Abstract
Background and aims
Prostate adenocarcinoma (PRAD) is a complex disease that can be driven by alterations in both coding and noncoding genes. Recent research has identified coding and non-coding genes that are considered to play important roles in prostate cancer evolution and which may be used as biomarkers for disease diagnosis, prognosis, and treatment. TP53 is a critical hub gene in prostate cancer. Advanced studies have demonstrated the crosstalk between coding and non-coding RNAs, particularly microRNAs (miRNAs).
Methods
In this study, we investigated the roundabout of TP53 and their regulatory miRNAs (miR-15a-5p, miR-34a-5p, and miR-141-3p) based on the TCGA data set. We validated an additional patient cohort of 28 matched samples of patients with PRAD at tissue and plasma level.
Results
Therefore, using the UALCAN online database, we evaluated the expression level in PRAD of these genes revealing overexpression of TP53. qRT-PCR validation step endorsed the expression level for these genes. Additionally, we evaluated the expression level of the four key miRNAs (miR-15a-5p, miR-34a-5p, and miR-141-3p) interconnected as a network at tissue and plasma levels.
Conclusions
Through these results, we demonstrated the essential function of TP53 and its associated miRNAs that play a significant role in tumor control, highlighting miRNAs’ potential as future therapeutic targets and biomarkers with important implications in managing prostate cancer.
Keywords: prostatic neoplasms, adenocarcinoma, microRNAs, biomarkers
Introduction
Prostate adenocarcinoma (PRAD) is the second most common cause of cancer death among men, particularly in Western countries [1,2]. While prostate-specific antigen (PSA) screening in combination with prostate biopsy has considerably improved the early diagnosis of prostate cancer, the sensitivity remains reduced, particularly for those expressing low PSA values. The prostate biopsy is also extremely invasive and might entail complications.
Advanced studies have demonstrated the crosstalk between coding and non-coding RNAs, particularly mRNA-microRNAs (miRNAs). miRNAs are short-length transcripts comprising 19–25 nucleotides that are highly stable in cells and do not degrade easily as the mRNAs play an essential role in cancer tumorigenesis and progression field [3–6]. Due to their short-conserved sequence, they can alter the physiological function of the coding genes by targeting the complementary mRNA sequences. This potential miRNA specificity to target coding genes will continue to generate interest for their potential clinical applications.
In cancer, miRNA alterations relate to the modulation of several signaling pathways involved in different hallmarks of cancer, from early carcinogenesis to late metastasis events. Among these, miRNAs’ fine-tuning of the TP53 axis has been widely reported in the literature [7–9]. This is the case of miR-15a which focuses on multiple oncogenic targets [8]. TP53 is a critical hub gene in the prostate cancer [10–12]. Expression levels are correlated with the PRAD recurrence [10]. TP53 gene regulates cell proliferation and differentiation [5], and mutations in this gene occur in only a small percentage of cases. However, alterations in other genes and signaling pathways that are regulated by TP53 may also affect the evolution of prostate cancer [13]. miR-34 is a family of microRNAs involved in regulating cell proliferation, differentiation, and apoptosis. miR-34 is a downstream target of TP53 and is often dysregulated in cancer. In particular, miR-34a has been shown to be a tumor suppressor and is frequently downregulated in prostate cancer [14]. One study found that miR-141-3p promotes prostate cancer cell proliferation, migration, and invasion by targeting HKF9 [15]. In addition, miR-141-3p has been investigated as a potential biomarker for prostate cancer. Several studies have shown that miR-141-3p is present at higher levels in the plasma of patients with prostate cancer compared to healthy individuals or patients with benign prostatic hyperplasia (BPH). This suggests that miR-141-3p may have the potential as a non-invasive diagnostic and prognostic biomarker for prostate cancer. Overall, miR-141-3p appears to play a role in prostate cancer development and progression and may have the potential as a biomarker and therapeutic target for the disease [16,17].
