Skip to main content
Erschienen in: BMC Cancer 1/2023

Open Access 01.12.2023 | Research

Functional role and epithelial to mesenchymal transition of the miR-590-3p/MDM2 axis in hepatocellular carcinoma

verfasst von: Alaa Ibrahem Youssef, Gehad Mahmoud Khaled, Asma Amleh

Erschienen in: BMC Cancer | Ausgabe 1/2023

Abstract

Background

There is considerable evidence that microRNAs (miRNAs) regulate several key tumor-associated genes/pathways and may themselves have a dual regulatory function either as tumor suppressors or oncogenic miRNA, depending on the tumor type. MicroRNA-590-3p (miR-590-3p) is a small non-coding RNA involved in the initiation and progression of numerous tumors. However, its expression pattern and biological role in hepatocellular carcinoma (HCC) are controversial.

Results

In the current work, computational and RT-qPCR analysis revealed that HCC tissues and cell lines exhibited miR-590-3p downregulation. Forced expression of miR-590-3p attenuated HepG2 cells proliferation, migration, and repressed EMT-related gene expression. Bioinformatic, RT-qPCR, and luciferase assays revealed that MDM2 is a direct functional target of miR-590-3p. Moreover, the knockdown of MDM2 mimicked the inhibitory effect of miR-590-3p in HepG2 cells.

Conclusion

We have identified not only novel targets for miR-590-3p in HCC, but also novel target genes for miR590-3p/MDM2 pathway in HCC like SNAIL, SLUG, ZEB1, ZEB2, and N-cadherin. Furthermore, these findings demonstrate a crucial role for MDM2 in the regulatory mechanism of EMT in HCC.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12885-023-10861-y.
Alaa Ibrahem Youssef and Gehad Mahmoud Khaled contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
DMEM
Dulbecco's Modified Eagle's Medium
FBS
Fetal bovine serum
PBS
Phosphate buffered saline
SDS
Sodium Dodecyl Sulfate
TBST
0.01% Tween-20 in 1X TBS
miRNA590-3p
miRNA590-3p
siRNA
small interfering RNA
SNAIL
Snail family zinc finger 1
N-cadherin
Neural Cadherin
MDM2
Murine double minute 2
ZEB
Zinc finger E-box-binding homeobox
EMT
Epithelial‐mesenchymal transition
HCC
Hepatocellular carcinoma
RT‐qPCR
Reverse transcription‐quantitative PCR
siMDM2
MDM2 siRNA
siNTC
negative siRNA

Background

Hepatocellular carcinoma (HCC) is the primary malignancy of liver cancer, and accounts for 75%-85% of all reported cases of liver cancer reported cases [1, 2]. The major causes of fatality in HCC patients are recurrence and metastasis, with 68% of patients with HCC develop this metastatic disease [35]. Hepatocellular carcinoma (HCC) is a major health issue in Egypt. The Egyptian National Cancer Registry (NCR) reports that HCC is the most frequently observed cancer in lower and middle Egypt, and the second most frequent in upper Egypt [6]. This difference in prevalence may be due to the higher prevalence of hepatitis C viral infection (HCV) in the Nile delta region (lower Egypt), which decreases as we move south [7]. In 2018, Egypt had one of the highest age-standardized mortality rates for HCC worldwide, along with Mongolia [8]. Therefore, elucidating the underlying mechanisms of HCC development and progression and identifying new biomarkers may help reduce the disease burden by enabling early diagnosis and highlighting avenues for the development of therapeutics, finally improving the prognosis of patients with liver cancer.
MicroRNAs comprise a group of small non-coding RNAs 18–22 nucleotides in length, which play a vital role in the regulation of gene expression [9]. There is growing evidence indicates that miRNA dysregulation can serve as a biomarker for the early diagnosis of HCC [10, 11]. miR-590-3p is a human intragenic miRNA located within the introns of the eukaryotic initiation factor 4H (EIF4H) gene. In the context of cancer, dysregulation of the miR-590-3p signature has been shown to play a dual regulatory role by enabling this miRNA to act as an oncomiR or tumor-suppressor depending on the specific tissue it is expressed in [1214]. However, the expression and mechanistic role of miR-590-3p in HCC remain elusive, with contradictory reports [1520] and warranting further study.
The proto-oncoprotein murine double minute 2 (MDM2) is overexpressed in several tumors, including HCC, and has been shown to have roles both dependent and independent of p53 [21, 22]. Recent reports have indicated that MDM2 expression in HCC is correlated with increased malignancy, epithelial-mesenchymal transition (EMT) progression, higher degree of invasiveness, and greater metastatic potential [2325]. However, the key roles of MDM2 and detailed mechanisms in HCC are still far from clear and to the best of our knowledge, the regulatory function of miR-590-3p towards MDM2 expression has not been investigated.
The present study investigates the role of mir-590-3p in HepG2 cells; specifically, whether miR-590-3p regulates the proliferation, migration, and EMT progression of HepG2 cells. To further explore the underlying mechanism, we selected MDM2 for further study because Insilco analysis and luciferase assay suggested it to be a direct functional target of miR-590-3p. In addition, we knocked down MDM2 expression to characterize its role in HepG2 cells. Together, the findings of these assays illustrate the role of the miR-590-3p/MDM2 axis in HCC.

Materials and methods

In silico analysis

The differential expression data of miR-590-3p and MDM2 between HCC and normal liver tissues were extracted from The Cancer Genome Atlas (TCGA) through Encyclopedia of RNA Interactomes (ENCORI) Pan-cancer analysis platform [26]. Potential downstream targets of miR-590-3p were predicted using the Condition-Specific miRNA Targets (CSmiRTar) database [27], which collects data from four common prediction tools (such as miRDB, TargetScan, microRNA.org and DIANA-microT) and then applies a functional filter to detect targets that are only expressed in a certain tissue and/ or a certain disease. TargetScan platform was used to feature the sequence alignment between miR-590-3p seed region and the downstream targets [28].

Cell culture

Two adherent hepatocellular carcinoma cell lines were used in this study: HepG2 and SNU449. HepG2 is a well differentiated, non-invasive HCC cell line. SNU449 cell line is a tumorigenic and more advanced stage of HCC with grade (II–III) [29]. HepG2 was obtained from NAWAH Scientific Inc., (Cairo, Egypt) and SNU449 was a kind gift from Dr Mehmet Ozturk from the Department of Molecular Biology and Genetics, Bilkent University, Turkey. HepG2 and SNU449 were grown in DMEM and RPMI 1640, respectively (LONZA, Bend, OR, USA), supplied with 10% fetal bovine serum (GIBCO, Grand Island, NY, USA) and 5% penicillin–streptomycin antibiotic (GIBCO, Grand Island, NY, USA). Cells were maintained in a humidified incubator supplied with 5% CO2 at 37 ̊Ϲ.

