA Methylation is Regulated by MicroRNAs and Promotes Reprogramming to Pluripotency. Cell Stem Cell. 2015; 16: 289–301.
[271]
Dhamodaran K, Subramani M, Ponnalagu M, Shetty R, Das D. Ocular stem cells: a status update! Stem Cell Research & Therapy. 2014; 5: 56.
[272]
Stern JH, Tian Y, Funderburgh J, Pellegrini G, Zhang K, Goldberg JL, et al. Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell. 2018; 22: 834–849.
[273]
Wang Y, Li Y, Toth JI, Petroski MD, Zhang Z, Zhao JC. N6-methyladenosine modification destabilizes developmental regulators in embryonic stem cells. Nature Cell Biology. 2014; 16: 191–198.
[274]
Wen J, Lv R, Ma H, Shen H, He C, Wang J, et al. Zc3h13 Regulates Nuclear RNA m6a Methylation and Mouse Embryonic Stem Cell Self-Renewal. Molecular Cell. 2018; 69: 1028–1038.e6.
[275]
Wu L, Pei Y, Zhu Y, Jiang M, Wang C, Cui W, et al. Association of N6-methyladenine DNA with plaque progression in atherosclerosis via myocardial infarction-associated transcripts. Cell Death & Disease. 2019; 10: 909.
[276]
Blenkinsop TA, Corneo B, Temple S, Stern JH. Ophthalmologic stem cell transplantation therapies. Regenerative Medicine. 2012; 7: 32–39.
[277]
Nazari H, Zhang L, Zhu D, Chader GJ, Falabella P, Stefanini F, et al. Stem cell based therapies for age-related macular degeneration: the promises and the challenges. Progress in Retinal and Eye Research. 2015; 48: 1–39.
[278]
Schwartz SD, Regillo CD, Lam BL, Eliott D, Rosenfeld PJ, Gregori NZ, et al. Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt’s macular dystrophy: follow-up of two open-label phase 1/2 studies. The Lancet. 2015; 385: 509–516.
[279]
Poulos J. The limited application of stem cells in medicine: a review. Stem Cell Research & Therapy. 2018; 9: 1.
[280]
Jin Z, Gao M, Deng W, Wu K, Sugita S, Mandai M, et al. Stemming retinal regeneration with pluripotent stem cells. Progress in Retinal and Eye Research. 2019; 69: 38–56.
[281]
Zhang C, Chen Y, Sun B, Wang L, Yang Y, Ma D, et al. M6a modulates haematopoietic stem and progenitor cell specification. Nature. 2017; 549: 273–276.
[282]
Li X, Xie J, Zhai Y, Fang T, Rao N, Hu S, et al. Differentiation of Stem Cells from Human Exfoliated Deciduous Teeth into Retinal Photoreceptor-Like Cells and Their Sustainability In Vivo. Stem Cells International. 2019; 2019: 2562981.
[283]
Sawyers CL. The cancer biomarker problem. Nature. 2008; 452: 548–552.
[284]
Tamhane M, Cabrera-Ghayouri S, Abelian G, Viswanath V. Review of Biomarkers in Ocular Matrices: Challenges and Opportunities. Pharmaceutical Research. 2019; 36: 40.
[285]
Zhang SY, Zhang SW, Liu L, Meng J, Huang Y. m6A-Driver: Identifying Context-Specific mRNA m6A Methylation-Driven Gene Interaction Networks. PLOS Computational Biology. 2016; 12: e1005287.
[286]
Deng X, Su R, Weng H, Huang H, Li Z, Chen J. RNA N6-methyladenosine modification in cancers: current status and perspectives. Cell Research. 2018; 28: 507–517.
[287]
Widagdo J, Zhao QY, Kempen MJ, Tan MC, Ratnu VS, Wei W, et al. Experience-Dependent Accumulation of N6-Methyladenosine in the Prefrontal Cortex Is Associated with Memory Processes in Mice. The Journal of Neuroscience. 2016; 36: 6771–6777.
[288]
De Jesus DF, Zhang Z, Kahraman S, Brown NK, Chen M, Hu J, et al. M6a mRNA methylation regulates human β-cell biology in physiological states and in type 2 diabetes. Nature Metabolism. 2019; 1: 765–774.
[289]
Li Q, Wang C, Dong W, Su Y, Ma Z. WTAP facilitates progression of endometrial cancer via CAV-1/NF-κB axis. Cell Biology International. 2021; 45: 1269–1277.