This study investigated the relationship between TP53 and key regulatory miRNAs (miR-15a-5p, miR-34a-5p and miR-141-3p) identified on bioinformatics analysis and then validated on an additional patient cohort, tissue and plasma (Figures 1a and 1b).
Figure 1a.
Flow chart of the study.
Figure 1b.
Flow chart for the patients included in the study.
Methods
In silico analysis based on TCGA dataset-mining analysis in PRAD
We performed a bioinformatic analysis based on the TCGA databases. TCGA is a user-friendly web resource for analyzing cancer data, furnishing data to analyze directly on the platform graphs and plots outlining gene expression and survival curves. The UALCAN and STARBASE databases generated data on miRNAs and their target genes for subgroup analysis of clinical and pathological features (Gleason score 7–9) [18,19].
mRNA-miRNA interactions
To assess the mRNA-miRNA interactions, we used miRNet, a valuable online tool that permits visual exploration of target interaction in a biological network context [20,21].
qRT-PCR data validation of TCGA data analysis
Patients and sample collection (Table I)
Table I.
Clinical characteristics of PRAD patients included in the qRT-PCR clinical study (tissue and plasma).
Patient no. | Age | PSA | cTNM | L | V | R | Gleason Score |
---|---|---|---|---|---|---|---|
1 | 75 | 5.5 | 3aXX | 0 | 0 | 0 | 7(3+4) |
2 | 63 | 7 | 3a0X | 0 | 0 | 1 | 7(3+4) |
3 | 68 | 13 | 30X | 0 | 0 | 0 | 7(3+4) |
4 | 64 | 6 | 2c0C | 0 | 0 | 0 | 7(4+3) |
5 | 64 | 9.25 | 2XX | 0 | 0 | 0 | 7(4+3) |
6 | 64 | 47 | 3b0X | 0 | 1 | 1 | 7(4+3) |
7 | 64 | 7.45 | 2b0X | 0 | 0 | 0 | 7(3+4) |
8 | 60 | 9 | 2c0X | 0 | 0 | 0 | 7(3+4) |
9 | 65 | 7 | 20X | 0 | 0 | 0 | 7(3+4) |
10 | 76 | 5.53 | 2cXX | 0 | 0 | 0 | 7(3+4) |
11 | 68 | 15 | 3b1X | 1 | 0 | 1 | 7(4+3) |
12 | 65 | 78 | 2c0X | 0 | 0 | 0 | 7(4+3) |
13 | 64 | 4.9 | 3a0X | 0 | 0 | 1 | 7(3+4) |
14 | 70 | 19 | 3b1X | 1 | 0 | 1 | 7(3+4) |
15 | 66 | 4.26 | 2cXX | 0 | 0 | 0 | 7(3+4) |
16 | 65 | 8.3 | 3aXX | 0 | 0 | 0 | 7(3+4) |
17 | 71 | 25 | 3a0X | 0 | 0 | 1 | 7(4+3) |
18 | 65 | 5.75 | 2c0x | 0 | 0 | 0 | 7(3+4) |
19 | 63 | 4.49 | 3aXX | 0 | 0 | 0 | 7(3+4) |
20 | 76 | 4.5 | 2XX | 0 | 0 | 1 | 7(3+4) |
21 | 69 | 8.9 | 3a00 | 0 | 0 | 1 | 7(3+4) |
22 | 58 | 8.09 | 3bXX | 1 | 0 | 0 | 7(4+3) |
23 | 70 | 1.9 | 3a0X | 1 | 0 | 1 | 7(3+4) |
24 | 74 | 7 | 20x | 1 | 0 | 0 | 7(4+3) |
25 | 54 | 19 | 3b1X | 1 | 1 | 0 | 8(4+4) |
26 | 63 | 19.71 | 3b0X | 0 | 0 | 1 | 9(4+5) |
27 | 68 | 46 | 3b001 | 0 | 0 | 1 | 9(5+4) |
28 | 64 | 9.5 | 20X | 1 | 0 | 0 | 9(4+5) |
cTNM: clinical TNM classification, T: Tumor classification, N: Node classification, M: Metastatic classification, L: lymph involvement; V: Vessel involvement.
