Abstract

RNA modifications include not only methylation modifications, such as m6A, but also acetylation modifications, which constitute a complex interaction involving “writers,” “readers,” and “erasers” that play crucial roles in growth, genetics, and disease. N4-acetylcytidine (ac4C) is an ancient and highly conserved RNA modification that plays a profound role in the pathogenesis of a wide range of diseases. This review provides insights into the functional impact of ac4C modifications in disease and introduces new perspectives for disease treatment. These studies provide important insights into the biological functions of post-transcriptional RNA modifications and their potential roles in disease mechanisms, offering new perspectives and strategies for disease treatment.

Introduction

Similar to DNA and proteins, RNA undergoes chemical modifications catalyzed by specific enzymes. As early as the 1950s, it was discovered that RNA could undergo modifications on the canonical ACUG bases. To date, over 170 RNA modifications have been identified [1], with N6-methyladenosine (m6A) methylation being the most prevalent and proven to play a crucial role in various advanced biological processes, such as RNA splicing, maturation, transport, and translation [2].

With the rapid development of high-throughput sequencing technologies, research on RNA modifications has become increasingly profound, and N4-acetylcytidine (ac4C) acetylation modification is gradually being elucidated. Although studies on understanding the “writer,” “reader,” and “eraser” aspects of its function have not reached the same depth as m6A modification [3–5], scientists have revealed the mechanism of action of ac4C acetyltransferase and found that this enzyme is closely related to the occurrence and development of various diseases [6, 7]. ac4C acetylation modification is mainly catalyzed by NAT10 acetyltransferase on the N4 position of cytidine in RNA and is widely present in various RNA molecules, including mRNA, tRNA, and rRNA. Among them, NAT10, as the only ac4C acetyltransferase in human cells, can precisely regulate the stability and translation of RNA by catalyzing the acetylation of rRNA [8], tRNA [9], and mRNA [10], which advances our understanding of the role of RNA acetylation modification in cellular physiology and pathology.

Numerous studies have substantiated the association between ac4C modification and the onset of cancer [11–14]. Additionally, the presence of ac4C modification has been implicated in the physiological mechanisms of various diseases, playing a pivotal role in disease treatment. In this concise review, we provide a detailed exploration of the molecular-level regulatory functions of ac4C acetylation and its roles in disease, with a prospective outlook on the involvement of NAT10 in disease therapy.

Modulatory function of ac4C RNA modification in molecular processes

The ac4C modification on mRNA exerts differential effects on gene expression and translation depending on its position. Within the coding sequence (CDS), the ac4C modification enhances stability through the formation of hydrogen bonds with internal N4-acetyl groups. Additionally, it interacts with homologous tRNA, thereby increasing translation rates [10, 15]. Conversely, the ac4C modification present in the 5’ untranslated region (5’UTR) may competitively inhibit annotated start codons, impeding protein synthesis. Particularly near strong AUG initiation codons, ac4C modification in the 5’UTR may also inhibit translation [16]. Moreover, the inhibitory effect of ac4C modification in the 5’UTR on translation may counteract the promoting effect observed in the CDS. Therefore, site-specificity must be considered in the development of therapeutic drugs.

In addition, ac4C modification plays a crucial role in ribosome synthesis. Depletion of ribosomal RNA cytidine acetyltransferase 1 (RRA1), homologous to NAT10, leads to significant accumulation of the 23S precursor of 18S rRNA, impacting ribosome synthesis [17]. In summary, ac4C acetylation on mRNA and rRNA collectively promotes gene expression (Fig. 1).

Functions of ac4C modifications in RNAs. The ac4C present in the mRNA CDS enhances translation rate through intramolecular hydrogen bonding and interaction with homologous tRNAs. ac4C near the 5’UTP AUG strong promoter represses translation, and ac4C on rRNAs promotes the assembly of ribosomes and facilitates translation.
Figure 1

Functions of ac4C modifications in RNAs. The ac4C present in the mRNA CDS enhances translation rate through intramolecular hydrogen bonding and interaction with homologous tRNAs. ac4C near the 5’UTP AUG strong promoter represses translation, and ac4C on rRNAs promotes the assembly of ribosomes and facilitates translation.

When studying ac4C acetylation modification on RNA, several detection methods have been discovered. Liquid chromatography-mass spectrometry (LC-MS) and high-performance LC-MS (HPLC-MS) are among the qualitative and quantitative methods for detecting ac4C modification in RNA, capable of identifying ac4C modifications within microorganisms and human cells. However, due to their inability to amplify signals, these techniques have limited sensitivity, posing challenges in detecting low-abundance ac4C modifications [18].

RNA modification immunoprecipitation-sequencing methods (such as RNA IP-seq) amplify signals. However, this method has a higher false positive rate and cannot accurately detect ac4C modifications at the nucleotide level or quantify their abundance at presumed modification sites[18]. In 2017, Sinclair et al. [19] developed specific affinity reagents for analyzing the expression spectrum of ac4C. Arango et al. [10] employed the acRIP-seq method, utilizing ac4C-binding protein antibodies to enrich ac4C mRNA, thereby mapping the locations of ac4C in the human transcriptome.

Additionally, Yan et al. [20] proposed a metabolic sequencing method, FAMseq, which does not require antibodies and utilizes fluoride ions to accurately obtain the acetylation profile of the entire transcriptome RNA. There are also emerging detection tools, such as iRNA-ac4C and LSA-ac4C, although these technologies currently cannot precisely determine the specific positions of ac4C [21–23].

