Next Article in Journal
Distinct Mechanisms of Target Search by Endonuclease VIII-like DNA Glycosylases
Next Article in Special Issue
Diabetic Macular Edema: Current Understanding, Molecular Mechanisms and Therapeutic Implications
Previous Article in Journal
PFN1 Inhibits Myogenesis of Bovine Myoblast Cells via Cdc42-PAK/JNK
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of N6-Methyladenosine Modification in Microvascular Dysfunction

Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing 210029, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(20), 3193; https://doi.org/10.3390/cells11203193
Submission received: 27 August 2022 / Revised: 24 September 2022 / Accepted: 28 September 2022 / Published: 11 October 2022
(This article belongs to the Special Issue What We Know about the Updated Information from Genes to Eye Disease)

Abstract

:
Microvascular dysfunction (MVD) has long plagued the medical field despite improvements in its prevention, diagnosis, and intervention. Microvascular lesions from MVD increase with age and further lead to impaired microcirculation, target organ dysfunction, and a mass of microvascular complications, thus contributing to a heavy medical burden and rising disability rates. An up-to-date understanding of molecular mechanisms underlying MVD will facilitate discoveries of more effective therapeutic strategies. Recent advances in epigenetics have revealed that RNA methylation, an epigenetic modification, has a pivotal role in vascular events. The N6-methylation of adenosine (m6A) modification is the most prevalent internal RNA modification in eukaryotic cells, which regulates vascular transcripts through splicing, degradation, translation, as well as translocation, thus maintaining microvascular homeostasis. Conversely, the disruption of the m6A regulatory network will lead to MVD. Herein, we provide a review discussing how m6A methylation interacts with MVD. We also focus on alterations of the m6A regulatory network under pathological conditions. Finally, we highlight the value of m6A regulators as prognostic biomarkers and novel therapeutic targets, which might be a promising addition to clinical medicine.

1. Introduction

Microvascular dysfunction (MVD) remains a major health issue worldwide despite decades of research on its diagnosis, treatment, and prognosis. Featured by lesions in microvasculature, MVD leads to microvascular complications in various organs and systems [1,2]. Endothelial cells (ECs), pericytes, and vascular smooth muscle cells (VSMCs) are major components of microvasculature, whose proliferation, coverage, and dysfunction are key predictors of vascular fragility [3,4]. The etiology of MVD is heterogeneous and polymorphic. Various pathogenic factors, including hypoxia, inflammation, and metabolic disorders, contribute to MVD occurrence and development (Figure 1) [5].
RNA methylation is a group of epigenetic modifications that modulate gene expression without altering nucleotide sequences. RNA methylation includes 7-methylguanosine (m7G), 5-methylcytosine (m5C), 5-hydroxymethylcytosine (hm5C), N1-methyladenosine (m1A), N6-methyladenosine (m6A), N6,2′-O-dimethyladenosine (m6Am), and 2′-O′ methylation (2′-OMe). Among all, m6A modification is the most prevalent, abundant, and typical form in eukaryotes. Reportedly, m6A modification regulates the vascular regulatory network by mediating metabolism of vascular cells and expression of vascular genes [6,7]. Impaired m6A regulatory network disrupts microvascular homeostasis, further leading to MVD [8,9].
In this review, we summarized and discussed the role of m6A modification in MVD, aiming to provide a better understanding into its pathogenesis. Three dominant pathological processes of MVD were investigated, including neovascularization, microvascular malformation, and microvascular remodeling. This review also highlighted the potential clinical applications of m6A regulators as prognostic biomarkers and therapeutic targets for MVD.

2. RNA m6A Methylation

M6A modification, first detected in the 1970s, is the most abundant biochemical modification in eukaryotic RNAs, accounting for 0.1–0.4% of mammalian adenosine [10]. M6A modification has been identified in various types of RNAs, including messenger RNAs (mRNAs), transfer RNAs (tRNAs), ribosomal RNAs, long noncoding RNAs (lncRNAs), circular RNAs (circRNAs), small nuclear RNAs (snRNAs), and microRNAs (miRNAs). M6A modification participates in almost every step of RNA metabolism, from its generation, splicing, and processing in the nucleus to its translation, stabilization, and degradation in the cytoplasm, serving as a bridge between transcription and translation [11].
The global m6A level is dynamically regulated by writers and erasers, namely RNA methylases and demethylases respectively (Figure 2). M6A writers include methyltransferase-like 3/14/16 (METTL3/14/16), Wilms tumor 1-associated protein (WTAP), zinc finger CCCH-type containing 13 (ZC3H13), Vir-like m6A methyltransferase associated protein (VIRMA), and RNA-binding motif protein 15 (RBM15) [12]. The METTL3-METTL14 heterodimer and its catalytically inactive partner WTAP constitute the nucleus methyltransferase complex (MTC), which installs m6A modification. VIRMA, RBM15, and ZC3H13 are regulatory enzymes that facilitate recruitment of MTC [13]. RBM15 and ZC3H13 bind to the MTC and direct it to target RNA sites [14]. VIRMA regulates selective m6A methylation on 3′-UTR [14]. Reportedly, METTL16 is an independent writer that modifies snRNAs, U6 snRNA, and lncRNAs, but only a few substrates of METTL16 have been confirmed [14]. M6A erasers include fat mass and obesity-associated protein (FTO) and a-ketoglutarate-dependent dioxygenase alkB homolog 5 (ALKBH5). Both of them belong to the Fe2+/α-ketoglutarate-dependent dioxygenases enzyme family, which recognizes adenine and cytosine methylation in RNAs [14]. ALKBH5 also affects the synthesis and splicing of mRNAs [15]. RNA m6A sites are further recognized by m6A readers. Identified m6A readers include YT521-B homology (YTH) domain-containing proteins (YTHDF1/2/3, YTHDC1/2), insulin-like growth factor 2 mRNA-binding-proteins (IGF2BP1/2/3), heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1), and hnRNPC (Figure 2) [16]. hnRNPs and YTHDC1 are nuclear readers. hnRNPC binds to structurally altered RNAs and mediates pre-mRNA processing [14]. hnRNPA2B1 plays a vital role in RNA splicing and primary miRNA processing [14]. YTHDC1 mediates alternative splicing and facilitates mRNA export to cytoplasm [13]. In contrast, YTHDF1/2/3, YTHDC2, and IGF2BP1/2/3 are cytoplasmic-distributed. YTHDF1 recognizes m6A sites near the stop codon and enables mRNA translation by recruiting eukaryotic initiation factor 3, whereas YTHDF2 transports target mRNAs to the cytoplasmic processing body and promotes their degradation [17]. YTHDF3 is a modulator of YTHDF1 and YTHDF2, which can both enhance and suppress their effects [14]. The IGF2BP proteins are co-localized with Hu antigen R to enhance stability of target RNA transcripts [14]. They are also reported to participate in DNA replication and cell cycle progression [18].

3. M6A Modifications in Pathological Neovascularization

Neovascularization is defined as the sprouting of ECs in response to stimuli to form new capillary branches. The following steps are involved in neovascularization: (1) recognition of physiological or pathological signals, such as hypoxia, inflammation, and metabolic dysregulation; (2) secretion of proteases, pro-angiogenic factors and cytokines, and their bindings to corresponding receptors; (3) metabolic changes of vascular cells; (4) maturation of newly-formed vessels [19]. Reportedly, dysregulated epigenetic modifications, including DNA methylation, histone modifications, and RNA methylation, contribute to neovascularization [20]. Herein, we have summarized associations between aberrantly changed expression of m6A regulators and pathological neovascularization in Table 1.

