Mir-455-3p Inhibits Hepatocellular Carcinoma Tumorigenesis, Lymphangiogenesis and Angiogenesis via Regulating Vascular Endothelial Growth Factor C as well as Vascular Endothelial Growth Factor Receptor-2

DOI: https://doi.org/10.21203/rs.3.rs-632152/v1

Abstract

Background: MicroRNAs (miRNAs), functioning as tumor suppressors or oncogenes, exert a considerable regulative influence in biological processes. Previous studies showed that miR-455-3p was significantly down-regulated in hepatocellular carcinoma (HCC), while the specific role of miR-455-3p in HCC is unclear.

Methods: QRT-PCR was used to detect miR-455-3p expression levels in HCC cells and tissues. We adopted colony formation, EdU, CCK-8, transwell assays and wound-healing to identify the influences of miR-455-3p on proliferation, invasion and migration in HHCC and HuH-7 cells. Western blot analysis, angiogenesis assays, dual-luciferase reporter gene assay and bioinformatics analysis were also adopted to investigate the underlying molecular mechanisms.

Results: MiR-455-3p was down-regulated in HCC tissues and cells. In vitro experiments identified miR-455-3p overexpression inhibited HCC cell proliferation, migration and invasion. Interestingly, it was identified that vascular endothelial growth factor C (VEGFC) was the downstream target gene of miR-455-3p. Additionally, overexpression of miR-455-3p in HCC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGFC, vascular endothelial growth factor receptor (VEGFR)-2, and subsequently decreased phosphatidylinositol 3-kinase (AKT) signaling pathway. Furthermore, in vivo restoration of miR-455-3p significantly suppressed tumorigenicity of HuH-7 cells in nude mice and inhibited both angiogenesis and lymphangiogenesis of tumor xenografts.

Conclusions: Our findings suggest that miR-455-3p could play a role in HCC tumorigenesis at least in part by modulation of angiogenesis and lymphangiogenesis through targeting VEGFC, and could simultaneously block AKT signaling pathways.

Background

As the sixth most ubiquitous cancer worldwide, liver cancer is placed the fourth prior reason of all cancer-related deaths, with nearly one million cases reported in 2016[1, 2]. Survival rates for patients with hepatocellular carcinoma are shockingly low, killing more than 700,000 people a year and leaving only 10 per cent alive two years after diagnosis in the UK[3]. According to statistics, 27,000 liver cancer patients died in the United States in 2016, and there's only a 15% 5-year survival rate[4]. Concerning the development of HCC, chronic hepatitis B and C virus infection were placed the most common risk factors[5]. While for most patients there isn’t any curative treatment[6].

MicroRNAs (miRNAs), about 19–24 nt in length, are endogenous non-coding small RNA exerting important influences in animals and plants, which is through targeting the target gene mRNA at its 3-terminal non-coding region thus causing transcriptional repression or regulating mRNA degradation[7, 8]. Numbers of studies have shown that miRNAs exert a considerable regulative influence in the progress of various diseases, especially tumors[9, 10]. Some miRNAs were found to be tumor suppressors in HCC, including miR-17-5p[11], miR-20a[12], miR-202[13], and microRNA-214[14]. In addition, HCC is a typical hypervascular tumor, whose growth and metastasis depend on angiogenesis[15]. However, miRNAs function as key modulators of angiogenesis and lymphangiogenesis[16, 17]. Therefore, we studied the pathogenesis of HCC from the perspective of miRNAs, which is helpful to find targets for early diagnosis and treatment.

Wentao Jiang and his colleagues analyzed the GSE98269 chip and found through further studies that in hepatocellular carcinoma, miR-455-3p was significantly down-regulated and could be used as a biomarker of HCC[18]. Lan et al. also anticipated low level of miR-455-3p with poor prognosis in HCC[19]. And investigations have demonstrated that miR-455-3p can inhibit the progression of a variety of tumors. For example, in prostate cancer microRNA-455-3p targets eIF4E, thus works as a tumor suppressor[20]. By targeting FAM83F, miR-455-3p also inhibits the proliferation and invasion of esophageal squamous cell carcinoma, as a prognostic marker[21]. Via Targeting TAZ, in pancreatic cancer cells miR-455-3p down-regulation correlates with proliferation and drug resistance[22]. However, the pathogenesis of miR-455-3p involvement in HCC remains unclear.