RNA Interference

The knockdown of the MDM2 gene (NCBI Reference Sequence: NG_016708.1) was achieved using ON-TARGETplus SMARTpool siRNA (siMDM2 SMART Pool; L-003279–00-0020) (Dharmacon, Lafayette, CO, USA). Target sequences of the MDM2 siRNAs are presented in Table 1. AllStars Negative Control siRNA (SI027280) (Qiagen, Hilden, Germany), hidden sequence.
Table 1
ON-TARGETplus SMARTpool siRNA sequences (5’-3’)
siRNA 1
GCCAGUAUAUUAUGACUAA
siRNA 2
GAACAAGAGACCCUGGUUA
siRNA 3
GAAUUUAGACAACCUGAAA
siRNA 4
GAUGAGAAGCAACAACAUA

Cell transfection

HepG2 cells were ectopically transfected with 40 nM of miR-590-3p mimics (Invitrogen, Waltham, MA, USA) or Allstar negative control siRNA (Qiagen, Hilden, Germany) using lipofectamine 3000 (Invitrogen, Waltham, MA, USA) in a 6-well plate with a seeding density of 4 × 105. For MDM2 knockdown, cells were transfected with 60 nM of MDM2 siRNA or Allstar negative control siRNA in a 12-well plate with a seeding density of 7 × 104. Cells were incubated for forty-eight hours before performing further experiments. All transfection experiments were done in an RNase-free environment.

Real Time Quantitative Polymerase Chain Reaction (RT-qPCR)

To assess miR-590-3p expression levels, total RNA-enriched small RNAs were extracted using miRNeasy mini kit (Qiagen, Hilden, Germany). MiRCURY LNA SYBR Green qPCR System (Qiagen, Hilden, Germany) was carried out to assess the levels of miR-590-3p in HepG2 and SNU449 cells. In brief, 10 ng of mature miRNA was polyadenylated and reverse-transcribed parallelly into a universal one first-strand cDNA. A UniSp6 RNA spike-in was used in the RT reaction to control efficient cDNA synthesis. RT-qPCR was performed using Locked Nucleic Acid (LNA) SYBR Green qPCR assays. Mir-103a-3p served as the endogenous normalizing control.
For assessing mRNA levels of downstream genes used in this study, total RNA was extracted from transfected cells using Trizol reagent (Invitrogen, Waltham, MA, USA) as per the manufacturer’s protocol. RT reactions were made using 2 μg of total RNA samples using RevertAid First Strand cDNA synthesis kit (Thermo Scientific, Waltham, MA, USA) according to the manufacturer’s instructions. 5 ng/reaction of cDNA along with 250–300 nM forward or reverse primers were used to carry out the qPCR using PowerUp SYBR Green kit (Applied Biosystems, Waltham, MA, USA). GAPDH was used for normalizing the expression of different genes. Table 2 lists the primers used in this study. All qPCR was done on Applied Biosystems 7500 system.
Table 2
RT-qPCR primers used in the RT-qPCR analysis
Gene
Primer sequence
hsa-mir-590-3p
5'UAAUUUUAUGUAUAAGCUAGU
hsa-mir-103a-3p
5'AGCAGCAUUGUACAGGGCUAUGA
GAPDH
F: 5’-AAGGTCATCCCTGAGCTGAAC- 3’
R: 5’-ACGCCTGCTTCACCACCTTCT-3’
MDM2
F: 5’-CCCAAGACAAAGAAGAGAGTGTGG-3’
R: 5’-CTGGGCAGGGCTTATTCCTTTTCT-3’
N-Cadherin
F: 5’-GCGTCTGTAGAGGCTTCTGGT-3’
R: 5’-TCTGCAGGCTCACTGCTCTC-3’
Vimentin
F: 5’-GAACGCCAGATGCGTGAAATG-3’
R: 5’-CCAGAGGGAGTGAATCCAGATTA-3’
SNAIL
F: ACTATGCCGCGCTCTTTCCT
R: GCTGCTGGAAGGTAAACTCTGG
SLUG
F: CAAGGCGTTTTCCAGACCCTG
R: AAGAAAAAGGCTTCTCCCCCGT
ZEB1
F: TGCTGGGAGGATGACACAGG
R: CTGCTTCATCTGCCTGAGCTT
ZEB2
F: TTCCTGGGCTACGACCATACC
R: CAAGCAATTCTCCCTGAAATCC

Transwell migration assay

To test the effect of mir-590-3p overexpression on the migration of HepG2 cells, Transwell assay was performed. Cells were collected forty-eight hours post-transfection, and almost (5 × 105) of mimic or negative control transfected cells were suspended in media supplemented with 1% FBS. For MDM2 knockdown, almost 150*103 cells/well of MDM2 siRNA or negative control.transfected cells were resuspended in serum-free media. Cell suspensions were loaded on the upper chamber of 8 μm cell culture translucent inserts (Greiner Bio-One, Frickenhausen, Germany) placed in a 24-well plate. Inside the 24-well plate, complete media with 10% FBS was added to serve as a chemotactic agent to allow for cell migration through the microporous filter. After 10 h, cells in the upper chamber of the insert were removed with cotton swabs. Migrated cells at the bottom of the insert were fixed with 4% formaldehyde and stained with DAPI (KPL, Gaithersburg، MD, USA) (1:1000 in PBS). Cells were examined under a fluorescent microscope at 20X magnification, and five random fields per insert were captured. Cells were counted using Fiji Image J software.

Colony formation assay

A colony-formation assay was conducted to test if mir-590-3p has a role in regulating HCC clonogenicity. Forty-eight hours post-transfection, HepG2 cells transfected with either miR-590-3p mimic or negative control were detached and seeded in a 6-well plate at a density of 500 cells. Cells were incubated to colonize for seven days. For MDM2 knockdown, transfected cells were incubated for ten days. When visible colonies were seen under the microscope, the experiment was terminated, and formed colonies (more than 50 cells/ colony) were fixed with 4% formaldehyde, stained with crystal violet, and manually counted.

Plasmid construction and luciferase reporter assay

Wild type (WT) and mutant (MUT) constructs of the 3′ untranslated region (3′UTR) of MDM2 were chemically synthesized. For the WT constructs, the miR-590-3p binding site in the 3′UTR of MDM2 was cloned into the pmirGLO dual luciferase miRNA target expression vector (Promega, Madison, WI, USA). For the MUT constructs, the binding region nucleotides in the 3′UTR of MDM2 was completely deleted. The forward and reverse primer sequences of both designed constructs flanked by SalI and NheI restriction sites are shown in Table 3. HepG2 cells were co-transfected with 50 ng of the WT or MUT constructs along with miR-590-3p mimics or negative control using Lipofectamine 3000 (Invitrogen, Waltham, MA, USA). Twenty-four hours after transfection, cell lysates were prepared, and the relative luciferase activity was normalized to the total protein content and tested through a dual-luciferase reporter assay system (Promega, Madison, WI, USA).
Table 3
The forward (F) and reverse (R) primer sequences for WT and MUT 3′UTR constructs of MDM2
MDM2 3’UTR
WT
F 5’ CTAGCGGCTAGTGATATATATAAAGTAAAATTTTCTTTGCAGG 3’
R3’ GCCGATCACTATATATATTTCATTTTAAAAGAAACGTCCAGCT 5’
MDM2 3’UTR
MUT
F5’ CTAGCGGCTAGTGATATATATAAAG–––-TTCTTTGCAGG 3’
R3’ GCCGATCACTATATATATTTC–––-AAGAAACGTCCAGCT 5’

