- Research Note
- Open access
- Published:
Epstein-Barr virus nuclear antigen 1 (EBNA1) increases the expression levels of MDM2 and MDM4 genes in HeLa cells: a review on MDM2 and MDM4 roles in cancer
BMC Research Notes volume 18, Article number: 221 (2025)
Abstract
Objective
Epstein-Barr virus nuclear antigen 1 (EBNA1) is a key viral protein expressed in all latency phases and EBV-associated tumors. It can modulate the expression of various host and viral genes. This study aimed to investigate the impact of EBNA1 on the expression levels of two cellular genes involved in p53 pathway regulation—MDM2 and MDM4—in HeLa cells. This investigation was conducted as part of our broader research on EBV-related oncogenic mechanisms.
Results
HeLa cells were transfected with either an EBNA1-expressing plasmid or a control plasmid. Gene expression levels of MDM2 and MDM4 were analyzed using real-time PCR. The results demonstrated a statistically significant increase in MDM4 expression in EBNA1-transfected cells compared to controls (p = 0.028). Although MDM2 expression was also elevated, the difference was not statistically significant (p = 0.11). These findings suggest that EBNA1 may play a role in cervical cancer development by upregulating genes that inhibit p53 tumor suppressor activity.
Introduction
Among females, cervical cancer (CC) is a commonly found disease that is connected to substantial levels of morbidity and mortality across the world [1]. There are numerous risk factors linked to the development of CC, and one of the primary causative factors is the infection of the human papillomavirus (HPV), which accounts for approximately 99.7% of all cases [2]. Additionally, various other factors have been identified as contributors to the increased risk of CC, such as smoking, immunosuppression, poor sexual health, and failure to attend regular screenings [3].
New clinical research has confirmed the association between Epstein–Barr virus (EBV) and high risk HPVs (HR-HPVs) in the development of CC [4, 5]. EBV, also identified as human herpesvirus-4, stands as the pioneering oncovirus in humans [6]. This virus is classified under the gamma-herpes viruses and is widely distributed among adults, primarily transmitted through saliva.
A published meta-analysis included 25 publications revealed that the pooled prevalence of EBV in CC was 43.63% [5]. This prevalence was significantly higher when compared to healthy controls, who had a prevalence of 19%. Additionally, the analysis found that the expression of EBV increased gradually from 27% in cervical intraepithelial neoplasia I (CIN I) to 35% in CIN2/3. The study also highlighted that EBV coinfection with HPV increased the risk of CC fourfold in EBV-positive women. Furthermore, EBV-positive women were found to have a higher incidence of precancerous cervical lesions compared to EBV-negative cases [5]. These findings suggest that EBV may actively contribute to the development and progression of CC, rather than being a passive participant.
Despite the fact that infected individuals serve as carriers of the EBV for the duration of their lives, it is worth mentioning that only a small proportion of these individuals actually experience the development of EBV-related cancers [7]. One of the most important late proteins of this virus, which is expressed in all viral latency phases, is EBV nuclear antigen-1 (EBNA1) [8]. It is indicated that EBNA1 as a transcription factor has the ability to alter the expression of genes upstream of both viral and cellular promoters [8].
Murine double minute 2 (MDM2) and MDM4 (MDMX) are two of the main regulators of p53 functions [9]. The p53 protein is known to be monoubiquitinated by MDM2, while polyubiquitination of p53 necessitates the development of a heterodimer between MDM2 and MDM4 [9]. MDM2 functions as an oncogene, and it is shown to be overexpressed in a range of human malignancies [10]. By engaging the transactivation domain and directly binding and degrading p21, MDM4 can effectively block p53 transcriptional activity [11]. In this regards, AlQarni et al. declare the connection between EBV infection and increased MDM2 expression [12]. Moreover, Hashemi and colleagues have recently documented the upregulation of MDM4 and MDM2 in MKN-45 cells subsequent to EBNA1 transfection [13].
Therefore, this research aimed to analyze how the EBV-EBNA1 protein affects the expression patterns of the cellular p53 inhibitory genes MDM2 and MDM4 in a CC cell line.