Central regulators of vasculogenesis, angiogenesis and lymphangiogenesis includes VEGFs (VEGFE, VEGFD, VEGFC, VEGFB, VEGFA, and PlGF, or placental growth factor) as well as their endothelial tyrosine kinase receptors[23]. Mature VEGFC binds VEGFR-3 and VEGFR-2 respectively, which is mostly located on lymphatic and vascular endothelial cells, after VEGFC undergoing extensive proteolytic processing[2426]. On tumor cells, including leukemic, skin, and gastric cells, VEGFC and its cognate receptors are expressed, and through autocrine signaling facilitates tumor progression partly[27, 28].

It was identified in this study that in HCC tissues and cells miR-455-3p was down-regulated and miR-455-3p works as a tumor suppressive miRNA by targeting VEGFC. Beside VEGFC and VEGFR-2 expression, ectopic expression of miR-455-3p led to inhibition of the cell invasion, migration and proliferation. Additionally, in HCC xenograft treatment model miR-455-3p overexpression curbed tumor growth, angiogenesis and lymphangiogenesis. In brief, we discovered the function of miR-455-3p in HCC, likely supplying a new sight on therapy for HCC patients.

Methods

Human HCC tissue samples

From August 2016 to September 2018, 40 paired HCC tissues as well as adjoining normal tissues were harvested from diagnosed patients in our hospital. All included patients had been diagnosed, and they didn’t receive treatment of radiotherapy or chemotherapy before. All included subjects inscribed an informed consent and the investigation got approval from Ethics Committee of Affiliated Hospital of Youjiang Medical University for Nationalities. We maintained these tissues in liquid nitrogen. 

Cell culture

Five HCC cell lines (HB611, HHCC, H-97, HuH-7, Li-7), normal liver cell line (LO2) and human umbilical vein endothelial cells (HUVECs) were acquired from Shang Hai Ze Ye Biotechnology Co., Ltd. (Shang Hai, China) and American Type Culture Collection (Manassas, VA, USA). We verified the authenticity of each cell line with short tandem repeat analysis. We preserved all cell lines in Dulbecco's modified Eagle's medium (DMEM; Gibco, USA), with 1% penicillin-streptomycin and 10% fetal bovine sera (FBS; Gibco, USA), and stored them in a 5% CO2 humidified incubator at 37℃. 

QRT-PCR (Quantitative reverse transcriptase-polymerase chain reaction)

For total RNA was extraction we used RNeasy Mini Kit (Qiagen, Germany) and for reversed transcription Quantitect Reverse Transcription Kit (Qiagen, Germany) was used, during which we refered to the manufacturer’s instructions. We performed amplification on a Lightcyle 480 platform (Roche, Germany) for each sample in triplicate. According to GAPDH levels, we normalized relative expression levels. Here followed primer sequences:

MiR-455-3p, 5’-ACACTCCAGCTGGGCACAUAUACGGGUAC-3’ (F), 

5’-CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGCGTCAGGT-3’(R); 

GAPDH, 5’-ATGGGGAAGGTGAAGGT-3’(F), 

5’-AAGCTTCCCGTTCTCAG-3’(R).

Western blot analysis

We prepared Protein lysates with RIPA buffer (Thermo Scientific Inc., Waltham, MA, USA) which contains 1% protease inhibitor and performed western blotting in triplicate experiments. We subjected protein lysates to electrophoresis on a 4% SDS-PAGE (sodium dodecyl sulfate polyacrylamide gel electrophoresis), and electrotransferred proteins to polyvinylidene fluoride membranes (Millipore, USA). With 5% non-fat dry milk in TBS, we incubated membranes and probed them with anti-VEGFC and anti-VEGFR-2 (sc-1881, sc-6251 respectively; Santa Cruz Biotechnology, Santa Cruz, CA, USA), anti-pAKT, anti-AKT and anti-β-actin (#4056, #9272, #4970, respectively; Cell Signaling Technology, USA) in TBST (0.1% Tween 20 in TBS). For detection of immunoreactive proteins, we used Horseradish peroxidase-conjugated anti-rabbit (or mouse) IgG (Cell Signaling Technology, USA) by chemiluminescence (Pierce® ECL kit)