3.1. M6A Modifications in Hypoxia-Related Neovascularization

Hypoxic effects are mediated by hypoxia-inducible factor (HIF), which combines with hypoxia-responsive elements (HREs) of target genes to regulate their expression [33]. There are three isoforms of HIF, including HIF-1, HIF-2, and HIF-3 [34]. HIFs are heterodimers composed of an α (HIF-1α, HIF-2α and HIF-3α) and a β (HIF-1β, HIF-2β and HIF-3β) subunit [35]. The C- and N-termini of α subunits have nuclear localization signals that direct them to nucleus to form adult HIFs [36]. Degradation of α subunits depends on prolyl hydroxylase domain-containing proteins (PHDs). Under normal conditions, PHDs target α subunits and mediate their polyubiquitination and degradation. However, activity of PHDs is disturbed upon hypoxia, thus interrupting the degradation of α subunits [37]. HIF-1α and HIF-2α share similar amino acid sequences and protein structures, and they regulate angiogenesis by targeting angiogenic factors (e.g., vascular endothelial growth factor (VEGF), angiopoietin-1/-2 (ANG-1/-2), transforming growth factor β (TGF-β), platelet-derived growth factor (PDGF)). However, the biological function of HIF-3 remains elusive [38,39].
Hypoxia could reprogram m6A epi-transcriptome, further reshaping downstream transcriptome and proteome that associate with neovascularization [40]. Increased METTL14 and ALKBH5 levels were detected in hypoxia-treated breast cancer cells, which led to upregulation of angiogenic transcripts, including TGF-β, matrix metallopeptidase 9 (MMP9), PDGF, and VEGFA. Conversely, METTL14/ALKBH5 knockdown reduced expression of angiogenic genes, thus inhibiting angiogenesis and cancer metastasis [23]. Hou et al. revealed a transcriptional inhibition of YTHDF2 by HIF-2α in hepatocellular carcinoma (HCC) cells. Suppressed expression of YTHDF2 not only promoted neovascularization through interleukin-11 (IL-11) and serpin family E member 2 (SERPINE2) but also led to microvascular malformation and remodeling. These adverse effects could be rescued by YTHDF2 upregulation [24]. Therefore, hypoxia primarily caused m6A changes, thus contributing to pathological neovascularization. Hypoxia-induced METTL3 downregulation in HCC promoted angiogenesis by upregulating expression of angiogenic genes, such as fibroblast growth factor, PDGF, and VEGFA, thus contributing to sorafenib resistance [25]. The Wnt signaling pathway is critical for vascular morphogenesis and endothelial specification [41]. Aberrantly activated Wnt signaling pathway is a leading cause of pathological neovascularization, particularly in wet age-related macular degeneration, diabetic retinopathy, and retinopathy of prematurity [42]. Yao et al. showed that METTL3 was upregulated in hypoxia-exposed retina [26]. METTL3 upregulation enhanced expression of LDL receptor related protein 6 (LRP6) and disheveled segment polarity protein 1 (DVL1) mRNAs, which promoted angiogenesis by activating Wnt signaling cascades [26].
Aberrantly changed expression of m6A regulators also facilitates HIFs generation and reprograms cellular metabolism, thus triggering neovascularization. In stomach cancer, IGF2BP3 directly targeted an m6A site in HIF-1α mRNA to upregulate its expression, leading to increased microvascular density and a poor outcome [22]. In HCC cells, METTL3, which was positively regulated by hepatitis B virus X-interacting protein (HBXIP), methylated HIF-1 mRNA to upregulate its expression, further contributing to the Warburg effect and angiogenesis [43]. Furthermore, in lung cancer, the crosstalk between polybromo 1 (PBRM1) and YTHDF2 was required for the effective synthesis of HIF-1 protein. YTHDF2 mediated RNA degradation in the cytoplasm under normal conditions, while it translocated into cell nucleus upon hypoxia to promote the cap-independent translation of HIF-1α mRNAs [21]. Collectively, these studies imply the critical role of m6A modification in hypoxia-induced neovascularization.

3.2. M6A Modifications in Inflammation-Related Pathological Neovascularization

Inflammation tends to induce irregularly shaped, leaky, and highly permeable angiogenesis rather than mature and functional vasculature [44]. Shan and colleagues detected altered expression of several m6A regulators, including FTO, METTL3, and METTL14, in mice with corneal neovascularization [27]. They further revealed that FTO promoted corneal neovascularization by inducing focal adhesion kinase (FAK) upregulation. In the alkali-burned corneal model, Yao et al. noticed that METTL3 knockdown restricted corneal neovascularization by inhibiting the Wnt pathway [26]. In HCC, YTHDF2 downregulation promoted neovascularization by accelerating the translation of inflammatory cytokines, such as IL-11 and SERPINE2 [24]. Similarly, lysine acetyltransferase 1 (KAT1) was poorly expressed in diabetic retinopathy, leading to YTHDF2 downregulation and inflammation-related neovascularization. YTHDF2 upregulation inhibited neovascularization and vascular leakage by degrading integrin subunit beta 1 (ITGB1) mRNAs and suppressing the FAK/PI3K/AKT signaling pathway [28]. These studies indicated the critical role of m6A modification in inflammation-related neovascularization.

3.3. Others

In this section, we present findings on the m6A-associated pathological angiogenesis in non-specific contexts. He et al. identified that decreased m6A level associated with reinforced angiogenesis and a poor survival rate in breast cancer [29]. They found that YTHDF3 promoted the binding between eukaryotic initiation factor 3 and angiogenic transcripts, such as VEGFA and epidermal growth factor receptor (EGFR), indicating its potential role as a therapeutic target in breast cancer [29]. Wang et al. found that METTL3 associated with angiogenesis and brain metastasis in lung cancer. Mechanistically, METTL3 promoted angiogenesis via facilitating the splicing of precursor miR-143-3p to generate its adult form, which positively regulated VEGFA expression [30]. Ma et al. identified miR-320b downregulation in lung cancer, which accelerated neovascularization through IGF2BP2-mediated thymidine kinase 1 (TK1) upregulation [45]. These results indicated that miRNAs and m6A regulators can be mutually regulated. In intrahepatic cholangiocarcinoma, FTO inhibited angiogenesis and tumor cell migration via upregulating C-C motif chemokine ligand 19 (CCL19) expression [31]. FTO also induced the apoptosis of intrahepatic cholangiocarcinoma cells by enhancing their sensitivity to cisplatin, indicating its potential role as a multipotent therapeutic target. In colorectal cancer/melanoma, ALKBH5 accelerated expression of angiogenic genes, such as VEGFA and TGFβ1, which weakened the efficacy of GVAX/anti–PD-1 therapy. These adverse effects could be rescued by the small-molecule ALKBH5 inhibitor (ALK-04) [32]. These studies revealed a critical role of m6A regulators in neovascularization and implied their potential therapeutic application in MVD.

4. M6A Modifications in Microvascular Malformation

Microvascular malformation mainly encompasses micro-venous malformation, arteriovenous malformation, lymphatic malformation, and mixed malformation [46]. Microvascular malformation, which can be congenital or acquired, arises from abnormal neovascularization, genetic mutations, and post-injury structural changes [46]. Endothelial dysplasia and incomplete pericyte coverage are two major characters of microvascular malformation [47]. Herein, we aim to discuss the association between m6A dysregulation and microvascular malformation (Table 2).