To validate the bioinformatic analysis, we initially collected samples from 71 patients with confirmed PRAD between 2018 – 2020 (Figure 1b). A database with clinical, pathological and molecular characteristics was created from all the patients. From each patient, tumor tissue and adjacent tissue were collected after the initial check by a pathologist. Additionally, blood, serum and plasma were collected. All the patients were selected from the Urological Surgery Department at the “Prof. Ion Chiricuta” Oncology Institute, Cluj-Napoca.
The tissues were collected immediately after the pathological examination and stored in liquid nitrogen at -170ºC. A trained pathologist selected the tissues to be further analyzed for molecular investigations, not to alter the pathological diagnosis of the tumors.
Two 5 ml vacutainers for peripheral blood collection (one with EDTA and one without anticoagulant), one for TriReagent and one for serum and plasma, were collected from all patients. All methods were performed to limit the blood hemolysis and exclusion from the cohort to be analyzed. The blood was prepared with TriReagent to conserve the mRNA and miRNA sequences. Coagulated blood was centrifuged at 3000 rpm for 10 minutes to obtain serum and plasma. We used RNAse-free tubes for serum and plasma storage at −80ºC. Repeated thawing of the frozen samples was avoided to limit the degradation of the biological material.
Ethical conditions
The present study followed all the regulations of the Declaration of Helsinki regarding human rights and informed consent for molecular biology research studies. Based on the data explained by the principal investigator, the “Prof. Ion Chiricuta” Institute of Oncology Ethical Committee approved the study with data anonymization for all results to be published. The approval number is 5991/26.06.2019.
A total of 28 patients were included in the qRT-PCR study; their data are shown in table I. The median age of the patients was 66.2±5.1 years old. All patients included were stage II and III, with a Gleason score between 7 and 9.
RNA isolation from tissue
The RNA isolation from tissue samples (tumor and normal adjacent) for genes and miRNAs was done using the TriReagent-based method, followed by the Nanodrop quality control step.
RNA isolation from plasma
For the RNA isolation from plasma samples (patients with PRAD and healthy subjects), we used Plasma/Serum Circulating and Exosomal RNA Purification kit (Slurry Format, cat no.42800, Norgene).
Gene expression quantification by qRT-PCR
The RNA was reverse-transcribed into cDNA for gene expression evaluation using a High-Capacity Reverse Transcription Kit (Applied Biosystems, Foster City, CA, USA). Subsequently, an amplification step has been performed using SYBR Select Master Mix (Applied Biosystems) on ViiATM7 System and specific primers for the target genes. The primer sequences are displayed in table II.
Table II.
Primer sequence and assay ID used for genes’ quantification.
Gene | Primer Sequences | |
---|---|---|
FW | RW | |
B2M | CACCCCCACTGAAAAAGATGAG | CCTCCATGATGCTGCTTACATG |
TP53 | CACCCCCACTGAAAAAGATGAG | CCTCCATGATGCTGCTTACATG |
TAZ | GCTGCAGACATCTGCTTCAC | TTCCCCTCATTCTCTGCTTG |
MALAT1 | AACTGCAGAGAGTTTGAGTGGTTTT | TGTCCTTATAGGCTGGCCATT |
The expression levels’ relative quantification was done using the 2−ΔΔCT method [3], and graphical representation was done using GraphPad Prism (version 9).
Tumor tissue/normal adjacent tissue and plasma miRNAs’ expression profiling
For miRNA quantification from tissue (tumoral versus adjacent normal tissue) and plasma (PRAD and healthy subjects miRNAs), we used 50 ng RNA for cDNA synthesis performed with TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems) and TaqMan Fast Advanced Master Mix (Applied Biosystems) on the same instrument, primers used for miRNA quantification are displayed in table III. The miRNAs were evaluated on the same patient cohort, and for normalization, U6 and RNU48 with the same method and graphical representation as for gene expression.
Table III.