Cell proliferation assay

Via the Cell Counting Kit-8 assay (CCK-8, Dojindo, Japan), we referred to the manufacturer’s protocol and performed cell proliferation assays. We seeded a sum of 1×103 cells per well and cultured them for 1, 2, 3, 4 and 5 days. We also added ten microlitres of the CCK-8 solution to each well, and incubated sample at 37℃ for 4 h. We read absorbance at 450 nm on a microplate reader (MultiSkan Spectrum). 

For EDU assay, we used EDU labeled solution (KeyGen Biotech, Nanjing, China) for cell culture for 2 h. As recommended by the manufacturer we performed EdU staining, after 15 min of cell fixation which was in 4% paraformaldehyde.

For colony-formation assays, with trypsin–collagenase we treated transfected HCC cells for digesting them into a single cell suspension, and we seeded them at a rate of 400 cells/well into 12-well plates before culturing them at 37℃ for 14 days in a 5% CO2 incubator. We then use PBS to wash the cells twice and for 15 min stained them with 2% crystal violet, then at room temperature we dried the plate. We counted the clone number which were formed in the plate. 

We performed all experiments thrice, consequently calculated the average of the results.

Wound-healing assay

We seeded cells in six-well plates and cultured them. After they reached confluence, we scratched confluent monolayer cells and washed them with PBS for three times for clearing suspension cells and cell debris. We added fresh serum-free medium and under normal conditions allowed the cells to close the wound for 24h. At the same position of the wound, with a computer-assisted microscope (Nikon) we took photographs.

Transwell assay

Using a Transwell chamber (8-µm pore size; Corning, USA), we performed cell invasion experiments. In the upper chamber we seeded HCC cells at a density of 105 cells per well, in which we also added DMEM containing 1% FBS. We precoated the upper chamber with Matrigel (Corning, USA, dilution ratio: 1:6) while in the lower chamber there was 600μL DMEM which contains 10% FBS. We fixed cells with 4% methanol, which were migrated or invaded to the lower surface of membrane, after incubation at 37℃ for 24 hours then we strained them with crystal violet. We selected 5 random microscopic fields and counted the number of cells in each field. Each experiment was carried out three times.

Generation of stable cell lines expressing miR-455-3p

From LO2 cell line genomic DNA, a DNA fragment which contains the miR-455-3p was amplified then into the pcDNA-copGFP vector (System Biosciences, USA) we cloned it. Name LV-miR-455-3p was given to the lentivirus vector expressing miR-455-3p. Via Lipofectamine2000 (Invitrogen, CA, USA) Lentiviral packaging plasmids and LV-miR-NC (as a negative control) were co-transfected following the manufacturer’s instruction in 293FT packaging cells. To infect HCC cells and HUVECs, we collected, filtered and used the lentivirus in the supernatant at the time forty-eight hours after transfection. Using qRT-PCR, we obtained stable clones and confirmed the expression of mature miR-455-3p after two weeks of antibiotic selection.

Tumor xenograft treatment model

We got approval of animal experimental procedures from the Institutional Animal Care and Use Committee of Affiliated Hospital of Youjiang Medical University for Nationalities. Of each BALB/c nude mouse (5–6 weeks of age, 20–22 g) we injected a suspension of HuH-7 cells (1×107) into the right flank subcutaneously. From the Laboratory Animal Center (Shanghai, China), we purchased all mice and maintained them under specific pathogen-free conditions in laminar flow cabinets. On day 7 post-implantation, when tumors formed, we randomly divided mice into two treatment groups: one is Lv-miR-455-3p group which was treated with lentivirus encoding miR-455-3p; another is Lv-NC which was treated with negative control lentivirus. After treatment with calipers two major axes, we measured tumor volumes every 7 days, and we calculated with the formula: V = 0.5×L (length) ×W2 (width). With all the experiments approval from Affiliated Hospital of Youjiang Medical University for Nationalities, we cared all animals with humane care referring to the criteria by the National Academy of Sciences and the National Institutes of Health, which outlines in the “Guide for the Care and Use of Laboratory Animals”.

