1. Introduction
Epstein-Barr virus (EBV) is a ubiquitous human gammaherpesvirus that establishes lifelong latent infection in more than 90% of the adult population worldwide [
1,
2]. EBV is considered an oncogenic virus owing to its ability to promote malignant transformation and stimulate tumor growth, and it has been implicated in several lymphoid and epithelial cancers including Burkitt’s lymphoma [
3], Hodgkin’s lymphoma [
4], post-transplant lymphoproliferative disorder [
5], nasopharyngeal carcinoma [
6], and gastric carcinoma [
7]. Following primary infection, EBV persists indefinitely in the host through the establishment of latent or lytic cycles of infection [
8]. The lytic stage of EBV is essential for horizontal viral spread and the maintenance of persistent infection, but its mechanistic role in viral oncogenesis remains unclear. The switch from EBV latency to lytic replication is widely demonstrated to be modulated by viral and host miRNAs [
9].
MicroRNAs (miRNAs) are short and non-coding RNA molecules, typically consisting of 18–24 nucleotides, that play crucial roles in gene regulation. MiRNAs exert gene silencing primarily through post-transcriptional mechanisms [
10]. Following their biogenesis, mature miRNAs are incorporated into the RNA-induced silencing complex (RISC), which facilitates the recognition and binding of the miRNA to its target messenger RNA (mRNA). This interaction is predominantly mediated through sequence complementarity between the “seed region” of the miRNA (nucleotides 2–7) and the 3′ untranslated region (3′ UTR) of the target mRNA [
11]. Upon binding, miRNAs can elicit gene silencing through two primary mechanisms: translational repression or mRNA degradation [
12]. Translational repression involves the inhibition of ribosome recruitment and/or translation initiation, resulting in reduced protein synthesis from the target mRNA [
13]. This mechanism is often observed when a miRNA exhibits imperfect complementarity to its target [
14]. Alternatively, miRNAs can induce mRNA degradation through the recruitment of deadenylation and decapping enzymes, leading to the rapid decay of the target mRNA [
15]. This mechanism typically occurs when the miRNA exhibits extensive complementarity to its target, often resulting in endonucleolytic cleavage of the mRNA [
16].
EBV encodes more than 44 mature miRNA sequences that have been experimentally identified [
8]. The latent-lytic switch of EBV, which is critically regulated by the interplay of viral and cellular miRNAs, plays a pivotal role in EBV-associated tumorigenesis [
17]. During the latent phase, EBV-encoded miRNAs, notably the miR-BARTs, function to repress the expression of viral immediate-early genes, such as
BZLF1 and
BRLF1, as well as host factors that facilitate lytic reactivation, thereby ensuring the maintenance of viral latency [
18,
19]. Conversely, cellular miRNAs exert a dualistic influence on the latent-lytic switch, either promoting or inhibiting it through direct targeting of viral transcripts or modulation of host signaling cascades. Hsa-miR-141 has been demonstrated to inhibit
FOXO3 expression, resulting in enhanced EBV lytic reactivation [
20]. Hsa-miR-190 targets and downregulates
NR4A3, a cellular immediate-early gene for EBV reactivation, and inhibits the expression of
BZLF1 and viral lytic DNA replication [
21]. Dysregulation of host miRNAs may accelerate the progression of EBV-associated malignancies by modulating key oncogenic pathways or dampening antitumor immune responses [
22,
23]. However, the specific cellular miRNAs involved in the regulatory roles of the latent-lytic transition remain underexplored.
Hsa-miR-7974 (miR-7974), a mature human miRNA, was first investigated and characterized in 2013 [
24]. Its genomic locus is located on chromosome 19, specifically at chr19:11495544–11495622. The functional role of miR-7974 in oncogenesis remains ambiguous. While studies have reported elevated expression levels of miR-7974 in breast cancer [
25] and colorectal cancer cells [
26], suggesting a potential oncogenic function, contrasting evidence has demonstrated its downregulation in SHG-8- treated glioblastoma samples, which target the oncogenes
CORO2A,
APC2, and
WNT7A [
27]. Furthermore, miR-7974 has been reported to be downregulated in glioma tissues, potentially by targeting
BOC. The downstream genes of
BOC,
POU2F1, and
TP53 are implicated in the transcriptional regulation of tumor-related genes [
28]. However, it is noteworthy that research on miR-7974 remains relatively limited compared to other well-studied miRNAs. The specific functions and regulatory networks of miR-7974 in EBV reactivation are not yet fully understood.