The role of ac4C acetylation modification in diseases

In recent years, ac4C acetylation modification has been implicated in the pathogenesis of various diseases. In neuroinflammation, the nucleoside metabolite ac4C induces the expression of alarmin high mobility group box-1 (HMGB1 protein), activating nuclear factor κB (NF-κB). This, in turn, upregulates NOD-like receptor thermal protein domain associated protein 3 (NLRP3) gene expression, maintaining the activation of NLRP3 inflammasomes and activating the immune function of microglial cells [24]. In Alzheimer’s disease (AD), ac4C modification on lncRNA is also speculated to be associated with AD [25], although the specific regulatory mechanisms require further in-depth investigation. Additionally, the co-increase of the nucleoside metabolite ac4C and adenine may be associated with hypertension [26]. This is hypothesized to be due to the activation of inflammatory cells and factors by the binding of ac4C and adenine, stimulating platelet and leukocyte generation, leading to elevated blood pressure. In sepsis, inhibition of NAT10 leads to restricted expression of UNC-52-like kinase 1 (ULK1), activating the STING-IRF3 signal and ultimately causing an increase in NLRP3 inflammasomes, leading to neutrophil apoptosis and septicemia [27].Furthermore, in systemic lupus erythematosus (SLE), NAT10, by mediating ac4C acetylation of certain SLE-related gene mRNAs, enhances their stability, ultimately activating immune cells and inflammatory responses [28].

These studies provide diverse perspectives for understanding the intrinsic mechanisms of diseases and offer new insights for overcoming them. Subsequently, we summarize the latest research on ac4C acetylation modification in the pathogenesis of various diseases, such as osteoporosis and periodontal diseases, to highlight its closely associated characteristics with diseases and explore its broad prospects as a potential therapeutic target (Fig. 2, Table 1).

The role of ac4C modifications in disease pathogenesis.
Figure 2

The role of ac4C modifications in disease pathogenesis.

Table 1

The role of ac4C acetylation modification in diseases

DiseaseEnzymeTargetDescriptionRef
Chronic neurogenic diseasesNAT10A2ARac4C activates BV2 microglia via A2AR signaling and maintains NLRP3 inflammatory vesicles[24]
HypertensionNAT10N.A.ac4C and adenine together promote NLRC4 expression, activating platelets and leukocytes to cause hypertension[26]
SepsisNAT10ULK1ac4C modification of ULK1 mRNA attenuates sepsis by increasing ULK1 transcription[27]
OsteoporosisNAT10RUNX2ac4C modification of RUNX2 mRNA increases RUNX2 expression to promote osteoblast differentiation[31]
PeriodontitisNAT10NOX2ac4C modification of NOX2 mRNA in macrophages enhances NOX2 expression to cause inflammatory responses[33]
Myocardial infarctionNAT10TfecHAAPIR promotes Tfec gene expression, while Tfec promotes Bik accumulation, leading to cardiomyocyte death.[38]
Heart failureNAT10CD47/ROCK2Upregulation of CD47 and ROCK2 mRNA ac4C modification promotes myocardial remodeling[39]
Pulmonary fibrosisNAT10TGFB1ac4C can accelerate lung fibrosis by increasing TGFB1 mRNA stability[41]
Corneal injuryNAT10mRNAsMSCs enhance mRNA ac4C acetylation and activate the ETV4/JUN/CCND2 signaling axis to promote corneal injury recovery[43]
Neuropathic painNAT10Syt9Activation of USF1 by nerve injury upregulates ac4C in Syt9 mRNA to increase SYT9 protein to trigger neuropathic pain[6]
Neuropathic painNAT10VegfaHNRNPK binding to Vegfa mRNA increases ac4C modification and upregulates VEGFA-induced neuropathic pain[45]
Premature ovarian failureNAT10P16ac4C acetylation increases P16 mRNA stability and promotes premature ovarian failure[7]
Intellectual disabilityTHUMPD1small RNAs/tRNA-SerCGAAberrant THUMPD1 expression leads to mental retardation syndrome caused by deletion of the ac4C modification in small RNAs and individually purified tRNAs-Ser-CGA[50]
Systemic lupus erythematosusNAT10CD4+T cell mRNAsSignificant differences in mRNA ac4C modification abundance of certain SLE-related genes in CD4+ T cells from SLE patients[28]
DiseaseEnzymeTargetDescriptionRef
Chronic neurogenic diseasesNAT10A2ARac4C activates BV2 microglia via A2AR signaling and maintains NLRP3 inflammatory vesicles[24]
HypertensionNAT10N.A.ac4C and adenine together promote NLRC4 expression, activating platelets and leukocytes to cause hypertension[26]
SepsisNAT10ULK1ac4C modification of ULK1 mRNA attenuates sepsis by increasing ULK1 transcription[27]
OsteoporosisNAT10RUNX2ac4C modification of RUNX2 mRNA increases RUNX2 expression to promote osteoblast differentiation[31]
PeriodontitisNAT10NOX2ac4C modification of NOX2 mRNA in macrophages enhances NOX2 expression to cause inflammatory responses[33]
Myocardial infarctionNAT10TfecHAAPIR promotes Tfec gene expression, while Tfec promotes Bik accumulation, leading to cardiomyocyte death.[38]
Heart failureNAT10CD47/ROCK2Upregulation of CD47 and ROCK2 mRNA ac4C modification promotes myocardial remodeling[39]
Pulmonary fibrosisNAT10TGFB1ac4C can accelerate lung fibrosis by increasing TGFB1 mRNA stability[41]
Corneal injuryNAT10mRNAsMSCs enhance mRNA ac4C acetylation and activate the ETV4/JUN/CCND2 signaling axis to promote corneal injury recovery[43]
Neuropathic painNAT10Syt9Activation of USF1 by nerve injury upregulates ac4C in Syt9 mRNA to increase SYT9 protein to trigger neuropathic pain[6]
Neuropathic painNAT10VegfaHNRNPK binding to Vegfa mRNA increases ac4C modification and upregulates VEGFA-induced neuropathic pain[45]
Premature ovarian failureNAT10P16ac4C acetylation increases P16 mRNA stability and promotes premature ovarian failure[7]
Intellectual disabilityTHUMPD1small RNAs/tRNA-SerCGAAberrant THUMPD1 expression leads to mental retardation syndrome caused by deletion of the ac4C modification in small RNAs and individually purified tRNAs-Ser-CGA[50]
Systemic lupus erythematosusNAT10CD4+T cell mRNAsSignificant differences in mRNA ac4C modification abundance of certain SLE-related genes in CD4+ T cells from SLE patients[28]