Dual-luciferase reporter gene assay

We cloned the 3’-UTR regions of human VEGFC mRNA, which is downstream of the luciferase reporter gene. We first amplified it by PCR and then transferred it into the pGL3-basic vector (Promega, Madison, WI, USA). The construct as Wt (wild-type) and named it VEGFC Wt. Using PCR and the site-directed mutagenesis kit (Takara), which acted as template, the mutated 3’-UTR was generated. Into the luciferase reporter we inserted the mutated sequence and named it VEGFC Mut. Then the Wt or Mut of VEGFC, firefly luciferase reporter and mock LV-miR-455-3p vector or miR-NC were co-transfected into HHCC and HuH-7 cells. 48 h after, we harvested cells and treated them with the Dual Luciferase Assay kit l.

In vitro angiogenesis assays

By tube formation assay, we determined the in vitro angiogenic activity of HUVECs, for a significant angiogenic property of HUVECs is capillary tube formation on Matrigel. After transfection, we serum-starved HUVECs in endothelial basal medium (EBM; Clonetics, CA, USA) with 0.2% BSA for 24 h by incubation. We harvested HUVECs and in a 12-well plate seeded 8×104 cells, which is coated with Matrigel basement membrane matrix (BD Biosciences, USA), after serum starvation. With a computer-assisted microscope (Nikon), we observed tube formation after 8 h of incubation. We defined tube formation as a tube-like structure with a length four times its width. We selected 10 random microscopic fields and took images of tube morphology at 100 magnification per sample. Using LAS software (Leica), we measured the number of tubes.

Statistical analyses

Using SPSS statistical software (16.0 for Windows), we performed statistical analyses. We displayed experimental data as means ± standard deviation (SD). Through Student’s t-test and one-way analysis of variance (ANOVA), we analysed the differences between groups. We performed all statistical tests two sided. P < 0.05 were considered significant statistically. We performed all experiments three times or more.

Results

In human HCC, miR-455-3p expression is down-regulated 

The GSE98269 chip was analyzed by Wen et al., and the fact that miR-455-3p was remarkably down-regulated in HCC was identified in further studies[18]. Using qRT-PCR, we assessed miR-455-3p level in 40 matched non-tumoral tissues and corresponding HCC specimens for determination whether miR-455-3p involves in the regulation of human HCC progress. Compared with non-tumoral tissues, miR-455-3p was remarkably down-regulated in tumor tissues as shown in Figure. 1A. Additionally, in human HCC cell lines (HB611, HHCC, H-97, HuH-7 and Li-7) and normal liver cell line (LO2), we also examined the miR-455-3p expression. In comparison with the LO2 cell line, miR-455-3p expression was examined remarkably lower in human HCC cell lines (Figure. 1B). 

Overexpression of miR-455-3p suppressed the proliferation, migration and invasion of HCC cells  

In HHCC and HuH-7 cells, we enhanced miR-455-3p expression to probe into the function of miR-455-3p, then observed the effects on the cell invasion, migration and proliferation. We displayed the transfection efficiency in Figure. 2A. Via CCK-8, EdU, and colony-formation assays, we examined cell proliferation. The growth rate of HHCC and HuH-7 cells was identified to remarkably decrease by miR-455-3p overexpression (Figure. 2B-D). We also adopted wound-healing assay for investigating the influence of miR-455-3p on cell migration. In comparison with miR-NC cells, miR-455-3p-overexpressing cells showed remarkably slower migration as shown in Figure. 2E. Besides, the invasiveness of miR-455-3p overexpressed cells was remarkably lower than that of the control cells, by transwell assay (Figure. 2F). 

miR-455-3p directly targets VEGFC gene

Using mirdb and Starbase, we searched candidate target genes of miR-455-3p and identified that VEGFC acted as a possible target of miR-455-3p. We displayed binding sequences of miR-455-3p and VEGFC in Figure. 3A. Then, in HHCC and HuH-7 cells we used dual-luciferase reporter gene assay for verifying the targeted binding relationship (Figure. 3B). The relative fluorescence activity of miR-455-3p mimic was remarkably decreased in VEGFC Wt group. However, co-transfection group with VEGFC Mut did not have this effect. Additionally, in miR-455-3p-overexpressing HCC cells the protein level of VEGFC was significantly inhibited, showed by western blot analysis (Figure. 3C). MiR-455-3p could target VEGFC was indicated by these results. 

