This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Hepatocellular carcinoma (HCC), representing approximately 90% of total primary liver cancer, is the second most frequent cause of cancer-associated deaths worldwide [1–3]. Several factors play essential roles during the progression of HCC, including the tumor microenvironment and genetic mutations [4–6]. Despite various treatment options, HCC is of high incidence and mortality due to rapid proliferation, cancer recurrence, and distant metastasis [7–10]. Thus, identification of novel treatment targets or useful biomarkers is badly needed to improve the clinical treatment outcomes of HCC.
MicroRNAs (miRNAs), a widespread small noncoding endogenous RNA, can interact with target messenger RNA (mRNA) to regulate target gene expression at the posttranscriptional level, influencing many important biological processes such as cell proliferation, invasion, and tumorigenesis [11–13]. Plenty of researches have shown that miRNA was closely intertwined with HCC occurrence and progression [14–16]. Furthermore, some studies suggest that miRNA play roles as either oncogenes or tumor suppressors, which can be used as predictive biomarkers or therapeutic targets in clinical applications [17–20].
hsa-miR-497-5p is located in human chromosome 17 (17p13.1) and derived from the miR-15 family [21]. hsa-miR-497-5p is widely distributed in the body, and its expression level is strongly associated with a poor prognosis and tumor progression [22–24]. It is shown that overexpression of hsa-miR-497-5p can regulate the expression of Smad3 and lead to cell cycle arrest in the G0/G1 phase [25–27]. Overexpression of miR-497-5p can reduce the content of insulin receptors and induce insulin resistance in HFD-MES rats [28–30]. Furthermore, the expression of miR-497-5p is associated with the occurrence and development of HCC. Zhang et al. reported that miR-497-5p was upregulated in HCC samples, and the high expression of miR-497-5p leads to the increase of tumor size and number. What is more, silencing of miR-497-5p could suppress HCC cell proliferation and migration [31]. On the contrary, Xu et al. reported that miR-497-5p was downregulated in HCC clinical samples and cell lines, and low expression of miR-497-5p was associated with aggressive tumor characteristics as well as patients’ prognosis in HCC patients [32]. Therefore, it is still controversial whether miR-497-5p is an oncogene or a tumor suppressor gene in HCC.
Here, we found that downregulation of miR-497-5p is closely associated with tumorigenesis and poor prognosis in HCC. The study is aimed at analyzing the expression of miR-497-5p and its potential target genes as well as its efficacy on overall survival (OS) and progression-free survival (PFS) in patients with HCC by bioinformatics analysis and reverse transcription quantitative polymerase chain reaction (RT-qPCR) to discuss its potential clinical significance in this disease.
2. Materials and Methods
2.1. Patients and Samples
Fresh HCC tumor tissue samples and adjacent normal tissues were collected via surgical resection between August 2004 and September 2014. A total of 328 primary HCC samples were collected, including tumor and adjacent nontumor liver tissues, 30 of which were paired with adjacent normal tissues. This study has been approved by the institutional research ethics committee of Shanghai Tenth People’s Hospital (Approval No. SHSY-IEC-2019 K10). All the participants provided written informed consent prior to investigation in the study.
2.2. Data Retrieval and Download
RNA expression data of HCC patients in TCGA (https://cancergenome.nih.gov/) and GEO (https://www.ncbi.nlm.nih.gov/gds/) databases were chosen for analysis. The sequencing data of miRNA expression of 372 HCC tissues and 50 normal tissues were procured from TCGA. miRNA data from three original GEO datasets (GSE36915, GSE97098, and GSE31383) were downloaded. The expression of hsa-miR-497-5p in HCC tissues and adjacent normal tissues was analyzed. The Cancer Cell Line Encyclopedia (CCLE, https://portals. http://broadinstitute.org/ccle) was used to verify the expression and DNA methylation of miR-497-5p in tumor cell lines.
2.3. DEGs Screening and Survival Analysis
The DEGs between HCC samples and noncancerous tissues were determined using the EdgeR software package [33].
2.4. RNA Isolation and Quantitative Reverse Transcriptase PCR
Total RNA was isolated from HCC cell lines, tumor samples, and normal tissue samples by the TRIzol reagent (Invitrogen, Waltham, MA, USA), according to the manufacturer’s guide. Obtaining cDNA from total RNA by reverse transcription and qRT-PCR of miR-497-5p was obtained using the TaqMan miRNA qRT-PCR Kit (HaiGene, Haerbin, China). Each sample shall be tested independently in at least triplicate. U6 small nuclear RNA was used for an internal control. The expression level of miR-497-5p and four potential target genes in each sample was calculated and expressed as ΔCt value, and the 2−ΔΔCT method was applied to analyze the relative hsa-miR-497-5p expression levels.
2.5. Cell Lines
Human liver cancer lines Huh7, Hep3B, and HepG2 were purchased from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China) and cultured in DMEM media (Invitrogen, Carlsbad, USA) and supplemented with 10% (
2.6. Cell Proliferation Assays
For the cell proliferation assays, 1,000 cells were seeded into 96-well plates to culture overnight. Next, the CCK8 (10 l) reagent was then added to each well, the absorbance (
2.7. Bioinformatics Analysis
Hierarchical clustering was performed using the multiple experiment viewer (MeV) 4.7.1 software programs (http://www.tm4.org/mev/). We used online target gene prediction software DIANA-miRPath (http://http://snf-515788.vm.okeanos.grnet.gr/), miRDB (http://mirdb.org/miRDB/), miRTarBase (http://mirtarbase.mbc.nctu.edu.tw/), and miRanda (http://www.microrna.org) to forecast several potential target genes of hsa-miR-497-5p. Subsequently, predicted target genes were subjected to KEGG pathway online analysis. The expression levels of four potential target genes in LIHC were obtained from GEPIA (http://gepia.cancer-pku.cn/).
2.8. Statistical Analysis
Statistical analysis was performed by GraphPad Prism (http://www.graphpad.com/) Software. Overall survival (OS) and progression-free survival (PFS) were evaluated by the Kaplan-Meier curves, and differences between survival rates were examined using the log-rank test. OS was calculated from the first day of diagnosis until the time of last follow-up or death. PFS was assessed as the interval from random assignment to the first documented disease progression or death because of any cause.
3. Results
3.1. Cluster Analysis of miRNA Expression in HCC
A total of 2,166 miRNA-seq data of 372 patients with LIHC cancer tissues (cancer) and 50 cases of adjacent normal tissue (normal) were downloaded from TCGA. The genes meet the criteria of
[figure omitted; refer to PDF]
In addition, there was a total of 372 LIHC samples with complete overall survival (OS) information; six of the top 20 DEGs are associated with the prognosis of patients (Figure 3).
[figures omitted; refer to PDF]
Next, the raw data were extracted from the GEO database (GEO accession no. GSE36915). In this present study, we examined the expression levels of miRNAs in tumor tissues and normal tissue from a group of 89 patients with HCC. The top ten DEGs miRNAs were screened out using MEV4.7.1 software (Figure 4(a)). As a result, we found that three DEGs (hsa-miR-497-5p, hsa-miR-139-5p, and hsa-miR-139-3p) were also as significant DEGs in the TCGA-LIHC database. Moreover, the three DEGs were also highly expressed in tumor tissues compared with normal tissue (Figures 4(b)–4(d)).