Through miRNA sequencing, we discovered that miR-7974 was expressed at lower levels in reactivated EBV-positive Burkitt’s lymphoma Raji cells and nasopharyngeal carcinoma C666-1 cells than in control cells, with subsequent validation by quantitative reverse transcription-polymerase chain reaction (RT-qPCR). Functional studies using miR-7974 mimics and inhibitors demonstrated that BZLF1 and BRLF1 are direct targets of miR-7974. Furthermore, GO analysis and KEGG pathway analysis were employed to identify biological pathways enriched among the predicted gene targets of the significantly regulated miRNAs, and we also constructed a miRNA–mRNA interaction network of the miR-7974 and EBV lytic genes. These results indicate important roles for miR-7974 in modulating the EBV lytic-latent cycle and contribute to a deeper understanding of the molecular mechanisms underlying EBV reactivation.
2. Materials and Methods
2.1. Cells and Cell Culture
The EBV-positive human Burkitt’s lymphoma cell line Raji was historically preserved in our laboratory. The human embryonic kidney cell line HEK293T was purchased from Cell Resource Center, Institute of Basic Medical Sciences, CAMS/PUMC (Beijing, China). The EBV-positive human nasopharyngeal carcinoma cell line C666-1 was purchased from Meisen CTCC (Hangzhou, China). The EBV-negative human nasopharyngeal carcinoma cell line HK-1 was purchased from CHI Scientific, Inc. (Shanghai, China). Raji and C666-1 cells were cultured in RPMI 1640 medium (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) and 1% penicillin-streptomycin (Gibco, Thermo Fisher Scientific, Waltham, MA, USA). HEK293T cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% FBS and 1% penicillin-streptomycin. HK-1 cells were cultured in a specialized medium provided by CHI Scientific, Inc., Maynard, MA, USA. All the cell lines were maintained at 37 °C in a humidified atmosphere containing 5% CO2.
2.2. Epstein-Barr Virus (EBV) Lytic Replication Induction
Raji cells were treated with 20 ng/mL 12-O-tetradecanoylphorbol-13-acetate (TPA; Cell Signaling Technology, Cat# 4174S, Danvers, MA, USA) for 48 h, and C666-1 cells were induced with 40 μmol/L Dp44mT (C7, EBV lytic cycle inducer-1; MedChemExpress, Cat# HY-149577, Monmouth Junction, NJ, USA) for 48 h to initiate EBV lytic replication.
2.3. Transcriptome Sequencing
RNA sequencing was performed, and libraries were constructed by Novogene Co., Ltd. (Beijing, China). The reactivated and untreated samples were processed and sequenced on a NovaSeq X Plus Series PE150 platform (Illumina, San Diego, CA, USA). Messenger RNA was purified from total RNA via poly-T oligo-attached magnetic beads. After fragmentation, the first strand cDNA was synthesized via random hexamer primers. Then, the second strand cDNA was subsequently synthesized via dUTP, instead of dTTP. The directional library was ready after end repair, A-tailing, adapter ligation, size selection, USER enzyme digestion, amplification, and purification. The library was checked with a Qubit and real-time PCR for quantification and a bioanalyzer for size distribution detection. After library quality control, different libraries were pooled based on the basis of the effective concentration and targeted data amount, and then subjected to Illumina sequencing.