[290]
Tang J, Wan Q, Lu J. The prognostic values of m6a RNA methylation regulators in uveal melanoma. BMC Cancer. 2020; 20: 674.
[291]
Shen F, Huang W, Huang JT, Xiong J, Yang Y, Wu K, et al. Decreased N(6)-methyladenosine in peripheral blood RNA from diabetic patients is associated with FTO expression rather than ALKBH5. The Journal of Clinical Endocrinology and Metabolism. 2015; 100: E148–E154.
[292]
Quiles-Jiménez A, Gregersen I, Mittelstedt Leal de Sousa M, Abbas A, Kong XY, Alseth I, et al. N6-methyladenosine in RNA of atherosclerotic plaques: an epitranscriptomic signature of human carotid atherosclerosis. Biochemical and Biophysical Research Communications. 2020; 533: 631–637.
[293]
Fu J, Cui X, Zhang X, Cheng M, Li X, Guo Z, et al. The Role of m6A Ribonucleic Acid Modification in the Occurrence of Atherosclerosis. Frontiers in Genetics.. 2021; 12: 733871.
[294]
Gong C, Fan Y, Liu J. METTL14 mediated m6a modification to LncRNA ZFAS1/ RAB22a: a novel therapeutic target for atherosclerosis. International Journal of Cardiology. 2021; 328: 177.
[295]
Chen M, Nie Z, Wen X, Gao Y, Cao H, Zhang S. M6a RNA methylation regulators can contribute to malignant progression and impact the prognosis of bladder cancer. Bioscience Reports. 2019; 39: BSR20192892.
[296]
Liu T, Li C, Jin L, Li C, Wang L. The Prognostic Value of m6A RNA Methylation Regulators in Colon Adenocarcinoma. Medical Science Monitor. 2019; 25: 9435–9445.
[297]
Wang Q, Chen C, Ding Q, Zhao Y, Wang Z, Chen J, et al. METTL3-mediated m6A modification of HDGF mRNA promotes gastric cancer progression and has prognostic significance. Gut. 2020; 69: 1193–1205.
[298]
Punzo C, Kornacker K, Cepko CL. Stimulation of the insulin/mTOR pathway delays cone death in a mouse model of retinitis pigmentosa. Nature Neuroscience. 2009; 12: 44–52.
[299]
Rodríguez-Muela N, Hernández-Pinto AM, Serrano-Puebla A, García-Ledo L, Latorre SH, de la Rosa EJ, et al. Lysosomal membrane permeabilization and autophagy blockade contribute to photoreceptor cell death in a mouse model of retinitis pigmentosa. Cell Death & Differentiation. 2015; 22: 476–487.
[300]
Besirli CG, Chinskey ND, Zheng Q, Zacks DN. Autophagy Activation in the Injured Photoreceptor Inhibits Fas-Mediated Apoptosis. Investigative Opthalmology & Visual Science. 2011; 52: 4193–4199.
[301]
Kunchithapautham K, Coughlin B, Lemasters JJ, Rohrer B. Differential Effects of Rapamycin on Rods and Cones during Light-Induced Stress in Albino Mice. Investigative Opthalmology & Visual Science. 2011; 52: 2967–2975.
[302]
Chen Y, Sawada O, Kohno H, Le Y, Subauste C, Maeda T, et al. Autophagy Protects the Retina from Light-induced Degeneration. Journal of Biological Chemistry. 2013; 288: 7506–7518.
[303]
Zhou Z, Doggett TA, Sene A, Apte RS, Ferguson TA. Autophagy supports survival and phototransduction protein levels in rod photoreceptors. Cell Death & Differentiation. 2015; 22: 488–498.
[304]
Ferguson TA, Laurie GW. Introduction to Autophagy in the Eye (or “what’s Eatin’ you?”). Experimental Eye Research. 2016; 144: 1–3.
[305]
Mitter SK, Rao HV, Qi X, Cai J, Sugrue A, Dunn WA, et al. Autophagy in the Retina: a Potential Role in Age-Related Macular Degeneration. Retinal Degenerative Diseases. 2012; 723: 83–90.
[306]
Boya P. Why autophagy is good for retinal ganglion cells? Eye. 2017; 31: 185–190.