Methods
Characterization of EBNA1-containing and control plasmids: transformation, verification, and quality assessment
In this study, we employed the pCEP4 plasmid, an EBV-based plasmid that includes the EBNA1 gene and a hygromycin B resistance gene, as well as a control plasmid that does not contain the EBNA1 gene. These plasmids were transformed into Top10 strain of Escherichia coli and propagated. To confirm the presence of the EBNA1 gene in the plasmid, we used enzyme digestion and colony PCR techniques. Following this verification, the plasmids were extracted, and their quality and concentrations were assessed using gel electrophoresis and spectrophotometry.
Establishment of stable EBNA1-expressing HeLa cell lines via transfection and hygromycin B selection
HeLa cells, a cervical adenocarcinoma cell line harboring HPV-18, were cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) at 37 °C with 5% CO2 in a 6-well plate. Upon reaching approximately 70% confluence, one group of cells was transfected with an EBNA1-containing plasmid, while another group received a mock plasmid. The transfection was performed using an optimized concentration of DNA-fectamine (BioBasic, Canada). Twenty-four hours post-transfection, the cells were exposed to 350 µg/ml hygromycin B to select for those with stable EBNA1 expression. These selected cells were subsequently cultured for 20 days with hygromycin B, undergoing multiple passages.
Extraction of total RNA, synthesis of cDNA, and confirmation of EBNA1 gene expression
Total RNA was extracted from selected HeLa cells using an RNA Isolation Kit (Dena Zist, Mashhad, Iran). The quality and quantity of the extracted RNA were evaluated through electrophoresis and spectrophotometry, respectively. RNase-free DNase (Sinaclon, Tehran, Iran) was used to remove any plasmid contamination. cDNA synthesis was performed using an EasycDNA Synthesis Kit (AddScript RT-PCR SYBR Master, AddBio, Sweden) with 1000 ng/µl of RNA from each sample. Finally, real-time PCR was utilized to confirm the expression of the EBNA1 gene.
Primer design and quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis
expression levels of the cellular genes MDM2, and MDM4 were evaluated using a qRT-PCR assay on an ABI Quant Studio 3TM (Applied Biosystems, Grand Island, NY, USA). The thermal cycling protocol included an initial denaturation at 95 °C for 15 min, followed by 40 cycles of denaturation at 95 °C for 15 s and annealing/extension at 62 °C. The beta-actin gene served as the reference gene for normalization (Table 1) [14].
Normalization of qRT-PCR Ct values and statistical analysis
Normalization of Ct values from the qRT-PCR runs was performed using the CtNorm algorithm, available at http://ctnorm.sums.ac.ir [15]. The means were compared using the Mann-Whitney U test in GraphPad Prism software, with statistical significance defined as a P-value of less than 0.05.
Results
Real-time PCR results of MDM4 gene expression following EBNA1 transfection
MDM4 cellular gene expression level was compared between EBNA1-transfected cells and controls (Fig. 1). Real-time PCR analysis indicated a significant three-fold increase in the expression level of this gene in the presence of EBNA1 (p = 0.028).
Real-time PCR results of the MDM2 gene expression following EBNA1 transfection
In Fig. 1, the expression of MDM2 gene was compared between EBNA1 transfected cells and control cells. Although EBNA1-transfected HeLa cells revealed a two-fold increase in the expression of this gene, this change was not significant (p = 0.11).
Discussion
An increased risk of CC has been observed in individuals with EBV/HR-HPV coinfection [5]. Studies indicate a possible collaboration between the EBV and the progression of CC, as evidenced by various research findings [16]. Among the different proteins encoded by EBV, EBNA1 is unique in being present across all viral latency stages. By binding to gene promoters, EBNA1 has the ability to influence the expression of a wide range of genes [8, 17].
In this study, we found that EBNA1 upregulated MDM2 oncogene expression in EBNA1-transfected HeLa cells compared to mock-transfected controls. Hashemi and colleagues demonstrated that EBNA1 has the ability to significantly increase the expression of p53-inhibiting genes such as MDM2 and MDM4 in a Burkitt’s Lymphoma Cell Line [18]. MDM2 is a crucial negative regulator of the p53 tumor suppressor protein. It acts as an E3 ubiquitin ligase, targeting the N-terminal trans-activation domain (TAD) of p53 for ubiquitination, and also serves as an inhibitor of p53’s transcriptional activation [19, 20]. Based on a study by Gnanasundram et al., EBV can modulate the MDM2-P53 pathways via EBNA1-induced mRNA translation stress [21]. A 2018 study further demonstrated that MDM2 is overexpressed in EµEBNA1 tumor cells, with its levels being associated with EBNA1 expression [22]. In this context, Alipour et al. observed that EBNA1 can increase the expression of HPV-18 E6 and E7 oncogenes, along with several cellular genes, in the HeLa cell line [23]. Our studies have shown that EBNA1 can boost MDM2 gene expression in cervical cells, reduce p53 activity, prevent cell death in infected cells, and contribute to the pathogenesis of EBV in CC.