Investigations have suggested that VEGFC could perform various biological actions so that facilitate tumor progression, including exerting autocrine stimulation influences on VEGFR-2 expression, vital for tumor angiogenesis and lymphangiogenesis[29, 30]. Thus, whether miR-455-3p mediated VEGFC inhibition could exert influence in expression of VEGFR-2 in HCC was analyzed. Compared with miR-NC cells the protein expressions of VEGFR-2 in miR-455-3p-overexpressing HCC cells were remarkably reduced (Figure. 3C). The phosphatidylinositol 3-kinase-AKT pathway, critical for cellular survival and growth, was activated by VEGFRs both in cancer cells and in endothelial cells. Thus, whether this pathway involves in miR-455-3p-caused growth suppression was furtherly examined. In HCC cells the phosphorylation of AKT was remarkably decreased by ectopic expression of miR-455-3p (Figure. 3D).

MiR-455-3p inhibited tumor growth, angiogenesis and lymphangiogenesis in tumor xenografts

With HuH-7 cells, we performed a xenograft experiment in vivo to probe into whether the expression of miR-455-3p have an effect on tumor growth. We displayed xenograft tumors in Figure. 4A. Mice treated with Lv-miR-455-3p were inhibited compared with that treated with Lv-NC in the tumor volume and weight (Figure. 4B, C). Next, in Lv-miR-455-3p-treated groups, VEGFC protein level were remarkably lower than in Lv-NC groups, which is showed by western blot analysis (Figure. 4D). And in Lv-miR-455-3p-treated groups the Ki-67 level were remarkably lower than that in Lv-NC groups, which is indicated by immunohistochemical staining (Figure. 4E). 

Then, we analyzed angiogenesis and lymphangiogenesis of tumors via immunohistochemical staining with anti-LYVE-1 antibodies and anti-CD34. Compared with Lv-NC, Lv-miR-455-3p treated groups showed remarkable reductions in the lymph and blood vessels, which is identified by quantitative analysis (Figure. 4F). These results indicated that miR-455-3p expression remarkably inhibited the tumor progression in vivo, and miR-455-3p seems to regulate tumorigenesis by inhibiting VEGFC-mediated angiogenesis and lymphangiogenesis.

Overexpression of miR-455-3p inhibited tube formation

For the reason that a significant angiogenic property of HUVECs is capillary tube formation on Matrigel, whether miR-455-3p inducing VEGFC up-regulation affected tube formation was furtherly investigated. After transfection with LV-miR-455-3p or LV-miR-NC for 48 h, we serum-starved HUVECs for 24 h before on a Matrigel-coated 12-well plate serum-starved cells the following day for 8 h, well-organized capillary-like structures was formed by LV-miR-NC-transfected HUVECs as seen in Figure. 5A. However, tube-forming activity was significantly impaired as a result of transfection with miR-455-3p. Our studies illustrated that in angiogenesis of HUVECs, miR-455-3p exerts negative influences. Before we examined whether miR-455-3p affects AKT phosphorylation, it is known that AKT works as a vital downstream pathway of VEGF/VEGFR signals. And in miR-455-3p-overexpressing HUVECs, VEGFC and his receptors (VEGFR-2) were negatively regulated (Figure. 5B). Compared with miR-NC-transfected HUVECs, AKT phosphorylation was significantly reduced in miR-455-3p-transfected HUVECs as displayed in Figure. 5C in miR-455-3p-transfected HUVECs. These results pointed out that an induced of VEGFC protein by miR-455-3p suppresses VEGFR-induced activation of the AKT signaling pathway as evident by reduction in AKT phosphorylation.