NAT10, N-acetyltransferase 10; ac4C, N4-acetylcytidine; tRNA, transfer RNA; mRNA, messenger RNA; A2AR, adenosine A2A receptor; NLRP3, NOD-like receptor thermal protein domain associated protein 3; NLRC4, NOD-like receptor family CARD-containing 4 protein; ULK1, UNC-51-like kinase 1; RUNX2, runt-related transcription factor 2; NOX2, the NADPH oxidase 2; Tfec, transcription factor EC; HAAPIR, heart-apoptosis-associated piRNA; Bik, BCL2-interacting killer; CD47, integrin-associated protein, IAP; ROCK2, rho-associated coiled-coil containing protein kinase 2; TGFB1, transforming growth factor beta 1; MSCs, marrow-derived mesenchymal stem cells; ETV4, ets translocation variant 4; CCND2, cyclin D2; Syt9, synaptotagmin 9; USF1, upstream transcription factor 1; Vegfa, vascular endothelial growth factor A; HNRNPK, heterogeneous nuclear ribonucleoprotein K, hnRNPK; P16, cyclin-dependent kinase inhibitor, MTS; THUMPD1, THUMP domain containing 1; SLE, systemic lupus erythematosus; N.A., not available.

Table 1

The role of ac4C acetylation modification in diseases

DiseaseEnzymeTargetDescriptionRef
Chronic neurogenic diseasesNAT10A2ARac4C activates BV2 microglia via A2AR signaling and maintains NLRP3 inflammatory vesicles[24]
HypertensionNAT10N.A.ac4C and adenine together promote NLRC4 expression, activating platelets and leukocytes to cause hypertension[26]
SepsisNAT10ULK1ac4C modification of ULK1 mRNA attenuates sepsis by increasing ULK1 transcription[27]
OsteoporosisNAT10RUNX2ac4C modification of RUNX2 mRNA increases RUNX2 expression to promote osteoblast differentiation[31]
PeriodontitisNAT10NOX2ac4C modification of NOX2 mRNA in macrophages enhances NOX2 expression to cause inflammatory responses[33]
Myocardial infarctionNAT10TfecHAAPIR promotes Tfec gene expression, while Tfec promotes Bik accumulation, leading to cardiomyocyte death.[38]
Heart failureNAT10CD47/ROCK2Upregulation of CD47 and ROCK2 mRNA ac4C modification promotes myocardial remodeling[39]
Pulmonary fibrosisNAT10TGFB1ac4C can accelerate lung fibrosis by increasing TGFB1 mRNA stability[41]
Corneal injuryNAT10mRNAsMSCs enhance mRNA ac4C acetylation and activate the ETV4/JUN/CCND2 signaling axis to promote corneal injury recovery[43]
Neuropathic painNAT10Syt9Activation of USF1 by nerve injury upregulates ac4C in Syt9 mRNA to increase SYT9 protein to trigger neuropathic pain[6]
Neuropathic painNAT10VegfaHNRNPK binding to Vegfa mRNA increases ac4C modification and upregulates VEGFA-induced neuropathic pain[45]
Premature ovarian failureNAT10P16ac4C acetylation increases P16 mRNA stability and promotes premature ovarian failure[7]
Intellectual disabilityTHUMPD1small RNAs/tRNA-SerCGAAberrant THUMPD1 expression leads to mental retardation syndrome caused by deletion of the ac4C modification in small RNAs and individually purified tRNAs-Ser-CGA[50]
Systemic lupus erythematosusNAT10CD4+T cell mRNAsSignificant differences in mRNA ac4C modification abundance of certain SLE-related genes in CD4+ T cells from SLE patients[28]
DiseaseEnzymeTargetDescriptionRef
Chronic neurogenic diseasesNAT10A2ARac4C activates BV2 microglia via A2AR signaling and maintains NLRP3 inflammatory vesicles[24]
HypertensionNAT10N.A.ac4C and adenine together promote NLRC4 expression, activating platelets and leukocytes to cause hypertension[26]
SepsisNAT10ULK1ac4C modification of ULK1 mRNA attenuates sepsis by increasing ULK1 transcription[27]
OsteoporosisNAT10RUNX2ac4C modification of RUNX2 mRNA increases RUNX2 expression to promote osteoblast differentiation[31]
PeriodontitisNAT10NOX2ac4C modification of NOX2 mRNA in macrophages enhances NOX2 expression to cause inflammatory responses[33]
Myocardial infarctionNAT10TfecHAAPIR promotes Tfec gene expression, while Tfec promotes Bik accumulation, leading to cardiomyocyte death.[38]
Heart failureNAT10CD47/ROCK2Upregulation of CD47 and ROCK2 mRNA ac4C modification promotes myocardial remodeling[39]
Pulmonary fibrosisNAT10TGFB1ac4C can accelerate lung fibrosis by increasing TGFB1 mRNA stability[41]
Corneal injuryNAT10mRNAsMSCs enhance mRNA ac4C acetylation and activate the ETV4/JUN/CCND2 signaling axis to promote corneal injury recovery[43]
Neuropathic painNAT10Syt9Activation of USF1 by nerve injury upregulates ac4C in Syt9 mRNA to increase SYT9 protein to trigger neuropathic pain[6]
Neuropathic painNAT10VegfaHNRNPK binding to Vegfa mRNA increases ac4C modification and upregulates VEGFA-induced neuropathic pain[45]
Premature ovarian failureNAT10P16ac4C acetylation increases P16 mRNA stability and promotes premature ovarian failure[7]
Intellectual disabilityTHUMPD1small RNAs/tRNA-SerCGAAberrant THUMPD1 expression leads to mental retardation syndrome caused by deletion of the ac4C modification in small RNAs and individually purified tRNAs-Ser-CGA[50]
Systemic lupus erythematosusNAT10CD4+T cell mRNAsSignificant differences in mRNA ac4C modification abundance of certain SLE-related genes in CD4+ T cells from SLE patients[28]