2.4. MicroRNA (miRNA) Deep Sequencing
MiRNA sequencing was performed, and libraries were constructed by Novogene Co., Ltd. (Beijing, China). The reactivated and untreated samples were processed and sequenced on an Illumina SE50 platform (Illumina, San Diego, CA, USA). The 3′ and 5′ adaptors were ligated to the 3′ and 5′ ends of the small RNA, respectively. Then, first-strand cDNA was synthesized after hybridization with reverse transcription primers. The double-stranded cDNA library was generated through PCR enrichment. After purification and size selection, libraries with insertions between 18 and 40 bp were ready for sequencing via Illumina sequencing with SE50. The library was checked with a Qubit and real-time PCR for quantification and a bioanalyzer for size distribution detection. The quantified libraries will be pooled and sequenced on Illumina platforms, according to the effective library concentration and data amount needed.
2.5. Differential MicroRNA (miRNA) Expression Analysis
Differential expression analysis was performed via the DESeq2 R package. For regulation calculations between the comparison, a p-value < 0.001 and a |log2(FoldChange)| > 1.5 were set as the thresholds for significantly differential expression.
2.6. In Silico Prediction of Viral and Cellular Targets of miR-7974
To identify direct targets of miR-7974, the computational prediction tools miRanda (
https://github.com/hacktrackgnulinux/miranda, accessed on 5 February 2025) and RNAhybrid (
https://bibiserv.cebitec.uni-bielefeld.de/rnahybrid, accessed on 5 February 2025) were utilized to predict potential miRNA binding sites within the EBV genome. For the prediction of potential cellular target genes of miR-7974, the miRDB (
https://mirdb.org, accessed on 10 February 2025) and TargetScan (
https://www.targetscan.org, accessed on 10 February 2025) databases were used. These platforms integrate sequence-based and context-specific features to predict miRNA–mRNA interactions, providing a comprehensive list of putative cellular targets for further experimental validation.
2.7. MiR-7974 Target Gene Enrichment for Biological Pathways, Disease Associations, and Gene Ontology (GO) Categories
To identify pathways and disease associations possibly regulated by miR-7974, the predicted genes were uploaded to the Functional Annotation Tools of DAVID Bioinformatic Resources 6.7 (
https://davidbioinformatics.nih.gov). An enrichment was considered significant when the
p-value was <0.05. Gene Ontology (GO) enrichment analysis and functional visualization were performed via ClueGO 2.5.9 and the CluePedia 1.5.9 plugin in the Cytoscape software 3.10.3.
2.8. Establishment of miRNA–mRNA Network
Nine key genes associated with EBV reactivation were first selected to construct the miRNA–mRNA regulatory network, which regulates the key genes involved in EBV reactivation. Cellular miRNAs predicted to target these genes were identified via both RNAhybrid and miRanda tools to analyze sequence complementarity and thermodynamic stability for miRNA–mRNA interactions. The predicted miRNA–mRNA pairs were then imported into Cytoscape, a bioinformatics software platform (3.10.3), to visualize and analyze the regulatory network.
2.9. Dual-Luciferase Reporter Assay
To investigate the effect of Hsa-miR-7974 on the expression of EBV immediate-early genes, the 3′ UTR sequences of BRLF1 and the corresponding mutants were cloned and inserted into the pmirGLO dual-luciferase miRNA target expression vector (Promega, Madison, WI, USA), which were named BRLF1-WT and BRLF1-MUT. HEK293T cells and HK-1 cells were seeded in a 96-well plate. After 24 h, the cells were co-transfected with the luciferase reporter vector and 20 nM miR-7974 mimics or scrambled miRNA (miR-NC). The relative luciferase activity was measured at 48 h post-transfection via a dual-luciferase reporter gene assay kit (Yeasen, Shanghai, China). For each sample, Renilla luciferase activity was normalized to firefly luciferase activity.