[307]
Wang X, Wu R, Liu Y, Zhao Y, Bi Z, Yao Y, et al. m6A mRNA methylation controls autophagy and adipogenesis by targeting Atg5 and Atg7. Autophagy. 2020; 16: 1221–1235.
[308]
Jin S, Zhang X, Miao Y, Liang P, Zhu K, She Y, et al. M6a RNA modification controls autophagy through upregulating ULK1 protein abundance. Cell Research. 2018; 28: 955–957.
[309]
Edgar JR. Q&A: What are exosomes, exactly? BMC Biology. 2016; 14: 46.
[310]
Li N, Zhao L, Wei Y, Ea VL, Nian H, Wei R. Recent advances of exosomes in immune-mediated eye diseases. Stem Cell Research & Therapy. 2019; 10: 278.
[311]
Klingeborn M, Dismuke WM, Bowes Rickman C, Stamer WD. Roles of exosomes in the normal and diseased eye. Progress in Retinal and Eye Research. 2017; 59: 158–177.
[312]
Abner JJ, Franklin JL, Clement MA, Hinger SA, Allen RM, Liu X, et al. Depletion of METTL3 alters cellular and extracellular levels of miRNAs containing m6a consensus sequences. Heliyon. 2021; 7: e08519.
[313]
Fujimura N. WNT/β-Catenin Signaling in Vertebrate Eye Development. Frontiers in Cell and Developmental Biology. 2016; 4: 138.
[314]
Gauthier AC, Liu J. Epigenetics and Signaling Pathways in Glaucoma. BioMed Research International. 2017; 2017: 5712341.
[315]
Lee M, Goraya N, Kim S, Cho S. Hippo-yap signaling in ocular development and disease. Developmental Dynamics. 2018; 247: 794–806.
[316]
Cavodeassi F, Creuzet S, Etchevers HC. The hedgehog pathway and ocular developmental anomalies. Human Genetics. 2019; 138: 917–936.
[317]
Zhang H, Zhang P, Long C, Ma X, Huang H, Kuang X, et al. m6A methyltransferase METTL3 promotes retinoblastoma progression via PI3K/AKT/mTOR pathway. Journal of Cellular and Molecular Medicine. 2020; 24: 12368–12378.
[318]
Zhou J, Du T, Li B, Rong Y, Verkhratsky A, Peng L. Crosstalk between MAPK/ERK and PI3K/AKT Signal Pathways during Brain Ischemia/Reperfusion. ASN Neuro. 2015; 7: 175909141560246.
[319]
Aksamitiene E, Kiyatkin A, Kholodenko B. Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: a fine balance. Biochemical Society Transactions. 2012; 40: 139–146.
[320]
Yu J, Shen L, Liu Y, Ming H, Zhu X, Chu M, et al. The m6a methyltransferase METTL3 cooperates with demethylase ALKBH5 to regulate osteogenic differentiation through NF-κB signaling. Molecular and Cellular Biochemistry. 2020; 463: 203–210.
[321]
Yang S, Wei J, Cui Y, Park G, Shah P, Deng Y, et al. M6a mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nature Communications. 2019; 10: 2782.
[322]
Xie Q, Wu TP, Gimple RC, Li Z, Prager BC, Wu Q, et al. N6-methyladenine DNA Modification in Glioblastoma. Cell. 2018; 175: 1228–1243.e1220.
[323]
Chai R, Wu F, Wang Q, Zhang S, Zhang K, Liu Y, et al. M6a RNA methylation regulators contribute to malignant progression and have clinical prognostic impact in gliomas. Aging. 2019; 11: 1204–1225.
[324]
Huang W, Kong F, Li R, Chen X, Wang K. Emerging Roles of mA RNA Methylation Regulators in Gynecological Cancer. Frontiers in Oncology. 2022; 12: 827956.
[325]
Vu LP, Pickering BF, Cheng Y, Zaccara S, Nguyen D, Minuesa G, et al. The N6-methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nature Medicine. 2017; 23: 1369–1376.
[326]
Niu Y, Lin Z, Wan A, Chen H, Liang H, Sun L, et al. RNA N6-methyladenosine demethylase FTO promotes breast tumor progression through inhibiting BNIP3. Molecular Cancer. 2019; 18: 46.
[327]
Cai J, Yang F, Zhan H, Situ J, Li W, Mao Y, et al. RNA m6A Methyltransferase METTL3 Promotes The Growth Of Prostate Cancer By Regulating Hedgehog Pathway. OncoTargets and Therapy. 2019; 12: 9143–9152.