NAT10, N-acetyltransferase 10; ac4C, N4-acetylcytidine; tRNA, transfer RNA; mRNA, messenger RNA; A2AR, adenosine A2A receptor; NLRP3, NOD-like receptor thermal protein domain associated protein 3; NLRC4, NOD-like receptor family CARD-containing 4 protein; ULK1, UNC-51-like kinase 1; RUNX2, runt-related transcription factor 2; NOX2, the NADPH oxidase 2; Tfec, transcription factor EC; HAAPIR, heart-apoptosis-associated piRNA; Bik, BCL2-interacting killer; CD47, integrin-associated protein, IAP; ROCK2, rho-associated coiled-coil containing protein kinase 2; TGFB1, transforming growth factor beta 1; MSCs, marrow-derived mesenchymal stem cells; ETV4, ets translocation variant 4; CCND2, cyclin D2; Syt9, synaptotagmin 9; USF1, upstream transcription factor 1; Vegfa, vascular endothelial growth factor A; HNRNPK, heterogeneous nuclear ribonucleoprotein K, hnRNPK; P16, cyclin-dependent kinase inhibitor, MTS; THUMPD1, THUMP domain containing 1; SLE, systemic lupus erythematosus; N.A., not available.

Osteoporosis

Osteoporosis, a common condition among middle-aged and elderly individuals, is notably exacerbated in patients who have undergone oophorectomy due to reduced estrogen secretion, which accelerates bone resorption and increases the likelihood of developing osteoporosis [29]. Previous studies have confirmed a close association between the occurrence of osteoporosis and epigenetic modifications [30]. NAT10 has been identified as a facilitator of acetylation on Runt-related transcription factor 2 (RUNX2) mRNA, thereby increasing its expression by extending its half-life. This promotion of RUNX2 acetylation by NAT10 stimulates the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts [31]. Notably, overexpression of NAT10 in mice subjected to bilateral oophorectomy has been found to reverse bone loss, mitigating the detrimental effects on the organism [31]. Consequently, NAT10 emerges as a potential direction for the prevention and treatment of osteoporosis.

Periodontal diseases

Periodontitis, a chronic inflammation caused by bacterial invasion of periodontal tissues within dental plaque, stands as a leading cause of adult tooth mobility and loss [32]. In the context of periodontitis, NAT10 mediates acetylation on the mRNA of NADPH oxidase 2 (NOX2) within macrophages, enhancing the expression of the NOX2 gene. This process promotes the generation of reactive oxygen species (ROS), subsequently accelerating the lipopolysaccharide (LPS)-induced NF-κB signaling pathway, culminating in heightened inflammatory responses. Deletion of NAT10 in mice results in reduced levels of IL-6 and TNF-α, while NAT10 overexpression increases the content of inflammatory cytokines [33]. Thus, NAT10 inhibitors have the potential to alleviate inflammatory reactions caused by macrophages during the pathological progression of periodontitis, offering a promising avenue for effective therapeutic interventions.

Furthermore, recent research has unveiled that NAT10, through ac4C acetylation, regulates the vascular endothelial growth factor A (VEGFA)-mediated PI3K/AKT signaling pathway, enhancing osteogenic development in human periodontal ligament stem cells [34]. During osteogenic differentiation, NAT10 overexpression results in elevated expression of osteogenic markers, increased alkaline phosphatase (ALP) activity, and enhanced osteogenic capability. These findings hold promise for applications in periodontal tissue engineering based on human periodontal ligament stem cells, providing assistance in the treatment of periodontal diseases [34].

Heart diseases

Myocardial infarction (MI) is a prevalent cardiac disease with a higher incidence in the elderly population [35, 36]. Piwi-interacting RNAs (piRNAs) are abundantly expressed in the heart, and their levels significantly increase under stressful conditions [37]. HAAPIR, a piRNA associated with cardiac apoptosis, targets NAT10 to mediate acetylation of transcription factor EC (Tfec), thereby increasing its gene expression. Concurrently, Tfec activates the pro-apoptotic factor BCL2-interacting killer (Bik), leading to Bik accumulation and subsequent cardiomyocyte death following MI [38]. Inhibition of NAT10 could potentially offer a therapeutic window for patients with MI.

Furthermore, heart failure patients commonly exhibit characteristic cardiac remodeling. NAT10 catalyzes acetylation of integrin-associated protein (CD47) and rho-associated coiled-coil containing protein kinase 2 (ROCK2) mRNA, enhancing the expression of proteins necessary for cardiac remodeling. In contrast, remodelin, a NAT10 inhibitor, can suppress cardiac fibrosis, providing effective prevention against cardiac functional impairment [39]. Thus, NAT10 emerges as a crucial target for the treatment of myocardial remodeling.

Pulmonary fibrosis

Long-term exposure to high concentrations of PM2.5 increases the risk of pulmonary fibrosis in the human respiratory system [40]. Experimental evidence reveals that ac4C modification and NAT10 levels increase in lung epithelial cells exposed to PM2.5, with the upregulation of NAT10 positively correlated with exposure duration and concentration. Further investigations confirm that NAT10 accelerates PM2.5-induced pulmonary fibrosis through ac4C-dependent stabilization of transforming growth factor beta 1 (TGFB1) mRNA. As an upstream driver, TGFB1 expedites the epithelial-mesenchymal transition (EMT) and fibrotic processes in lung epithelial cells. Simultaneously, inhibiting NAT10 significantly protects against PM2.5-induced EMT and fibrosis [41]. The experimental findings suggest that NAT10 could be a potential target for blocking PM2.5-induced pulmonary epithelial transformation and fibrosis.

Corneal injury

Corneal injury represents the most common type of trauma affecting the eyes, involving various factors, such as infection, allergic reactions, and more. Mesenchymal stem cells (MSCs) with potent differentiation and repair capabilities are found in various tissues [42]. Research suggests that MSCs enhance mRNA ac4C acetylation by stimulating the expression of NAT10, subsequently activating the ETV4/JUN/CCND2 signaling axis and increasing its stability. This ultimately promotes the repair of corneal injuries [43]. Therefore, ac4C acetylation modification emerges as a crucial therapeutic avenue for treating corneal injuries.