Western blotting

Transfected cells were resuspended and lysed using the CelLytic™ M Cell lysis Reagent (Sigma Aldrish, Burlington, MA, USA). Equal amounts (15 μg) of proteins were loaded on 10% SDS–Polyacrylamide gel and separated by electrophoresis at 150 V for an hour. SDS gels were blotted to nitrocellulose membranes (GE Healthcare, Chicago, Illinois, USA). Membranes were then left for overnight blocking at 4 C in 5% nonfat dry milk diluted in 1X Tris-buffered saline with 0.01% Tween 20 (TBST). Following blocking, membranes were incubated with primary antibodies. Membranes were then washed by TBST and incubated with alkaline phosphatase-conjugated secondary goat anti-rabbit IgG or goat anti-mouse IgG antibodies (KPL, Gaithersburg، MD, USA) (1:10,000 in 5% non-fat milk). Finally, membranes were washed and incubated with the chromogenic substrate BCIP/NBT (KPL, Gaithersburg، MD, USA) until protein bands were visible with eye. Primary antibodies mentioned here are Anti-GAPDH (MA5-15,738, 1:10,000) (Invitrogen, Waltham, MA, USA) and MDM2 (E-AB-31995, 1:200) (Elabscience Biotechnology Inc., Houston, Texas, USA).

Data analysis

The relative expression of miR-590-3p in HepG2 and SNU449 and the differential expression levels of downstream genes in different transfection conditions were analyzed using the comparative ΔΔCT method [30]. Fiji ImageJ software [31] was used to analyze Transwell assay results. Data presented as the mean ± standard error of the mean (SEM) from three independent experiments unless specified otherwise. For statistical analysis, an unpaired Students’ t-test was used to measure differences between two experimental groups. A probability value (P-value) < 0.05 was considered a significant result. Statistical analyses were established using GraphPad Prism 6 statistical packages [32].

Results

miR-590-3p is a downregulated miRNA in hepatocellular carcinoma

Comparison of miR-590-3p expression in HCC and non-cancerous liver tissues revealed a very significant (P > 0.0001) downregulation of miR-5909-3p expression in 370 HCC samples compared with 50 healthy counterparts, with a fold change of (0.75) (Fig. 1A). Examination of miR-590-3p levels in two cell lines representing an early and an advanced stage of HCC using RT qPCR revealed the expression of miR-590-3p to be significantly lower in SNU449 cells compared with HepG2 cells (P < 0.01) (Fig. 1B).

Restoration of miR-590-3p expression inhibits cellular clonogenicity, migration, and epithelial-mesenchymal transition in HepG2 cells

After establishing a miR-590-3p transient transfection using miR-590-3p mimics in the HepG2 cell line, miR-590-3p levels post-transfection were dramatically higher in miR-590-3p mimic transfected cells than in the negative control (Fig. 2A). Overexpression of miR-590-3p caused a significant decrease in the number of colonies of HepG2 cells compared with the negative control (Fig. 2B). The Transwell migration assay (Fig. 2C) revealed the cellular migration capacity of HepG2 cells to be significantly reduced when miR-590-3p was overexpressed. Having observed that miR-590-3p overexpression exerted a critical role in suppressing HepG2 cell motility, we investigated its effect on EMT. Bioinformatic analyses and RT qPCR revealed that the transcript levels of four EMT transcription factors: SNAIL, SLUG, ZEB1, and ZEB2 were dramatically decreased in miR-590-3p-mimics-transfected HepG2 cells compared with the control group (Figures S1 and 2D). In addition, a significant reduction was found in the levels of the mesenchymal marker N-cadherin, but not Vimentin, in HepG2 cells after overexpressing miR-590-3p (Fig. 2E). Consistently with qPCR results, a complementary binding between the seed region of miR-590-3p and N-cadherin was shown, but no sequence alignment was found with Vimentin (Fig. S2).

MDM2 is a specific downstream target gene of miR-590-3p in hepatocellular carcinoma

The CSmiRTar prediction algorithm identified five validated target genes that are liver-tissue and HCC-disease specific (Table 4). Among these, we selected MDM2 for further investigation based on a previous study that reported a functional network between MDM2 and miR-590-3p in HepG2 cells [33]. Specifically, miR-590-3p was identified as one of 33 P53-regulated miRNAs after doxorubicin treatment, and MDM2 was shown to be a direct target gene of miR-590-3p. However, our study was primarily interested in exploring the P53-independent effects of MDM2, particularly those related to EMT.
Table 4
Alignment features of five experimentally validated potential targets of miR-590-3p in hepatocellular carcinoma predicted by the CSmiRTar algorithm
Gene Name
Average Normalized Scorea
Validation method
References
SETD7
0.555
PAR-CLIPb
[34]
RACGAP1
0.444
PAR-CLIPb
[34]
YWAHZ
0.399
PAR-CLIPb
[35]
RAPGEF1
0.383
PAR-CLIPb
[36]
MDM2
0.239
PAR-CLIPb
[37]
aAverage of scores from four prediction databases (higher scores indicate better predictions)
bPAR-CLIP refers to Photoactivatable Ribonucleoside-enhanced Cross-linking Immunoprecipitation, a technique used to identify RNA targets of RNA-binding proteins
To further investigate the potential role of MDM2 in HCC, we analyzed TCGA data and found that MDM2 expression was significantly upregulated in HCC samples compared to healthy samples (fold change = 1.34, P < 0.001) (Fig. 3A). Additionally, we conducted functional experiments in HepG2 cells and found that overexpression of miR-590-3p significantly downregulated MDM2 mRNA levels (P < 0.05) (Fig. 3B). We confirmed the target site of miR-590-3p on MDM2 mRNA using TargetScan, which identified a sequence alignment in the seed region of miR-590-3p (Fig. 3C). Finally, to directly test if there is a mechanistic interaction between miR-590-3p and MDM2, we performed luciferase reporter assays and found that co-transfection of miR-590-3p mimics and WT constructs of the MDM2 3′UTR significantly decreased luciferase activity compared to the negative control (P < 0.01), while no change was observed in the MDM2 3′UTR MUT group (Fig. 3D). Taken together, our results provide evidence that miR-590-3p directly targets MDM2 and regulates its expression in HCC cells.

Knockdown of MDM2 has similar effects as miR-590-3p overexpression in HepG2 cells

To characterize the role of MDM2 in HepG2 cells, we explored the effect of MDM2 silencing on cell proliferation, migration, and EMT progression of HepG2 cells. Figure 4A&B illustrates that the on-target smart pool siRNAs induced remarkably strong and detectable knockdown (more than 80% at the mRNA level and almost 70% at the protein level) (Full length blot S3 A&B). The clonogenic cell survival and Transwell assays revealed that MDM2 gene silencing impaired HepG2 cell proliferation and migration (Fig. 4C&D). The RT qPCR data indicated that MDM2 silencing significantly suppressed the transcript levels of EMT transcription factors such as SNAIL, SLUG, ZEB1, and ZEB2, and mesenchymal genes, such as N-cadherin and Vimentin (Fig. 4E&F).

