Introduction
Pediatric acute lymphoblastic leukemia (pALL) is a highly aggressive and life-threatening hematological cancer in children [
1]. Despite notable progress in improving survival rates, multidrug resistance (MDR) remains a major challenge in pALL treatment [
2]. Several mechanisms have been identified or proposed as contributory factors to cancer development, among which dysregulation of miRNAs emerges as a pivotal determinant [
3]. miR-326, a tumor suppressor miRNA, plays a role in tumor induction through regulating ABC transporters and signaling molecules involved in apoptosis and cell cycle [
4,
5]. Downregulation of miR-326 in pALL patients was associated with adverse outcomes, including overall survival and disease progression, particularly in cases of relapse and MDR [
6].
The P53 signaling pathway plays a crucial role in the development of pALL.
TP53 mutations are associated with poor treatment response and reduced overall survival rates in pALL. However, these mutations are rare in pediatric B-cell precursor pALL, occurring in only 2–4% of cases at diagnosis and 12% during relapse. Consequently, P53 inactivation is more likely due to regulatory abnormalities rather than
TP53 mutations [
7,
8]. Significantly, P53 plays a pivotal role in regulating cellular apoptosis through essential interactions with key effectors, such as
Bcl-xl,
Bcl-2, and
Bax [
9]. Furthermore, the induction of cell cycle arrest can be attributed to the P53-dependent activation of P21 or the suppression of
CCND1 through its interference with the Hedgehog (Hh) signaling pathway, which is widely recognized as a crucial pathway in the development of cancer stem cells. Notably, our previous research effectively elucidated a remarkable correlation between the upregulation of
SMO, a pivotal constituent of the Hh pathway, and the development of resistance in pALL [
10].
The objective of the present study was to provide, for the first time, in silico and empirical evidence substantiating the regulatory network of miR-326 in pALL and its possible cross talk with the tumor suppressor gene TP53. The outcomes have the potential to facilitate the identification of molecular therapeutic targets associated with pALL.
Materials and methods
Multilayer functional enrichment analysis of miR-326 interactions networks data collection
The PubMed database was searched for articles published until October 15, 2022, containing experimental data related to has-miR-326 (URS00000A939F_9606). The search utilized keywords such as "miR-326", "hsa-miR-326," and "microRNA-326," resulting in a total of 391 articles. Selection criteria were then applied, including reporter assay, western blot, and RT-qPCR evidence. The final selection process included articles that described experimentally verified interactions between miR-326 and mRNA transcripts of proteins, lncRNAs, or circRNAs, irrespective of cancer type or biological process. After screening, 207 articles were chosen for further annotation, all of which satisfied the specified selection criteria and provided sufficient experimental evidence of miR-326 interactions (Supplemental Table
S1).
Functional enrichment analysis of miR-326: targets interaction network
The miR-326 target interaction network was constructed using 118 unique experimentally verified interactions from the selected 207 articles. Functional enrichment analysis was performed to gain insights into the biological processes associated with miR-326's target interactions.
For the analysis, Cytoscape plugins, BiNGO [
11], and GOlorize [
12] were utilized. The BiNGO settings selected for analysis included 'Cluster from Network', 'Overrepresentation', and 'No Visualization'. The Hypergeometric statistical test was employed to determine the significance of enriched categories, and the Benjamini & Hochberg false discovery rate (FDR) correction was applied to account for multiple testing. Enriched categories with an FDR-adjusted P value ≤ 0.05 were considered significant.
The reference set for functional enrichment analysis consisted of human entities annotated to the Gene Ontology (GO) term 'biological process' or its descendants. The Gene Ontology file (go-basic.obo) (released 22nd December 2022) was obtained from the GO Consortium website (
http://geneontology.org/page/download-ontology). Additionally, gene association files for human proteins (goa human.gaf) and ncRNAs (goa human rna.gaf) were downloaded from the European Bioinformatics Institute ftp site (
ftp://ftp.ebi.ac.uk/pub/databases/GO/goa/HUMAN/) (released 4th May 2022) and merged into a single file named 'gene association.goa human'. No evidence codes were excluded during the analysis process to ensure comprehensive coverage.