Neuropathic pain

Neuropathic pain (NPP), resulting from neurological injuries caused by trauma or diseases, has been identified as a complex condition that significantly impacts individuals [44]. Recent studies indicate that NAT10 plays a role in the formation of NPP through multiple pathways, presenting itself as a promising new target for treating neuropathic pain. In damaged dorsal root ganglia, the activation of upstream transcription factor 1 (USF1) promotes NAT10 expression, upregulating ac4C in synaptotagmin 9 (Syt9) mRNA and increasing the ac4C site and SYT9 protein levels. Inducing these physiological processes in the absence of injury can lead to the occurrence of neuropathic pain-like behaviors. Therefore, USF1-regulated NAT10 targets Syt9 and ac4C in peripheral nociceptive neurons, modulating neuropathic pain [6].

Another study revealed that nerve injury increases NAT10 expression in dorsal horn neurons and the interaction between NAT10 and vascular endothelial growth factor A (Vegfa) mRNA. In spared nerve injuny (SNI), heterogeneous nuclear ribonucleoprotein K (HNRNPK) binding to Vegfa mRNA facilitated NAT10 recruitment. Elevated NAT10 expression, through increased ac4C modification, enhanced the translation efficiency of Vegfa mRNA, resulting in upregulation of VEGFA. Upregulated VEGFA enhances central sensitization and neuropathic pain induced by SNI or AAV-hSyn-NAT10. Blocking this cascade reaction may provide a novel therapeutic approach for patients with neuropathic pain [45].

Premature ovarian failure

Premature ovarian insufficiency (POI), a dysfunctional ovarian condition occurring in females under the age of 40, although not highly prevalent, has profound and far-reaching effects on affected individuals [46, 47]. Granulosa cells play a crucial role in the growth and development of follicles, promoting this process through the secretion of hormones [48]. Cyclin-dependent kinase inhibitor P16 is associated with cellular senescence, and interferes with the cellular proliferation process by inhibiting the expression of Cyclin D1 (CCND1). In a mouse model of premature ovarian insufficiency, NAT10 has been identified to promote acetylation of P16 mRNA, thereby inhibiting the proliferation and differentiation of ovarian granulosa cells by increasing their stability, ultimately contributing to the onset of premature ovarian insufficiency [7]. Therefore, NAT10 exhibits the potential for therapeutic intervention in the context of premature ovarian insufficiency.

Intellectual disability

THUMPD1, a tRNA-binding protein found on human serine and leucine tRNAs, plays a crucial role in the acetylation process [49]. Biallelic mutations in the THUMPD1 gene are associated with intellectual disability syndrome, characterized by developmental delays, behavioral abnormalities, impaired hearing and vision, and facial dysmorphisms. Experimental evidence confirms that targeted THUMPD1 knockout results in the loss of ac4C modification in small RNAs and tRNAs. In summary, dysregulated THUMPD1 expression leads to tRNA acetylation deficiency, impairing protein function, and ultimately giving rise to a syndromic form of intellectual disability [50].

Viral Diseases

Following infection with influenza A virus, the ac4C modification of DAZAP1 in the host cells is significantly enriched in the 5’UTR. Meanwhile, decreased expression of NAT10 in infected host cells can inhibit viral growth [51]. These findings suggest that by decreasing the expression of host cell NAT10, viral replication and pathogenicity can be effectively reduced, resulting in an antiviral effect [51].

In HIV-1 (human immunodeficiency virus type 1), the expression level of NAT10 is abnormally high. Studies have found that inhibiting NAT10 expression not only reduces the ac4C modification of viral RNA and the expression of HIV-1 genes simultaneously, but also inhibits HIV-1 replication without affecting normal cellular physiological functions. This result highlights the importance of ac4C modification as a potential target for antiviral drug development. Additionally, mutations in the ac4C site on the HIV-1 env gene are part of the viral messenger RNA 3’UTR encoding gag, leading to a significant decrease in gag protein expression and accelerating virus spread [52]. HIV-1 Tat is a virus-encoded transactivator that activates viral transcription. NAT10, as one of the host proteins associated with HIV-1 Tat, can help HIV-1 latency in Jurkat cells by inhibiting Tat-mediated HIV-1 transcription [53].

On the other hand, studies have shown that ac4C modification can enhance the translation efficiency and stability of viral RNA, thereby enhancing virus replication and pathogenicity [51]. For example, in the 5’UTR of enterovirus 71 mRNA, ac4C modification enhances the recruitment of PCBP2 to the internal ribosome entry site (IRES) selectively, promoting the translation of viral RNA and enhancing virus infectivity. These findings provide new insights into viral biology and important biomarkers and targets for the development of antiviral strategies [54].

Discussion

In the 1970s, ac4C acetylation modification was first discovered on tRNA-ser and tRNA-leu in eukaryotic organisms and subsequently detected in rRNA and mRNA [8, 55]. As an acetylation modification identified on mRNA, ac4C exhibits robust biological functions. In this review, we delve into the role of ac4C modification in RNA post-transcriptional regulation, emphasizing its significance in the mechanisms underlying various diseases, particularly neurological and cardiac disorders. The ubiquity and specificity of ac4C modification reveal its crucial role as a biological regulatory mechanism, especially in the context of diseases. The presence of ac4C modification on different RNA types, such as mRNA and rRNA, underscores its importance in biology, while its specificity in specific disease contexts emphasizes the importance of considering the type and location of modifications when studying the relationship between RNA modification and diseases.

Regarding therapeutic potential, NAT10, identified as the sole ac4C modification catalyzing enzyme found in the human body [8], plays a pivotal role in the development of various diseases. Therefore, the development of inhibitors or activators targeting NAT10, particularly in the treatment of heart diseases and neuropathic pain, represents a promising strategy. For instance, in MI, NAT10 mediates the acetylation of Tfec and activates the expression of the pro-apoptotic factor Bik [38]. Furthermore, remodelin, which inhibits NAT10 expression, can reduce cardiac remodeling and treat premature aging syndrome. Simultaneously, in neuropathic pain, NAT10 is involved in NPP formation through various pathways, such as its interaction with USF1 and VEGFA mRNA. Targeting NAT10 can modulate key molecules like Syt9 and VEGFA, providing new strategies for treatment.