4.1. M6A Modifications in Hypoxia-Related Microvascular Malformation

M6A modification participates in hypoxia-related microvascular malformation by triggering incomplete pericyte coverage [48]. YTHDF2 positively regulates pericyte coverage by degrading m6A-containing IL-11 and SERPINE2 mRNAs [24]. YTHDF2 expression was suppressed in HIF-2α-treated HCC cells, which inhibited pericyte coverage and generated aberrant microvasculature. The HIF-2α blockade (PT2385) upregulated YTHDF2 expression, thus reversing the subsequent microvascular abnormalities in HCC [24]. Malignant tumors tend to obtain sufficient blood perfusion through vasculogenic mimicry, a vasculature-like structure formed by tumor cells instead of ECs [49]. Qiao and colleagues identified METTL3 upregulation in HCC, which facilitated both angiogenesis and vasculogenic mimicry [50]. Mechanistically, METTL3 aberrantly activated the Hippo pathway to generate vasculogenic mimicry, and upregulated angiogenic transcripts, such as vascular endothelial growth factor receptor 1/2 (VEGFR1/2) and matrix metallopeptidase 2/9 (MMP2/9), to promote angiogenesis [50]. Collectively, these studies implied a critical role of m6A modification in hypoxia-induced microvascular remodeling.

4.2. M6A Modifications in Inflammation-Related Microvascular Malformation

In diabetic retinopathy, METTL3 upregulation was detected in pericytes treated with inflammatory stimuli, such as tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) [51]. METTL3 impaired viability, proliferation, and differentiation of pericytes via inhibiting the protein kinase C (PKC)/FAT4/PDGFRA axis in a YTHDF2-dependent manner. Conversely, Suo et al. detected that METTL3-specific deletion in pericytes promoted their coverage and suppressed diabetic microvascular complications [51]. In diabetic nephropathy, METTL14 was found to inhibit expression of α-klotho gene (an anti-inflammatory gene) and its encoding protein, leading to upregulation of inflammatory cytokines (TNF-α, IL-6) and microvascular malformation [52]. Therefore, a single m6A regulator may affect various downstream genes and set off a chain effect.

4.3. Others

Arteriovenous malformation is a vascular variation caused by the lack of capillary beds between venules and arterioles [53,54]. Wang et al. detected reduced METTL3 expression in arteriovenous malformation, which inhibited synergistic function of deltex E3 ubiquitin ligase 3L/1 (DTX1/3L) as Notch blockers, leading to aberrantly activated Notch signaling pathway and capillary malformation. These adverse effects could be restored by the Notch antagonist DAPT [55]. WTAP was also found downregulated in arteriovenous malformation, which caused capillary malformation through destabilizing desmoplakin (DSP), a critical component that maintains the integrity of vascular wall [56]. The Akt/mTOR signaling pathway is critical for endothelial differentiation [57]. In zebrafish embryos, METTL3 deletion in ECs upregulated the expression of PH domain and leucine rich repeat protein phosphatase 2 (PHLPP2), which promoted Akt dephosphorylation and suppressed the Akt/mTOR signaling pathway, thus leading to microvascular malformation [58]. Consistently, METTL3 deletion in bone mesenchymal stem cells also caused Akt dephosphorylation during osteogenic differentiation, thus inhibiting vascular normalization [59,60]. These microvascular defects were salvaged by Akt1 overexpression and/or the Akt activator SC79 [61]. These studies indicated a critical role of m6A modification in regulating Akt phosphorylation (Table 2).

5. M6A Modifications in Microvascular Remodeling

Microvascular remodeling is defined as structural or functional adaptations of the microvasculature. Either neovascularization or microvascular malformation can progress into microvascular remodeling (Figure 1) [62]. Herein, we have summarized associations between aberrantly changed expression of m6A regulators and microvascular remodeling in Table 3.

5.1. M6A Modifications in Hypoxia-Related Microvascular Remodeling

Hypoxia-induced microvascular remodeling is primarily driven by HIF-2α [63]. Hou et al. identified that HIF-2α suppressed YTHDF2 expression in HCC. The reduced YTHDF2 level further provoked microvascular reconstruction by upregulating expression of IL-11 and SERPINE2 [24]. Pulmonary arterial hypertension is a lethal disease driven by progressive microvascular remodeling [64]. Proliferation of VSMCs is the main character of pulmonary arterial hypertension, manifested by concentric vasoconstriction and extracellular matrix deposition. METTL14 upregulation was observed in hypoxia-treated VSMCs, leading to progressive microvascular remodeling [65]. However, the downstream regulatory mechanism of METTL14-induced microvascular malformation remains elusive [65]. Proliferation of VSMCs depends on phosphatase and tensin homolog (PTEN), an endogenous inhibitor of PI3K/Akt/mTOR signaling cascades [66]. METTL3 upregulation in hypoxia-treated VSMCs mediated the degradation of PTEN mRNAs through YTHDF2 recognition. Thus, aberrant proliferation and migration of VSMCs occurred through Akt hyperphosphorylation, contributing to microvascular remodeling [67].

5.2. M6A Modifications in Inflammation-Related Microvascular Remodeling

Inflammation-related microvascular remodeling is driven by migration of inflammatory cells, which is mediated by adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin [62]. In atherosclerosis, METTL3 promoted microvascular remodeling by upregulating the expression of NLR family pyrin domain containing 1 (NRLP1), a gene generating inflammasomes, with YTHDF1 as the reader [68]. Moreover, METTL3 aggravated endothelial inflammation by inhibiting the expression of the anti-inflammatory protein KLF transcription factor 4 (KLF4) [68]. However, in TNF-α-treated ECs, METTL3 knockdown mitigated monocyte adhesion and microvascular remodeling [68]. In addition, METTL14 was also found upregulated in TNF-α-treated ECs, facilitating FOXO1 translation through YTHDF1 recognition [69]. FOXO1 then acted upon promoter regions of VCAM-1 and ICAM-1 mRNAs and promoted their transcriptions, contributing to microvascular remodeling.

5.3. M6A Modifications in Metabolism-Related Microvascular Remodeling

Metabolic disorders, such as dysregulation of glucose and lipid metabolism, also associate with microvascular remodeling [70]. VSMC dysfunction and intimal hyperplasia are two typical features of microvascular remodeling [71]. FTO upregulation in VSMCs was detected in type 2 diabetes mellitus, which triggered intimal hyperplasia through disturbing mRNA stability of smooth muscle 22 alpha (SM22α) [71]. YTHDC2 promoted circYTHDC2 expression in VSMCs under high glucose. CircYTHDC2 then inhibited the expression of ten-eleven translocation 2 (TET2), a gene positively regulating VSMC plasticity, thus contributing to VSMC dysfunction and microvascular remodeling. Metformin, a first-line hypoglycemic drug, alleviated YTHDF2-mediated microvascular remodeling by arresting cell cycle and inducing cell apoptosis [72,73].
Another leading cause of microvascular remodeling is dysregulated lipid metabolism. Macrophages take up oxidized lipoproteins and transform into foam cells, which cause endothelial dysfunction and extracellular matrix deposition, thus contributing to microvascular remodeling [74]. Gong et al. speculated that in atherosclerosis METTL14 promoted lncRNA ZFAS1 expression, an ncRNA that caused dyslipidemia. LncRNA ZFAS1 then elevated ADAM10/RAB22A expression to inhibit cholesterol efflux and facilitate microvascular remodeling [75]. The scavenger receptor CD36 is the primary transporter mediating lipid uptake and is directly targeted by PPARγ [76]. FTO inhibited foam cell formation by reducing CD36 and PPARγ levels. FTO also facilitated intracellular cholesterol efflux by upregulating ATP-binding cassette transporter A1 (ABCA1) expression, implying its potential role in preventing microvascular remodeling [76] (Table 3).