Discussion

Dysregulation of particular miRNAs has been reported in several types of cancer, including HCC [38]. The present study demonstrated that miR-590-3p is downregulated in HCC tissues and in different stages of HCC cell lines. Herein, we used HepG2 and SNU449 cell lines which are commonly used in HCC research, and their characteristics are well-described in the literature. HepG2 represent an early stage of HCC with an epithelial appearance, while SNU449 is considered an intermediate stage of HCC [29, 39]. We hypothesized that since miR-590-3p is downregulated in HCC compared to normal individuals, its expression is expected to decrease further as the tumor advances from an early stage to a later stage of the disease. Our findings revealed an almost 90-fold decrease in the expression level of miR-590-3p in SNU449 as compared to HepG2 cells. These results highlight the potential of miR-590-3p as a diagnostic and a prognostic factor in HCC, as it is able to differentiate between normal tissues and HCC, as well as between different stages of HCC.
However, it is important to consider the limitations of this study, such as the fact that miR-590-3p expression was only tested in HCC cell lines. Including normal liver cell lines would have provided a more comprehensive understanding of miR-590-3p expression in HCC tissue compared to normal tissue. Nonetheless, our study provides valuable insights into the function of miR-590-3p at different stages of HCC, which can guide future investigations into the potential clinical applications of this miRNA.
It is widely accepted that miRNAs play a significant role in cancer pathogenesis by regulating many biological processes such as proliferation, metastasis, and apoptosis [40]. In the Current study, the restoration of miR-590-3p by ectopic transfection of miRNA mimics reduces the clonogenicity and migration rates of the HepG2 cells. Moreover, overexpression of miR-590-3p caused a marked reduction in the transcript levels of EMT transcription factors (SNAIL, SLUG, ZEB1, and ZEB2) and the EMT mesenchymal marker, N-cadherin. Interestingly, the expression of Vimentin, another EMT mesenchymal marker, was not significantly affected by miR-590-3p overexpression. Collectively, these results suggest that miR-590-3p has tumor suppressive effects on HCC.
There is growing evidence indicating that miR-590-3p plays a role as a tumor suppressor or an oncomiR in several cancers. For example, miR-590-3p reduced migration, invasion and EMT in glioblastoma multiforme by targeting ZEB1 and ZEB2 [13], and in intrahepatic cholangiocarcinoma by targeting ZEB2 [41]. Similarly, the suppressive effects of miR-590-3p on apoptosis have been reported in the context of osteosarcoma [42] and breast cancer [43]. In contrast, miR-590-3p causes increased cellular proliferation and metastasis in some cancers, such as prostate [44], ovarian [14], gastric [45], and colorectal cancer [12]. However, the role of miR-590- 3p role in HCC remains unclear. Some studies have suggested that miR-590-3p targets the CHFR3/p-STAT3/p53 axis or activates the PI3K-AKT pathway, leading to a malignant phenotype [17, 18]. However, other reports have found that restoring miR-590-3p expression suppressed HCC growth via interaction with the TEAD1 target gene or the EED gene [15, 16]. Two other studies suggested miR-590-3p played an intermediate tumor suppressor role in the LINC0016/miR-590-3p/ROCK and PART1/miR-590-3p/HMGB2 [19, 20]. These conflicting findings suggest a complex role for miR-590-3p in HCC pathogenesis, and further research is necessary to fully understand its mechanisms of action.
The discrepancies in the literature related to miR-590-3p likely arise from the fact that miRNAs can regulate multiple target genes, which can affect differential tumorigenic pathways. However, the concept that a single miRNA could function both as a tumor suppressor and an oncomiR in the same cancer is a relatively novel scientific idea.
The present study adds evidence to the tumor-suppressive role of miR-590-3p in HCC. It demonstrates the inhibition of three important cancer hallmarks: clonogenicity, migration, and EMT. Our findings support previous studies [15, 16, 19, 20] that suggest miR-590-3p has tumor suppressor effects in HCC. However, our results conflict with those of [17, 18], which suggest that miR-590-3p functions as an oncomiR in HCC. Thus, further research is required to reconcile these conflicting findings and fully understand the role of miR-590-3p in HCC.
A particularly interesting finding of the present study is the non-significant change in Vimentin gene expression in response to miR-590-3p overexpression. Because EMT is not a complete process in all cancers, a population of cancer cells will express mixed and different EMT markers [46]. Also, the desynchronization between EMT phenotype and marker gene expression has been reported before [47], with expression of some EMT markers remaining unchanged until after three days of treatment while the mesenchymal phenotype was detectable after just one day after treatment. Since Vimentin expression did not significantly change in our experiment, it is possible that Vimentin is not regulated by miR-590-3p as an EMT marker, or there may be intermediate players between miR-590-3p and Vimentin that slow down the signaling to a later stage of the EMT cascade. Further research is needed to clarify the role of miR-590-3p in regulating Vimentin expression in EMT.
The function of a particular miRNA in a certain cancer is mainly attributed to the function of its target genes [48]. Bioinformatic analysis revealed MDM2 as a promising miR-590-3p candidate gene with a differential expression pattern between normal liver and HCC tissues. The upregulation of MDM2 in HCC tissues demonstrates a negative correlation with miR-590-3p downregulation, that was identified before, and was further corroborated by the reduction of mRNA MDM2 levels after overexpressing miR-590-3p. The mechanistic interaction between MDM2 and miR-590-3p, established through the luciferase experiment, further confirms that MDM2 is a direct novel target of miR-590-3p in HCC. Collectively, these results suggest that there is a strong relationship between miR-590-3p and MDM2 that might be contributed, at least in part, to the suppressive effects of miR-590-3p on HCC discussed before.
We further examined the above hypothesis by knocking down MDM2 to gain insight into its biological role in HCC. MDM2 is a major oncogene, which is expressed at high levels in several tumors including HCC [21, 22, 49, 50]. The main function of MDM2 is to influence the p53 tumor suppressor function through different mechanisms [51]. However, there is growing evidence suggesting that MDM2 interacts with certain proteins that contribute to cell proliferation, migration, EMT progression, and metastasis, independent of p53 [22, 24, 52, 53].
We observed that knocking down MDM2 suppressed HepG2 cell proliferation and migration, in line with these observations that have shown that MDM2 possesses proliferation- and migration-promoting activities in HCC and several other human cancers, independent of p53 [24, 25, 54]. Further evidence from studies on breast cancer suggests that MDM2 promotes cell motility and invasiveness of breast cancer cells by targeting E-cadherin for degradation and inducing MMP9 expression [55, 56]. Thus, the possibility that the MMPs and E-cadherin proteins are involved in the regulative effect of MDM2 on HepG2 cell migration cannot be excluded.
We demonstrated that MDM2 positively regulates SNAIL genes at the mRNA level, in contrast to previous studies that have shown MDM2 to negatively regulate SNAIL and SLUG at the protein level [57, 58]. Most previous studies focused on the protein targets of MDM2, and there is little information about the mRNA targets. Recent findings, however, demonstrated that MDM2 is also capable of binding to and stabilizing the mRNA of certain genes, including the EMT transcription factor, SLUG [5962]. Notably, Jung et al. suggested that the expression of SLUG protein is determined by a balance between MDM2-mediated accumulation of SLUG mRNA and MDM2-mediated degradation of SLUG protein, with p53 required for the latter function [62]. In support of this assumption, our data implicate MDM2 serves as a positive regulator of the SNAIL family at the mRNA level in HepG2 cells. To the best of our knowledge, the current work is the first description of the regulative effect of MDM2 on SNAIL genes at the mRNA level in HCC.
The data presented here implies that MDM2 regulates the expression of ZEB factors in HCC. Consistent with this, MDM2 overexpression has been found to enhance the expression of EMT markers, such as ZEB1, via the B‐Raf signaling pathway in glioma, lung cancer, and breast cancer cells [63]. In addition, recent evidence has been published suggesting that MDM2 can regulate ZEB1 and ZEB2 by activating Smad2 and Smad3, resulting in EMT progression [50]. To the best of our knowledge, this is the first report of ZEB2 regulation by MDM2 in the context of HCC. Our experimental results also suggested that silencing MDM2 mimics the tumor-suppressive activity of miR-590-3p in HepG2 cells, indicating that miR-590-3p plays a tumor-suppressor role in HepG2 by targeting MDM2.
It is important to note that siRNA-mediated knockdown of MDM2 can result in off-target effects, which can complicate the interpretation of results. This study utilized the On-TARGETplus SMARTpool siMDM2, a highly efficient and widely recognized approach to reduce off-target effects. Overall, our findings shed new light on the biological role of MDM2 in HCC and provided insights into potential therapeutic targets for this disease.