Meanwhile, research on the relationship between ac4C modification on RNA and diseases is still limited. First, the specific regulatory mechanisms of ac4C modification remain unclear. Currently, only one “writer” enzyme, NAT10, has been identified in humans, with no other enzymes or “reader” and “eraser” functions similar to NAT10 found yet. Second, the detailed involvement of ac4C modification in the pathogenesis of many diseases has not been fully elucidated. Additionally, the presence of ac4C modification is not the sole pathogenic factor in disease occurrence. Therefore, the significance of ac4C modification in the development of diseases requires further evaluation.

A deeper understanding of the specific mechanisms of ac4C modification in different diseases, particularly how it affects RNA stability, translation efficiency, or interaction with specific proteins, will help reveal the subtle regulatory mechanisms of RNA modification in disease occurrence. This insight offers new perspectives for future treatments.

In summary, our research further confirms the significant role of RNA modification, especially ac4C modification, in the occurrence and development of diseases. These findings not only provide new approaches for the diagnosis and treatment of diseases but also hold crucial implications for understanding the molecular mechanisms of diseases. By thoroughly investigating this modification, we can offer new strategies and methods for disease treatment.

Key Points
  • Complex interplay of post-transcriptional RNA modifications: describes the intricate process of post-transcriptional RNA modifications involving “writers,” “readers,” and “erasers,” playing crucial roles in growth, heredity, and disease.

  • Significance of N4-acetylcytidine (ac4C): emphasizes the profound effects of ac4C as an ancient and highly conserved RNA modification in the pathogenesis of various diseases.

  • In-depth exploration of ac4C modification in disease: delves into the functional implications of ac4C modification in disease, revealing its critical role in the pathogenesis of different conditions.

  • Introduction of novel perspectives on disease treatment: highlights the introduction of fresh perspectives and strategies for disease treatment based on the study of ac4C modification, providing innovative approaches for addressing diseases.

  • Revelation of biological functions of post-transcriptional RNA modifications: indicates that these investigations not only offer critical insights into the biological functions of post-transcriptional RNA modifications but also unveil their potential roles in disease mechanisms.

  • In summary, the text underscores the importance of post-transcriptional RNA modifications, particularly the ac4C modification in disease, and introduces new perspectives and strategies for disease therapy, shedding light on the biological functions of these modifications.

Acknowledgements

The Review Figure created with BioRender.com

Author contributions

Y.H. and J.B. were responsible for literature selection and retrieval. K.Y.W. and T.T.N. drafted the manuscript. W.H.O.Y. and Y.J.X. compiled and summarized the table and figures. Z.J.H. and X.H.Z. reviewed and revised the manuscript. L.L. funded our project. All authors approved the final draft.

Conflict of interest: None declared.

Funding

The Science and Technology Project of Shenzhen (JCYJ20190808123611319).

Data availability

The authors verify that the data underpinning the study’s conclusions can be found both in the main text and the supplementary resources provided.

Author Biographies

Keyu Wan, M.D., from the First Medical College of Nanchang University, focuses on exploring the basic theories of medicine and participates in many experimental projects, working to gain a deeper understanding of the molecular mechanisms of diseases.

Tiantian Nie, M.D., from the First Medical College of Nanchang University, has accumulated experience in clinical practice, focuses on disease diagnosis and treatment, and participates in clinical trials and case studies.

Wenhao Ouyang, M.D., from Sun Yat-sen Memorial Hospital of Sun Yat-sen University, has been dedicated to tumor research for many years and has published several important papers on tumor development, treatment and prevention, making significant contributions to the field of medical oncology.

Yunjing Xiong, M.D., from the Second Medical College of Nanchang University, focuses on the study and practice of experimental techniques and participates in a variety of experimental projects in an effort to explore the molecular mechanisms and therapeutic approaches of diseases.

Jing Bian, M.D., from the First Medical College of Nanchang University, focuses on oncology research and participates in various experimental projects in an effort to explore the molecular mechanisms of tumor development.

Ying Huang, M.D., from the First Medical College of Nanchang University, is actively involved in clinical and experimental projects to explore innovative therapeutic approaches.

Li Ling, M.D., from Shenzhen Hospital of Southern Medical University, focuses on optimizing patient care, promoting the application of precision medicine in clinical practice, and improving outcomes through personalized treatment plans.

Zhenjun Huang, M.D., from Sun Yat-sen Memorial Hospital, Sun Yat-sen University, focuses on cancer biomarkers and targeted therapies, and is committed to discovering new therapies.

Xianhua Zhu, M.D., a clinical specialist in cardiovascular surgery from the Second Affiliated Medical College of Nanchang University, has made significant achievements in disease treatment and has published several research papers on cancer and cardiovascular disease with outstanding contributions.

References

1.

Boccaletto
 
P
,
Stefaniak
 
F
,
Ray
 
A
. et al.  
MODOMICS: a database of RNA modification pathways. 2021 update
.
Nucleic Acids Res
 
2022
;
50
:
D231
5
.

2.

Zhao
 
BS
,
Roundtree
 
IA
,
He
 
C
.
Post-transcriptional gene regulation by mRNA modifications
.
Nat Rev Mol Cell Biol
 
2017
;
18
:
31
42
.

3.

Deng
 
L-J
,
Deng
 
WQ
,
Fan
 
SR
. et al.  
m6A modification: recent advances, anticancer targeted drug discovery and beyond
.
Mol Cancer
 
2022
;
21
:
52
.

4.

Wang
 
S
,
Lv
 
W
,
Li
 
T
. et al.  
Dynamic regulation and functions of mRNA m6A modification
.
Cancer Cell Int
 
2022
;
22
:
48
.

5.

Jiang
 
X
,
Liu
 
B
,
Nie
 
Z
. et al.  
The role of m6A modification in the biological functions and diseases
.
Sig Transduct Target Ther
 
2021
;
6
:
74
.

6.

Zhang M, Yang K, Wang QH. et al.  

The cytidine N-Acetyltransferase NAT10 participates in peripheral nerve injury-induced neuropathic pain by stabilizing SYT9 expression in primary sensory neurons
.
J Neurosci
2023;
43
:3009–27.