As presented in Figure. 6, down-regulated of miR-455-3p promoted the level of VEGFC, VEGFR-2, and AKT phosphorylation. Led to an increase of the cell proliferation, migration, invasion, angiogenesis and lymphangiogenesis.

Discussion

Nowadays, increasing evidences have demonstrated that miRNAs work as regulators of the tumor phenotype via regulating the expression of signaling pathways and critical genes, which involves in tumorigenesis and other malignant processes[31, 32]. It was showed in our study that in HCC cell lines and tissues miR-455-3p expression was remarkably down-regulated. However, only a small amount of patient tissues was included in our study, and it is necessary to conduct further study with larger amount to confirm its clinical importance in HCC.

Lentivirus-mediated miR-455-3p-overexpression HHCC and HuH-7 cells were established to research the functions of miR-455-3p in HCC, then we conducted a series of experiments in vivo and in vitro. It was demonstrated clearly in our results that HCC cell proliferation, colony formation, migration and invasion was remarkably suppressed by miR-455-3p in vitro, as well in nude mice HCC xenograft, tumor growth was suppressed. MiR-455-3p was implied by these results as an inhibitor of HCC tumorigenesis.

Through bioinformatics analysis, western blot assay and dual-luciferase reporter gene assay, our indicated that miR-455-3p could target the 3′UTR of VEGFC. In addition, VEGFC, VEGFR-2 and the phosphorylation of AKT, as well as angiogenesis and lymphangiogenesis were decreased by miR-455-3p overexpression in HCC cells. All data provided a basis for new anti-HCC therapies, among which anti-angiogenesis and anti-lymphangiogenesis approaches are significant.

Conclusion

In conclusion, it was showed that in HCC, miR-455-3p was down-regulated, and it worked as a tumor suppressor, directly targeting VEGFC. MiR-455-3p ectopic expression inhibited tumor angiogenesis, progression and lymphangiogenesis, and could simultaneously inhibit AKT signal pathway. In a word, these conclusions may supply a tactic for targeting the interaction of miR-455-3p/VEGFC, as well as a new treatment for HCC.

Declarations

Ethics approval and consent to participate

This study was conducted according to the World Medical Association Declaration of Helsinki and approved by the Medical Ethic Committee of Affiliated Hospital of Youjiang Medical University for Nationalities. All participants provided written informed consent prior to their inclusion in the study. All animal experiments were approved by the Institutional Animal Care and Use Committee of Affiliated Hospital of Youjiang Medical University for Nationalities.

Consent for publication

Not applicable.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Competing interests

The authors declare that they have no competing interests.

Funding

This work was supported by Key Laboratory of the Ministry of Education Project for Early Prevention and Treatment of Regional High-risk Tumors [grant No. GKE2018-03 and No. GKE2019-09].

Authors’ contributions

Sufang Zhou designed the research and revised the manuscript; Xiaoyun Bin and Jianchu Wang performed the experiments and drafted the manuscript; Libai Lu and  Guanbin Ye collected the data and did the analysis; All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