[figures omitted; refer to PDF]
To further identify the dependability of the miRNA genes, the same analysis was used to verify the next dataset from the NCBI GEO database (GEO accession no. GSE31383). The top twenty DEGs were selected between HCC and control samples (Figure 4(e)). Our results demonstrated that the expression level of miR-497-5p was significantly higher (
3.3. The Levels of miR-497-5p Expression in HCC Samples
The levels of miR-497-5p expression were evaluated in HCC samples (
[figures omitted; refer to PDF]
To evaluate the miR-497-5p expression levels divided between HCC and normal tissues, we detected the expression in 30 pairs of tumor tissues and adjacent nonneoplastic liver tissues using qRT-PCR. In particular, the present results indicated a significantly lower miR-497-5p in human HCC specimens compared with adjacent noncancerous specimens (
Subsequently, further analysis of the Pearson correlation between miR-497-5p in HCC tumor tissues in contrast with adjacent normal tissues was negatively correlated (
3.4. Association between miR-497-5p Expression and Clinical Features with HCC Prognosis
To further identify the prognostic significance of miR-497-5p expression in HCC, GraphPad software was used to assess the association of miR-497-5p expression between progress-free survival (PFS) and overall survival (OS). The results show that lower miR-497-5p expression was significantly correlated with poor PFS (Figure 5(d)) and OS (Figure 5(e)). Hence, miR-497-5p levels were dramatically related to an increased probability of PFS and OS in patients with HCC.
Our results further indicated that tumor diameter was significantly correlated with decreased duration of PFS (Figure 5(f)) and OS (Figure 5(g)) of patients with HCC. Survival analysis revealed that miR-497-5p levels and tumor diameter were also found to be concerned with PFS and OS. It was observed that patients with low miR-497-5p levels and large tumor diameter (≥5 cm) were associated with shorter PFS and OS than those with high miR-497-5p expression and small tumor diameter (Figures 5(h) and 5(i)).
3.5. Expression and Biological Role of miR-497-5p in HCC In Vitro
To explore the expression and biological role of miR-497-5p expression in HCC in vitro, we performed DNA methylation analysis of miR-497-5p in cancer cell lines from the Cancer Cell Line Encyclopedia (CCLE) dataset and found that DNA methylation analysis of miR-497-5p was low in most tumor cell lines (Figure 6(a)) and liver cancer cell lines (Figure 6(b)). Expression levels of miR-497-5p in liver cancer cell lines Huh7, Hep3B, and HepG2 were significantly lower than that in normal liver cell line LO2 from GEO dataset GSE71108 (Figure 6(c)).
[figures omitted; refer to PDF]
To investigate the biological role of miR-497-5p in HCC cells, we performed gain-of-function studies using miR-497-5p overexpressed plasmid in Huh7, Hep3B, and HepG2 cells (Figure 6(d)). As shown in Figure 6(e), overexpression of miR-497-5p significantly inhibited the growth rate of Huh7, Hep3B, and HepG2 cells transfected with the miR-497-5p overexpressed plasmid compared with the negative control-transfected cells.
3.6. Association between miR-497-5p Target Gene Expression and Their Clinical Features with HCC Prognosis
Next, we searched for potential target genes of miR-497-5p using four publicly available target gene prediction databases including DIANA-miRPath, miRDB, miRTarBase, and miRanda to predict the potential target genes of miR-497-5p.
The result showed that there were 125 commonly predicted target genes in at least 3 prediction results. Subsequently, predicted target genes were subjected to KEGG pathway analysis, and the results revealed some important cancer and stem-related pathways including the HIPPO signaling pathway, TGFb signaling pathway, and Wnt signaling pathway (Figure 7(a)), in which 4 genes (ACTG1, CSNK1D, PPP1CC, and BIRC5) were clustered in the HIPPO signaling pathway. Moreover, the mRNA expression levels of ACTG1, CSNK1D, PPP1CC, and BIRC5 were significantly lower in the miR-497-5p overexpression group when compared to the control group (Figure 7(b)), which suggested that ACTG1, CSNK1D, PPP1CC, and BIRC5 were regulated by miR-497-5p and were negatively correlated with miR-497-5p.
[figures omitted; refer to PDF]
Next, we analyzed the expression levels of ACTG1, CSNK1D, PPP1CC, and BIRC5 in HCC tissue samples in normal liver (
We then evaluated the prognostic value of ACTG1, CSNK1D, PPP1CC, and BIRC5 expression for patients with HCC. The OS in HCC patients with a low expression of ACTG1, CSNK1D, PPP1CC, or BIRC5 was prolonged relative to patients with high expression of ACTG1, CSNK1D, PPP1CC, or BIRC5 (Figure 7(e)). Our results indicated that ACTG1, CSNK1D, PPP1CC, and BIRC5 expression was positively correlated with lower OS in HCC patients.
4. Discussion
HCC is the fifth most common form of cancer type in the world [33–36]. The traditional treatment strategy for HCC is radical resection, but the rate of recurrence is very high, and the 5-year survival rate is low [37–41]. Thus, the prognosis of HCC is still unsatisfactory. Over the past years, due to the combination of the application of molecular biology techniques and tumor histological examination, great progress has been made in the disease [42–46]. miRNAs are considered an ideal biomarker compared with other biomarkers, miRNAs with sufficient sensitivity in clinical diagnosis and treatment [47–50].
Up to now, many studies have demonstrated that miRNA expression profiles in HCC and nontumor tissues are significantly different [51–54]. Certain miRNAs such as miR-21, miR-221, and miR-222 are frequently overexpressed in HCC [55–62]. These miRNAs have been demonstrated to promote oncogenesis by negatively regulating important tumor-suppressive protein-coding genes in HCC [63–67]. On the other hand, miRNAs such as miR-122, miR-125b, miR-139, miR-101, and let-7 are recurrently downregulated in HCC [68–72]. These miRNAs exert tumor-suppressive effects by repressing oncogenes in HCC [73–75]. miRNA deregulation is an early event that can be detected in premalignant liver dysplastic nodules and can further accumulate during liver carcinogenesis [76–79].
Interestingly, additional studies showed the existence of a large number of stable miRNAs in human serum/plasma, which laid the foundation for studying the role of serum/plasma microRNAs in the diagnosis and prognosis of HCC [80–83]. Differential expression of several serum microRNAs, including miR-16, miR122, miR-21, miR-223, miR-25, miR-375, and let-7f, in patients with HCC, patients with hepatitis B, and healthy individuals was reported recently [84–87].
The aberrant expression of miR-497-5p plays an essential role in tumorigenesis. miR-497-5p has been proven to be downregulated in NSCLC, gastric cancer, colorectal cancer, ovarian cancer, renal cell carcinoma, and melanoma [88–95]. Furthermore, in vitro and in vivo studies revealed that miR-497-5p overexpression suppressed HCC cell proliferation, colony formation, and metastasis [96]. Thus, for a more comprehensive understanding of the clinical value of miR-497-5p in HCC, we use bioinformatics and qRT-PCR assays to investigate the expression level of miR-497-5p in HCC tissues, as well as its association with overall survival and progress-free survival.