2.10. Stem-Loop Quantitative Real-Time Polymerase Chain Reaction (PCR) for MicroRNA (miRNA) Analysis
MiRNAs were extracted from the cell samples via the miRNeasy Micro Kit (Qiagen, Cat# 217084, Hilden, Germany), following the manufacturer’s protocol. The reverse transcription primers were pre-treated as follows to guarantee a stem-loop structure: they were incubated at gradient temperatures of 95, 80, 70, 60, 50, 40, 30, and 20 °C, for 30 s each. cDNA was synthesized via the PrimeScript RT Reagent Kit with gDNA Eraser (TaKaRa, Cat# RR047A, Kyoto, Japan) according to the manufacturer’s instructions. Before reverse transcription, the mixture was incubated at 16 °C for 15 min to anneal the primers. The sequences of the primers used were as follows: miR-7974 stem-loop RT primer: GTCTGTATGCTTGTTCTCGTCTCTGTGTCATCCCTCAAGCATACAGACGGGCTC, forward primer: TACAGGCTGTGATGCTCTCCT, reverse primer: TATGCTTGTTCTCGTCTCTGTGTC; and snRNA U6 stem-loop RT/reverse primer: AACGCTTCACGAATTTGCGT, forward primer: CTCGCTTCGGCAGCACA. Reverse transcription was carried out in a T100 thermal cycler (Bio-Rad, Hercules, CA, USA) for 15 min at 37 °C, 5 s at 85 °C, and then held at 4 °C. Real-time quantitative PCR was performed via an SYBR Green Premix Pro Taq HS qPCR Kit (Accurate Biotechnology, Guangzhou, China) on an Archimed X6 System (RocGene, Beijing, China). All amplifications were performed in triplicate, and values were normalized to the value for an endogenous control, snRNA U6.
2.11. Transfection of MicroRNA (miRNA) Mimics and Inhibitors
The mimic, the inhibitor, and the scrambled control were purchased from Beijing TsingKe Biotech Co., Ltd. (Beijing, China). In this study, C666-1 cells were seeded 24 h prior to transfection in a 6-well-plate containing 2 mL of RPMI 1640 medium. The cells were transfected with negative controls, miR-7974 mimic or miR-7974 inhibitor via the Lipofectamine RNAiMAX (Invitrogen, Cat# 13778030, Thermo Fisher Scientific, Waltham, MA, USA) transfection agent, according to the manufacturer’s protocol. The final concentrations of the miRNA mimic and inhibitor were 10 nM and 50 nM, respectively. After 24 h, the cells were treated with 40 μmol/L C7 compound for 48 h to induce the lytic cycle of EBV.
2.12. Quantitative Reverse Transcription-Polymerase Chain Reaction (RT-qPCR)
Total RNA was extracted from the cell samples via an RNeasy Mini Kit (Qiagen, Cat# 74106), following the manufacturer’s protocol. The quality of the extracted RNA was determined on an FC-3100 spectrophotometer (Lifereal Biotechnology, Hangzhou, China). Total RNA reverse transcription was performed via the PrimeScript RT Reagent Kit with gDNA Eraser (TaKaRa, Cat# RR047A, Japan) to generate cDNA. RT-qPCR was carried out via the use of an SYBR Green Premix Pro Taq HS qPCR Kit (Accurate Biotechnology, China) on an Archimed X6 System (RocGene, China). The sequences of the primers used were as follows: BZLF1 forward: TTGGGCACATCTGCTTCAACAGGA, reverse: AATGCCGGGCCAAGTTTAAGCAAC; BRLF1 forward: GAACATACCTTCCCGGCTATC, reverse: GAGCGATGAGAGACCCATATTC; BMRF1 forward: GCCGTTGAGGCCCACGTTGT, reverse: TGGGAATGGCAGGCGAGGGT; and GAPDH forward: GACACCCACTCCTCCACCTTT, reverse: ACCACCCTGTTGCTGTAGCC. The PCR conditions were 95 °C for 30 s, followed by 40 cycles at 95 °C for 5 s, 60 °C for 30 s, with fluorescence measured continuously. The relative mRNA expression levels of BZLF1, BRLF1, and BMRF1, normalized to that of GAPDH as an internal control, were calculated via the 2−ΔΔCt method.