References

  1. Santopaolo F, Lenci I, Milana M, Manzia TM, Baiocchi L. Liver transplantation for hepatocellular carcinoma: Where do we stand? World J Gastroenterol. 2019;25(21):2591–602.
  2. Anwanwan D, Singh SK, Singh S, Saikam V, Singh R. Challenges in liver cancer and possible treatment approaches. Biochim Biophys Acta Rev Cancer. 2020;1873(1):188314.
  3. Affo S, Yu LX, Schwabe RF. The Role of Cancer-Associated Fibroblasts and Fibrosis in Liver Cancer. Annu Rev Pathol. 2017;12:153–86.
  4. Ryerson AB, Eheman CR, Altekruse SF, Ward JW, Jemal A, Sherman RL, Henley SJ, Holtzman D, Lake A, Noone AM, Anderson RN, Ma J, Ly KN, et al. Annual Report to the Nation on the Status of Cancer, 1975–2012, featuring the increasing incidence of liver cancer. Cancer. 2016; 122(9):1312–1337.
  5. Li CL, Ho MC, Lin YY, Tzeng ST, Chen YJ, Pai HY, Wang YC, Chen CL, Lee YH, Chen DS, Yeh SH, Chen PJ. Cell-free virus-host chimera DNA from Hepatitis B virus integration sites as a circulating biomarker of hepatocellular cancer. Hepatology. 2020.
  6. Gerbes A, Zoulim F, Tilg H, Dufour JF, Bruix J, Paradis V, Salem R, Peck-Radosavljevic M, Galle PR, Greten TF, Nault JC, Avila MA. Gut roundtable meeting paper: selected recent advances in hepatocellular carcinoma. Gut. 2018;67(2):380–8.
  7. Swida-Barteczka A, Szweykowska-Kulinska Z. Micromanagement of Developmental and Stress-Induced Senescence: The Emerging Role of MicroRNAs. Genes (Basel). 2019; 10(3).
  8. Chen X, Xie D, Zhao Q, You ZH. MicroRNAs and complex diseases: from experimental results to computational models. Brief Bioinform. 2019;20(2):515–39.
  9. Norouzi M, Yasamineh S, Montazeri M, Dadashpour M, Sheervalilou R, Abasi M, Pilehvar-Soltanahmadi Y. Recent advances on nanomaterials-based fluorimetric approaches for microRNAs detection. Mater Sci Eng C Mater Biol Appl. 2019;104:110007.
  10. Qadir MI, Faheem A, miRNA. A Diagnostic and Therapeutic Tool for Pancreatic Cancer. Crit Rev Eukaryot Gene Expr. 2017;27(3):197–204.
  11. Liu DL, Lu LL, Dong LL, Liu Y, Bian XY, Lian BF, Xie L, Wen D, Gao DM, Ke AW, Fan J, Wu WZ. miR-17-5p and miR-20a-5p suppress postoperative metastasis of hepatocellular carcinoma via blocking HGF/ERBB3-NF-kappaB positive feedback loop. Theranostics. 2020;10(8):3668–83.
  12. Tipanee J, Di Matteo M, Tulalamba W, Samara-Kuko E, Keirsse J, Van Ginderachter JA, Chuah MK, VandenDriessche T. Validation of miR-20a as a Tumor Suppressor Gene in Liver Carcinoma Using Hepatocyte-Specific Hyperactive piggyBac Transposons. Mol Ther Nucleic Acids. 2020;19:1309–29.
  13. Wang J, Chen J, Sun F, Wang Z, Xu W, Yu Y, Ding F, Shen H. miR-202 functions as a tumor suppressor in hepatocellular carcinoma by targeting HK2. Oncol Lett. 2020;19(3):2265–71.
  14. Long LM, He BF, Huang GQ, Guo YH, Liu YS, Huo JR. microRNA-214 functions as a tumor suppressor in human colon cancer via the suppression of ADP-ribosylation factor-like protein 2. Oncol Lett. 2015;9(2):645–50.
  15. Zhang C, Wang N, Tan HY, Guo W, Chen F, Zhong Z, Man K, Tsao SW, Lao L, Feng Y. Direct inhibition of the TLR4/MyD88 pathway by geniposide suppresses HIF-1alpha-independent VEGF expression and angiogenesis in hepatocellular carcinoma. Br J Pharmacol. 2020.
  16. Bao L, Chau C, Bao J, Tsoukas MM, Chan LS. IL-4 dysregulates microRNAs involved in inflammation, angiogenesis and apoptosis in epidermal keratinocytes. Microbiol Immunol. 2018;62(11):732–6.
  17. Hunter S, Nault B, Ugwuagbo KC, Maiti S, Majumder M. Mir526b and Mir655 Promote Tumour Associated Angiogenesis and Lymphangiogenesis in Breast Cancer. Cancers (Basel). 2019; 11(7).