In our present study, the raw datasets from the GEO database and TCGA database were used to analyze miR-497-5p expression in HCC patients. The results demonstrated that the miR-497-5p expression level was significantly downregulated in HCC compared with adjacent normal tissues. A further correlation study showed that HCC with low miR-497-5p expression was significantly correlated with shorter OS and poor prognosis. In addition, we used quantitative real-time PCR to detect the expression of hsa-miR-497-5p in HCC biopsies and paracancerous tissues. This finding is consistent with our previous analysis data; the expression level of miR-497-5p in HCC was significantly downregulated contrasted with adjacent noncancerous tissues. Moreover, low expression of hsa-miR-497-5p correlated with poor clinicopathologic features, such as shorter OS and PFS. Furthermore, the correlation between hsa-miR-497-5p expression and clinical characters shows hsa-miR-497-5p expression downregulation and positive association with tumor diameter and negative association with OS and PFS. Our conclusions are consistent with previous studies; hsa-miR-497-5p is considered a tumor suppressor, which can inhibit the proliferation and invasion of tumor cells in HCC and NSCLC [97–99]. These results elucidated that the role of hsa-miR-497-5p exerts tumor-suppressive effects on clinical HCC samples.
In summary, our present study clarified the clinical and prognostic significance of hsa-miR-497-5p in HCC. The expression of hsa-miR-497-5p not only plays a significant role in the development and progression of HCC but also may serve as a potential diagnostic biomarker for HCC patients. The results also implied that hsa-miR-497-5p may be a promising and specific therapeutic target inductor of overexpression; cellular levels of miR-497-5p may be a new clinical therapeutic strategy for the treatment of HCC.
Authors’ Contributions
Lin-Lin Tian, Bin Qian, Xiao-Hui Jiang, and Yu-Shan Liu contributed equally to this work.
Acknowledgments
This study was supported partly by grants from the National Natural Science Foundation of China (81972214, 81772932, 81472202, 81201535, 81302065, 81671716, 81372175, and 81472209), Fundamental Research Funds for the Central Universities (22120170212), Shanghai Natural Science Foundation (20ZR1472400), Shanghai Municipal Commission of Health and Family Planning (201540228), Wu Jieping Medical Foundation (320.6750.14326), and Nantong Science and Technology Project (JC2018125).
[1] N. Furukawa, A. S. Popel, "Therapeutic peptides that immunoactivate the tumor microenvironment," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1875, article 188486, 2021.
[2] Q. L. Chen, C. F. Xie, K. L. Feng, D. Y. Cui, S. L. Sun, J. C. Zhang, C. M. Xiong, J. H. Huang, Z. Chong, "MicroRNAs carried by exosomes promote epithelial-mesenchymal transition and metastasis of liver cancer cells," American Journal of Translational Research, vol. 12 no. 10, pp. 6811-6826, 2020.
[3] M. Shao, Q. Yang, W. Zhu, H. Jin, J. Wang, J. Song, Y. Kong, X. Lv, "LncHOXA10 drives liver TICs self-renewal and tumorigenesis via HOXA10 transcription activation," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0921-y, 2018.
[4] Y. Kim, M. Jo, J. Schmidt, X. Luo, T. P. Prakash, T. Zhou, S. Klein, X. Xiao, N. Post, Z. Yin, A. R. MacLeod, "Enhanced potency of GalNAc-conjugated antisense oligonucleotides in hepatocellular cancer models," Molecular Therapy, vol. 27 no. 9, pp. 1547-1557, DOI: 10.1016/j.ymthe.2019.06.009, 2019.
[5] L. Suksanpaisan, R. Xu, M. Z. Tesfay, C. Bomidi, S. Hamm, R. Vandergaast, N. Jenks, M. B. Steele, A. Ota-Setlik, H. Akhtar, A. Luckay, R. Nowak, K. W. Peng, J. H. Eldridge, D. K. Clarke, S. J. Russell, R. M. Diaz, "Preclinical development of oncolytic immunovirotherapy for treatment of HPV POS cancers," Oncolytics, vol. 10,DOI: 10.1016/j.omto.2018.05.001, 2018.
[6] H. Wang, W. Chen, M. Jin, L. Hou, X. Chen, R. Zhang, J. Zhang, J. Zhu, "CircSLC3A2 functions as an oncogenic factor in hepatocellular carcinoma by sponging miR-490-3p and regulating PPM1F expression," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0909-7, 2018.
[7] Y. F. Yang, M. F. Zhang, Q. H. Tian, J. Fu, X. Yang, C. Z. Zhang, H. Yang, "SPAG5 interacts with CEP55 and exerts oncogenic activities via PI3K/AKT pathway in hepatocellular carcinoma," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0872-3, 2018.
[8] B. A. Fifield, J. Talia, C. Stoyanovich, M. J. Elliott, M. K. Bakht, A. Basilious, J. P. Samsoondar, M. Curtis, K. F. Stringer, L. A. Porter, "Cyclin-like proteins tip regenerative balance in the liver to favour cancer formation," Carcinogenesis, vol. 41 no. 6, pp. 850-862, DOI: 10.1093/carcin/bgz164, 2020.
[9] M. Yu, H. Luo, M. Fan, X. Wu, B. Shi, S. Di, Y. Liu, Z. Pan, H. Jiang, Z. Li, "Development of GPC3-specific chimeric antigen receptor-engineered natural killer cells for the treatment of hepatocellular carcinoma," Molecular Therapy, vol. 26 no. 2, pp. 366-378, DOI: 10.1016/j.ymthe.2017.12.012, 2018.
[10] E. Y. Peng, Y. Shu, Y. Wu, F. Zeng, S. Tan, Y. Deng, Y. Deng, H. Chen, L. Zhu, H. Xu, "Presence and diagnostic value of circulating tsncRNA for ovarian tumor," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0910-1, 2018.
[11] M. Ma, Y. Zhang, M. Weng, Y. Hu, Y. Xuan, Y. Hu, K. Lv, "lncRNA GCAWKR promotes gastric cancer development by scaffolding the chromatin modification factors WDR5 and KAT2A," Molecular Therapy, vol. 26 no. 11, pp. 2658-2668, DOI: 10.1016/j.ymthe.2018.09.002, 2018.
[12] K. Horita, H. Kurosaki, M. Nakatake, N. Kuwano, T. Oishi, H. Itamochi, S. Sato, H. Kono, M. Ito, K. Hasegawa, T. Harada, T. Nakamura, "lncRNA UCA1-mediated Cdc42 signaling promotes oncolytic Vaccinia virus cell- to-cell spread in ovarian cancer," Oncolytics, vol. 13, pp. 35-48, DOI: 10.1016/j.omto.2019.03.003, 2019.
[13] Y. Huang, B. Xiang, Y. Liu, Y. Wang, H. Kan, "LncRNA CDKN2B-AS1 promotes tumor growth and metastasis of human hepatocellular carcinoma by targeting let-7c-5p/NAP1L1 axis," Cancer Letters, vol. 437, pp. 56-66, DOI: 10.1016/j.canlet.2018.08.024, 2018.
[14] H. Liu, H. Deng, Y. Zhao, C. Li, Y. Liang, "LncRNA XIST/miR-34a axis modulates the cell proliferation and tumor growth of thyroid cancer through MET-PI3K-AKT signaling," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-0950-9, 2018.
[15] Q. Li, C. Dong, J. Cui, Y. Wang, X. Hong, "Over-expressed lncRNA HOTAIRM1 promotes tumor growth and invasion through up-regulating HOXA1 and sequestering G9a/EZH2/Dnmts away from the HOXA1 gene in glioblastoma multiforme," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-0941-x, 2018.
[16] R. Wang, Z. Ma, L. Feng, Y. Yang, C. Tan, Q. Shi, M. Lian, S. He, H. Ma, J. Fang, "LncRNA MIR31HG targets HIF1A and P21 to facilitate head and neck cancer cell proliferation and tumorigenesis by promoting cell-cycle progression," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0916-8, 2018.