Conclusion

This study presents a novel miR-590-3p/MDM2 axis (Fig. 5) through which the precise role of miR-590-3p in EMT regulation in HCC can be identified. Our findings suggest that the MDM2 oncogene plays a significant role in HepG2 proliferation, migration, and EMT progression and that the tumor suppressive effect of miR-590-3p in HCC is due to interaction, at least in part, with MDM2. We believe that our findings could have important implications for HCC treatment considering the substantial role of the miR-590-3p/MDM2 axis in HCC pathogenesis, implicating it as an axis worthy of clinical investigation. Future studies on the correlation between miR-590-3p and MDM2 expression profiles with the clinico-pathological patterns in HCC might provide more insights into the significance of the miR-590-3p/ MDM2 axis as a potential therapeutic target of HCC. Finally, our results shed light on the importance of exploring more mRNA targets of MDM2 involved in HCC progression, which may help in understanding the metastasis of HCC through MDM2.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat 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(3):209–49.PubMedCrossRef 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(3):209–49.PubMedCrossRef
2.
Zurück zum Zitat Suresh D, Srinivas AN, Kumar DP. Etiology of Hepatocellular Carcinoma: Special Focus on Fatty Liver Disease. Front Oncol. 2020;10:2673.CrossRef Suresh D, Srinivas AN, Kumar DP. Etiology of Hepatocellular Carcinoma: Special Focus on Fatty Liver Disease. Front Oncol. 2020;10:2673.CrossRef
3.
Zurück zum Zitat Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primer. 2021;7(1):1–28. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, et al. Hepatocellular carcinoma. Nat Rev Dis Primer. 2021;7(1):1–28.
4.
Zurück zum Zitat Schütte K, Schinner R, Fabritius MP, Möller M, Kuhl C, Iezzi R, et al. Impact of Extrahepatic Metastases on Overall Survival in Patients with Advanced Liver Dominant Hepatocellular Carcinoma: A Subanalysis of the SORAMIC Trial. Liver Cancer. 2020;9(6):771–86.PubMedPubMedCentralCrossRef Schütte K, Schinner R, Fabritius MP, Möller M, Kuhl C, Iezzi R, et al. Impact of Extrahepatic Metastases on Overall Survival in Patients with Advanced Liver Dominant Hepatocellular Carcinoma: A Subanalysis of the SORAMIC Trial. Liver Cancer. 2020;9(6):771–86.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Ezzat R, Eltabbakh M, El Kassas M. Unique situation of hepatocellular carcinoma in Egypt: A review of epidemiology and control measures. World J Gastrointest Oncol. 2021;13(12):1919–38.PubMedPubMedCentralCrossRef Ezzat R, Eltabbakh M, El Kassas M. Unique situation of hepatocellular carcinoma in Egypt: A review of epidemiology and control measures. World J Gastrointest Oncol. 2021;13(12):1919–38.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Ibrahim AS, Khaled HM, Mikhail NN, Baraka H, Kamel H. Cancer Incidence in Egypt: Results of the National Population-Based Cancer Registry Program. J Cancer Epidemiol. 2014;21(2014):e437971. Ibrahim AS, Khaled HM, Mikhail NN, Baraka H, Kamel H. Cancer Incidence in Egypt: Results of the National Population-Based Cancer Registry Program. J Cancer Epidemiol. 2014;21(2014):e437971.
8.
9.
Zurück zum Zitat Acunzo M, Romano G, Wernicke D, Croce CM. MicroRNA and cancer – A brief overview. Adv Biol Regul. 2015;1(57):1–9.CrossRef Acunzo M, Romano G, Wernicke D, Croce CM. MicroRNA and cancer – A brief overview. Adv Biol Regul. 2015;1(57):1–9.CrossRef
10.
Zurück zum Zitat Jin Y, Wong YS, Goh BKP, Chan CY, Cheow PC, Chow PKH, et al. Circulating microRNAs as Potential Diagnostic and Prognostic Biomarkers in Hepatocellular Carcinoma. Sci Rep. 2019;9(1):10464.PubMedPubMedCentralCrossRef Jin Y, Wong YS, Goh BKP, Chan CY, Cheow PC, Chow PKH, et al. Circulating microRNAs as Potential Diagnostic and Prognostic Biomarkers in Hepatocellular Carcinoma. Sci Rep. 2019;9(1):10464.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Mizuguchi Y, Takizawa T, Yoshida H, Uchida E. Dysregulated miRNA in progression of hepatocellular carcinoma: A systematic review. Hepatol Res. 2016;46(5):391–406.PubMedCrossRef Mizuguchi Y, Takizawa T, Yoshida H, Uchida E. Dysregulated miRNA in progression of hepatocellular carcinoma: A systematic review. Hepatol Res. 2016;46(5):391–406.PubMedCrossRef
12.
13.
Zurück zum Zitat Pang H, Zheng Y, Zhao Y, Xiu X, Wang J. miR-590-3p suppresses cancer cell migration, invasion and epithelial–mesenchymal transition in glioblastoma multiforme by targeting ZEB1 and ZEB2. Biochem Biophys Res Commun. 2015;468(4):739–45.PubMedCrossRef Pang H, Zheng Y, Zhao Y, Xiu X, Wang J. miR-590-3p suppresses cancer cell migration, invasion and epithelial–mesenchymal transition in glioblastoma multiforme by targeting ZEB1 and ZEB2. Biochem Biophys Res Commun. 2015;468(4):739–45.PubMedCrossRef
15.
Zurück zum Zitat Ge X, Gong L. MiR-590-3p suppresses hepatocellular carcinoma growth by targeting TEAD1. Tumour Biol J Int Soc Oncodevelopmental Biol Med. 2017;39(3):1010428317695947.CrossRef Ge X, Gong L. MiR-590-3p suppresses hepatocellular carcinoma growth by targeting TEAD1. Tumour Biol J Int Soc Oncodevelopmental Biol Med. 2017;39(3):1010428317695947.CrossRef
16.