2.13. Epstein-Barr Virus (EBV) DNA Viral Load
Viral-derived DNA was measured to determine the EBV viral load. Briefly, pelleted cells were harvested, and the total DNA was extracted via the QIAamp DNA Mini Kit (Cat# 51304, Qiagen, USA), according to the kit’s handbook. The quality of the extracted RNA was determined on an FC-3100 spectrophotometer (Lifereal Biotechnology, China). To quantify EBV copy numbers, the given standard samples (from 107 copies/µL to 10 copies/µL) were first used to create the standard curve according to the manufacturer’s instructions by using an EBV Probe quantitative PCR Kit (Cat# 15-60800, BINGENE, Beijing, China). Then, 40 ng (2 µL) of each DNA sample was added to PCR tubes, and the PCR was performed as follows (Archimed X6 System, China): 95 °C for 10 min, 95 °C for 15 s, and 60 °C for 60 s (data collection) for 40 cycles. The EBV copy number of each sample can be calculated by the corresponding threshold (Ct) cycle with the aid of a standard curve.
2.14. Western Blot Analysis
Total protein was extracted from cell samples lysed in RIPA buffer (Abcam, Cat# ab156034, Waltham, MA, USA) containing a 1% protease inhibitor cocktail. The protein concentration was determined via a BCA assay. Proteins were separated by 10% SDS-PAGE (Beyotime, Shanghai, China) and transferred to 0.45 μM PVDF membranes (Millipore, Billerica, MA, USA) via a semidry transfer system (Bio-Rad, USA). The membranes were blocked with 5% (w/v) non-fat dried milk in TBST for 1 h at room temperature and then incubated overnight at 4 °C with the following primary antibodies: ZEBRA (1:250; Santa Cruz Biotechnology, Cat# sc-53904, Dallas, TX, USA), Ea-D (1:500; Sigma-Aldrich, Cat# MAB8186, Burlington, MA, USA), and α-Actinin (1:5000; Proteintech, Cat# 11312-2-AP, Wuhan, China). After washing, the membranes were incubated for 1 h at room temperature with HRP-conjugated secondary antibodies (rabbit, 1:20,000, Abcam, Cat# ab205718; mouse, 1:20,000, Abcam, Cat# ab205719). Immunoreactive bands were visualized via enhanced chemiluminescence (Pierce Plus-ECL; Revvity, Waltham, MA, USA) and captured with a CLINX ChemiScope 6200 touch imaging system (Qinxiang, Hangzhou, China). Band intensities were quantified via densitometry via ImageJ software (v1.54 m).
2.15. Statistical Analysis
Statistical analyses were performed via R (version 4.4.2) and GraphPad Prism (version 10.3.1). The results are presented as the means ± standard deviations (SDs) of at least three independent experiments, and a two-tailed Student’s t-test was used to demonstrate statistical significance. Differences were considered as statistically significant when the p-value was <0.05.
4. Discussion
EBV, a ubiquitous human gammaherpesvirus, establishes lifelong latent infection in more than 90% of the global adult population. Its ability to switch between latent and lytic phases is crucial for viral persistence and pathogenesis, particularly in EBV-associated malignancies such as nasopharyngeal carcinoma, Burkitt’s lymphoma, and Hodgkin’s lymphoma. The transition from latency to lytic replication is tightly regulated by both viral and host factors, including miRNAs. MiRNAs are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation, often by binding to the 3′ UTR of target mRNAs, leading to mRNA degradation or translational repression. While EBV-encoded miRNAs have been extensively studied, the role of host miRNAs in regulating EBV reactivation remains poorly understood. A study by Cramer et al. (2014) demonstrated that miR-190 promotes host cell survival and prevents EBV from entering the lytic life cycle [
21]. In this study, we identified miR-7974 as a novel regulator of EBV reactivation, demonstrating its ability to directly target and suppress the expression of two key viral lytic genes,
BZLF1 and
BRLF1.