[17] J. M. Santos, V. Cervera-Carrascon, R. Havunen, S. Zafar, M. Siurala, S. Sorsa, M. Anttila, A. Kanerva, A. Hemminki, "Adenovirus coding for interleukin-2 and tumor necrosis factor alpha replaces lymphodepleting chemotherapy in adoptive T cell therapy," Molecular Therapy, vol. 26 no. 9, pp. 2243-2254, DOI: 10.1016/j.ymthe.2018.06.001, 2018.
[18] C. Zeng, S. Liu, S. Lu, X. Yu, J. Lai, Y. Wu, S. Chen, L. Wang, Z. Yu, G. Luo, Y. Li, "The c-Myc-regulated lncRNA NEAT1 and paraspeckles modulate imatinib-induced apoptosis in CML cells," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0884-z, 2018.
[19] S. Xie, X. Yu, Y. Li, H. Ma, S. Fan, W. Chen, G. Pan, W. Wang, H. Zhang, J. Li, Z. Lin, "Upregulation of lncRNA ADAMTS9-AS2 promotes salivary adenoid cystic carcinoma metastasis via PI3K/Akt and MEK/Erk signaling," Molecular Therapy, vol. 26 no. 12, pp. 2766-2778, DOI: 10.1016/j.ymthe.2018.08.018, 2018.
[20] Y. Wang, X. Zhang, Z. Wang, Q. Hu, J. Wu, Y. Li, X. Ren, T. Wu, X. Tao, X. Chen, X. Li, J. Xia, B. Cheng, "LncRNA-p23154 promotes the invasion-metastasis potential of oral squamous cell carcinoma by regulating Glut1-mediated glycolysis," Cancer Letters, vol. 434, pp. 172-183, DOI: 10.1016/j.canlet.2018.07.016, 2018.
[21] P. Magini, E. Scarano, I. Donati, A. Sensi, L. Mazzanti, A. Perri, F. Tamburrino, P. Mongelli, A. Percesepe, P. Visconti, A. Parmeggiani, M. Seri, C. Graziano, "Challenges in the clinical interpretation of small de novo copy number variants in neurodevelopmental disorders," Gene, vol. 706, pp. 162-171, DOI: 10.1016/j.gene.2019.05.007, 2019.
[22] L. Chai, X. J. Kang, Z. Z. Sun, M. F. Zeng, S. R. Yu, Y. Ding, J. Q. Liang, T. T. Li, J. Zhao, "miR-497-5p, miR-195-5p and miR-455-3p function as tumor suppressors by targeting hTERT in melanoma A375 cells," Cancer Management and Research, vol. Volume 10, pp. 989-1003, DOI: 10.2147/CMAR.S163335, 2018.
[23] J. Gu, Y. Wang, X. Wang, D. Zhou, C. Shao, M. Zhou, Z. He, "Downregulation of lncRNA GAS5 confers tamoxifen resistance by activating miR-222 in breast cancer," Cancer Letters, vol. 434,DOI: 10.1016/j.canlet.2018.06.039, 2018.
[24] L. Y. Sun, X. J. Li, Y. M. Sun, W. Huang, K. Fang, C. Han, Z. H. Chen, X. Q. Luo, Y. Q. Chen, W. T. Wang, "LncRNA ANRIL regulates AML development through modulating the glucose metabolism pathway of AdipoR1/AMPK/SIRT1," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0879-9, 2018.
[25] H. Xu, G. F. Chen, Y. S. Ma, H. W. Zhang, Y. Zhou, G. H. Liu, D. Y. Chen, J. Ping, Y. H. Liu, X. Mou, D. Fu, "Hepatic proteomic changes and Sirt1/AMPK signaling activation by oxymatrine treatment in rats with non-alcoholic steatosis," Frontiers in Pharmacology, vol. 11,DOI: 10.3389/fphar.2020.00216, 2020.
[26] J. Guo, W. Liu, Z. Zeng, J. Lin, X. Zhang, L. Chen, "Tgfb3 and Mmp13 regulated the initiation of liver fibrosis progression as dynamic network biomarkers," Journal of Cellular and Molecular Medicine, vol. 25, pp. 67-879, 2020.
[27] S. Yoshida, S. Miyagawa, S. Fukushima, T. Kawamura, N. Kashiyama, F. Ohashi, T. Toyofuku, K. Toda, Y. Sawa, "Maturation of human induced pluripotent stem cell-derived cardiomyocytes by soluble factors from human mesenchymal stem cells," Molecular Therapy, vol. 26 no. 11, pp. 2681-2695, DOI: 10.1016/j.ymthe.2018.08.012, 2018.
[28] O. Hemminki, M. Oksanen, K. Taipale, I. Liikanen, A. Koski, T. Joensuu, A. Kanerva, A. Hemminki, "Oncograms visualize factors influencing long-term survival of cancer patients treated with adenoviral oncolytic immunotherapy," Molecular Therapy—Oncolytics, vol. 9, pp. 41-50, DOI: 10.1016/j.omto.2018.04.003, 2018.
[29] J. Dai, Y. Su, S. Zhong, L. Cong, B. Liu, J. Yang, Y. Tao, Z. He, C. Chen, Y. Jiang, "Exosomes: key players in cancer and potential therapeutic strategy," Signal Transduction and Targeted Therapy, vol. 5 no. 1,DOI: 10.1038/s41392-020-00261-0, 2020.
[30] Y. S. Ma, X. F. Wang, F. Yu, T. M. Wu, J. B. Liu, Y. J. Zhang, Q. Xia, Z. Y. Jiang, Q. L. Lin, D. Fu, "Inhibition of USP14 and UCH37 deubiquitinating activity by b-AP15 as a potential therapy for tumors with _p53_ deficiency," Signal Transduction and Targeted Therapy, vol. 5 no. 1,DOI: 10.1038/s41392-020-0143-9, 2020.
[31] Y. Zhang, Z. Zhu, S. Huang, Q. Zhao, C. Huang, Y. Tang, C. Sun, Z. Zhang, L. Wang, H. Chen, M. Chen, W. Ju, X. He, "lncRNA XIST regulates proliferation and migration of hepatocellular carcinoma cells by acting as miR-497-5p molecular sponge and targeting PDCD4," Cancer Cell International, vol. 19 no. 1,DOI: 10.1186/s12935-019-0909-8, 2019.
[32] G. Xu, Z. Li, Z. P. Huang, F. C. Brunicardi, F. Jia, C. Song, H. J. Zou, R. F. Sun, "miR-497-5p inhibits cell proliferation and metastasis in hepatocellular carcinoma by targeting insulin-like growth factor 1," Molecular Genetics & Genomic Medicine, vol. 7 no. 10, article e00860,DOI: 10.1002/mgg3.860, 2019.
[33] Y. S. Ma, X. F. Wang, Y. J. Zhang, P. Luo, H. D. Long, L. Li, H. Q. Yang, R. T. Xie, C. Y. Jia, G. X. Lu, Z. Y. Chang, J. J. Zhang, S. B. Xue, Z. W. Lv, F. Yu, Q. Xia, D. Fu, "Inhibition of USP14 deubiquitinating activity as a potential therapy for tumors with _p53_ deficiency," Molecular Therapy—Oncolytics, vol. 16, pp. 147-157, DOI: 10.1016/j.omto.2019.12.013, 2020.