Primer sequence and assay ID used for miRNAs’ quantification.
miRNA Assay | Sequence | Assay ID |
---|---|---|
miR-15a-5p | UAGCAGCACAUAAUGGUUUGUG | 000389 |
miR-34a-5p | UGGCAGUGUCUUAGCUGGUUGU | 000426 |
miR-141-3p | UAACACUGUCUGGUAAAGAUGG | 000463 |
U6 snRNA | GTGCTCGCTTCGGCAGCACATATACTAAAATTGGAACGATACAGAGAAGATTAGCATGGCCCCTGCGCAAGGATGACACGCAAATTCGTGAAGCGTTCCATATTTT | 001973 |
RNU48 | GATGACCCCAGGTAACTCTGAGTGTGTCGCTGATGCCATCACCGCAGCGCTCTGACC | 001006 |
Results
TP53 expression levels in PRAD
Using the UALCAN online tool, the analysis revealed that the mRNA expression level of TP53 has been overexpressed in PRAD versus normal adjacent tissue (Figure 2).
Figure 2. TP53 expression level in PRAD.
(A) The expression level of TP53, generated using the UALCAN online tool, graphical representation as “primary tumor” and normal adjacent tissue displayed as “normal” (* p ≤ 0.05).
Validation of TP53 gene by qRT-PCR
To validate the observed alteration of the TP53 genes in PRAD, qRT-PCR was performed, wherein B2M was used as an endogenous control for the normalization of qRT-PCR data using the 2−ΔΔCT method. These transcripts were validated using 28 matched pairs of tumor and adjacent normal tissues of PRAD with a Gleason score between 7–9, revealing the overexpression of the TP53 gene (Figure 3).
Figure 3.
TP53 expression levels in PRAD tumor tissue (TT, n=28) versus normal adjacent tissue (NT, n=28). Scatter plots demonstrate the overexpression of TP53 in tumor tissues versus normal adjacent tissues. To normalize the gene expression data, B2M data were analyzed using the 2−ΔΔCT method (**p≤ 0.01).
TP53 interactions network with key miRNAs in prostate cancer
TP53 was interconnected with key regulatory miRNAs, miR-15a-5p, miR-25-3p miR-34a-5p (Figure 4), via BCL2 genes, network interaction generated using miRNET online tool. miR-141-is interconnected indirectly to with TP53 via BCL2 genes.
Figure 4. TP53-miRNA interaction network.
mRNA-miRNA interaction network for TP53, a key suppressor gene revealed direct interconnection with miR-15a-5p, miR-34a-5p, indirectly via BCL2 with miR-141-3p.
Evaluation of the expression level of miR-15a, miR-34a-5p and miR-141-3p
We evaluated the expression level for miR-15a, miR-34a-5p and miR-141-3p, revealing an increased level in tumor tissue versus normal adjacent tissue in the TCGA patient cohort, with emphasis on no important alteration considering the Gleason score (Figure 5). Then the expression levels for these transcripts were validated in our patient cohort (Figure 6), data being in agreement with those from TCGA.
Figure 5.
miR-15a-5p, miR-34a-5p, and miR-141-3p expression level considering Gleason score in TCGA patient cohort, revealing overexpression of these transcripts for the cases with Gleason 6 and Gleason 7-1.
Figure 6.
miR-15a-5p, miR-34a-5p and miR-141-3p expression levels in tumor tissues (TT) versus normal adjacent tissues (TN) for cases with Gleason score 7–10. Scatter plots demonstrate the overexpression of miR-15a-5p, miR-34a-5p, and miR-141-3p in tumor tissues (TT) versus normal adjacent tissues (TN). For normalization of the gene expression data, U6 and RNU48 were analyzed based on the 2−ΔΔCT method (* p ≤ 0.05 and *** p ≤ 0.001).
To validate the selected miRNAs, we investigated the expression level in plasma of the same patients’ cohort versus healthy controls, confirming the increased expression in PRAD plasma versus healthy controls (Figure 7). Furthermore, these findings suggest that these miRNAs are useful both as biomarkers and therapeutic targets in PRAD.