We also found that HeLa cells transfected with the EBNA1 plasmid exhibited significantly higher levels of MDM4 transcripts compared to cells transfected with a mock plasmid. This gene encodes a nuclear protein that features a p53 binding domain at the N-terminus and a RING finger domain at the C-terminus, displaying structural similarities to the p53-binding protein MDM2. Both proteins interact with the p53 tumor suppressor protein to inhibit its activity and have been found to be overexpressed in various human cancers [24]. Unlike MDM2, which targets p53 for degradation, this protein inhibits p53 by binding to its transcriptional activation domain. Additionally, it interacts with MDM2 through its RING finger domain, preventing the degradation of MDM2. So MDM4 can reverse MDM2-targeted degradation of p53 while maintaining suppression of p53 transactivation and apoptotic functions [25, 26]. Consistent with our findings, Hashemi et al. showed that the presence of EBV-EBNA1 protein in MKN-45 cells led to a significant increase in the expression of both MDM2 and MDM4 genes [13]. Consequently, the overexpression of MDM2 and MDM4 mediated by EBNA1 may be associated with p53 suppression in CC cells.
Although this study focused on HeLa cells, which are HPV-18 positive, we acknowledge that the use of a single cell line may limit the generalizability of our findings. To determine whether EBNA1-mediated upregulation of MDM2 and MDM4 is cell-line-specific or influenced by HPV subtype, future studies should examine additional cervical cancer cell lines. These include HPV-16–positive lines such as CaSki or HCB-514, and other HPV-18–positive lines like TMCC-1. Such comparative analyses could clarify whether the observed regulatory effects are broadly applicable across HPV-driven cervical cancers or specific to certain genetic backgrounds.
On the basis of these results, we hypothesized that EBNA1 would be related to the degree of MDM2 and MDM4 expression as well as the likelihood of developing CC.
E6 from high-risk HPV types is an oncoprotein that promotes host cell transformation by targeting p53 for degradation [27]. Additionally, studies have indicated that the E6 protein from HPV-16 hinders p53 function, which aids in tumor progression [28]. Our findings also reveal that EBNA1 expression significantly elevates the levels of two p53-inhibiting genes, MDM4 and MDM2, suggesting that EBNA1, along with the E6 oncoprotein, may further suppress p53. Supporting this, our previous research demonstrated that the EBV-EBNA1 protein can enhance the expression of HPV-18 E6 and E7 oncogenes [29].
In this study, we confirmed the elevated expression of two important cellular p53 inhibitory genes at the mRNA level by real-time PCR technique. Previous studies have shown that genetic variants in the MDM2 gene may influence p53 activity and cancer susceptibility [30,31,32,33]. Researchers have suggested that this genetic polymorphism may contribute to an increased risk of cancer [34]. For instance, Hong et al. demonstrated that the 309GG genotype of MDM2 is linked to a higher risk of esophageal squamous cell carcinoma [35]. Additionally, the 309G allele has been reported to be associated with the early diagnosis of estrogen receptor-positive breast cancer [36]. Furthermore, the presence of the 285 C allele of rs117039649 has been linked to a reduced risk of breast, ovarian, and endometrial cancer (EC) in patients with the 309G allele, indicating that it might counteract the effect of SNP309 in MDM2 [36].
Several studies have also indicated that these two significant polymorphisms, rs2279744 and rs117039649, may be linked to an increased susceptibility to gynecological cancers (GCs) [37, 38]. In a meta-analysis, Zhang et al. demonstrated that both rs2279744 (SNP309) and rs117039649 (SNP285) are associated with the risk of GCs. Additionally, researchers in another meta-analysis found that polymorphisms at the rs2279744 loci of the MDM2 gene are linked to an increased risk of EC [39]. The del1518 variant has been reported to be associated with a reduced risk of EC among individuals with the SNP309TT genotype, in both dominant and recessive models [40]. In a hospital-based case-control study, Miedl et al. reported that the SNP285 and SNP309 genetic variants of the MDM2 gene are associated with an increased risk of breast cancer (BC) in Central European women. Moreover, these polymorphisms have also been linked to an earlier onset of BC and its prognosis [31].