Identifying miR-326 regulatory networks related to ALL
We conducted a kinases-TF regulatory network analysis to identify regulators of miR-326 target genes in ALL. The analysis included a transcription factor (TF) enrichment analysis using the ChEA tool from the Enrichr database and a kinase enrichment analysis using the KEA tool of Enrichr [
13]. Both analyses used a statistical significance threshold of adjusted P value ≤ 0.05. Additionally, we obtained STRING network with ALL associated protein from the STRING disease query database with confidence score of 0.4 and maximum number of proteins 1500 [
14]. By performing a Venn diagram analysis on the enriched TFs, kinases, and ALL-related networks, we identified a shared gene set, providing insights into potential regulatory mechanisms influenced by miR-326 in ALL. ClueGO plug-in was used to perform GO functional enrichment and KEGG pathway enrichment analysis of the differentially expressed genes (DEGs) in Cytoscape.
Visualization and analysis of the miR-326-associated ceRNA network
The miR-326-associated ceRNA network was visualized and analyzed using Cytoscape. The network integration included experimentally validated data from luciferase assays for circRNA: miR-326, lncRNA: miR-326, and miR-326: mRNA interactions. Additionally, protein–protein interactions from the STRING algorithm were incorporated to illustrate the interactions between miR-326 and circRNAs, lncRNAs, and mRNAs.
Cell culture and transfection
The RN95 cells, a TP53 wild-type precursor B-ALL cell line, were previously established and authenticated in our laboratory [
15]. These cells were cultured in RPMI-1640 medium (Bio Idea, Tehran, Iran) supplemented with 10% heat-inactivated FBS (Fetal Bovine Serum) (Bio Idea, Tehran, Iran) and 1% penicillin/streptomycin, in a humidified incubator at 37 °C under 5% CO2. The electroporation system (Eppendorf Multiporator®, Germany) was used to transfect RN95 cells with 250 nM of the miR-326 mimic and scrambled miR-326 negative control (referred to as scrambled control, Exiqon, Denmark). In brief, during the exponential growth phase, cells were suspended in RPMI-1640 medium supplemented with 0.5% FBS and 2.5 × 106 cells were transfected under optimized conditions (250 V, 40 ms width and one pulse). Mock control cells were transfected in the same conditions without any oligonucleotide. Following electroporation, the transfected cells were placed in 6-well plates (2.5 × 106 cells/well) and allowed to recover in 4 ml of RPMI 1640 supplemented with 10% FBS at 37 °C and 5% CO2. The transfected cells were used for further analysis at 48 h after transfection.
Total RNA was extracted from the transfected cells, 48 h after transfection, using TRIzol reagent (Invitrogen, Carlsbad, CA) according to the manufacturer’s protocol. cDNA synthesis and RT-qPCR were then carried out to identify relative gene expression levels of miR-326 as previously described [
6]. Primers for miR-326 and RNU6 small nuclear RNA (as endogenous control for data normalization) were purchased from Qiagen (Exiqon, Denmark) as part of their validated primer sets. The exact sequences are proprietary but have been validated by Qiagen for RT-qPCR experiments. After cDNA synthesis using PrimeScript™ RT reagent kit (Takara, Tokyo, Japan), RT-qPCR was performed to assess relative gene expression levels of
ABCC1,
ABCB1,
ABCA3,
CCND1,
SMO,
Bcl-xl,
Bcl-2,
Bax,
SNHG1, and H19 using SYBR Premix Ex TaqII (Takara, Tokyo, Japan) and the specific primers as previously described [
10,
16,
17]. GAPDH was used as a house keeping gene for data normalization (Supplementary Table
S2).