Zurück zum Zitat He B, Lu P, Guan L, Li T, Zhang L, Zhu QG, et al. Identifying key regulating miRNAs in hepatocellular carcinomas by an omics’ method. Oncotarget. 2017;8(61):103919–30.PubMedPubMedCentralCrossRef He B, Lu P, Guan L, Li T, Zhang L, Zhu QG, et al. Identifying key regulating miRNAs in hepatocellular carcinomas by an omics’ method. Oncotarget. 2017;8(61):103919–30.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Wan Z, Li X, Luo X, Wang B, Zhou X, Chen A. The miR-590-3p/CFHR3/STAT3 signaling pathway promotes cell proliferation and metastasis in hepatocellular carcinoma. Aging. 2022;14(14):5783–99.PubMedPubMedCentralCrossRef Wan Z, Li X, Luo X, Wang B, Zhou X, Chen A. The miR-590-3p/CFHR3/STAT3 signaling pathway promotes cell proliferation and metastasis in hepatocellular carcinoma. Aging. 2022;14(14):5783–99.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Yang H, Zheng W, Zhao W, Guan C, An J. Roles of miR-590-5p and miR-590-3p in the development of hepatocellular carcinoma. Nan Fang Yi Ke Da Xue Xue Bao. 2013;33(6):804–11.PubMed Yang H, Zheng W, Zhao W, Guan C, An J. Roles of miR-590-5p and miR-590-3p in the development of hepatocellular carcinoma. Nan Fang Yi Ke Da Xue Xue Bao. 2013;33(6):804–11.PubMed
19.
Zurück zum Zitat Bao MH, Li GY, Huang XS, Tang L, Dong LP, Li JM. Long Noncoding RNA LINC00657 Acting as a miR-590-3p Sponge to Facilitate Low Concentration Oxidized Low-Density Lipoprotein-Induced Angiogenesis. Mol Pharmacol. 2018;93(4):368–75.PubMedCrossRef Bao MH, Li GY, Huang XS, Tang L, Dong LP, Li JM. Long Noncoding RNA LINC00657 Acting as a miR-590-3p Sponge to Facilitate Low Concentration Oxidized Low-Density Lipoprotein-Induced Angiogenesis. Mol Pharmacol. 2018;93(4):368–75.PubMedCrossRef
20.
Zurück zum Zitat Pu J, Tan C, Shao Z, Wu X, Zhang Y, Xu Z, et al. Long Noncoding RNA PART1 Promotes Hepatocellular Carcinoma Progression via Targeting miR-590-3p/HMGB2 Axis. OncoTargets Ther. 2020;16(13):9203–11.CrossRef Pu J, Tan C, Shao Z, Wu X, Zhang Y, Xu Z, et al. Long Noncoding RNA PART1 Promotes Hepatocellular Carcinoma Progression via Targeting miR-590-3p/HMGB2 Axis. OncoTargets Ther. 2020;16(13):9203–11.CrossRef
21.
Zurück zum Zitat Shaikh MF, Morano WF, Lee J, Gleeson E, Babcock BD, Michl J, et al. Emerging Role of MDM2 as Target for Anti-Cancer Therapy: A Review. Ann Clin Lab Sci. 2016;46(6):627–34.PubMed Shaikh MF, Morano WF, Lee J, Gleeson E, Babcock BD, Michl J, et al. Emerging Role of MDM2 as Target for Anti-Cancer Therapy: A Review. Ann Clin Lab Sci. 2016;46(6):627–34.PubMed
23.
Zurück zum Zitat Cao H, Chen X, Wang Z, Wang L, Xia Q, Zhang W. The role of MDM2–p53 axis dysfunction in the hepatocellular carcinoma transformation. Cell Death Discov. 2020;6(1):1–14.CrossRef Cao H, Chen X, Wang Z, Wang L, Xia Q, Zhang W. The role of MDM2–p53 axis dysfunction in the hepatocellular carcinoma transformation. Cell Death Discov. 2020;6(1):1–14.CrossRef
24.
Zurück zum Zitat Ranjan A, Bera K, Iwakuma T. Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis. Hepatoma Res. 2016;2:114–21.PubMedPubMedCentralCrossRef Ranjan A, Bera K, Iwakuma T. Murine double minute 2, a potential p53-independent regulator of liver cancer metastasis. Hepatoma Res. 2016;2:114–21.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Wang W, Hu B, Qin JJ, Cheng JW, Li X, Rajaei M, et al. A novel inhibitor of MDM2 oncogene blocks metastasis of hepatocellular carcinoma and overcomes chemoresistance. Genes Dis. 2019;6(4):419–30.PubMedPubMedCentralCrossRef Wang W, Hu B, Qin JJ, Cheng JW, Li X, Rajaei M, et al. A novel inhibitor of MDM2 oncogene blocks metastasis of hepatocellular carcinoma and overcomes chemoresistance. Genes Dis. 2019;6(4):419–30.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat 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(D1):D92-7.PubMedCrossRef 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(D1):D92-7.PubMedCrossRef
27.
Zurück zum Zitat Wu WS, Tu BW, Chen TT, Hou SW, Tseng JT. CSmiRTar: Condition-Specific microRNA targets database. de Bruin RAM, editor. PLOS One. 2017;12(7):e0181231.PubMedPubMedCentralCrossRef Wu WS, Tu BW, Chen TT, Hou SW, Tseng JT. CSmiRTar: Condition-Specific microRNA targets database. de Bruin RAM, editor. PLOS One. 2017;12(7):e0181231.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Jung CW, Song TJ, Lee KO, Choi SB, Kim WB, Suh SO, et al. Characterization of hepatocellular carcinoma cell lines based on cell adhesion molecules. Int J Mol Med. 2012;29(6):1158–64.PubMed Jung CW, Song TJ, Lee KO, Choi SB, Kim WB, Suh SO, et al. Characterization of hepatocellular carcinoma cell lines based on cell adhesion molecules. Int J Mol Med. 2012;29(6):1158–64.PubMed
30.
Zurück zum Zitat Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods San Diego Calif. 2001;25(4):402–8.PubMedCrossRef Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods San Diego Calif. 2001;25(4):402–8.PubMedCrossRef
31.
Zurück zum Zitat Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9(7):676–82.PubMedCrossRef Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9(7):676–82.PubMedCrossRef
32.
Zurück zum Zitat Motulsky H. Christopoulos A. Fitting Models to Biological Data Using Linear and Nonlinear Regression: A Practical Guide to Curve Fitting. Oxford University Press; 2004. p. 353. Motulsky H. Christopoulos A. Fitting Models to Biological Data Using Linear and Nonlinear Regression: A Practical Guide to Curve Fitting. Oxford University Press; 2004. p. 353.
33.