[34] J. Fahrer, "Switching off DNA repair--how colorectal cancer evades targeted therapies through adaptive mutability," Signal Transduction and Targeted Therapy, vol. 5 no. 1,DOI: 10.1038/s41392-020-0120-3, 2020.
[35] M. C. Ballesteros-Briones, E. Martisova, E. Casales, N. Silva-Pilipich, M. Buñuales, J. Galindo, U. Mancheño, M. Gorraiz, J. J. Lasarte, G. Kochan, D. Escors, A. R. Sanchez-Paulete, I. Melero, J. Prieto, R. Hernandez-Alcoceba, S. Hervas-Stubbs, C. Smerdou, "Short-term local expression of a PD-L1 blocking antibody from a self- replicating RNA vector induces potent antitumor responses," Molecular Therapy, vol. 27 no. 11, pp. 1892-1905, DOI: 10.1016/j.ymthe.2019.09.016, 2019.
[36] Y. J. Kim, D. S. Baek, S. Lee, D. Park, H. N. Kang, B. C. Cho, Y. S. Kim, "Dual-targeting of EGFR and neuropilin-1 attenuates resistance to EGFR-targeted antibody therapy in KRAS-mutant non-small cell lung cancer," Cancer Letters, vol. 466, pp. 23-34, DOI: 10.1016/j.canlet.2019.09.005, 2019.
[37] Y. S. Ma, Z. J. Wu, H. W. Zhang, B. Cai, T. Huang, H. D. Long, H. Xu, Y. Z. Zhao, Y. Z. Yin, S. B. Xue, L. Li, C. L. Liu, R. T. Xie, L. L. Tian, J. B. Liu, X. M. Wu, D. Fu, "Dual regulatory mechanisms of expression and mutation involving metabolism- related genes FDFT1 and UQCR5 during CLM," Molecular Therapy—Oncolytics, vol. 14, pp. 172-178, DOI: 10.1016/j.omto.2019.04.008, 2019.
[38] D. A. Fergusson, N. L. Wesch, G. J. Leung, J. L. MacNeil, I. Conic, J. Presseau, K. D. Cobey, J. S. Diallo, R. Auer, J. Kimmelman, N. Kekre, N. el-Sayes, R. Krishnan, B. A. Keller, C. Ilkow, M. M. Lalu, "Assessing the completeness of reporting in preclinical oncolytic virus therapy studies," Molecular Therapy—Oncolytics, vol. 14, pp. 179-187, DOI: 10.1016/j.omto.2019.05.004, 2019.
[39] D. Rodrigues, T. Souza, D. G. J. Jennen, L. Lemmens, J. C. S. Kleinjans, T. M. de Kok, "Drug-induced gene expression profile changes in relation to intestinal toxicity: state-of-the-art and new approaches," Cancer Treatment Reviews, vol. 77, pp. 57-66, DOI: 10.1016/j.ctrv.2019.06.004, 2019.
[40] T. K. Leslie, A. D. James, F. Zaccagna, J. T. Grist, S. Deen, A. Kennerley, F. Riemer, J. D. Kaggie, F. A. Gallagher, F. J. Gilbert, W. J. Brackenbury, "Sodium homeostasis in the tumour microenvironment," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1872 no. 2, article 188304,DOI: 10.1016/j.bbcan.2019.07.001, 2019.
[41] C. Jimenez Calvente, H. Del Pilar, M. Tameda, C. D. Johnson, A. E. Feldstein, "MicroRNA 223 3p negatively regulates the NLRP3 inflammasome in acute and chronic liver injury," Molecular Therapy, vol. 28 no. 2, pp. 653-663, DOI: 10.1016/j.ymthe.2019.09.013, 2020.
[42] W. T. Liu, Y. Wang, J. Zhang, F. Ye, X. H. Huang, B. Li, Q. Y. He, "A novel strategy of integrated microarray analysis identifies CENPA, CDK1 and CDC20 as a cluster of diagnostic biomarkers in lung adenocarcinoma," Cancer Letters, vol. S0304-3835, pp. 30246-30255, 2018.
[43] Y. S. Ma, Z. W. Lv, F. Yu, Z. Y. Chang, X. L. Cong, X. M. Zhong, G. X. Lu, J. Zhu, D. Fu, "MicroRNA-302a/d inhibits the self-renewal capability and cell cycle entry of liver cancer stem cells by targeting the E2F7/AKT axis," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-0927-8, 2018.
[44] Y. S. Ma, F. Yu, X. M. Zhong, G. X. Lu, X. L. Cong, S. B. Xue, W. T. Xie, L. K. Hou, L. J. Pang, W. Wu, W. Zhang, L. L. Cong, T. Liu, H. D. Long, R. Sun, H. Y. Sun, Z. W. Lv, C. Y. Wu, D. Fu, "miR-30 family reduction maintains self-renewal and promotes tumorigenesis in NSCLC-initiating cells by targeting oncogene TM4SF1," Molecular Therapy, vol. 26 no. 12, pp. 2751-2765, DOI: 10.1016/j.ymthe.2018.09.006, 2018.
[45] D. Lv, Q. H. Ma, J. J. Duan, H. B. Wu, X. L. Zhao, S. C. Yu, X. W. Bian, "Optimized dissociation protocol for isolating human glioma stem cells from tumorspheres via fluorescence-activated cell sorting," Cancer Letters, vol. 377 no. 1, pp. 105-115, DOI: 10.1016/j.canlet.2016.04.022, 2016.
[46] H. Jia, H. Qi, Z. Gong, S. Yang, J. Ren, Y. Liu, M. Y. Li, G. G. Chen, "The expression of FOXP3 and its role in human cancers," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1871 no. 1, pp. 170-178, DOI: 10.1016/j.bbcan.2018.12.004, 2019.
[47] X. Xin, M. Wu, Q. Meng, C. Wang, Y. Lu, Y. Yang, X. Li, Q. Zheng, H. Pu, X. Gui, T. Li, J. Li, S. Jia, D. Lu, "Long noncoding RNA HULC accelerates liver cancer by inhibiting PTEN via autophagy cooperation to miR15a," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0843-8, 2018.
[48] B. Liu, S. Jiang, M. Li, X. Xiong, M. Zhu, D. Li, L. Zhao, L. Qian, L. Zhai, J. Li, H. Lu, S. Sun, J. Lin, Y. Lu, X. Li, M. Tan, "Proteome-wide analysis of USP14 substrates revealed its role in hepatosteatosis via stabilization of FASN," Nature Communications, vol. 9 no. 1, article 4770,DOI: 10.1038/s41467-018-07185-y, 2018.
[49] X. Wu, H. Luo, B. Shi, S. Di, R. Sun, J. Su, Y. Liu, H. Li, H. Jiang, Z. Li, "Combined antitumor effects of sorafenib and GPC3-CAR T cells in mouse models of hepatocellular carcinoma," Molecular Therapy, vol. 27 no. 8, pp. 1483-1494, DOI: 10.1016/j.ymthe.2019.04.020, 2019.
[50] H. Fan, P. Lv, T. Mu, X. Zhao, Y. Liu, Y. Feng, J. Lv, M. Liu, H. Tang, "LncRNA n335586/miR-924/CKMT1A axis contributes to cell migration and invasion in hepatocellular carcinoma cells," Cancer Letters, vol. 429, pp. 89-99, DOI: 10.1016/j.canlet.2018.05.010, 2018.