7.

Geng Z, Liu P, Yuan L. et al.  

Electroacupuncture attenuates ac4C modification of P16 mRNA in the ovarian granulosa cells of a mouse model premature ovarian failure
.
Acupunct Med
2023;
41
:27–37.

8.

Ito S, Horikawa S, Suzuki T. et al.  

Human NAT10 is an ATP-dependent RNA acetyltransferase responsible for N4-acetylcytidine formation in 18 S ribosomal RNA (rRNA)
.
J Biol Chem
2014;
289
:35724–30.

9.

Kawai G, Hashizume T, Miyazawa T. et al.  

Conformational characteristics of 4-acetylcytidine found in tRNA
.
Nucleic Acids Symp Ser
1989;61–62.

10.

Arango D, Sturgill D, Alhusaini N. et al.  

Acetylation of cytidine in mRNA promotes translation efficiency
.
Cell
2018;
175
:1872–1886.e24.

11.

Zheng X, Wang Q, Zhou Y. et al.  

N-acetyltransferase 10 promotes colon cancer progression by inhibiting ferroptosis through N4-acetylation and stabilization of ferroptosis suppressor protein 1 (FSP1) mRNA
.
Cancer Commun (Lond)
2022;
42
:1347–66.

12.

Wei W, Zhang S, Han H. et al.  

NAT10-mediated ac4C tRNA modification promotes EGFR mRNA translation and gefitinib resistance in cancer
.
Cell Rep
2023;
2
:112810.

13.

Li Q, Liu X, Jin K. et al.  

NAT10 is upregulated in hepatocellular carcinoma and enhances mutant p53 activity
.
BMC Cancer
2017;
17
:605.

14.

Yang Q, Lei X, He J. et al.  

N4-acetylcytidine drives glycolysis addiction in gastric cancer via NAT10/SEPT9/HIF-1α positive feedback loop
.
Adv Sci (Weinh)
2023;
10
:e2300898.

15.

Kumbhar BV, Kamble AD, Sonawane KD.

Conformational preferences of modified nucleoside N(4)-acetylcytidine, ac4C occur at ‘wobble’ 34th position in the anticodon loop of tRNA
.
Cell Biochem Biophys
2013;
66
:797–816.

16.

Arango D, Sturgill D, Yang R. et al.  

Direct epitranscriptomic regulation of mammalian translation initiation through N4-acetylcytidine
.
Mol Cell
2022;
82
:2797–2814.e11.

17.

Ito S, Akamatsu Y, Noma A. et al.  

A single acetylation of 18 S rRNA is essential for biogenesis of the small ribosomal subunit in Saccharomyces cerevisiae
.
J Biol Chem
2014;
289
:26201–12.

18.

Thalalla Gamage S, Sas-Chen A, Schwartz S. et al.  

Quantitative nucleotide resolution profiling of RNA cytidine acetylation by ac4C-seq
.
Nat Protoc
2021;
16
:2286–2307.

19.

Sinclair WR, Arango D, Shrimp JH. et al.  

Profiling cytidine acetylation with specific affinity and reactivity
.
ACS Chem Biol
2017;
12
:2922–6.

20.

Yan S, Lu Z, Yang W. et al.  

Antibody-free fluorine-assisted metabolic sequencing of RNA N4-acetylcytidine
.
J Am Chem Soc
2023;
145
:22232–42.

21.

Su W, Xie XQ, Liu XW. et al.  

iRNA-ac4C: a novel computational method for effectively detecting N4-acetylcytidine sites in human mRNA
.
Int J Biol Macromol
2023;
227
:1174–81.

22.

Lai F-L, Gao F.

LSA-ac4C: a hybrid neural network incorporating double-layer LSTM and self-attention mechanism for the prediction of N4-acetylcytidine sites in human mRNA
.
Int J Biol Macromol
2023;
253
:126837.

23.

Zhao W, Zhou Y, Cui Q. et al.  

PACES: prediction of N4-acetylcytidine (ac4C) modification sites in mRNA
.
Sci Rep
2019;
9
:11112.

24.

Duan J, Zhang Q, Hu X. et al.  

N4-acetylcytidine is required for sustained NLRP3 inflammasome activation via HMGB1 pathway in microglia
.
Cell Signal
2019;
58
:44–52.

25.

Ma Y, Li W, Fan C. et al.  

Comprehensive analysis of long non-coding RNAs N4-acetylcytidine in Alzheimer’s disease mice model using high-throughput sequencing
.
J Alzheimers Dis
2022;
90
:1659–75.

26.

Furman D, Chang J, Lartigue L. et al.  

Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states
.
Nat Med
2017;
23
:174–84.

27.

Zhang H, Chen Z, Zhou J. et al.  

NAT10 regulates neutrophil pyroptosis in sepsis via acetylating ULK1 RNA and activating STING pathway
.
Commun Biol
2022;
5
:916.

28.

Guo G, Shi X, Wang H. et al.  

Epitranscriptomic N4-acetylcytidine profiling in CD4+ T cells of systemic lupus erythematosus
.
Front Cell Dev Biol
2020;
8
:842.

29.

M K, Jm S.

Osteoporosis as a model for the long-term clinical consequences of the menopause
.
Progress in cardiovascular diseases
1995;
38
.

30.

Marini
 
F
,
Cianferotti
 
L
,
Brandi
 
ML
.
Epigenetic mechanisms in bone biology and osteoporosis: can they drive therapeutic choices?
 
Int J Mol Sci
 
2016
;
17
:
1329
.

31.

Yang
 
W
,
Li
 
HY
,
Wu
 
YF
. et al.  
ac4C acetylation of RUNX2 catalyzed by NAT10 spurs osteogenesis of BMSCs and prevents ovariectomy-induced bone loss
.
Mol Ther Nucleic Acids
 
2021
;
26
:
135
47
.

32.

Slots J. Periodontitis: facts, fallacies and the future.

Periodontol 2000
2017;
75
:7–23.

33.