Zurück zum Zitat Yang Y, Liu W, Ding R, Xiong L, Dou R, Zhang Y, et al. Comprehensive Expression Profiling and Functional Network Analysis of p53-Regulated MicroRNAs in HepG2 Cells Treated with Doxorubicin. PLoS One. 2016;11(2):e0149227.PubMedPubMedCentralCrossRef Yang Y, Liu W, Ding R, Xiong L, Dou R, Zhang Y, et al. Comprehensive Expression Profiling and Functional Network Analysis of p53-Regulated MicroRNAs in HepG2 Cells Treated with Doxorubicin. PLoS One. 2016;11(2):e0149227.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Hafner M, Landthaler M, Burger L, Khorshid M, Hausser J, Berninger P, et al. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell. 2010;141(1):129–41.PubMedPubMedCentralCrossRef Hafner M, Landthaler M, Burger L, Khorshid M, Hausser J, Berninger P, et al. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell. 2010;141(1):129–41.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.PubMedCrossRef Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495(7441):333–8.PubMedCrossRef
36.
Zurück zum Zitat Gottwein E, Corcoran DL, Mukherjee N, Skalsky RL, Hafner M, Nusbaum JD, et al. Viral microRNA targetome of KSHV-infected primary effusion lymphoma cell lines. Cell Host Microbe. 2011;10(5):515–26.PubMedPubMedCentralCrossRef Gottwein E, Corcoran DL, Mukherjee N, Skalsky RL, Hafner M, Nusbaum JD, et al. Viral microRNA targetome of KSHV-infected primary effusion lymphoma cell lines. Cell Host Microbe. 2011;10(5):515–26.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Kishore S, Jaskiewicz L, Burger L, Hausser J, Khorshid M, Zavolan M. A quantitative analysis of CLIP methods for identifying binding sites of RNA-binding proteins. Nat Methods. 2011;8(7):559–64.PubMedCrossRef Kishore S, Jaskiewicz L, Burger L, Hausser J, Khorshid M, Zavolan M. A quantitative analysis of CLIP methods for identifying binding sites of RNA-binding proteins. Nat Methods. 2011;8(7):559–64.PubMedCrossRef
38.
Zurück zum Zitat Liu LN, Li DD, Xu HX, Zheng SG, Zhang XP. Role of microRNAs in hepatocellular carcinoma. Front Biosci Landmark Ed. 2015;1(20):1056–67. Liu LN, Li DD, Xu HX, Zheng SG, Zhang XP. Role of microRNAs in hepatocellular carcinoma. Front Biosci Landmark Ed. 2015;1(20):1056–67.
39.
Zurück zum Zitat Costantini S, Di Bernardo G, Cammarota M, Castello G, Colonna G. Gene expression signature of human HepG2 cell line. Gene. 2013;518(2):335–45.PubMedCrossRef Costantini S, Di Bernardo G, Cammarota M, Castello G, Colonna G. Gene expression signature of human HepG2 cell line. Gene. 2013;518(2):335–45.PubMedCrossRef
40.
Zurück zum Zitat Vasuri F, Visani M, Acquaviva G, Brand T, Fiorentino M, Pession A, et al. Role of microRNAs in the main molecular pathways of hepatocellular carcinoma. World J Gastroenterol. 2018;24(25):2647–60.PubMedPubMedCentralCrossRef Vasuri F, Visani M, Acquaviva G, Brand T, Fiorentino M, Pession A, et al. Role of microRNAs in the main molecular pathways of hepatocellular carcinoma. World J Gastroenterol. 2018;24(25):2647–60.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Zu C, Liu S, Cao W, Liu Z, Qiang H, Li Y, et al. MiR-590-3p suppresses epithelial-mesenchymal transition in intrahepatic cholangiocarcinoma by inhibiting SIP1 expression. Oncotarget. 2017;8(21):34698–708.PubMedPubMedCentralCrossRef Zu C, Liu S, Cao W, Liu Z, Qiang H, Li Y, et al. MiR-590-3p suppresses epithelial-mesenchymal transition in intrahepatic cholangiocarcinoma by inhibiting SIP1 expression. Oncotarget. 2017;8(21):34698–708.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Wang WT, Qi Q, Zhao P, Li CY, Yin XY, Yan RB. miR-590-3p is a novel microRNA which suppresses osteosarcoma progression by targeting SOX9. Biomed Pharmacother. 2018;107:1763–9.PubMedCrossRef Wang WT, Qi Q, Zhao P, Li CY, Yin XY, Yan RB. miR-590-3p is a novel microRNA which suppresses osteosarcoma progression by targeting SOX9. Biomed Pharmacother. 2018;107:1763–9.PubMedCrossRef
43.
Zurück zum Zitat Abdolvahabi Z, Nourbakhsh M, Hosseinkhani S, Hesari Z, Alipour M, Jafarzadeh M, et al. MicroRNA-590-3P suppresses cell survival and triggers breast cancer cell apoptosis via targeting sirtuin-1 and deacetylation of p53. J Cell Biochem. 2019;120(6):9356–68.PubMedCrossRef Abdolvahabi Z, Nourbakhsh M, Hosseinkhani S, Hesari Z, Alipour M, Jafarzadeh M, et al. MicroRNA-590-3P suppresses cell survival and triggers breast cancer cell apoptosis via targeting sirtuin-1 and deacetylation of p53. J Cell Biochem. 2019;120(6):9356–68.PubMedCrossRef
44.
Zurück zum Zitat Chen H, Luo Q, Li H. MicroRNA-590-3p promotes cell proliferation and invasion by targeting inositol polyphosphate 4-phosphatase type II in human prostate cancer cells. Tumour Biol J Int Soc Oncodevelopmental Biol Med. 2017;39(3):1010428317695941.CrossRef Chen H, Luo Q, Li H. MicroRNA-590-3p promotes cell proliferation and invasion by targeting inositol polyphosphate 4-phosphatase type II in human prostate cancer cells. Tumour Biol J Int Soc Oncodevelopmental Biol Med. 2017;39(3):1010428317695941.CrossRef
45.
Zurück zum Zitat Zhang J, Jin M, Chen X, Zhang R, Huang Y, Liu H, et al. Loss of PPM1F expression predicts tumour recurrence and is negatively regulated by miR-590-3p in gastric cancer. Cell Prolif. 2018;51(4):e12444.PubMedPubMedCentralCrossRef Zhang J, Jin M, Chen X, Zhang R, Huang Y, Liu H, et al. Loss of PPM1F expression predicts tumour recurrence and is negatively regulated by miR-590-3p in gastric cancer. Cell Prolif. 2018;51(4):e12444.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Song J, Wang W, Wang Y, Qin Y, Wang Y, Zhou J, et al. Epithelial-mesenchymal transition markers screened in a cell-based model and validated in lung adenocarcinoma. BMC Cancer. 2019;19(1):680.PubMedPubMedCentralCrossRef Song J, Wang W, Wang Y, Qin Y, Wang Y, Zhou J, et al. Epithelial-mesenchymal transition markers screened in a cell-based model and validated in lung adenocarcinoma. BMC Cancer. 2019;19(1):680.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Harrandah AM, Mora RA, Chan EKL. Emerging microRNAs in cancer diagnosis, progression, and immune surveillance. Cancer Lett. 2018;01(438):126–32.CrossRef Harrandah AM, Mora RA, Chan EKL. Emerging microRNAs in cancer diagnosis, progression, and immune surveillance. Cancer Lett. 2018;01(438):126–32.CrossRef