Western blotting
Forty-eight hours after transfection, total protein was extracted using a homemade lysis buffer. The buffer was prepared by dissolving 144 g Glycine, 30 g Trisbase, and 10 g SDS in 800 ml dH2O, adjusting the volume to 1 L without pH adjustment (final pH ~ 8.3). The buffer was stored at room temperature for up to six months and diluted to 1 × with dH2O before use. The protein concentration was quantified by Bradford assay (Bio-Rad) according to the standard protocol.Equal amounts of total proteins (35 µg) were separated by SDS–PAGE (Sodium Dodecyl Sulfate–Polyacrylamide Gel Electrophoresis) using gradient gel and transferred to nitrocellulose membranes (Amersham Biosciences). After blocking with 10% BSA (Bovine Serum Albumin) (w/v) in TBST (Tris-Buffered Saline with Tween-20) for 1 h, membranes were incubated at 4 °C overnight with the mouse anti-human primary antibodies, Mdm2 (1:300, Merck Millipore), p21 (1:100, Calbiochem), p53 (1:250, Santacruz Biotechnology, INC.), and Actin (1:250, Santacruz Biotechnology, INC). Membranes were then treated with goat anti-mouse IgG-HRP (Horseradish Peroxidase) secondary antibody (1:1000, Dako) at room temperature for 1 h. Finally, proteins were visualized on sensitive X-ray films in a dark room using ECL (Enhanced Chemiluminescence) solution (Bio-rad, Hercules, CA). The visualized bands were quantified and analyzed for intensity using Image J software (National Institute of Health, USA). Band intensities were quantified using ImageJ software. Regions of interest for each band were carefully selected to exclude extra signals, and normalization was performed against Actin as the loading control. Despite minor saturation in the Actin signal, the analysis was conducted rigorously and in line with standard practices in Western blot experiments.
Statistical analysis
The data in this study were presented as mean ± standard error of mean. To compare multiple groups, ordinary one-way analysis of variance (One-Way ANOVA) was employed. The findings represent the outcomes of two independent experiments conducted in duplicate. Student's t-test was performed for pairwise comparisons between two groups. GraphPad Prism 9.3.1 (GraphPad Software, San Diego, CA) software was utilized for data analysis. A significance level of P value < 0.05 was employed to determine statistical significance.
Discussion
The present study offers a comprehensive examination of the functional implication of miR-326 in cellular processes and its regulatory networks within the context of ALL. The research is primarily focused on elucidating the gene ontology of miR-326 and its molecular targets, with two specific objectives in mind: first to illuminate the biological processes associated with drug resistance modulated by miR-326, and second, to demonstrate an overlap analysis of enriched functional gene sets (GO terms) linked to miR-326. The gene ontology analysis revealed a significant role for miR-326 in the regulation of signal transduction through the P53 class mediator. Notably, the P53 protein exerts a profound influence on various signaling pathways involved in multidrug resistance (MDR), including Hh signaling, apoptosis induction, cell cycle arrest, migration regulation, and adhesion modulation. Although there is no specific study that has directly explored the association between P53 and miR-326, the functional enrichment analysis of miR-326-target interactions suggested the involvement of miR-326 in the regulation of this signaling pathway (Figs.
1 and
2).
Gene ontology analysis of the miR-326 target interaction network also reveals involvement of
ABCC1 in cell migration, a key indicator of epithelial-mesenchymal transition (EMT) and MDR (Fig.
1). As an ATP-dependent membrane transporter,
ABCC1 not only enhances cancer cell progression but also contributes to MDR by facilitating cell migration [
20]. Our RT-qPCR findings further support these observations, demonstrating a significant downregulation of drug resistance-related genes
ABCC1 and
ABCB1, followed by the upregulation of miR-326 (Fig.
3). Notably, cancer stem cells (CSCs) are recognized contributors to drug resistance and disease recurrence, primarily due to their heightened expression of drug resistance-associated genes, including ABC transporters [
21‐
24]. Besides, the level of cellular P53 significantly regulates the expression of
ABCC1 and
ABCB1 [
25]. Such intricate interplay emphasizes the role of miR-326 in modulating key pathways associated with both cell migration and drug resistance, providing valuable insights into potential therapeutic interventions.