The expression of MDM2 and MDM4 proteins is being investigated across various cancer cell lines and tissues. In this line, it has been confirmed that the MDM4 protein is overexpressed in several human tumor cell lines [41, 42]. Additionally, tissues from soft-tissue sarcomas exhibit significantly higher levels of MDM4 protein compared to healthy controls [43]. Moreover, Danovi et al. revealed overexpression of the MDM2 protein in BC tissues [44]. Analysis of MDM2 and MDM4 proteins also showed higher steady state levels in retinoblastoma than control group [31].
Taken together, it is now evident that in many human cancers, p53 activity is inhibited by high levels of MDM proteins. Therefore, inactivating MDM proteins with therapeutic molecules might be a valuable strategy for tumor treatment [32]. Several studies suggest that small-molecule inhibitors targeting MDM2 and MDM4 could restore p53 activity and offer therapeutic benefits [11, 45].
Other inhibitors of MDM4 and MDM2, such as 5-fluorouracil, have also demonstrated anti-tumor activity in colon and gastric cancer cells [46]. Researchers found that malignant rhabdoid tumor (MRT) cells were particularly sensitive to MDM2 inhibition by idasanutlin (specific to MDM2) and to dual inhibition of MDM2/4 by ATSP-7041, compared to other p53 wild-type cancer cell lines. These compounds led to a significant upregulation of the p53 pathway in MRT cells, and this sensitivity was eliminated by CRISPR-Cas9-mediated inactivation of TP53 [32]. Furthermore, park et al., found that simultaneous inhibition of both MDM2 and MDM4 in virus-positive Merkel cell carcinoma led to an increased p53 response [47]. It has also been reported that trametinib, which inhibits MDM4/MDM2, enhanced G1 cell cycle arrest and induced apoptosis in colon and gastric cancer cells [26]. Recently, Langenbach et al. demonstrated that MDM2 inhibitors induce the production of MHC-II and IL-15, suggesting a novel approach to disrupting the immunosuppressive tumor microenvironment [48].
Limitations
-
The study was conducted using only a single cervical cancer cell line (HeLa), which may not fully represent the biological diversity of HPV-positive or HPV-negative cervical cancers.
-
Normal cervical epithelial cells were not included as a control group, limiting our ability to compare gene expression levels against a physiological baseline.
-
Gene expression analysis was restricted to the mRNA level using real-time PCR. No protein-level validation (e.g., Western blotting or immunohistochemistry) was performed to confirm whether changes in transcription translated into functional protein changes.
-
Functional assays were not included in this study. Therefore, the biological consequences of EBNA1-induced gene expression changes—such as their effects on cell proliferation, apoptosis, or migration—remain to be determined.
-
Only a limited number of target genes were evaluated. A broader transcriptomic or proteomic analysis would be valuable to identify other potential pathways regulated by EBNA1.
-
As this was an in vitro study, the results may not fully reflect in vivo conditions. Further animal model or tissue-based studies are needed to validate these findings in the context of cervical cancer progression.
Conclusion
In conclusion, our results suggest that the EBV-EBNA1 protein can elevate the expression levels of the MDM2 and MDM4 genes in HeLa cells. This leads to significant suppression of the p53 protein and enhanced cancer cell survival, potentially accelerating the onset of CC. Additionally, findings from other studies highlight the critical role of MDM2 and MDM4 in cancer development, indicating that inhibiting their function with various molecules could be considered a therapeutic strategy for treating different types of cancer. Further research is recommended to clarify these findings.
Data availability
The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
References
Luckett R, Feldman S. Impact of 2-, 4-and 9-valent HPV vaccines on morbidity and mortality from cervical cancer. Hum Vaccines Immunotherapeutics. 2016;12(6):1332–42.
Okunade KS. Human papillomavirus and cervical cancer. J Obstet Gynaecol. 2020;40(5):602–8.
Cohen PA, et al. Cervical cancer. Lancet. 2019;393(10167):169–82.
Shi Y, et al. Co-infection of Epstein-Barr virus and human papillomavirus in human tumorigenesis. Chin J Cancer. 2016;35:16.
de Lima MAP, et al. Association between Epstein-Barr virus (EBV) and cervical carcinoma: A meta-analysis. Gynecol Oncol. 2018;148(2):317–28.