[51] W. Zhou, L. Gong, Q. Wu, C. Xing, B. Wei, T. Chen, Y. Zhou, S. Yin, B. Jiang, H. Xie, L. Zhou, S. Zheng, "PHF8 upregulation contributes to autophagic degradation of E-cadherin, epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-0890-4, 2018.
[52] H. Dong, W. Wang, S. Mo, R. Chen, K. Zou, J. Han, F. Zhang, J. Hu, "SP1-induced lncRNA AGAP2-AS1 expression promotes chemoresistance of breast cancer by epigenetic regulation of MyD88," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-0875-3, 2018.
[53] H. Yuan, Z. Chen, S. Bai, H. Wei, Y. Wang, R. Ji, Q. Guo, Q. Li, Y. Ye, J. Wu, Y. Zhou, L. Qiao, "Molecular mechanisms of lncRNA SMARCC2/miR-551b-3p/TMPRSS4 axis in gastric cancer," Cancer Letters, vol. 418, pp. 84-96, DOI: 10.1016/j.canlet.2018.01.032, 2018.
[54] X. N. Li, Z. J. Wang, C. X. Ye, B. C. Zhao, Z. L. Li, Y. Yang, "RNA sequencing reveals the expression profiles of circRNA and indicates that circDDX17 acts as a tumor suppressor in colorectal cancer," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-1006-x, 2018.
[55] G. Zhang, S. Li, J. Lu, Y. Ge, Q. Wang, G. Ma, Q. Zhao, D. Wu, W. Gong, M. du, H. Chu, M. Wang, A. Zhang, Z. Zhang, "LncRNA MT1JP functions as a ceRNA in regulating FBXW7 through competitively binding to miR-92a-3p in gastric cancer," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0829-6, 2018.
[56] V. Reebye, K. W. Huan, K. Czysz, S. Ciriello, S. Dorman, I. Reccia, H. S. Lai, L. Peng, N. Kostomitsopoulos, J. Nicholls, R. S. Habib, D. A. Tomalia, P. Saetrom, E. Wilkes, P. Cutillas, D. Blakey, J. J. Rossi, N. Habib, "Liver activation of hepatocellular nuclear factor-4 α by small activating rna rescues dyslipidemia and improves metabolic profile," Molecular Therapy--Nucleic Acids, vol. 19, pp. 361-370, DOI: 10.2139/ssrn.3304286, 2018.
[57] Y. Ma, J. Zhang, L. Wen, A. Lin, "Membrane-lipid associated lncRNA: a new regulator in cancer signaling," Cancer Letters, vol. 419, pp. 27-29, DOI: 10.1016/j.canlet.2018.01.008, 2018.
[58] M. Kang, M. Ren, Y. Li, Y. Fu, M. Deng, C. Li, "Exosome-mediated transfer of lncRNA PART1 induces gefitinib resistance in esophageal squamous cell carcinoma via functioning as a competing endogenous RNA," Journal of Experimental & Clinical Cancer Research, vol. 37 no. 1,DOI: 10.1186/s13046-018-0845-9, 2018.
[59] G. L. Rogers, H. Y. Chen, H. Morales, P. M. Cannon, "Homologous recombination-based genome editing by clade F AAVs is inefficient in the absence of a targeted DNA break," Molecular Therapy, vol. 27 no. 10, pp. 1726-1736, DOI: 10.1016/j.ymthe.2019.08.019, 2019.
[60] J. M. Matés, J. A. Campos-Sandoval, J. L. Santos-Jiménez, J. Márquez, "Dysregulation of glutaminase and glutamine synthetase in cancer," Cancer Letters, vol. 467, pp. 29-39, DOI: 10.1016/j.canlet.2019.09.011, 2019.
[61] A. He, Y. Liu, Z. Chen, J. Li, M. Chen, L. Liu, X. Liao, Z. Lv, Y. Zhan, C. Zhuang, J. Lin, W. Huang, H. Mei, "Over-expression of long noncoding RNA BANCR inhibits malignant phenotypes of human bladder cancer," Journal of Experimental & Clinical Cancer Research, vol. 35 no. 1,DOI: 10.1186/s13046-016-0397-9, 2016.
[62] Y. Tang, Y. He, P. Zhang, J. Wang, C. Fan, L. Yang, F. Xiong, S. Zhang, Z. Gong, S. Nie, Q. Liao, X. Li, X. Li, Y. Li, G. Li, Z. Zeng, W. Xiong, C. Guo, "LncRNAs regulate the cytoskeleton and related Rho/ROCK signaling in cancer metastasis," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0825-x, 2018.
[63] X. Zhao, J. Sun, Y. Chen, W. Su, H. Shan, Y. Li, Y. Wang, N. Zheng, H. Shan, H. Liang, "lncRNA PFAR promotes lung fibroblast activation and fibrosis by targeting miR-138 to regulate the YAP1-twist axis," Molecular Therapy, vol. 26 no. 9, pp. 2206-2217, DOI: 10.1016/j.ymthe.2018.06.020, 2018.
[64] B. Yue, C. Liu, H. Sun, M. Liu, C. Song, R. Cui, S. Qiu, M. Zhong, "A positive feed-forward loop between LncRNA-CYTOR and Wnt/ β -catenin signaling promotes metastasis of colon cancer," Molecular Therapy, vol. 26 no. 5, pp. 1287-1298, DOI: 10.1016/j.ymthe.2018.02.024, 2018.
[65] M. Boesch, F. Baty, H. Rumpold, S. Sopper, D. Wolf, M. H. Brutsche, "Fibroblasts in cancer: defining target structures for therapeutic intervention," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1872 no. 1, pp. 111-121, DOI: 10.1016/j.bbcan.2019.06.003, 2019.
[66] Y. Liu, J. Feng, M. Sun, G. Yang, H. Yuan, Y. Wang, Y. Bu, M. Zhao, S. Zhang, X. Zhang, "Long non-coding RNA HULC activates HBV by modulating HBx/STAT3/miR-539/APOBEC3B signaling in HBV-related hepatocellular carcinoma," Cancer Letters, vol. 454, pp. 158-170, DOI: 10.1016/j.canlet.2019.04.008, 2019.
[67] Z. Huang, J. Wen, J. Yu, J. Liao, S. Liu, N. Cai, H. Liang, X. Chen, Z. Ding, B. Zhang, "MicroRNA-148a-3p inhibits progression of hepatocelluar carcimoma by repressing SMAD2 expression in an Ago2 dependent manner," Journal of Experimental & Clinical Cancer Research, vol. 39 no. 1,DOI: 10.1186/s13046-020-01649-0, 2020.
[68] C. Tomasetti, J. Poling, N. J. Roberts, N. R. London, M. E. Pittman, M. C. Haffner, A. Rizzo, A. Baras, B. Karim, A. Kim, C. M. Heaphy, A. K. Meeker, R. H. Hruban, C. A. Iacobuzio-Donahue, B. Vogelstein, "Cell division rates decrease with age, providing a potential explanation for the age-dependent deceleration in cancer incidence," Proceedings of the National Academy of Sciences of the United States of America, vol. 116 no. 41, pp. 20482-20488, DOI: 10.1073/pnas.1905722116, 2019.
[69] M. Alečković, S. S. McAllister, K. Polyak, "Metastasis as a systemic disease: molecular insights and clinical implications," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1872 no. 1, pp. 89-102, DOI: 10.1016/j.bbcan.2019.06.002, 2019.