Figure 7.
miR-15a-5p, miR-34a-5p and miR-141-3p expression levels at plasma levels in PRAD (n=28) versus healthy controls (n=30) by qRT-PCR. For normalization of the miRNA expression data, U6 and RNU4 were analyzed based on the 2−ΔΔCT method (* p ≤ 0.05, **p≤ 0.01**** p ≤ 0.0001).
Discussion
Although earlier studies have reported progress in elucidating the potential molecular mechanism in prostate cancer development, the fundamental knowledge of the TP53 network signaling remains undeciphered regarding its prognostic value and related biological processes in PRAD. Most of the studies are related to the mutation status of TP53 [10,22,23], with less information related to the expression level for this gene, as in the present study. However, further research is needed to fully understand the role of this axis in prostate cancer and develop effective therapeutic strategies.
Therefore, miR-141-3p has been proposed as a potential prostate cancer diagnosis, prognosis, and treatment biomarker. Its detection in blood or urine samples could be used as a non-invasive diagnostic or prognostic tool, and its inhibition may be a potential therapeutic strategy for prostate cancer treatment [15].
miR-15a-5p is presented in the literature as an important transcript related to the androgen signaling [24], representing an important therapeutic target for this pathology [25]. miR-15a is involved in multiple biological processes, overexpression of miR-15a [26,27] along with miR-16 inhibiting the TGFβ signaling pathway [28], regulated cell proliferation and invasion by Wnt/β-Catenin signaling pathway [29], or immune invasion and malignant progression of PRAD via up-regulating PD-L1 [30].
TP53 transcriptionally controls miR-34a expression, often altered in cancer [31]. miR-34a-5p transcript is overexpressed in PRAD. It correlated with STK4 (gene correlated with disease-free survival) [32,33]. miR-34a is transcriptionally regulated via TP63, TP73, and other transcription factors, such as STAT3, and MYC are involved [34]. The expression level may dynamically change in EMT, hypoxia, and inflammation [34]. The overexpression of this transcript mainly in high-grade tumors [26]. Meanwhile, other studies reveal the downregulation of this transcript [27,35].
Another key element is represented by miR-141 with prognostic value in PRAD [36]. Literature data report it to be related to outcomes and aggressive tumor characteristics in PRAD [16,37]. The high expression of this transcript at tissue and plasma levels is related to disease progression and metastatic disease [37]. MiR-141 was significantly higher in the plasma of patients with advanced PRAD than in the matched controls [38]. miR-141 was significantly up-regulated in the serum samples of metastatic tumors versus localized tumor samples [39]. Another study demonstrated the prognostic role of miR-141 along with miR-21 and miR-375 in the PCa diagnosis [40]. Despite this, the role of this transcript remains controversial in PRAD, considering the interconnection with epigenetic factors [41]. Overall, these findings suggest that the dysregulation of BCL2 and miR-141 may contribute to the development and progression of prostate cancer and that targeting these molecules may be a potential therapeutic strategy for the disease, but no study investigated the relationship between miR-141 and Bcl2 in prostate cancer as yet. Additional investigation needs to be done to validate this.
Conclusions
Although various gene expression studies have already been performed, there have not been any detailed analyses of TP53 and their targeting transcripts (miR-15a, miR-34a-5p, and miR-141-3p) that can be used for prognostics for overall survival in PRAD. The validation of these miRNAs in tumor tissues and liquid biopsy opens a new field of investigation in the near future, focusing on the genes targeted by these miRNAs and their potential role in the disease evolution and how this can be controlled. While at the beginning of their validation in liquid biopsies from cancer patients, these short noncoding sequences gain more territory to become concrete molecules as biomarkers. Overall, these findings suggest that a panel of coding and noncoding genes may be used as biomarkers for prostate cancer evolution and may help guide personalized treatment strategies for patients with this disease. Further research is needed to validate these findings and to develop effective diagnostic and therapeutic approaches based on this gene panel.