Mui UN, Haley CT, Tyring SK. Viral oncology: molecular biology and pathogenesis. J Clin Med. 2017;6(12):111.
Blanco R et al. Role of Epstein-Barr virus and human papillomavirus coinfection in cervical cancer: epidemiology, mechanisms and perspectives. Pathogens, 2020. 9(9).
Jiang L, et al. EBNA1-targeted inhibitors: novel approaches for the treatment of Epstein-Barr virus-associated cancers. Theranostics. 2018;8(19):5307–19.
Wang X, Jiang X. Mdm2 and MdmX partner to regulate p53. FEBS Lett. 2012;586(10):1390–6.
Rayburn E, et al. MDM2 and human malignancies: expression, clinical pathology, prognostic markers, and implications for chemotherapy. Curr Cancer Drug Targets. 2005;5(1):27–41.
Li Q, Lozano G. Molecular pathways: targeting Mdm2 and Mdm4 in cancer therapy. Clin Cancer Res. 2013;19(1):34–41.
AlQarni S et al. Lymphomas driven by Epstein-Barr virus nuclear antigen-1 (EBNA1) are dependant upon Mdm2. (1476–5594 (Electronic)).
Hashemi SMA, et al. A new insight into p53-Inhibiting genes in Epstein–Barr Virus-Associated gastric adenocarcinoma. Iran Biomed J. 2023;27(1):4–4.
Dowran R, et al. Analysis of TLR7, SOCS1 and ISG15 immune genes expression in the peripheral blood of responder and non-responder patients with chronic hepatitis C. Gastroenterol Hepatol Bed Bench. 2017;10(4):272.
Ramezani A. CtNorm: real time PCR cycle of threshold (Ct) normalization algorithm. J Microbiol Methods. 2021;187:106267.
Blanco R, et al. Role of epstein-barr virus and human papillomavirus coinfection in cervical cancer: epidemiology, mechanisms and perspectives. Pathogens. 2020;9(9):685.
Canaan A, et al. EBNA1 regulates cellular gene expression by binding cellular promoters. Proc Natl Acad Sci U S A. 2009;106(52):22421–6.
Hashemi SMA, et al. EBNA1 upregulates P53-Inhibiting genes in Burkitt’s lymphoma cell line. Rep Biochem Mol Biology. 2023;11(4):672.
Momand J, et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell. 1992;69(7):1237–45.
Oliner JD, et al. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature. 1993;362(6423):857–60.
Gnanasundram SV, et al. MDM2’s dual mRNA binding domains co-ordinate its oncogenic and tumour suppressor activities. Nucleic Acids Res. 2020;48(12):6775–87.
AlQarni S, et al. Lymphomas driven by Epstein-Barr virus nuclear antigen-1 (EBNA1) are dependant upon Mdm2. Oncogene. 2018;37(29):3998–4012.
Alipour AH, et al. Epstein-Barr virus nuclear antigen 1 increases the expression of viral oncogenes and cellular genes in the HeLa cell line. Int J Mol Cell Med. 2022;11(4):346.
Marine JC, Jochemsen AG. MDMX (MDM4), a promising target for p53 reactivation therapy and beyond. Cold Spring Harb Perspect Med, 2016. 6(7).
Cheng Q, Chen J. Mechanism of p53 stabilization by ATM after DNA damage. Cell Cycle. 2010;9(3):472–8.
Wang X, et al. Enhanced G1 arrest and apoptosis via MDM4/MDM2 double knockdown and MEK Inhibition in wild–type TP53 colon and gastric cancer cells with aberrant KRAS signaling. Oncol Lett. 2021;22(1):1–12.
Yim EK, Park JS. The role of HPV E6 and E7 oncoproteins in HPV-associated cervical carcinogenesis. (2005–9256 (Electronic)).
Chang YF et al. HPV16 E6 promotes the progression of HPV Infection-Associated cervical Cancer by upregulating Glucose-6-Phosphate dehydrogenase expression. (2234-943X (Print)).
Alipour AH, et al. Epstein-Barr virus nuclear antigen 1 increases the expression of HPV type 18 E6 and E7 oncogenes and BIRC5/ C-MYC cellular genes in the Hela cell line. Ijmcmed. 2022;11(4):0–0.