[70] L. Rimassa, R. Danesi, T. Pressiani, P. Merle, "Management of adverse events associated with tyrosine kinase inhibitors: improving outcomes for patients with hepatocellular carcinoma," Cancer Treatment Reviews, vol. 77, pp. 20-28, DOI: 10.1016/j.ctrv.2019.05.004, 2019.
[71] D. W. Ho, Y. M. Tsui, K. M. Sze, L. K. Chan, T. T. Cheung, E. Lee, P. C. Sham, S. K. Tsui, T. K. Lee, I. O. Ng, "Single-cell transcriptomics reveals the landscape of intra-tumoral heterogeneity and stemness-related subpopulations in liver cancer," Cancer Letters, vol. 459, pp. 176-185, DOI: 10.1016/j.canlet.2019.06.002, 2019.
[72] Y. Li, X. Han, H. Feng, J. Han, "Long noncoding RNA OIP5-AS1 in cancer," Clinica Chimica Acta, vol. 499, pp. 75-80, DOI: 10.1016/j.cca.2019.08.031, 2019.
[73] J. R. Conejo-Garcia, "Breaking barriers for T cells by targeting the EPHA2/TGF- β /COX-2 axis in pancreatic cancer," The Journal of Clinical Investigation, vol. 130, pp. 3521-3523, 2019.
[74] W. Z. Ding, Q. F. Ni, Y. T. Lu, L. L. Kong, J. J. Yu, L. W. Tan, L. B. Kong, "MicroRNA-497 regulates cell proliferation in hepatocellular carcinoma," Oncology Letters, vol. 11 no. 2, pp. 1081-1088, DOI: 10.3892/ol.2015.3981, 2016.
[75] L. Zhang, Z. Yu, Y. Xian, X. Lin, "MicroRNA-497 inhibits cell proliferation and induces apoptosis by targeting YAP1 in human hepatocellular carcinoma," FEBS Open Bio, vol. 6 no. 2, pp. 155-164, DOI: 10.1002/2211-5463.12032, 2016.
[76] M. Tahmasebi-Birgani, H. Ansari, V. Carloni, "Defective mitosis-linked DNA damage response and chromosomal instability in liver cancer," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1872 no. 1, pp. 60-65, DOI: 10.1016/j.bbcan.2019.05.008, 2019.
[77] G. Li, K. Wang, J. Wang, S. Qin, X. Sun, H. Ren, "miR-497-5p inhibits tumor cell growth and invasion by targeting SOX5 in non-small-cell lung cancer," Journal of Cellular Biochemistry, vol. 120 no. 6, pp. 10587-10595, DOI: 10.1002/jcb.28345, 2019.
[78] Z. Chen, Z. Liu, M. Zhang, W. Huang, Z. Li, S. Wang, C. Zhang, B. Dong, J. Gao, L. Shen, "EPHA2 blockade reverses acquired resistance to afatinib induced by EPHA2-mediated MAPK pathway activation in gastric cancer cells and avatar mice," International Journal of Cancer, vol. 145 no. 9, pp. 2440-2449, DOI: 10.1002/ijc.32313, 2019.
[79] R. Lévêque, C. Corbet, L. Aubert, M. Guilbert, C. Lagadec, E. Adriaenssens, J. Duval, P. Finetti, D. Birnbaum, N. Magné, V. Chopin, F. Bertucci, X. le Bourhis, R. A. Toillon, "ProNGF increases breast tumor aggressiveness through functional association of TrkA with EphA2," Cancer Letters, vol. 449, pp. 196-206, DOI: 10.1016/j.canlet.2019.02.019, 2019.
[80] J. Y. Li, T. Xiao, H. M. Yi, H. Yi, J. Feng, J. F. Zhu, W. Huang, S. S. Lu, Y. H. Zhou, X. H. Li, Z. Q. Xiao, "S897 phosphorylation of EphA2 is indispensable for EphA2-dependent nasopharyngeal carcinoma cell invasion, metastasis and stem properties," Cancer Letters, vol. 444, pp. 162-174, DOI: 10.1016/j.canlet.2018.12.011, 2019.
[81] M. de Robertis, T. Mazza, C. Fusilli, L. Loiacono, M. L. Poeta, M. Sanchez, E. Massi, G. Lamorte, M. G. Diodoro, E. Pescarmona, E. Signori, G. Pesole, A. L. Vescovi, J. Garcia-Foncillas, V. M. Fazio, "EphB2 stem-related and EphA2 progression-related miRNA-based networks in progressive stages of CRC evolution clinical significance and potential miRNA drivers," Molecular Cancer, vol. 17 no. 1,DOI: 10.1186/s12943-018-0912-z, 2018.
[82] I. Nikas, H. S. Ryu, S. Theocharis, "Viewing the Eph receptors with a focus on breast cancer heterogeneity," Cancer Letters, vol. 434, pp. 160-171, DOI: 10.1016/j.canlet.2018.07.030, 2018.
[83] Y. Shi, D.-D. Zhang, J.-B. Liu, X.-L. Yang, R. Xin, C.-Y. Jia, H.-M. Wang, G.-X. Lu, P.-Y. Wang, Y. Liu, Z.-J. Li, J. Deng, Q.-L. Lin, L. Ma, S.-S. Feng, X.-Q. Chen, X.-M. Zheng, Y.-F. Zhou, Y.-J. Hu, H.-Q. Yin, L.-L. Tian, L.-P. Gu, Z.-W. Lv, F. Yu, W. Li, Y.-S. Ma, F. Da, "Comprehensive analysis to identify DLEU2L/TAOK1 axis as a prognostic biomarker in hepatocellular carcinoma," Molecular Therapy - Nucleic Acids, vol. 23, pp. 702-718, DOI: 10.1016/j.omtn.2020.12.016, 2021.
[84] B. Zhang, X. Wang, J. Deng, H. Zheng, W. Liu, S. Chen, J. Tian, F. Wang, "p53-dependent upregulation of miR-16-2 by sanguinarine induces cell cycle arrest and apoptosis in hepatocellular carcinoma," Cancer LettersJournal of Experimental & Clinical Cancer Research, vol. 459, pp. 50-58, DOI: 10.1016/j.canlet.2019.05.042, 2019.
[85] J. Lu, T. Tan, L. Zhu, H. Dong, R. Xian, "Hypomethylation Causes MIR21 Overexpression in Tumors," Molecular Therapy - Oncolytics, vol. 18, pp. 47-57, DOI: 10.1016/j.omto.2020.05.011, 2020.
[86] H. L. Wang, P. F. Liu, J. Yue, W. H. Jiang, Y. L. Cui, H. Ren, H. Wang, Y. Zhuang, Y. Liu, D. Jiang, Q. Dong, H. Zhang, J. H. Mi, Z. M. Xu, C. J. Tian, Z. Z. Zhang, X. W. Wang, M. N. Su, W. Lu, "Somatic gene mutation signatures predict cancer type and prognosis in multiple cancers with pan-cancer 1000 gene panel," Cancer Letters, vol. 470, pp. 181-190, DOI: 10.1016/j.canlet.2019.11.022, 2020.
[87] H. He, Y. Wang, P. Ye, D. Yi, Y. Cheng, H. Tang, Z. Zhu, X. Wang, S. Jin, "Long noncoding RNA ZFPM2-AS1 acts as a miRNA sponge and promotes cell invasion through regulation of miR-139/GDF10 in hepatocellular carcinoma," Journal of Experimental & Clinical Cancer Research, vol. 39 no. 1,DOI: 10.1186/s13046-020-01664-1, 2020.