Acknowledgements
Vlad Horia Schitcu received an internal grant from the host institution no 7690/101/15.04.2016.
Footnotes
Informed Consent Statement
Informed consent was obtained from all subjects involved in the study.
References
- 1.Zhu S, Min Z, Qiao X, Chen S, Yang J, Zhang X, et al. Expression profile-based screening for critical genes reveals S100A4, ACKR3 and CDH1 in docetaxel-resistant prostate cancer cells. Aging (Albany NY) 2019;11:12754–12772. doi: 10.18632/aging.102600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209–249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 3.Groza IM, Braicu C, Jurj A, Zanoaga O, Lajos R, Chiroi P, et al. Cancer-Associated Stemness and Epithelial-to-Mesenchymal Transition Signatures Related to Breast Invasive Carcinoma Prognostic. Cancers (Basel) 2020;12:3053. doi: 10.3390/cancers12103053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Busuioc C, Ciocan-Cartita CA, Braicu C, Zanoaga O, Raduly L, Trif M, et al. Epithelial-Mesenchymal Transition Gene Signature Related to Prognostic in Colon Adenocarcinoma. J Pers Med. 2021;11:476. doi: 10.3390/jpm11060476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Braicu C, Pileczki V, Irimie A, Berindan-Neagoe I. p53siRNA therapy reduces cell proliferation, migration and induces apoptosis in triple negative breast cancer cells. Mol Cell Biochem. 2013;381:61–68. doi: 10.1007/s11010-013-1688-5. [DOI] [PubMed] [Google Scholar]
- 6.Jurj A, Zanoaga O, Braicu C, Lazar V, Tomuleasa C, Irimie A, et al. A Comprehensive Picture of Extracellular Vesicles and Their Contents. Molecular Transfer to Cancer Cells. Cancers (Basel) 2020;12:298. doi: 10.3390/cancers12020298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Pudova EA, Krasnov GS, Nyushko KM, Kobelyatskaya AA, Savvateeva MV, Poloznikov AA, et al. miRNAs expression signature potentially associated with lymphatic dissemination in locally advanced prostate cancer. BMC Medical Genomics. 2020;13(Suppl 8):129. doi: 10.1186/s12920-020-00788-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bonci D, Coppola V, Musumeci M, Addario A, Giuffrida R, Memeo L, et al. The miR-15a-miR-16-1 cluster controls prostate cancer by targeting multiple oncogenic activities. Nat Med. 2008;14:1271–1277. doi: 10.1038/nm.1880. [DOI] [PubMed] [Google Scholar]
- 9.Liu J, Zhang C, Zhao Y, Feng Z. MicroRNA Control of p53. J Cell Biochem. 2017;118:7–14. doi: 10.1002/jcb.25609. [DOI] [PubMed] [Google Scholar]
- 10.Sun J, Zhang K, Cai Z, Li K, Zhao C, Fan C, et al. Identification of critical pathways and hub genes in TP53 mutation prostate cancer by bioinformatics analysis. Biomark Med. 2019;13:831–840. doi: 10.2217/bmm-2019-0141. [DOI] [PubMed] [Google Scholar]
- 11.Collavin L, Lunardi A, Del Sal G. p53-family proteins and their regulators: hubs and spokes in tumor suppression. Cell Death Differ. 2010;17:901–911. doi: 10.1038/cdd.2010.35. [DOI] [PubMed] [Google Scholar]
- 12.Gong Z, Zhang H, Ge Y, Wang P. Long noncoding RNA MIAT regulates TP53 ubiquitination and expedites prostate adenocarcinoma progression by recruiting TBL1X. Biochim Biophys Acta Mol Cell Res. 2023;1870:119527. doi: 10.1016/j.bbamcr.2023.119527. [DOI] [PubMed] [Google Scholar]