6. Discussion

MVD and its regulatory network have long been investigated. Various pathogenic factors, including hypoxia, inflammation, and metabolic disorders, contribute to MVD occurrence and development. RNA m6A modification is a post-transcriptional modification, which regulates all steps of RNA metabolism (splicing, maturation, export, translation, degradation). Herein, we summarized the role of m6A modification in MVD, aiming to provide a better understanding into its pathogenesis. M6A regulators participate in MVD pathogenesis by altering m6A status of vascular transcripts, thus mediating their expression. In turn, expression patterns of m6A regulators could also be changed by various pathogenic factors contributing to MVD. We also summarized the promising application of m6A modification in therapeutic strategies for MVD.
Roles and regulatory mechanisms of m6A regulators vary with their subcellular locations and in different diseases. Reportedly, stress induced the translocation of YTHDF2 from cytoplasm to nucleus, and unlike the role of cytoplasmic YTHDF2 in mediating RNA degradation, the endonuclear YTHDF2 promoted the cap-independent mRNA translation of HIF-1α, thus contributing to neovascularization [21,78]. M6A regulators may also play opposite roles in different diseases or pathogenesis. For instance, METTL3 promoted neovascularization in stomach cancer, but suppressed expression of angiogenic factors in sorafenib-resistant HCC [25,79]. FTO showed a pro-angiogenic role in diabetic retinopathy, but an anti-angiogenic role in intrahepatic cholangiocarcinoma [28,31]. The diversity is probably due to the distinct downstream regulatory network of METTL3 in different pathological processes. In addition, m6A modifications have been detected in various types of RNAs, while their roles in mediating metabolism of noncoding RNAs that associate with MVD are largely unknown. More investigations are warranted to reveal the complex biological/pathological effects and regulatory mechanisms of m6A modification.
Targeting m6A modification might be a promising therapeutic option for MVD. In colorectal cancer/melanoma, the ALKBH5 inhibitor ALK-04 downregulated expression of VEGFA and TGFβ1, thus inhibiting angiogenesis and enhancing efficacy of anti–PD-1 therapy [32]. Excitingly, in recent years, demethylation/methylation drugs, such as decitabine and azacitidine, have been developed, which have been clinically applied for the treatment of myelodysplastic syndrome and acute myeloid leukemia [80]. Both drugs are cytidine analogues that inhibit DNA methylation and restore normal function of tumor suppressor genes. Unlike decitabine, which only incorporates into DNA, azacitidine could be phosphorylated and incorporate into DNA/RNA, thus altering RNA synthesis and processing [81]. Reportedly, effects and sensitivities of antineoplastic drugs are enhanced by m6A regulators. In intrahepatic cholangiocarcinoma, FTO promoted cisplatin sensitivity to inhibit angiogenesis and accelerate the apoptosis of tumor cells [31]. ALKBH5 sensitized pancreatic ductal adenocarcinoma cells to gemcitabine by activating the Wnt pathway [82]. Moreover, in pancreatic cancer, suppressed METTL3 expression improved the efficacy of anti-cancer agents, such as gemcitabine, 5-fluorouracil, and cisplatin. These studies further suggested the potential clinical application of m6A modification in therapeutic strategies [83]. However, more investigations are needed to explore the role of m6A modification in MVD, thus helping with the development of prognostic and therapeutic strategies for MVD.