Multiple pharmacological agents, including romidepsin [
29], NEO-212 [
30], valproic acid [
31], and suberoylanilide hydroxamic acid (SAHA) [
32], have been reported to effectively induce lytic reactivation in C666-1 cells. Notably, the choice of lytic inducer may influence experimental outcomes, as different agents can activate distinct signaling pathways. Whether downregulation of miR-7974 is a conserved feature of EBV solubility reactivation or a drug-specific artifact is a question worth considering. Therefore, when we designed the protocol for this study, we did consider the drug-specific effects. We employed two cell model systems: Raji cells treated with TPA, and C666-1 cells treated with C7. In both systems, successful EBV lytic replication was induced, accompanied by significant reduction in miR-7974 expression (as shown in
Table 1 and
Figure 3 and
Figure 4). This suggested that the changes in miR-7974 expression in EBV-infected Raji cells are attributable primarily to viral reactivation rather than TPA-mediated cellular effects. Thus, we can similarly assume that the situation in C666-1 cells is also consistent. And, therefore, we suggest that downregulation of miR-7974 is a conserved feature of EBV lytic reactivation rather than a drug-specific artifact. In addition, C7 has been extensively documented as an effective lytic inducer in epithelial-derived cell lines, including NPC models [
33,
34]. Our preliminary experiments also confirmed that C7 induced a consistent and reproducible lytic cycle response in C666-1 cells, as measured by the expression of early lytic genes and viral genome replication; therefore, we selected C7 as the primary lytic inducer for C666-1 cells.
We have identified miR-7974 as a critical regulator of EBV reactivation, demonstrating its ability to directly target and suppress the expression of two pivotal viral lytic genes,
BZLF1 and
BRLF1. We observed a significant downregulation of
BZLF1 and
BRLF1 following transfection with miR-7974 mimics, and a corresponding upregulation upon miR-7974 inhibition. The direct interaction between miR-7974 and the 3′ UTR of
BRLF1 was further validated via dual-luciferase reporter assays, and the downregulation of the mRNA expression of the
BZLF1 and
BRLF1 genes following miR-7974 transfection provided robust mechanistic evidence for this regulatory relationship. These findings further support the role of miR-7974 as a negative regulator of EBV lytic gene expression and viral replication. The inverse correlation between the miR-7974 level and viral load underscores the potential of miR-7974 as a therapeutic target for controlling EBV replication and associated diseases. Zhang et al. (2010) reported that Hsa-199a-3p and Hsa-210-3p suppress hepatitis B virus (HBV) replication by targeting the HBV pre-S1 region [
35], similar to the mechanism of action of miR-7974. Future studies should explore the mechanisms by which miR-7974 modulates viral gene expression and its broader implications for EBV latency and reactivation.
Our results align with and extend previous findings on the regulatory roles of miRNAs in the EBV lytic-latent switch. EBV reactivation is precisely regulated by an intricate crosstalk network of viral and host miRNAs [
9]. For example, EBV-encoded miR-BARTs have been shown to repress
BZLF1 and
BRLF1 expression, thereby promoting viral latency. Lung et al. (2018) suggested that BART5-5p, BART7-3p, BART9-3p, and BART14-3p control the ATM signaling pathway in NPC cells. By suppressing these endogenous miR-BARTs in EBV-positive NPC cells, a novel function of miR-BARTs in inhibiting Zta-induced lytic reactivation was discovered [
36]. Jung et al. (2014) identified miR-BART20-5p as a potent regulator of EBV latency through specific targeting of the
BRLF1 3′ UTR, effectively blocking viral reactivation [
18]. Barth et al. (2008) identified miR-BART2-mediated suppression of
BALF5 (viral DNA polymerase), with miRNA overexpression achieving 40–50% BALF5 protein reduction and 20% decrease in virion production, effectively maintaining viral latency [
37]. Ellis-Connell et al. (2010) reported that miR-200b and miR-429 serve as lytic inducers that counteract ZEB1/ZEB2-mediated repression of the
BZLF1 Zp promoter across EBV-infected cell types [
38]. Chen et al. (2021) identified miR-142 as a critical regulator that suppresses EBV reactivation by modulating the BCR-initiated SOS1/Ras/Raf/Mek/Erk signaling cascade [
20]. Cao et al. (2018) demonstrated that ultraviolet (UV) irradiation and hypoxia induce EBV reactivation in lymphoma cells, and miR-18a serves as a key mediator which promotes EBV reactivation by suppressing the ATM-dependent DNA damage response (DDR) pathway [
39]. However, in contrast to these previously characterized miRNAs, miR-7974 has been implicated in various cancer contexts. Its role in EBV biology, as revealed in this study, adds a novel dimension to its functional repertoire and highlights its potential as a key player in viral–host interactions. The functional role of miR-7974 in cancer appears to be highly context-dependent. While some studies have reported its upregulation in breast and colorectal cancers, suggesting a potential oncogenic function, others have reported its downregulation in glioma, where it acts as a tumor suppressor by targeting oncogenes such as
CORO2A,
APC2,
WNT7A, and
BOC [
27]. This duality underscores the tissue-specific and context-dependent nature of the miRNA function, which may be influenced by the unique molecular and cellular environments of different cancer types.