On the other hand, RT-qPCR demonstrated a downregulation of
SMO, the pivotal member of the Hh pathway, in RN95 cells transfected with exogenous miR-326 mimic (Supplemental Figure
S8). These results aligned with our prior investigations, which established a positive correlation between miR-326 and
SMO expression levels in patients diagnosed with B-ALL [
10]. Previous studies implicated increased Hh pathway activity and elevated
ABCC1 expression in the development of drug resistance in multiple myeloma cell lines. Furthermore, it was highlighted that components of the Hh signaling pathway play a critical role in promoting the survival of cancer stem cells [
26]. Collectively, it is hypothesized that the miR-326-mediated inhibition of ABC transporters could conceivably hinder disease recurrence and the retrieval of leukemia stem cells in B-ALL.
The Western blot analysis was utilized to investigate the interplay between miR-326 and P53 in the treatment-resistant pALL cell line, RN95. The results demonstrated that the introduction of exogenous miR-326-mimic led to an elevation in the expression levels of P53 (Fig.
4). Consequently, the heightened activity of P53 on the cell cycle was validated through increased expression levels of Mdm2 and P21 proteins, as well as the downregulation of
CCND1 gene (Fig.
4B, C). To substantiate the functional impact of P53 on apoptosis, the upregulation of apoptotic Bax and the reduced expression levels of antiapoptotic
Bcl-2 and
Bcl-xl genes were observed (Fig.
4C).
In silico studies revealed that miR-326 indirectly affects P53 by inhibiting the mRNA translation of AKT1/2 (Figs.
2 and
4D). The association between PI3K/AKT upregulation and unfavorable prognosis in pre-B-ALL, which includes drug resistance and decreased apoptosis, provides support for these findings [
27]. Additionally, prior research has demonstrated a decrease in phosphorylated AKT following transfection with the miR-326 mimic in pancreatic cancer [
28]. Consequently, we proposed that in the cytarabine-resistant RN95 cells, miR-326 may impede Mdm2 phosphorylation by inhibiting AKT1/2. This may prevent the nuclear translocation of Mdm2, thereby triggering P53 activation (Figs.
4C and
6). This hypothesis was substantiated by the analogous protective effect observed in drug-resistant ALL cells, where PTEN played a crucial role in safeguarding P53 activity through the suppression of PI3K, thus effectively inhibiting MDM2 [
29] (Also shown in Fig.
4D). Further investigation is imperative to elucidate the underlying mechanisms by which miR-326 exerts its influence on P53, a pivotal regulator of apoptosis.
Finally, the results from RT-qPCR and in silico assays demonstrated that miR-326 exerts a downregulatory effect on the stability of H19 and SNHG1 in B-ALL cells (Fig.
5). It is noteworthy that H19 has been implicated in leukemia cell proliferation and apoptosis, while SNHG1 has been associated with drug resistance in liver cancer [
30,
31]. In the context of CD133 + liver cancer stem cells, elevated H19 expression has been shown to correlate with increased levels of MDR1 and Gst-π proteins. Importantly, inhibition of H19 through the induction of ROS production has been observed to enhance apoptosis and impede the MAPK/ERK pathway, consequently reducing drug resistance in CD133 + CSCs [
32]. Further investigation is required to elucidate the underlying mechanisms responsible for the reduced expression of H19 and SNHG1.
Although we acknowledge that additional experiments, such as the use of miRNA inhibitors, would have strengthened our findings, the lack of sufficient funding and resources in the current phase prevented us from performing these experiments. We aim to address this limitation in future studies to better validate the role of ABC transporters. Additionally, although the MDM2-p53-p21 pathway was explored, due to constraints related to experiment repetition and antibody availability, the data presented here is limited. We plan to investigate this pathway further in subsequent experiments, where we can fully optimize the methodology. Additionally, it is necessary to assess the in vivo efficacy of miR-326.
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.