- 13.Huang H, Tang Y, Li P, Ye X, Chen W, Xie H, et al. Significance of TP53 and immune-related genes to prostate cancer. Transl Androl Urol. 2021;10:1754–1768. doi: 10.21037/tau-21-179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Navarro F, Lieberman J. miR-34 and p53: New Insights into a Complex Functional Relationship. PLoS One. 2015;10:e0132767. doi: 10.1371/journal.pone.0132767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Li JZ, Li J, Wang HQ, Li X, Wen B, Wang YJ. MiR-141-3p promotes prostate cancer cell proliferation through inhibiting kruppel-like factor-9 expression. Biochem Biophys Res Commun. 2017;482:1381–1386. doi: 10.1016/j.bbrc.2016.12.045. [DOI] [PubMed] [Google Scholar]
- 16.Zedan AH, Osther PJS, Assenholt J, Madsen JS, Hansen TF. Circulating miR-141 and miR-375 are associated with treatment outcome in metastatic castration resistant prostate cancer. Sci Rep. 2020;10:227. doi: 10.1038/s41598-019-57101-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Huang S, Wa Q, Pan J, Peng X, Ren D, Huang Y, et al. Downregulation of miR-141-3p promotes bone metastasis via activating NF-κB signaling in prostate cancer. J Exp Clin Cancer Res. 2017;36:173. doi: 10.1186/s13046-017-0645-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVSK, et al. UALCAN: A Portal for Facilitating Tumor Subgroup Gene Expression and Survival Analyses. Neoplasia. 2017;19:649–658. doi: 10.1016/j.neo.2017.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014;42(Database issue):D92–D97. doi: 10.1093/nar/gkt1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.von Stechow L, Delgado AS, editors. Computational Cell Biology Methods and protocols. Humana Press; New York: 2018. [Google Scholar]
- 21.Fan Y, Siklenka K, Arora SK, Ribeiro P, Kimmins S, Xia J. miRNet - dissecting miRNA-target interactions and functional associations through network-based visual analysis. Nucleic Acids Res. 2016;44:W135–41. doi: 10.1093/nar/gkw288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pizurica M, Larmuseau M, Van der Eecken K, de Schaetzen van Brienen L, Carrillo-Perez F, Isphording S, et al. Whole slide imaging-based prediction of TP53 mutations identifies an aggressive disease phenotype in prostate cancer. Cancer Res. 2023 Jun 23;:CAN-22-3113. doi: 10.1158/0008-5472.CAN-22-3113. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Liu Z, Guo H, Zhu Y, Xia Y, Cui J, Shi K, et al. TP53 alterations of hormone-naïve prostate cancer in the Chinese population. Prostate Cancer Prostatic Dis. 2021;24:482–491. doi: 10.1038/s41391-020-00302-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Liu B, Qian D, Zhou W, Jiang H, Xiang Z, Wu D. A Novel Androgen-Induced lncRNA FAM83H-AS1 Promotes Prostate Cancer Progression via the miR-15a/CCNE2 Axis. Front Oncol. 2021;10:620306. doi: 10.3389/fonc.2020.620306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ghaffari M, Kalantar SM, Hemati M, Dehghani Firoozabadi A, Asri A, Shams A, et al. Co-delivery of miRNA-15a and miRNA-16-1 using cationic PEGylated niosomes downregulates Bcl-2 and induces apoptosis in prostate cancer cells. Biotechnol Lett. 2021;43:981–994. doi: 10.1007/s10529-021-03085-2. [DOI] [PubMed] [Google Scholar]
- 26.Walter BA, Valera VA, Pinto PA, Merino MJ. Comprehensive microRNA Profiling of Prostate Cancer. J Cancer. 2013;4:350–357. doi: 10.7150/jca.6394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kristensen H, Thomsen AR, Haldrup C, Dyrskjøt L, Høyer S, Borre M, et al. Novel diagnostic and prognostic classifiers for prostate cancer identified by genome-wide microRNA profiling. Oncotarget. 2016;7:30760–30771. doi: 10.18632/oncotarget.8953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Jin W, Chen F, Wang K, Song Y, Fei X, Wu B. miR-15a/miR-16 cluster inhibits invasion of prostate cancer cells by suppressing TGF-β signaling pathway. Biomed Pharmacother. 2018;104:637–644. doi: 10.1016/j.biopha.2018.05.041. [DOI] [PubMed] [Google Scholar]