  18. Jiang W, Zhang L, Guo Q, Wang H, Ma M, Sun J, Chen C. Identification of the Pathogenic Biomarkers for Hepatocellular Carcinoma Based on RNA-seq Analyses. Pathol Oncol Res. 2019;25(3):1207–13.
  19. Lan Y, Han J, Wang Y, Wang J, Yang G, Li K, Song R, Zheng T, Liang Y, Pan S, Liu X, Zhu M, Liu Y, et al. STK17B promotes carcinogenesis and metastasis via AKT/GSK-3beta/Snail signaling in hepatocellular carcinoma. Cell Death Dis. 2018;9(2):236.
  20. Zhao Y, Yan M, Yun Y, Zhang J, Zhang R, Li Y, Wu X, Liu Q, Miao W, Jiang H. MicroRNA-455-3p functions as a tumor suppressor by targeting eIF4E in prostate cancer. Oncol Rep. 2017;37(4):2449–58.
  21. Yang H, Wei YN, Zhou J, Hao TT, Liu XL. MiR-455-3p acts as a prognostic marker and inhibits the proliferation and invasion of esophageal squamous cell carcinoma by targeting FAM83F. Eur Rev Med Pharmacol Sci. 2017;21(14):3200–6.
  22. Zhan T, Huang X, Tian X, Chen X, Ding Y, Luo H, Zhang Y. Downregulation of MicroRNA-455-3p Links to Proliferation and Drug Resistance of Pancreatic Cancer Cells via Targeting TAZ. Mol Ther Nucleic Acids. 2018;10:215–26.
  23. Lohela M, Bry M, Tammela T, Alitalo K. VEGFs and receptors involved in angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol. 2009;21(2):154–65.
  24. Su CM, Tang CH, Chi MJ, Lin CY, Fong YC, Liu YC, Chen WC, Wang SW. Resistin facilitates VEGF-C-associated lymphangiogenesis by inhibiting miR-186 in human chondrosarcoma cells. Biochem Pharmacol. 2018;154:234–42.
  25. Muhammad A, Funmilola A, Aimola IA, Ndams IS, Inuwa MH, Nok AJ. Kolaviron shows anti-proliferative effect and down regulation of vascular endothelial growth factor-C and toll like receptor-2 in Wuchereria bancrofti infected blood lymphocytes. J Infect Public Health. 2017;10(5):661–6.
  26. Zhao B, Smith G, Cai J, Ma A, Boulton M. Vascular endothelial growth factor C promotes survival of retinal vascular endothelial cells via vascular endothelial growth factor receptor-2. Br J Ophthalmol. 2007;91(4):538–45.
  27. Yeh YW, Cheng CC, Yang ST, Tseng CF, Chang TY, Tsai SY, Fu E, Chiang CP, Liao LC, Tsai PW, Yu YL, Su JL. Targeting the VEGF-C/VEGFR3 axis suppresses Slug-mediated cancer metastasis and stemness via inhibition of KRAS/YAP1 signaling. Oncotarget. 2017;8(3):5603–18.
  28. Pepper MS, Mandriota SJ, Jeltsch M, Kumar V, Alitalo K. Vascular endothelial growth factor (VEGF)-C synergizes with basic fibroblast growth factor and VEGF in the induction of angiogenesis in vitro and alters endothelial cell extracellular proteolytic activity. J Cell Physiol. 1998;177(3):439–52.
  29. Feng Y, Hu J, Ma J, Feng K, Zhang X, Yang S, Wang W, Zhang J, Zhang Y. RNAi-mediated silencing of VEGF-C inhibits non-small cell lung cancer progression by simultaneously down-regulating the CXCR4, CCR7, VEGFR-2 and VEGFR-3-dependent axes-induced ERK, p38 and AKT signalling pathways. Eur J Cancer. 2011;47(15):2353–63.
  30. Bower NI, Vogrin AJ, Le Guen L, Chen H, Stacker SA, Achen MG, Hogan BM. Vegfd modulates both angiogenesis and lymphangiogenesis during zebrafish embryonic development. Development. 2017;144(3):507–18.
  31. Yang N, Zhu S, Lv X, Qiao Y, Liu YJ, Chen J. MicroRNAs: Pleiotropic Regulators in the Tumor Microenvironment. Front Immunol. 2018;9:2491.
  32. Vasuri F, Visani M, Acquaviva G, Brand T, Fiorentino M, Pession A, Tallini G, D'Errico A, de Biase D. Role of microRNAs in the main molecular pathways of hepatocellular carcinoma. World J Gastroenterol. 2018;24(25):2647–60.