[328]
Ma JZ, Yang F, Zhou CC, Liu F, Yuan JH, Wang F, et al. METTL14 suppresses the metastatic potential of hepatocellular carcinoma by modulating N6 -methyladenosine-dependent primary MicroRNA processing. Hepatology. 2017; 65: 529–543.
[329]
Jin D, Guo J, Wu Y, Du J, Yang L, Wang X, et al. M6a mRNA methylation initiated by METTL3 directly promotes YAP translation and increases YAP activity by regulating the MALAT1-miR-1914-3p-YAP axis to induce NSCLC drug resistance and metastasis. Journal of Hematology & Oncology. 2019; 12: 135.
[330]
Tang B, Yang Y, Kang M, Wang Y, Wang Y, Bi Y, et al. M6a demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Molecular Cancer. 2020; 19: 3.
[331]
Li T, Hu P, Zuo Z, Lin J, Li X, Wu Q, et al. METTL3 facilitates tumor progression via an m6a-IGF2BP2-dependent mechanism in colorectal carcinoma. Molecular Cancer. 2019; 18: 112.
[332]
Zhou J, Wang J, Hong B, Ma K, Xie H, Li L, et al. Gene signatures and prognostic values of m6a regulators in clear cell renal cell carcinoma – a retrospective study using TCGA database. Aging. 2019; 11: 1633–1647.
[333]
Zhang C, Fu J, Zhou Y. A Review in Research Progress Concerning m6A Methylation and Immunoregulation. Frontiers in Immunology. 2019; 10: 922.
[334]
Li X, Tang J, Huang W, Wang F, Li P, Qin C, et al. The M6a methyltransferase METTL3: acting as a tumor suppressor in renal cell carcinoma. Oncotarget. 2017; 8: 96103–96116.
[335]
Selberg S, Blokhina D, Aatonen M, Koivisto P, Siltanen A, Mervaala E, et al. Discovery of Small Molecules that Activate RNA Methylation through Cooperative Binding to the METTL3-14-WTAP Complex Active Site. Cell Reports. 2019; 26: 3762–3771.e5.
[336]
Lux A, Llacer H, Heussen FMA, Joussen AM. Non-responders to bevacizumab (Avastin) therapy of choroidal neovascular lesions. British Journal of Ophthalmology. 2007; 91: 1318–1322.
[337]
Suzuki M, Nagai N, Izumi-Nagai K, Shinoda H, Koto T, Uchida A, et al. Predictive factors for non-response to intravitreal ranibizumab treatment in age-related macular degeneration. British Journal of Ophthalmology. 2014; 98: 1186–1191.
[338]
Garbo S, Zwergel C, Battistelli C. M6a RNA methylation and beyond – the epigenetic machinery and potential treatment options. Drug Discovery Today. 2021; 26: 2559–2574.
[339]
Huang Y, Yan J, Li Q, Li J, Gong S, Zhou H, et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Research. 2015; 43: 373–384.
[340]
YEBRA MJ, SANCHEZ J, MARTIN CG, HARDISSON C, BARBES C. The effect of sinefungin and synthetic analogues on RNA and DNA methyltransferases from Streptomyces. The Journal of Antibiotics. 1991; 44: 1141–1147.
[341]
Richon VM, Johnston D, Sneeringer CJ, Jin L, Majer CR, Elliston K, et al. Chemogenetic Analysis of Human Protein Methyltransferases. Chemical Biology & Drug Design. 2011; 78: 199–210.
[342]
Yadav MK, Park SW, Chae SW, Song JJ. Sinefungin, a natural nucleoside analogue of S-adenosylmethionine, inhibits Streptococcus pneumoniae biofilm growth. BioMed Research International. 2014; 2014: 156987.
[343]
Tzelepis K, De Braekeleer E, Yankova E, Rak J, Aspris D, Domingues AF, et al. Pharmacological Inhibition of the RNA m6a Writer METTL3 as a Novel Therapeutic Strategy for Acute Myeloid Leukemia. Blood. 2019; 134: 403–403.
[344]
Huang Y, Su R, Sheng Y, Dong L, Dong Z, Xu H, et al. Small-Molecule Targeting of Oncogenic FTO Demethylase in Acute Myeloid Leukemia. Cancer Cell. 2019; 35: 677–691.e10.