- 29.Cui Y, Yang Y, Ren L, Yang J, Wang B, Xing T, et al. miR-15a-3p Suppresses Prostate Cancer Cell Proliferation and Invasion by Targeting SLC39A7 Via Downregulating Wnt/β-Catenin Signaling Pathway. Cancer Biother Radiopharm. 2019;34:472–479. doi: 10.1089/cbr.2018.2722. [DOI] [PubMed] [Google Scholar]
- 30.Chen QH, Li B, Liu DG, Zhang B, Yang X, Tu YL. LncRNA KCNQ1OT1 sponges miR-15a to promote immune evasion and malignant progression of prostate cancer via up-regulating PD-L1. Cancer Cell Int. 2020;20:394. doi: 10.1186/s12935-020-01481-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Slabáková E, Culig Z, Remšík J, Souček K. Alternative mechanisms of miR-34a regulation in cancer. Cell Death Dis. 2017;8:e3100. doi: 10.1038/cddis.2017.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhang W, Dong Y, Sartor O, Zhang K. Deciphering the Increased Prevalence of TP53 Mutations in Metastatic Prostate Cancer. Cancer Inform. 2022;21:11769351221087046. doi: 10.1177/11769351221087046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang X, Sun Q. TP53 mutations, expression and interaction networks in human cancers. Oncotarget. 2017;8:624–643. doi: 10.18632/oncotarget.13483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li WJ, Wang Y, Liu R, Kasinski AL, Shen H, Slack FJ, et al. MicroRNA-34a: Potent Tumor Suppressor, Cancer Stem Cell Inhibitor, and Potential Anticancer Therapeutic. Front Cell Dev Biol. 2021;9:640587. doi: 10.3389/fcell.2021.640587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Casanova-Salas I, Rubio-Briones J, Calatrava A, Mancarella C, Masiá E, Casanova J, et al. Identification of miR-187 and miR-182 as biomarkers of early diagnosis and prognosis in patients with prostate cancer treated with radical prostatectomy. J Urol. 2014;192:252–259. doi: 10.1016/j.juro.2014.01.107. [DOI] [PubMed] [Google Scholar]
- 36.Ye Y, Yuan XH, Wang JJ, Wang YC, Li SL. The diagnostic value of miRNA-141 in prostate cancer: A systematic review and PRISMA-compliant meta-analysis. Medicine (Baltimore) 2020;99:e19993. doi: 10.1097/MD.0000000000019993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Richardsen E, Andersen S, Melbø-Jørgensen C, Rakaee M, Ness N, Al-Saad S, et al. MicroRNA 141 is associated to outcome and aggressive tumor characteristics in prostate cancer. Sci Rep. 2019;9:386. doi: 10.1038/s41598-018-36854-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci U S A. 2008;105:10513–10518. doi: 10.1073/pnas.0804549105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Khorasani M, Teimoori-Toolabi L, Farivar TN, Asgari M, Abolhasani M, Shahrokh H, et al. Aberrant expression of miR-141 and nuclear receptor small heterodimer partner in clinical samples of prostate cancer. Cancer Biomark. 2018;22:19–28. doi: 10.3233/CBM-170696. [DOI] [PubMed] [Google Scholar]
- 40.Porzycki P, Ciszkowicz E, Semik M, Tyrka M. Combination of three miRNA (miR-141, miR-21, and miR-375) as potential diagnostic tool for prostate cancer recognition. Int Urol Nephrol. 2018;50:1619–1626. doi: 10.1007/s11255-018-1938-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Arrighetti N, Beretta GL. miRNAs as Therapeutic Tools and Biomarkers for Prostate Cancer. Pharmaceutics. 2021;13:380. doi: 10.3390/pharmaceutics13030380. [DOI] [PMC free article] [PubMed] [Google Scholar]