49.
Zurück zum Zitat Chen Y, Wang DD, Wu YP, Su D, Zhou TY, Gai RH, et al. MDM2 promotes epithelial–mesenchymal transition and metastasis of ovarian cancer SKOV3 cells. Br J Cancer. 2017;117(8):1192–201.PubMedCrossRef Chen Y, Wang DD, Wu YP, Su D, Zhou TY, Gai RH, et al. MDM2 promotes epithelial–mesenchymal transition and metastasis of ovarian cancer SKOV3 cells. Br J Cancer. 2017;117(8):1192–201.PubMedCrossRef
50.
Zurück zum Zitat Tang Y, Xuan Y, Qiao G, Ou Z, He Z, Zhu Q, et al. MDM2 promotes epithelial–mesenchymal transition through activation of Smad2/3 signaling pathway in lung adenocarcinoma. OncoTargets Ther. 2019;27(12):2247–58.CrossRef Tang Y, Xuan Y, Qiao G, Ou Z, He Z, Zhu Q, et al. MDM2 promotes epithelial–mesenchymal transition through activation of Smad2/3 signaling pathway in lung adenocarcinoma. OncoTargets Ther. 2019;27(12):2247–58.CrossRef
53.
Zurück zum Zitat Saadatzadeh MR, Elmi AN, Pandya PH, Bijangi-Vishehsaraei K, Ding J, Stamatkin CW, et al. The Role of MDM2 in Promoting Genome Stability versus Instability. Int J Mol Sci. 2017;18(10):2216.PubMedPubMedCentralCrossRef Saadatzadeh MR, Elmi AN, Pandya PH, Bijangi-Vishehsaraei K, Ding J, Stamatkin CW, et al. The Role of MDM2 in Promoting Genome Stability versus Instability. Int J Mol Sci. 2017;18(10):2216.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Brekman A, Singh KE, Polotskaia A, Kundu N, Bargonetti J. A p53-independent role of Mdm2 in estrogen-mediated activation of breast cancer cell proliferation. Breast Cancer Res. 2011;13(1):R3.PubMedPubMedCentralCrossRef Brekman A, Singh KE, Polotskaia A, Kundu N, Bargonetti J. A p53-independent role of Mdm2 in estrogen-mediated activation of breast cancer cell proliferation. Breast Cancer Res. 2011;13(1):R3.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Yang JY, Zong CS, Xia W, Wei Y, Ali-Seyed M, Li Z, et al. MDM2 Promotes Cell Motility and Invasiveness by Regulating E-Cadherin Degradation. Mol Cell Biol. 2006;26(19):7269–82.PubMedPubMedCentralCrossRef Yang JY, Zong CS, Xia W, Wei Y, Ali-Seyed M, Li Z, et al. MDM2 Promotes Cell Motility and Invasiveness by Regulating E-Cadherin Degradation. Mol Cell Biol. 2006;26(19):7269–82.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Lim SO, Kim H, Jung G. p53 inhibits tumor cell invasion via the degradation of snail protein in hepatocellular carcinoma. FEBS Lett. 2010;584(11):2231–6.PubMedCrossRef Lim SO, Kim H, Jung G. p53 inhibits tumor cell invasion via the degradation of snail protein in hepatocellular carcinoma. FEBS Lett. 2010;584(11):2231–6.PubMedCrossRef
58.
Zurück zum Zitat Wang SP, Wang WL, Chang YL, Wu CT, Chao YC, Kao SH, et al. p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat Cell Biol. 2009;11(6):694–704.PubMedCrossRef Wang SP, Wang WL, Chang YL, Wu CT, Chao YC, Kao SH, et al. p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat Cell Biol. 2009;11(6):694–704.PubMedCrossRef
59.
Zurück zum Zitat Zhou S, Gu L, He J, Zhang H, Zhou M. MDM2 Regulates Vascular Endothelial Growth Factor mRNA Stabilization in Hypoxia. Mol Cell Biol. 2011;31(24):4928–37.PubMedPubMedCentralCrossRef Zhou S, Gu L, He J, Zhang H, Zhou M. MDM2 Regulates Vascular Endothelial Growth Factor mRNA Stabilization in Hypoxia. Mol Cell Biol. 2011;31(24):4928–37.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Gnanasundram SV, Malbert-Colas L, Chen S, Fusée L, Daskalogianni C, Muller P, et al. MDM2’s dual mRNA binding domains co-ordinate its oncogenic and tumour suppressor activities. Nucleic Acids Res. 2020;48(12):6775–87.PubMedPubMedCentralCrossRef Gnanasundram SV, Malbert-Colas L, Chen S, Fusée L, Daskalogianni C, Muller P, et al. MDM2’s dual mRNA binding domains co-ordinate its oncogenic and tumour suppressor activities. Nucleic Acids Res. 2020;48(12):6775–87.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Faruq O, Zhao D, Shrestha M, Vecchione A, Zacksenhaus E, Chang H. Targeting an MDM2/MYC Axis to Overcome Drug Resistance in Multiple Myeloma. Cancers. 2022;14(6):1592.PubMedPubMedCentralCrossRef Faruq O, Zhao D, Shrestha M, Vecchione A, Zacksenhaus E, Chang H. Targeting an MDM2/MYC Axis to Overcome Drug Resistance in Multiple Myeloma. Cancers. 2022;14(6):1592.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Jung CH, Kim J, Park JK, Hwang SG, Moon SK, Kim WJ, et al. Mdm2 increases cellular invasiveness by binding to and stabilizing the Slug mRNA. Cancer Lett. 2013;335(2):270–7.PubMedCrossRef Jung CH, Kim J, Park JK, Hwang SG, Moon SK, Kim WJ, et al. Mdm2 increases cellular invasiveness by binding to and stabilizing the Slug mRNA. Cancer Lett. 2013;335(2):270–7.PubMedCrossRef
63.
Zurück zum Zitat Ou M, Xu X, Chen Y, Li L, Zhang L, Liao Y, et al. MDM2 induces EMT via the B-Raf signaling pathway through 14-3-3. Oncol Rep. 2021;46(1):1–19.CrossRef Ou M, Xu X, Chen Y, Li L, Zhang L, Liao Y, et al. MDM2 induces EMT via the B-Raf signaling pathway through 14-3-3. Oncol Rep. 2021;46(1):1–19.CrossRef
Metadaten
Titel
Functional role and epithelial to mesenchymal transition of the miR-590-3p/MDM2 axis in hepatocellular carcinoma
verfasst von
Alaa Ibrahem Youssef
Gehad Mahmoud Khaled
Asma Amleh
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2023
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-023-10861-y

Weitere Artikel der Ausgabe 1/2023

BMC Cancer 1/2023 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.