Zhang
 
Z
,
Zhang
 
Y
,
Cai
 
Y
. et al.  
NAT10 regulates the LPS-induced inflammatory response via the NOX2-ROS-NF-κB pathway in macrophages
.
Biochim Biophys Acta Mol Cell Res
 
2023
;
1870
:
119521
.

34.

Cui
 
Z
,
Xu
 
Y
,
Wu
 
P
. et al.  
NAT10 promotes osteogenic differentiation of periodontal ligament stem cells by regulating VEGFA-mediated PI3K/AKT signaling pathway through ac4C modification
.
Odontology
 
2023
;
111
:
870
82
.

35.

Rodgers
 
JL
,
Jones
 
J
,
Bolleddu
 
SI
. et al.  
Cardiovascular risks associated with gender and aging
.
J Cardiovasc Dev Dis
 
2019
;
6
:
19
.

36.

Roth
 
GA
,
Mensah
 
GA
,
Johnson
 
CO
. et al.  
Global Burden of Cardiovascular Diseases and Risk Factors, 1990-2019: Update from the GBD 2019 Study
.
J Am Coll Cardiol
 
2020
;
76
:
2982
3021
.

37.

Rajan
 
KS
,
Velmurugan
 
G
,
Gopal
 
P
. et al.  
Abundant and altered expression of PIWI-interacting RNAs during cardiac hypertrophy
.
Heart Lung Circ
 
2016
;
25
:
1013
20
.

38.

Wang
 
K
,
Zhou
 
LY
,
Liu
 
F
. et al.  
PIWI-Interacting RNA HAAPIR regulates cardiomyocyte death after myocardial infarction by promoting NAT10-mediated ac4C acetylation of Tfec mRNA
.
Adv Sci (Weinh)
 
2022
;
9
:
e2106058
.

39.

Shi
 
J
,
Yang
 
C
,
Zhang
 
J
. et al.  
NAT10 is involved in cardiac remodeling through ac4C-mediated transcriptomic regulation
.
Circ Res
 
2023
;
133
:
989
1002
.

40.

Kim
 
K-H
,
Kabir
 
E
,
Kabir
 
S
.
A review on the human health impact of airborne particulate matter
.
Environ Int
 
2015
;
74
:
136
43
.

41.

Shenshen
 
W
,
Yin
 
L
,
Han
 
K
. et al.  
NAT10 accelerates pulmonary fibrosis through N4-acetylated TGFB1-initiated epithelial-to-mesenchymal transition upon ambient fine particulate matter exposure
.
Environ Pollut
 
2023
;
322
:
121149
.

42.

Liu
 
H
,
Zhang
 
J
,
Liu
 
C-Y
. et al.  
Bone marrow mesenchymal stem cells can differentiate and assume corneal keratocyte phenotype
.
J Cell Mol Med
 
2012
;
16
:
1114
24
.

43.

Fei
 
X
,
Cai
 
Y
,
Lin
 
F
. et al.  
Amniotic fluid mesenchymal stem cells repair mouse corneal cold injury by promoting mRNA N4-acetylcytidine modification and ETV4/JUN/CCND2 signal axis activation
.
Hum Cell
 
2021
;
34
:
86
98
.

44.

Van Hecke O, Austin SK, Khan RA. et al.  

Neuropathic pain in the general population: a systematic review of epidemiological studies
.
Pain
2014;
155
:654–62.

45.

Xu
 
T
,
Wang
 
J
,
Wu
 
Y
. et al.  
ac4C enhances the translation efficiency of Vegfa mRNA and mediates central sensitization in spinal dorsal horn in neuropathic pain
.
Adv Sci (Weinh)
 
2023
;
10
:
e2303113
.

46.

Lu
 
C
,
Qin
 
C
,
Fu
 
Z
. et al.  
Metabolic dysregulation in patients with premature ovarian insufficiency revealed by integrated transcriptomic, methylomic and metabolomic analyses
.
Clin Transl Med
 
2022
;
12
:
e1006
.

47.

Goswami
 
D
,
Conway
 
GS
.
Premature ovarian failure
.
Horm Res
 
2007
;
68
:
196
202
.

48.

Nelson
 
LR
,
Bulun
 
SE
.
Estrogen production and action
.
J Am Acad Dermatol
 
2001
;
45
:
S116
24
.

49.

Sharma
 
S
,
Langhendries
 
JL
,
Watzinger
 
P
. et al.  
Yeast Kre33 and human NAT10 are conserved 18S rRNA cytosine acetyltransferases that modify tRNAs assisted by the adaptor Tan1/THUMPD1
.
Nucleic Acids Res
2015;
43
:2242–58.

50.

Broly
 
M
,
Polevoda
 
BV
,
Awayda
 
KM
. et al.  
THUMPD1 bi-allelic variants cause loss of tRNA acetylation and a syndromic neurodevelopmental disorder
.
Am J Hum Genet
2022;
109
:587–600.

51.

Furuse
 
Y
.
RNA modifications in genomic RNA of influenza A virus and the relationship between RNA modifications and viral infection
.
Int J Mol Sci
2021;
22
:9127.

52.

Tsai
 
K
,
Jaguva Vasudevan
 
AA
,
Martinez Campos
 
C
. et al.  
Acetylation of cytidine residues boosts HIV-1 gene expression by increasing viral RNA stability
.
Cell Host Microbe
2020;
28
:306–312.e6.

53.

Jean
 
MJ
,
Power
 
D
,
Kong
 
W
. et al.  
Identification of HIV-1 tat-associated proteins contributing to HIV-1 transcription and latency
.
Viruses
2017;
9
:67.

54.

Hao
 
H
,
Liu
 
W
,
Miao
 
Y
. et al.  
N4-acetylcytidine regulates the replication and pathogenicity of enterovirus 71
.
Nucleic Acids Res
2022;
50
:9339–54.

55.

Stern
 
L
,
Schulman
 
LH
.
The role of the minor base N4-acetylcytidine in the function of the Escherichia coli noninitiator methionine transfer RNA
.
J Biol Chem
1978;
253
:6132–9.

Author notes

Keyu Wan, Tiantian Nie and Wenhao Ouyang have contributed equally to this work and share first authorship.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com