Zhang J, Zhang Y, Zhang Z. Association of rs2279744 and rs117039649 promoter polymorphism with the risk of gynecological cancer: a meta-analysis of case–control studies. Medicine. 2018;97(2):e9554.
Miedl H, et al. Association of the MDM2 SNP285 and SNP309 genetic variants with the risk, age at onset and prognosis of breast cancer in central European women: A hospital-based case-control study. Int J Mol Sci. 2019;20(3):509.
Howard TP, et al. MDM2 and MDM4 are therapeutic vulnerabilities in malignant rhabdoid tumors. Cancer Res. 2019;79(9):2404–14.
Bond GL, et al. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell. 2004;119(5):591–602.
Walsh CS, et al. Association between a functional single nucleotide polymorphism in the MDM2 gene and sporadic endometrial cancer risk. Gynecol Oncol. 2007;104(3):660–4.
Hong Y, et al. The role of P53 and MDM2 polymorphisms in the risk of esophageal squamous cell carcinoma. Cancer Res. 2005;65(20):9582–7.
Karni-Schmidt O, Lokshin M, Prives C. The roles of MDM2 and MDMX in cancer. Annu Rev Pathol. 2016;11:617–44.
Xue Z, Zhu X, Teng Y. Relationship between murine double minute 2 (MDM2) T309G polymorphism and endometrial cancer risk: a meta-analysis. Med Sci Monitor: Int Med J Experimental Clin Res. 2016;22:3186.
Ma Y-Y, et al. The MDM2 309T > G polymorphism and ovarian cancer risk: a meta-analysis of 1534 cases and 2211 controls. PLoS ONE. 2013;8(1):e55019.
Das AP, et al. Meta-analysis of 49 SNPs covering 25,446 cases and 41,106 controls identifies polymorphisms in hormone regulation and DNA repair genes associated with increased endometrial Cancer risk. Genes. 2023;14(3):741.
Gansmo LB, et al. MDM2 promoter polymorphism del1518 (rs3730485) and its impact on endometrial and ovarian cancer risk. BMC Cancer. 2017;17:1–6.
Ramos YF, et al. Aberrant expression of HDMX proteins in tumor cells correlates with wild-type p53. Cancer Res. 2001;61(5):1839–42.
Stad R, et al. Hdmx stabilizes Mdm2 and p53. J Biol Chem. 2000;275(36):28039–44.
Bartel F, et al. Significance of HDMX-S (or MDM4) mRNA splice variant overexpression and HDMX gene amplification on primary soft tissue sarcoma prognosis. Int J Cancer. 2005;117(3):469–75.
Danovi D, et al. Amplification of Mdmx (or Mdm4) directly contributes to tumor formation by inhibiting p53 tumor suppressor activity. Molecular and cellular biology; 2004.
Vassilev LT, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science. 2004;303(5659):844–8.
Imanishi M, et al. Augmented antitumor activity of 5-fluorouracil by double knockdown of MDM 4 and MDM 2 in colon and gastric cancer cells. Cancer Sci. 2019;110(2):639–49.
Park DE et al. Dual inhibition of MDM2 and MDM4 in virus-positive Merkel cell carcinoma enhances the p53 response. Proceedings of the National Academy of Sciences. 2019;116(3):1027–1032.
Langenbach M, et al. MDM2 Inhibition enhances immune checkpoint inhibitor efficacy by increasing IL15 and MHC class II production. Mol Cancer Res. 2023;21(8):849–64.
Acknowledgements
The authors would like to express their gratitude to Shiraz University of Medical Sciences for providing the necessary facilities and resources to conduct this study. Additionally, we appreciate the support of our colleagues for their insightful discussions and technical assistance.
Funding
This research received no specific grant from any funding agency, commercial, or not-for-profit sectors.
Author information
Authors and Affiliations
Contributions
SMA.H. and AH.A. contributed equally to conducting the research and writing the manuscript, S.E. assisted in data collection and statistical analysis, A.F. contributed to laboratory experiments and result validation, J.S. supervised the study, provided conceptual guidance, and finalized the manuscript. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Hashemi, S.M.A., Alipour, A.H., Emamifar, S. et al. Epstein-Barr virus nuclear antigen 1 (EBNA1) increases the expression levels of MDM2 and MDM4 genes in HeLa cells: a review on MDM2 and MDM4 roles in cancer. BMC Res Notes 18, 221 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07275-3
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13104-025-07275-3