Author Contributions

X.C. and Y.-R.Z. designed the study. Y.-R.Z., J.-N.W., Y.W., H.-J.Z., R.-X.S. and J.-D.J. drafted the manuscript. X.C. and Q.-H.L. critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by National Natural Science Foundation of China (82070974 to X.C., 81970821 and 82271100 to Q.-H.L.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Horton, W.B.; Barrett, E.J. Microvascular Dysfunction in Diabetes Mellitus and Cardiometabolic Disease. Endocr. Rev. 2021, 42, 29–55. [Google Scholar] [CrossRef] [PubMed]
  2. Marseglia, A.; Fratiglioni, L.; Kalpouzos, G.; Wang, R.; Bäckman, L.; Xu, W. Prediabetes and diabetes accelerate cognitive decline and predict microvascular lesions: A population-based cohort study. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2019, 15, 25–33. [Google Scholar] [CrossRef] [PubMed]
  3. Suhrs, H.E.; Schroder, J.; Bové, K.B.; Mygind, N.D.; Frestad, D.; Michelsen, M.M.; Lange, T.; Gustafsson, I.; Kastrup, J.; Prescott, E. Inflammation, non-endothelial dependent coronary microvascular function and diastolic function-Are they linked? PLoS ONE 2020, 15, e0236035. [Google Scholar] [CrossRef] [PubMed]
  4. Virdis, A.; Colucci, R.; Bernardini, N.; Blandizzi, C.; Taddei, S.; Masi, S. Microvascular Endothelial Dysfunction in Human Obesity: Role of TNF-α. J. Clin. Endocrinol. Metab. 2019, 104, 341–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Wong, B.W.; Marsch, E.; Treps, L.; Baes, M.; Carmeliet, P. Endothelial cell metabolism in health and disease: Impact of hypoxia. EMBO J. 2017, 36, 2187–2203. [Google Scholar] [CrossRef] [PubMed]
  6. Yang, C.; Hu, Y.; Zhou, B.; Bao, Y.; Li, Z.; Gong, C.; Yang, H.; Wang, S.; Xiao, Y. The role of m(6)A modification in physiology and disease. Cell Death Dis. 2020, 11, 960. [Google Scholar] [CrossRef] [PubMed]
  7. Garbo, S.; Zwergel, C.; Battistelli, C. m6A RNA methylation and beyond—The epigenetic machinery and potential treatment options. Drug Discov. Today 2021, 26, 2559–2574. [Google Scholar] [CrossRef]
  8. Wang, X.; Ma, R.; Zhang, X.; Cui, L.; Ding, Y.; Shi, W.; Guo, C.; Shi, Y. Crosstalk between N6-methyladenosine modification and circular RNAs: Current understanding and future directions. Mol. Cancer 2021, 20, 121. [Google Scholar] [CrossRef]
  9. Gu, Y.; Wu, X.; Zhang, J.; Fang, Y.; Pan, Y.; Shu, Y.; Ma, P. The evolving landscape of N(6)-methyladenosine modification in the tumor microenvironment. Mol. Ther. 2021, 29, 1703–1715. [Google Scholar] [CrossRef]
  10. Hu, B.B.; Wang, X.Y.; Gu, X.Y.; Zou, C.; Gao, Z.J.; Zhang, H.; Fan, Y. N(6)-methyladenosine (m(6)A) RNA modification in gastrointestinal tract cancers: Roles, mechanisms, and applications. Mol. Cancer 2019, 18, 178. [Google Scholar] [CrossRef]
  11. Shi, H.; Wei, J.; He, C. Where, When, and How: Context-Dependent Functions of RNA Methylation Writers, Readers, and Erasers. Mol. Cell 2019, 74, 640–650. [Google Scholar] [CrossRef] [PubMed]
  12. Oerum, S.; Meynier, V.; Catala, M.; Tisné, C. A comprehensive review of m6A/m6Am RNA methyltransferase structures. Nucleic Acids Res. 2021, 49, 7239–7255. [Google Scholar] [CrossRef] [PubMed]
  13. Qin, Y.; Li, L.; Luo, E.; Hou, J.; Yan, G.; Wang, D.; Qiao, Y.; Tang, C. Role of m6A RNA methylation in cardiovascular disease (Review). Int. J. Mol. Med. 2020, 46, 1958–1972. [Google Scholar] [CrossRef] [PubMed]
  14. Uddin, M.B.; Wang, Z.; Yang, C. The m(6)A RNA methylation regulates oncogenic signaling pathways driving cell malignant transformation and carcinogenesis. Mol. Cancer 2021, 20, 61. [Google Scholar] [CrossRef]
  15. Alarcon, C.R.; Lee, H.; Goodarzi, H.; Halberg, N.; Tavazoie, S.F. N6-methyladenosine marks primary microRNAs for processing. Nature 2015, 519, 482–485. [Google Scholar] [CrossRef] [Green Version]
  16. Shi, H.; Chai, P.; Jia, R.; Fan, X. Novel insight into the regulatory roles of diverse RNA modifications: Re-defining the bridge between transcription and translation. Mol. Cancer 2020, 19, 78. [Google Scholar] [CrossRef]
  17. Fu, Y.; Dominissini, D.; Rechavi, G.; He, C. Gene expression regulation mediated through reversible m⁶A RNA methylation. Nat. Rev. Genet. 2014, 15, 293–306. [Google Scholar] [CrossRef]
  18. 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. J. Exp. Clin. Cancer Res. 2020, 39, 203. [Google Scholar] [CrossRef]
  19. Zimna, A.; Kurpisz, M. Hypoxia-Inducible Factor-1 in Physiological and Pathophysiological Angiogenesis: Applications and Therapies. BioMed Res. Int. 2015, 2015, 549412. [Google Scholar] [CrossRef] [Green Version]
  20. Cho, W.C.; Jour, G.; Aung, P.P. Role of angiogenesis in melanoma progression: Update on key angiogenic mechanisms and other associated components. Semin. Cancer Biol. 2019, 59, 175–186. [Google Scholar] [CrossRef]
  21. Shmakova, A.; Frost, M.; Batie, M.; Kenneth, N.S.; Rocha, S. PBRM1 Cooperates with YTHDF2 to Control HIF-1α Protein Translation. Cells 2021, 10, 1425. [Google Scholar] [CrossRef]
  22. Jiang, L.; Li, Y.; He, Y.; Wei, D.; Yan, L.; Wen, H. Knockdown of m6A Reader IGF2BP3 Inhibited Hypoxia-Induced Cell Migration and Angiogenesis by Regulating Hypoxia Inducible Factor-1α in Stomach Cancer. Front. Oncol. 2021, 11, 711207. [Google Scholar] [CrossRef]
  23. Panneerdoss, S.; Eedunuri, V.K.; Yadav, P.; Timilsina, S.; Rajamanickam, S.; Viswanadhapalli, S.; Abdelfattah, N.; Onyeagucha, B.C.; Cui, X.; Lai, Z.; et al. Cross-talk among writers, readers, and erasers of m(6)A regulates cancer growth and progression. Sci. Adv. 2018, 4, eaar8263. [Google Scholar] [CrossRef] [Green Version]
  24. Hou, J.; Zhang, H.; Liu, J.; Zhao, Z.; Wang, J.; Lu, Z.; Hu, B.; Zhou, J.; Zhao, Z.; Feng, M.; et al. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol. Cancer 2019, 18, 163. [Google Scholar] [CrossRef] [Green Version]
  25. Lin, Z.; Niu, Y.; Wan, A.; Chen, D.; Liang, H.; Chen, X.; Sun, L.; Zhan, S.; Chen, L.; Cheng, C.; et al. RNA m(6) A methylation regulates sorafenib resistance in liver cancer through FOXO3-mediated autophagy. EMBO J. 2020, 39, e103181. [Google Scholar] [CrossRef]
  26. Yao, M.D.; Jiang, Q.; Ma, Y.; Liu, C.; Zhu, C.Y.; Sun, Y.N.; Shan, K.; Ge, H.M.; Zhang, Q.Y.; Zhang, H.Y.; et al. Role of METTL3-Dependent N(6)-Methyladenosine mRNA Modification in the Promotion of Angiogenesis. Mol. Ther. 2020, 28, 2191–2202. [Google Scholar] [CrossRef]
  27. Shan, K.; Zhou, R.M.; Xiang, J.; Sun, Y.N.; Liu, C.; Lv, M.W.; Xu, J.J. FTO regulates ocular angiogenesis via m(6)A-YTHDF2-dependent mechanism. Exp. Eye Res. 2020, 197, 108107. [Google Scholar] [CrossRef]
  28. Qi, Y.; Yao, R.; Zhang, W.; Cui, Q. KAT1 triggers YTHDF2-mediated ITGB1 mRNA instability to alleviate the progression of diabetic retinopathy. Pharmacol. Res. 2021, 170, 105713. [Google Scholar] [CrossRef]