Comprehensive bioinformatics analyses (GO enrichment, KEGG pathway, and disease association analyses) revealed the broader regulatory network of miR-7974. GO analysis revealed significant enrichment in biological processes related to membrane dynamics, cellular adhesion, system development, and signal transduction—key pathways critically implicated in EBV reactivation. These findings suggest that miR-7974 may regulate EBV reactivation not only through direct viral gene targeting, but also by modulating host genes involved in viral envelope glycoprotein processing, assembly, and intercellular viral dissemination. Zheng et al. (2023) reported that the miR-7974 inhibitor reduced the osteogenic differentiation of dental follicle stem cells by targeting
FKBP15, potentially impacting skeletal development [
40]. According to the research by Kumar et al. (2020), miR-7974 was predicted to regulate several gene clusters (
NOG,
S1PR1,
MRAS,
PDGFC, and
BMP5) involved in early neural induction of human embryonic stem cells and induced pluripotent stem cells [
41]. KEGG analysis highlighted the enrichment of key signaling pathways, including the “MAPK signaling pathway”, “NF-κB signaling pathway”, and “IL-10 signaling pathway”, all of which are pivotal in regulating EBV latency and lytic cycle transitions. Notably, disease association analysis identified “nasopharyngeal neoplasms”, “lymphoproliferative disorders”, and “leukemia” as prominent hits, reflecting the well-established links between EBV and these malignancies. According to the research by Gao et al. (2019), the PKC, NF-κB, p38 MAPK, PI3K, and ERK signaling pathways have been implicated in EBV reactivation [
42]. Nanbo et al. (2012) reported that ERK and NF-κB pathway inhibitors effectively suppressed both lytic induction and cell-to-cell EBV transmission [
43]. Chen et al. (2023) reported that miR-7974 can regulate the immune processes by modulating several pathways, such as the mTOR and PI3K-Akt signaling pathway [
44]. Dong et al. (2020) reported that the pathway enrichment analyses of the miR-7974 target genes were partly associated with the cytokine–cytokine receptor interaction [
45]. These findings are generally consistent with the miR-7974-related pathways identified in this study.
Several critical questions remain to be addressed in future studies. First, the precise molecular mechanisms by which miR-7974 regulates BZLF1 and BRLF1 expression, including the identification of specific binding sites and the involvement of RNA-induced silencing complex (RISC) components, warrant further investigation. Second, the impact of miR-7974 on other EBV lytic genes and the overall viral life cycle needs to be systematically explored. Third, the clinical relevance of miR-7974 expression levels in EBV-associated malignancies and their correlation with disease progression and patient outcomes should be investigated to validate its potential as a biomarker or therapeutic target.
Our study has certain limitations. While we have demonstrated the direct targeting of BZLF1 and BRLF1 by miR-7974, the broader impact of miR-7974 on host cellular pathways that may indirectly influence EBV reactivation remains to be fully elucidated. Additionally, further research is needed to explore the effects of miR-7974 on the protein levels of ZEBRA and Rta, as well as the downstream consequences on viral replication and particle production. Future studies should also investigate the interplay between miR-7974 and other miRNAs or host factors that may synergistically regulate EBV reactivation.