[345]
Su R, Dong L, Li Y, Gao M, Han L, Wunderlich M, et al. Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion. Cancer Cell. 2020; 38: 79–96.e11.
[346]
Niu Y, Wan A, Lin Z, Lu X, Wan G. N6-Methyladenosine modification: a novel pharmacological target for anti-cancer drug development. Acta Pharmaceutica Sinica B. 2018; 8: 833–843.
[347]
Su R, Dong L, Li C, Nachtergaele S, Wunderlich M, Qing Y, et al. R-2HG Exhibits Anti-tumor Activity by Targeting FTO/m6a/MYC/CEBPA Signaling. Cell. 2018; 172: 90–105.e23.
[348]
Yankova E, Blackaby W, Albertella M, Rak J, De Braekeleer E, Tsagkogeorga G, et al. Small-molecule inhibition of METTL3 as a strategy against myeloid leukaemia. Nature. 2021; 593: 597–601.
[349]
Bedi RK, Huang D, Eberle SA, Wiedmer L, Śledź P, Caflisch A. Small‐Molecule Inhibitors of METTL3, the Major Human Epitranscriptomic Writer. ChemMedChem. 2020; 15: 744–748.
[350]
Moroz‐Omori EV, Huang D, Kumar Bedi R, Cheriyamkunnel SJ, Bochenkova E, Dolbois A, et al. METTL3 Inhibitors for Epitranscriptomic Modulation of Cellular Processes. ChemMedChem. 2021; 16: 3035–3043.
[351]
Liu X, Zhou J, Mao Y, Ji Q, Qian S. Programmable RNA N6-methyladenosine editing by CRISPR-Cas9 conjugates. Nature Chemical Biology. 2019; 15: 865–871.
[352]
Bajaj J, Hamilton M, Shima Y, Chambers K, Spinler K, Van Nostrand EL, et al. An in vivo genome-wide CRISPR screen identifies the RNA-binding protein Staufen2 as a key regulator of myeloid leukemia. Nature Cancer. 2020; 1: 410–422.
[353]
Li J, Chen Z, Chen F, Xie G, Ling Y, Peng Y, et al. Targeted mRNA demethylation using an engineered dCas13b-ALKBH5 fusion protein. Nucleic Acids Research. 2020; 48: 5684–5694.
[354]
Ianniello Z, Paiardini A, Fatica A. N6-Methyladenosine (m6A): A Promising New Molecular Target in Acute Myeloid Leukemia. Frontiers in Oncology. 2019; 9: 251.
[355]
Cully M. Chemical inhibitors make their RNA epigenetic mark. Nature Reviews Drug Discovery. 2019; 18: 892–894.
[356]
Flynn RA, Pedram K, Malaker SA, Batista PJ, Smith BAH, Johnson AG, et al. Small RNAs are modified with N-glycans and displayed on the surface of living cells. Cell. 2021; 184: 3109–3124.e22.
[357]
Møller HD, Mohiyuddin M, Prada-Luengo I, Sailani MR, Halling JF, Plomgaard P, et al. Circular DNA elements of chromosomal origin are common in healthy human somatic tissue. Nature Communications. 2018; 9: 1069.
[358]
Paulsen T, Kumar P, Koseoglu MM, Dutta A. Discoveries of Extrachromosomal Circles of DNA in Normal and Tumor Cells. Trends in Genetics. 2018; 34: 270–278.
[359]
Morton AR, Dogan-Artun N, Faber ZJ, MacLeod G, Bartels CF, Piazza MS, et al. Functional Enhancers Shape Extrachromosomal Oncogene Amplifications. Cell. 2019; 179: 1330–1341.e1313.
[360]
Wu R, Liu Y, Zhao Y, Bi Z, Yao Y, Liu Q, et al. m6A methylation controls pluripotency of porcine induced pluripotent stem cells by targeting SOCS3/JAK2/STAT3 pathway in a YTHDF1/YTHDF2-orchestrated manner. Cell Death & Disease. 2019; 10: 171.
[361]
Zhang Z, Luo K, Zou Z, Qiu M, Tian J, Sieh L, et al. Genetic analyses support the contribution of mRNA N6-methyladenosine (m6A) modification to human disease heritability. Nature Genetics. 2020; 52: 939–949.
Share
Back to top