  29. Chang, G.; Shi, L.; Ye, Y.; Shi, H.; Zeng, L.; Tiwary, S.; Huse, J.T.; Huo, L.; Ma, L.; Ma, Y.; et al. YTHDF3 Induces the Translation of m(6)A-Enriched Gene Transcripts to Promote Breast Cancer Brain Metastasis. Cancer Cell 2020, 38, 857–871.e7. [Google Scholar] [CrossRef]
  30. Wang, H.; Deng, Q.; Lv, Z.; Ling, Y.; Hou, X.; Chen, Z.; Dinglin, X.; Ma, S.; Li, D.; Wu, Y.; et al. N6-methyladenosine induced miR-143-3p promotes the brain metastasis of lung cancer via regulation of VASH1. Mol. Cancer 2019, 18, 181. [Google Scholar] [CrossRef]
  31. Rong, Z.X.; Li, Z.; He, J.J.; Liu, L.Y.; Ren, X.X.; Gao, J.; Mu, Y.; Guan, Y.D.; Duan, Y.M.; Zhang, X.P.; et al. Downregulation of Fat Mass and Obesity Associated (FTO) Promotes the Progression of Intrahepatic Cholangiocarcinoma. Front. Oncol. 2019, 9, 369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Li, N.; Kang, Y.; Wang, L.; Huff, S.; Tang, R.; Hui, H.; Agrawal, K.; Gonzalez, G.M.; Wang, Y.; Patel, S.P.; et al. ALKBH5 regulates anti-PD-1 therapy response by modulating lactate and suppressive immune cell accumulation in tumor microenvironment. Proc. Natl. Acad. Sci. USA 2020, 117, 20159–20170. [Google Scholar] [CrossRef]
  33. Tirpe, A.A.; Gulei, D.; Ciortea, S.M.; Crivii, C.; Berindan-Neagoe, I. Hypoxia: Overview on Hypoxia-Mediated Mechanisms with a Focus on the Role of HIF Genes. Int J. Mol. Sci. 2019, 20, 6140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Serocki, M.; Bartoszewska, S.; Janaszak-Jasiecka, A.; Ochocka, R.J.; Collawn, J.F.; Bartoszewski, R. miRNAs regulate the HIF switch during hypoxia: A novel therapeutic target. Angiogenesis 2018, 21, 183–202. [Google Scholar] [CrossRef] [Green Version]
  35. Taylor, C.T.; Colgan, S.P. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat. Rev. Immunol. 2017, 17, 774–785. [Google Scholar] [CrossRef]
  36. Lee, J.W.; Ko, J.; Ju, C.; Eltzschig, H.K. Hypoxia signaling in human diseases and therapeutic targets. Exp. Mol. Med. 2019, 51, 1–13. [Google Scholar] [CrossRef] [Green Version]
  37. Watts, E.R.; Walmsley, S.R. Inflammation and Hypoxia: HIF and PHD Isoform Selectivity. Trends Mol. Med. 2019, 25, 33–46. [Google Scholar] [CrossRef] [Green Version]
  38. Knutson, A.K.; Williams, A.L.; Boisvert, W.A.; Shohet, R.V. HIF in the heart: Development, metabolism, ischemia, and atherosclerosis. J. Clin. Investig. 2021, 131, e137557. [Google Scholar] [CrossRef]
  39. Maynard, M.A.; Evans, A.J.; Hosomi, T.; Hara, S.; Jewett, M.A.; Ohh, M. Human HIF-3alpha4 is a dominant-negative regulator of HIF-1 and is down-regulated in renal cell carcinoma. FASEB J. 2005, 19, 1396–1406. [Google Scholar] [CrossRef]
  40. Wang, Y.J.; Yang, B.; Lai, Q.; Shi, J.F.; Peng, J.Y.; Zhang, Y.; Hu, K.S.; Li, Y.Q.; Peng, J.W.; Yang, Z.Z.; et al. Reprogramming of m(6)A epitranscriptome is crucial for shaping of transcriptome and proteome in response to hypoxia. RNA Biol. 2021, 18, 131–143. [Google Scholar] [CrossRef]
  41. Jiang, L.; Yin, M.; Wei, X.; Liu, J.; Wang, X.; Niu, C.; Kang, X.; Xu, J.; Zhou, Z.; Sun, S.; et al. Bach1 Represses Wnt/β-Catenin Signaling and Angiogenesis. Circ. Res. 2015, 117, 364–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Wang, Z.; Liu, C.H.; Huang, S.; Chen, J. Wnt Signaling in vascular eye diseases. Prog. Retin. Eye Res. 2019, 70, 110–133. [Google Scholar] [CrossRef]
  43. Yang, N.; Wang, T.; Li, Q.; Han, F.; Wang, Z.; Zhu, R.; Zhou, J. HBXIP drives metabolic reprogramming in hepatocellular carcinoma cells via METTL3-mediated m6A modification of HIF-1α. J. Cell Physiol. 2021, 236, 3863–3880. [Google Scholar] [CrossRef]
  44. Zhang, Q.; Cao, X. Epigenetic Remodeling in Innate Immunity and Inflammation. Annu. Rev. Immunol. 2021, 39, 279–311. [Google Scholar] [CrossRef]
  45. Ma, Y.S.; Shi, B.W.; Guo, J.H.; Liu, J.B.; Yang, X.L.; Xin, R.; Shi, Y.; Zhang, D.D.; Lu, G.X.; Jia, C.Y.; et al. microRNA-320b suppresses HNF4G and IGF2BP2 expression to inhibit angiogenesis and tumor growth of lung cancer. Carcinogenesis 2021, 42, 762–771. [Google Scholar] [CrossRef] [PubMed]
  46. Lawton, M.T.; Rutledge, W.C.; Kim, H.; Stapf, C.; Whitehead, K.J.; Li, D.Y.; Krings, T.; terBrugge, K.; Kondziolka, D.; Morgan, M.K.; et al. Brain arteriovenous malformations. Nat. Rev. Dis. Primers 2015, 1, 15008. [Google Scholar] [CrossRef] [PubMed]
  47. Sadick, M.; Müller-Wille, R.; Wildgruber, M.; Wohlgemuth, W.A. Vascular Anomalies (Part I): Classification and Diagnostics of Vascular Anomalies. Rofo Fortschr. Auf Dem Geb. der Rontgenstrahlen der Nukl. 2018, 190, 825–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Darden, J.; Payne, L.B.; Zhao, H.; Chappell, J.C. Excess vascular endothelial growth factor-A disrupts pericyte recruitment during blood vessel formation. Angiogenesis 2019, 22, 167–183. [Google Scholar] [CrossRef] [PubMed]
  49. Wei, X.; Chen, Y.; Jiang, X.; Peng, M.; Liu, Y.; Mo, Y.; Ren, D.; Hua, Y.; Yu, B.; Zhou, Y.; et al. Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments. Mol. Cancer 2021, 20, 7. [Google Scholar] [CrossRef] [PubMed]
  50. Qiao, K.; Liu, Y.; Xu, Z.; Zhang, H.; Zhang, H.; Zhang, C.; Chang, Z.; Lu, X.; Li, Z.; Luo, C.; et al. RNA m6A methylation promotes the formation of vasculogenic mimicry in hepatocellular carcinoma via Hippo pathway. Angiogenesis 2021, 24, 83–96. [Google Scholar] [CrossRef] [PubMed]
  51. Suo, L.; Liu, C.; Zhang, Q.Y.; Yao, M.D.; Ma, Y.; Yao, J.; Jiang, Q.; Yan, B. METTL3-mediated N (6)-methyladenosine modification governs pericyte dysfunction during diabetes-induced retinal vascular complication. Theranostics 2022, 12, 277–289. [Google Scholar] [CrossRef] [PubMed]
  52. Li, M.; Deng, L.; Xu, G. METTL14 promotes glomerular endothelial cell injury and diabetic nephropathy via m6A modification of α-klotho. Mol. Med. 2021, 27, 106. [Google Scholar] [CrossRef]
  53. Tu, J.; Li, Y.; Hu, Z.; Chen, Z. Radiosurgery inhibition of the Notch signaling pathway in a rat model of arteriovenous malformations. J. Neurosurg. 2014, 120, 1385–1396. [Google Scholar] [CrossRef] [Green Version]
  54. ZhuGe, Q.; Zhong, M.; Zheng, W.; Yang, G.Y.; Mao, X.; Xie, L.; Chen, G.; Chen, Y.; Lawton, M.T.; Young, W.L.; et al. Notch-1 signalling is activated in brain arteriovenous malformations in humans. Brain 2009, 132, 3231–3241. [Google Scholar] [CrossRef] [Green Version]
  55. Wang, L.J.; Xue, Y.; Huo, R.; Yan, Z.; Xu, H.; Li, H.; Wang, J.; Zhang, Q.; Cao, Y.; Zhao, J.Z. N6-methyladenosine methyltransferase METTL3 affects the phenotype of cerebral arteriovenous malformation via modulating Notch signaling pathway. J. Biomed. Sci. 2020, 27, 62. [Google Scholar] [CrossRef]
  56. Wang, L.J.; Xue, Y.; Li, H.; Huo, R.; Yan, Z.; Wang, J.; Xu, H.; Wang, J.; Cao, Y.; Zhao, J.Z. Wilms’ tumour 1-associating protein inhibits endothelial cell angiogenesis by m6A-dependent epigenetic silencing of desmoplakin in brain arteriovenous malformation. J. Cell Mol. Med. 2020, 24, 4981–4991. [Google Scholar] [CrossRef] [Green Version]