[88] L. Wang, J. Guo, J. Zhou, D. Wang, X. Kang, L. Zhou, "NF-kappaB maintains the stemness of colon cancer cells by downregulating miR-195-5p/497-5p and upregulating MCM2," Journal of Experimental & Clinical Cancer Research, vol. 39 no. 1,DOI: 10.1186/s13046-020-01704-w, 2020.
[89] Y. Zhang, X. Li, J. Zhang, L. Mao, "E6 hijacks KDM5C/lnc_000231/miR-497-5p/CCNE1 axis to promote cervical cancer progression," Journal of Cellular and Molecular Medicine, vol. 24 no. 19, pp. 11422-11433, DOI: 10.1111/jcmm.15746, 2020.
[90] Y. Xiao, R. M. Najeeb, D. Ma, K. Yang, Q. Zhong, Q. Liu, "Upregulation of CENPM promotes hepatocarcinogenesis through mutiple mechanisms," Journal of Experimental & Clinical Cancer Research, vol. 38 no. 1,DOI: 10.1186/s13046-019-1444-0, 2019.
[91] K. J. Livak, T. D. Schmittgen, "Analysis of relative gene expression data using real-time quantitative PCR and the 2− ΔΔ _C_ T method," Methods, vol. 25 no. 4, pp. 402-408, DOI: 10.1006/meth.2001.1262, 2001.
[92] P. Dong, Y. Xiong, J. Yue, D. Xu, K. Ihira, Y. Konno, N. Kobayashi, Y. Todo, H. Watari, "Long noncoding RNA NEAT1 drives aggressive endometrial cancer progression via miR-361-regulated networks involving STAT3 and tumor microenvironment-related genes," Journal of Experimental & Clinical Cancer Research, vol. 38 no. 1,DOI: 10.1186/s13046-019-1306-9, 2019.
[93] Y. S. Ma, Z. J. Wu, R. Z. Bai, H. Dong, B. X. Xie, X. H. Wu, X. S. Hang, A. N. Liu, X. H. Jiang, G. R. Wang, J. J. Jiang, W. H. Xu, X. P. Chen, G. H. Tan, D. Fu, J. B. Liu, Q. Liu, "DRR1 promotes glioblastoma cell invasion and epithelial-mesenchymal transition via regulating AKT activation," Cancer Letters, vol. 423, pp. 86-94, DOI: 10.1016/j.canlet.2018.03.015, 2018.
[94] Y. Chen, H. Zhao, H. Li, X. Feng, H. Tang, C. Qiu, J. Zhang, B. Fu, "LINC01234/microRNA-31-5p/MAGEA3 axis mediates the proliferation and chemoresistance of hepatocellular carcinoma cells," Molecular Therapy--Nucleic Acids, vol. 19, pp. 168-178, DOI: 10.1016/j.omtn.2019.10.035, 2020.
[95] Y. Zhang, J. Song, Z. Zhao, M. Yang, M. Chen, C. Liu, J. Ji, D. Zhu, "Single-cell transcriptome analysis reveals tumor immune microenvironment heterogenicity and granulocytes enrichment in colorectal cancer liver metastases," Cancer Letters, vol. 470, pp. 84-94, DOI: 10.1016/j.canlet.2019.10.016, 2020.
[96] Y. Chen, C. Peng, J. Chen, D. Chen, B. Yang, B. He, W. Hu, Y. Zhang, H. Liu, L. Dai, H. Xie, L. Zhou, J. Wu, S. Zheng, "WTAP facilitates progression of hepatocellular carcinoma via m6A-HuR-dependent epigenetic silencing of ETS1," Molecular Cancer, vol. 18 no. 1,DOI: 10.1186/s12943-019-1053-8, 2019.
[97] J. P. Unfried, G. Serrano, B. Suárez, P. Sangro, V. Ferretti, C. Prior, L. Boix, J. Bruix, B. Sangro, V. Segura, P. Fortes, "Identification of coding and long noncoding RNAs differentially expressed in tumors and preferentially expressed in healthy tissues," Cancer Research, vol. 79 no. 20, pp. 5167-5180, DOI: 10.1158/0008-5472.CAN-19-0400, 2019.
[98] S. Vivekanadhan, D. Mukhopadhyay, "Divergent roles of Plexin D1 in cancer," Biochimica Et Biophysica Acta. Reviews on Cancer, vol. 1872 no. 1, pp. 103-110, DOI: 10.1016/j.bbcan.2019.05.004, 2019.
[99] B. Y. Tam, K. Chiu, H. Chung, C. Bossard, J. D. Nguyen, E. Creger, B. W. Eastman, C. C. Mak, M. Ibanez, A. Ghias, J. Cahiwat, L. Do, S. Cho, J. Nguyen, V. Deshmukh, J. Stewart, C. W. Chen, C. Barroga, L. Dellamary, S. K. KC, T. J. Phalen, J. Hood, S. Cha, Y. Yazici, "The CLK inhibitor SM08502 induces anti-tumor activity and reduces Wnt pathway gene expression in gastrointestinal cancer models," Cancer Letters, vol. 473, pp. 186-197, DOI: 10.1016/j.canlet.2019.09.009, 2020.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2021 Lin-Lin Tian et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
Background. MicroRNAs (miRNAs) have been demonstrated to exhibit important regulatory roles in multiple malignancies, including hepatocellular carcinoma (HCC). hsa-miR-497-5p was reported to involve in cancer progression and poor prognosis in many kinds of tumors. However, the expression and its clinical significance of hsa-miR-497-5p in HCC remain unclear. Methods. In the present study, we investigated the expression of hsa-miR-497-5p in HCC and analyzed the correction of clinical features with prognosis. The expression levels of hsa-miR-497-5p and potential target genes were analyzed in HCC and adjacent noncancerous tissues using The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) datasets. Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was used to analyze hsa-miR-497-5p levels in 328 HCC tissues and 30 paired adjacent noncancer tissues. Overall survival (OS) and progression-free survival (PFS) of patients with HCC were assessed using the Kaplan-Meier method and the log-rank test. Results. The hsa-miR-497-5p expression levels were decreased, and its target genes ACTG1, CSNK1D, PPP1CC, and BIRC5 were upregulated in HCC tissues compared with normal tissues. Lower levels of hsa-miR-497-5p expression and higher levels of the four target genes were significantly associated with higher tumor diameter. Moreover, patients with lower hsa-miR-497-5p expression and higher target genes levels had shorter OS. Conclusion. The expression levels of hsa-miR-497-5p may play an important regulatory role in HCC and are closely correlated with HCC progression and poor prognosis in patients. The hsa-miR-497-5p may be a specific therapeutic target for the treatment of HCC.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details



1 Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China; Department of Microbiology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541001, China
2 Department of General Surgery, Shanghai Eighth People’s Hospital, Shanghai 200235, China
3 Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Nantong University, Nantong 226631, China
4 Department of Pathology, Nantong Tumor Hospital, Nantong 226631, China
5 Department of Pediatric Surgery, Shanghai Children’s Hospital, Shanghai Jiao Tong University, China
6 Central Laboratory for Medical Research, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
7 Department of Microbiology, Faculty of Basic Medical Sciences, Guilin Medical University, Guilin 541001, China
8 Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China