  57. Karar, J.; Maity, A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 2011, 4, 51. [Google Scholar] [CrossRef] [Green Version]
  58. Parial, R.; Li, H.; Li, J.; Archacki, S.; Yang, Z.; Wang, I.Z.; Chen, Q.; Xu, C.; Wang, Q.K. Role of epigenetic m(6) A RNA methylation in vascular development: Mettl3 regulates vascular development through PHLPP2/mTOR-AKT signaling. FASEB J. 2021, 35, e21465. [Google Scholar] [CrossRef]
  59. 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. Int. J. Mol. Sci. 2019, 20, 551. [Google Scholar] [CrossRef] [Green Version]
  60. Chen, X.; Hua, W.; Huang, X.; Chen, Y.; Zhang, J.; Li, G. Regulatory Role of RNA N(6)-Methyladenosine Modification in Bone Biology and Osteoporosis. Front. Endocrinol. 2019, 10, 911. [Google Scholar] [CrossRef]
  61. Jiang, W.; Zhu, P.; Huang, F.; Zhao, Z.; Zhang, T.; An, X.; Liao, F.; Guo, L.; Liu, Y.; Zhou, N.; et al. The RNA Methyltransferase METTL3 Promotes Endothelial Progenitor Cell Angiogenesis in Mandibular Distraction Osteogenesis via the PI3K/AKT Pathway. Front. Cell Dev. Biol. 2021, 9, 720925. [Google Scholar] [CrossRef] [PubMed]
  62. Rizzoni, D.; De Ciuceis, C.; Szczepaniak, P.; Paradis, P.; Schiffrin, E.L.; Guzik, T.J. Immune System and Microvascular Remodeling in Humans. Hypertension 2022, 79, 691–705. [Google Scholar] [CrossRef]
  63. Koh, M.Y.; Powis, G. Passing the baton: The HIF switch. Trends Biochem. Sci. 2012, 37, 364–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Thompson, A.A.R.; Lawrie, A. Targeting Vascular Remodeling to Treat Pulmonary Arterial Hypertension. Trends Mol. Med. 2017, 23, 31–45. [Google Scholar] [CrossRef]
  65. Zhou, X.L.; Huang, F.J.; Li, Y.; Huang, H.; Wu, Q.C. SEDT2/METTL14-mediated m6A methylation awakening contributes to hypoxia-induced pulmonary arterial hypertension in mice. Aging 2021, 13, 7538–7548. [Google Scholar] [CrossRef]
  66. Mourani, P.M.; Garl, P.J.; Wenzlau, J.M.; Carpenter, T.C.; Stenmark, K.R.; Weiser-Evans, M.C. Unique, highly proliferative growth phenotype expressed by embryonic and neointimal smooth muscle cells is driven by constitutive Akt, mTOR, and p70S6K signaling and is actively repressed by PTEN. Circulation 2004, 109, 1299–1306. [Google Scholar] [CrossRef] [PubMed]
  67. Qin, Y.; Qiao, Y.; Li, L.; Luo, E.; Wang, D.; Yao, Y.; Tang, C.; Yan, G. The m(6)A methyltransferase METTL3 promotes hypoxic pulmonary arterial hypertension. Life Sci. 2021, 274, 119366. [Google Scholar] [CrossRef]
  68. Chien, C.S.; Li, J.Y.; Chien, Y.; Wang, M.L.; Yarmishyn, A.A.; Tsai, P.H.; Juan, C.C.; Nguyen, P.; Cheng, H.M.; Huo, T.I.; et al. METTL3-dependent N(6)-methyladenosine RNA modification mediates the atherogenic inflammatory cascades in vascular endothelium. Proc. Natl. Acad. Sci. USA 2021, 118, e2025070118. [Google Scholar] [CrossRef]
  69. Jian, D.; Wang, Y.; Jian, L.; Tang, H.; Rao, L.; Chen, K.; Jia, Z.; Zhang, W.; Liu, Y.; Chen, X.; et al. METTL14 aggravates endothelial inflammation and atherosclerosis by increasing FOXO1 N6-methyladeosine modifications. Theranostics 2020, 10, 8939–8956. [Google Scholar] [CrossRef] [PubMed]
  70. Wu, J.; Frazier, K.; Zhang, J.; Gan, Z.; Wang, T.; Zhong, X. Emerging role of m(6) A RNA methylation in nutritional physiology and metabolism. Obes. Rev. 2020, 21, e12942. [Google Scholar] [CrossRef]
  71. Zhang, B.F.; Wu, Z.H.; Deng, J.; Jin, H.J.; Chen, W.B.; Zhang, S.; Liu, X.J.; Wang, W.T.; Zheng, X.T. M(6)A methylation-mediated elevation of SM22α inhibits the proliferation and migration of vascular smooth muscle cells and ameliorates intimal hyperplasia in type 2 diabetes mellitus. Biol. Chem. 2021, 403, 317–329. [Google Scholar] [CrossRef] [PubMed]
  72. Yuan, J.; Liu, Y.; Zhou, L.; Xue, Y.; Lu, Z.; Gan, J. YTHDC2-Mediated circYTHDC2 N6-Methyladenosine Modification Promotes Vascular Smooth Muscle Cells Dysfunction Through Inhibiting Ten-Eleven Translocation 2. Front. Cardiovasc. Med. 2021, 8, 686293. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, R.; Jin, Y.; Tang, W.H.; Qin, L.; Zhang, X.; Tellides, G.; Hwa, J.; Yu, J.; Martin, K.A. Ten-eleven translocation-2 (TET2) is a master regulator of smooth muscle cell plasticity. Circulation 2013, 128, 2047–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Messner, B.; Bernhard, D. Smoking and cardiovascular disease: Mechanisms of endothelial dysfunction and early atherogenesis. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 509–515. [Google Scholar] [CrossRef] [Green Version]
  75. Gong, C.; Fan, Y.; Liu, J. METTL14 mediated m6A modification to LncRNA ZFAS1/ RAB22A: A novel therapeutic target for atherosclerosis. Int. J. Cardiol. 2021, 328, 177. [Google Scholar] [CrossRef]
  76. Mo, C.; Yang, M.; Han, X.; Li, J.; Gao, G.; Tai, H.; Huang, N.; Xiao, H. Fat mass and obesity-associated protein attenuates lipid accumulation in macrophage foam cells and alleviates atherosclerosis in apolipoprotein E-deficient mice. J. Hypertens. 2017, 35, 810–821. [Google Scholar] [CrossRef] [PubMed]
  77. Zhang, B.Y.; Han, L.; Tang, Y.F.; Zhang, G.X.; Fan, X.L.; Zhang, J.J.; Xue, Q.; Xu, Z.Y. METTL14 regulates M6A methylation-modified primary miR-19a to promote cardiovascular endothelial cell proliferation and invasion. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7015–7023. [Google Scholar] [CrossRef] [PubMed]
  78. Lee, B.; Lee, S.; Shim, J. YTHDF2 Suppresses Notch Signaling through Post-transcriptional Regulation on Notch1. Int. J. Biol. Sci. 2021, 17, 3776–3785. [Google Scholar] [CrossRef] [PubMed]
  79. Wang, Q.; Chen, C.; Ding, Q.; Zhao, Y.; Wang, Z.; Chen, J.; Jiang, Z.; Zhang, Y.; Xu, G.; Zhang, J.; et al. METTL3-mediated m(6)A modification of HDGF mRNA promotes gastric cancer progression and has prognostic significance. Gut 2020, 69, 1193–1205. [Google Scholar] [CrossRef] [PubMed]
  80. Pollyea, D.A.; Pratz, K.; Letai, A.; Jonas, B.A.; Wei, A.H.; Pullarkat, V.; Konopleva, M.; Thirman, M.J.; Arellano, M.; Becker, P.S.; et al. Venetoclax with azacitidine or decitabine in patients with newly diagnosed acute myeloid leukemia: Long term follow-up from a phase 1b study. Am. J. Hematol. 2021, 96, 208–217. [Google Scholar] [CrossRef]
  81. Scott, L.J. Azacitidine: A Review in Myelodysplastic Syndromes and Acute Myeloid Leukaemia. Drugs 2016, 76, 889–900. [Google Scholar] [CrossRef] [PubMed]
  82. Tang, B.; Yang, Y.; Kang, M.; Wang, Y.; Wang, Y.; Bi, Y.; He, S.; Shimamoto, F. m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol. Cancer 2020, 19, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Taketo, K.; Konno, M.; Asai, A.; Koseki, J.; Toratani, M.; Satoh, T.; Doki, Y.; Mori, M.; Ishii, H.; Ogawa, K. The epitranscriptome m6A writer METTL3 promotes chemo- and radioresistance in pancreatic cancer cells. Int. J. Oncol. 2018, 52, 621–629. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathogenic factors and pathological processes of MVD. Pathogenic factors, such as hypoxia, inflammation, and metabolic dysregulation, contribute to MVD. Pathological processes of MVD include neovascularization, microvascular malformation, and microvascular remodeling. Both neovascularization and microvascular malformation can be structurally and functionally remodeled in response to physical and chemical stimuli.
Figure 1. Pathogenic factors and pathological processes of MVD. Pathogenic factors, such as hypoxia, inflammation, and metabolic dysregulation, contribute to MVD. Pathological processes of MVD include neovascularization, microvascular malformation, and microvascular remodeling. Both neovascularization and microvascular malformation can be structurally and functionally remodeled in response to physical and chemical stimuli.
Cells 11 03193 g001
Figure 2. The process and molecular functions of RNA m6A methylation. M6A modification is dynamically installed by writers (METTL3, METTL4, METTL14, WTAP, RBM15, ZC3H13) and removed by erasers (FTO and ALKBH5). M6A sites are recognized by readers. hnRNPs and YTHDC1 are nuclear readers. hnRNPC binds to structurally altered RNAs and mediates pre-mRNA processing. hnRNPA2B1 regulates RNA splicing and primary miRNA processing. YTHDC1 mediates alternative splicing and facilitates mRNA export to cytoplasm. YTHDF1/2/3, YTHDC2, and IGF2BP1/2/3 are cytoplasmic-distributed. YTHDF1 enables mRNA translation by recruiting eukaryotic initiation factor 3, whereas YTHDF2 transports target mRNAs to the cytoplasmic processing body and promotes their degradation. YTHDF3 is a modulator of YTHDF1 and YTHDF2, which can both enhance and suppress their effects. IGF2BPs enhance stability of target RNA transcripts.
Figure 2. The process and molecular functions of RNA m6A methylation. M6A modification is dynamically installed by writers (METTL3, METTL4, METTL14, WTAP, RBM15, ZC3H13) and removed by erasers (FTO and ALKBH5). M6A sites are recognized by readers. hnRNPs and YTHDC1 are nuclear readers. hnRNPC binds to structurally altered RNAs and mediates pre-mRNA processing. hnRNPA2B1 regulates RNA splicing and primary miRNA processing. YTHDC1 mediates alternative splicing and facilitates mRNA export to cytoplasm. YTHDF1/2/3, YTHDC2, and IGF2BP1/2/3 are cytoplasmic-distributed. YTHDF1 enables mRNA translation by recruiting eukaryotic initiation factor 3, whereas YTHDF2 transports target mRNAs to the cytoplasmic processing body and promotes their degradation. YTHDF3 is a modulator of YTHDF1 and YTHDF2, which can both enhance and suppress their effects. IGF2BPs enhance stability of target RNA transcripts.
Cells 11 03193 g002
Table 1. Molecular mechanisms of m6A modification in pathological neovascularization.
Table 1. Molecular mechanisms of m6A modification in pathological neovascularization.
Pathological
Process
DiseaseM6A RegulatorsModel SystemMechanismReference
Human TissueAnimal ModelCell Line
hypoxialung cancerYTHDF2↑ promote HIF-1 expression[21]
stomach cancerIGF2BP3↑ promote HIF-1 expression[22]
breast cancerMETTL14
/ALKBH5↑
increase TGFβ1 expression[23]
HCCYTHDF2↓stabilize IL-11 and SERPINE2 mRNA[24]
METTL3↓increase PDGF and VEGF expression[25]
oxygen-induced retinopathyMETTL3↑ activate the Wnt pathway[26]
inflammationHCCYTHDF2↓stabilize IL-11 and SERPINE2 mRNA[24]
corneal neovascularizationFTO↑ increase FAK expression[27]
METTL3↑ activate the Wnt signaling pathway[26]
diabetic retinopathyYTHDF2↓ activate FAK/PI3K/AKT pathway[28]
othersbreast cancerYTHDF3↑enhance translation of VEGF[29]
lung cancerMETTL3↑increase VEGFA expression[30]
intrahepatic cholangiocarcinomaFTO↓increase CCL19 expression[31]
colorectal cancer/melanomaALKBH5↑promote VEGF expression[32]
Abbreviations: HCC, human hepatocellular carcinoma; IL-11, interleukin-11; SERPINE2, serpin family E member 2; FAK, focal adhesion kinase; VEGF, vascular endothelial growth factor; TGF-β, transforming growth factor β; CCL19, C-C motif chemokine ligand 19; ↑, upregulation; ↓, downregulation; √, the experimental model was included.
Table 2. Molecular mechanisms of m6A modification in microvascular malformation.
Table 2. Molecular mechanisms of m6A modification in microvascular malformation.
Pathological
Process
DiseaseM6A RegulatorsModel SystemMechanismReference
Human TissueAnimal ModelCell Line
hypoxiaHCC YTHDF2↓stabilize IL-11 and SERPINE2 mRNA[24]
METTL3↑activate Hippo pathway[50]
inflammationdiabetic nephropathyMETTL14↑decrease α-klotho expression[52]
diabetic retinopathyMETTL3↑ suppress PKC/FAT4/PDGFRA pathway[51]
othersarteriovenous malformationMETTL3↓ activate the Notch pathway[55]
WTAP↓ block the Wnt pathway[56]
model system (endothelial cells)METTL3↓ inhibit the PI3K/AKT pathway[58]
model system (bone mesenchymal stem cells)METTL3↓ inhibit the PI3K/AKT pathway[59,60]
Abbreviations: ↑, upregulation; ↓, downregulation; √, the experimental model was included.
Table 3. Molecular mechanisms of m6A modification in microvascular remodeling.
Table 3. Molecular mechanisms of m6A modification in microvascular remodeling.
Pathological ProcessDiseaseM6A RegulatorsModel SystemMechanismReference
Human TissueAnimal ModelCell Line
hypoxiaHCCYTHDF2↓stabilize IL-11 and SERPINE2 mRNA[24]
pulmonary arterial hypertensionMETTL3↑ degrade PETN mRNAs[67]
METTL14↑ cooperate with SETD2[65]
inflammationatherosclerosisMETTL3↑ increase NLRP1 and decrease KLF4 expression[68]
METTL14↑ increase VCAM-A and ICAM-1 expression[69,77]
metabolismtype 2 diabetes mellitusFTO↑ destabilize SM22α mRNAs [71]
YTHDC2↑ inhibit TET2 expression[72]
atherosclerosisFTO↑reduce CD36 and PPARγ level[76]
Abbreviations: NRLP1, NLR family pyrin domain containing 1; KLF4, KLF transcription factor 4; ICAM-1, intercellular adhesion molecule 1; VCAM-1, vascular cell adhesion molecule 1; SM22α, smooth muscle 22 alpha; PTEN, phosphatase and tensin homolog; ABCA1, ATP-binding cassette transporter A1; CD36, CD36 molecule; ↑, upregulation; ↓, downregulation; √, the experimental model was included.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, Y.-R.; Ji, J.-D.; Wang, J.-N.; Wang, Y.; Zhu, H.-J.; Sun, R.-X.; Liu, Q.-H.; Chen, X. The Role of N6-Methyladenosine Modification in Microvascular Dysfunction. Cells 2022, 11, 3193. https://doi.org/10.3390/cells11203193

AMA Style

Zhang Y-R, Ji J-D, Wang J-N, Wang Y, Zhu H-J, Sun R-X, Liu Q-H, Chen X. The Role of N6-Methyladenosine Modification in Microvascular Dysfunction. Cells. 2022; 11(20):3193. https://doi.org/10.3390/cells11203193

Chicago/Turabian Style

Zhang, Ye-Ran, Jiang-Dong Ji, Jia-Nan Wang, Ying Wang, Hong-Jing Zhu, Ru-Xu Sun, Qing-Huai Liu, and Xue Chen. 2022. "The Role of N6-Methyladenosine Modification in Microvascular Dysfunction" Cells 11, no. 20: 3193. https://doi.org/10.3390/cells11203193

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

Article Metrics

Back to TopTop