Hostname: page-component-848d4c4894-75dct Total loading time: 0 Render date: 2024-05-06T03:05:37.553Z Has data issue: false hasContentIssue false

miR-19-3p Targets PTEN to Regulate Cervical Cancer Cell Proliferation, Invasion, and Autophagy

Published online by Cambridge University Press:  01 January 2024

Wei Wang*
Affiliation:
Medical Laboratory, Fuyang City People’s Hospital, Fuyang 236004, Anhui, China
Lu Liu
Affiliation:
College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi 830052, Xinjiang, China
Yongjian Tian
Affiliation:
Medical Laboratory, Fuyang City People’s Hospital, Fuyang 236004, Anhui, China
*
Correspondence should be addressed to Wei Wang; wangwei_fuyang@163.com
Rights & Permissions [Opens in a new window]

Abstract

Background. Cervical cancer is the second most common cancer among women worldwide. Extensive studies have shown that microRNAs (miRNA/miR) can regulate the formation, progression, and metastasis of cancer. The purpose of this study was to investigate the effect of miR-19-3p on the proliferation, invasion, and autophagy of cervical cancer cells and to explore the underlying mechanism. Methods. SiHa and HeLa cells were transfected with miR-19-3p mimic and inhibitor. miR-19-3p and PTEN expression were detected using real-time quantitative PCR and western blot, respectively. The binding between miR-19-3p and PTEN was predicted using Targetscan7.2 and verified by a dual-luciferase reporter gene assay. The effects of miR-19-3p on cell invasion and proliferation were evaluated by Transwell assays and MTT, respectively. The effect of miR-19-3p on autophagy was observed using fluorescence microscopy. Results. The expression of miR-19-3p in cervical cancer tissues and SiHa and HeLa cells was significantly upregulated, whereas the expression of PTEN was significantly downregulated. PTEN was one of the direct targets of miR-19-3p. The miR-19-3p mimic significantly reduced the apoptosis rate and autophagy and promoted cell proliferation and invasion of the SiHa and HeLa cells. Conclusion. In summary, miR-19b-3p can target PTEN to regulate the proliferation, invasion, and autophagy of cervical cancer cells. Our findings indicate the potential of miR-19-3p as a target for cervical cancer treatment in the future.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BY
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.
Copyright
Copyright © 2023 Wei Wang et al.

1. Introduction

Cervical cancer is the second leading cause of cancer-related deaths among women worldwide [Reference Sung, Ferlay and Siegel1, Reference Overmeer2]. Persistent infection with high-risk human papillomavirus (HPV) is considered the main risk factor for cervical cancer [Reference Hausen3]. About 50% of cervical cancers are related to persistent HPV16 infection, and about 20% are related to persistent HPV18 infection [Reference Syrjänen, Mäntyjärvi and Väyrynen4Reference De Vuyst, Clifford, Nascimento, Madeleine and Franceschi6]. However, only ∼15% of women with persistent high-risk HPV infection will develop cervical cancer, which indicates that other factors are involved in the regulation of the occurrence and development of cervical cancer [Reference Crow7]. The HPV vaccine has no protective effect on patients who are already infected with HPV or have cervical lesions. Therefore, understanding the pathogenesis of cervical cancer is important for developing therapeutic targets for this type of cancer.

MicroRNAs (miRNA/miR) are small noncoding single-stranded RNAs (19–25 nucleotides in length) [Reference Bracken, Scott and Goodall8]. miRNA silences its target genes via base-pairing with complementary sequences of 3′-untranslated region (3′-UTR) in the mRNAs of the target gene, thereby inhibiting translation or inducing mRNA degradation [Reference Dhawan, Scott, Harris and Buffa9, Reference Wang, Zhang, Zhang and Xu10]. In this way, miRNA is involved in many biological processes, including cell proliferation, apoptosis, cell differentiation, and development [Reference Rotondo, Mazziotta, Lanzillotti, Tognon and Martini11Reference Li, Wang and Wen15]. Accumulating evidence supports the importance of miRNA in cervical cancer. For example, Wang et al. reported that miRNA signatures can be useful for the screening, diagnosis, and prognosis of cervical cancer [Reference Wang, Zhang, Zhang and Xu10, Reference Nascimento, Gally, Borges, Campos and Kaneto16, Reference Aftab, Poojary and Seshan17]. Hu et al. and Park et al. reported that miR-200a, miR-9, and miR-944 can be used to predict the survival rate of cervical cancer [Reference Hu, Schwarz and Lewis18, Reference Park, Kim and Eom19]. The aberrant expression of miR-466 and miR-34a is closely associated with the occurrence and development of cervical cancer [Reference Sun, Shen, Gong, Zhou, Sheng and Duan20, Reference Liu, Wang and Chen21]. miR-21 can promote the proliferation of HeLa cells by targeting programmed cell death [Reference Yao, Xu, Zhang, Zhou and Qu22]. It has also been shown that miR-361 targets HSP90 to inhibit the invasion and epithelial-mesenchymal transition (EMT) of SiHa and HeLa cells [Reference Xu, Dong and Xiong23]. There is increasing evidence about miRNA as a new biomarker and therapeutic target for cervical cancer. Upregulation and downregulation of miR-19b have been reported in various cancers, including rectal cancer [Reference Su, Zang, Wang and Ding24], breast cancer [Reference Zhou, Zhang, Shi and Fan25], lung cancer [Reference Qiu, Gu, Li, Nie and Yu26], and pancreatic cancer [Reference Song, Sun, Xia, Chen and Yang27], and miR-19b has been identified as a key regulatory molecule in the mechanism of cancer development. However, the role of miR-19b-3p in cervical cancer remains unknown.

In this study, SiHa and HeLa cells were transfected with miR-19-3p mimics and inhibitors to detect cell proliferation, invasion, and autophagy. The potential target of miR-19-3p was determined using the bioinformatics analysis tool TargetScan7.2 and verified using a dual-luciferase reporter assay. This study will clarify the molecular mechanism of miR-19b-3p in regulating the occurrence and development of cervical cancer. We present the following article in accordance with the MDAR reporting checklist.

2. Materials and Methods

2.1. Tissue Samples

In total, 20 pairs of cervical cancer tissues (tumour) and adjacent tissue (normal) samples were obtained from HPV16/HPV18-positive cervical cancer patients (age: 53.20 ± 10.21 years) who were admitted to Fuyang City People’s Hospital from July 2019 to June 2020. For control, normal cervical exfoliated cells were collected from 10 healthy subjects who were HPV negative and without cervical lesions. The study was approved by the Institutional Ethics Board of Fuyang City People’s Hospital (no. (2020)0026), and informed consent was obtained from all individual participants.

2.2. Cell Culture and Transfection

Human cervical cancer cell lines SiHa and HeLa were cultured with DMEM medium (11885092, Gibco, USA) containing 10% fetal bovine serum (FBS) (CCS30013.01HI, MRC, Australia), 1% penicillin/streptomycin (15140122, Gibco, USA), and 2 mM glutamine. The cells were then cultured in a 5% CO2 incubator at 37°C and were passaged when confluence reached 90%.

Next, the cells were transfected with miR-19-3p mimic (mimic), miR-19-3p mimic negative control (mimic-NC), miR-19-3p inhibitor (inhibitor), and miR-inhibitor negative control (inhibitor-NC), which were all obtained from Sangon Biotech (Shanghai) Co., Ltd., using Lipofectamine 2000 (11668019, Invitrogen, USA).

2.3. Cell Proliferation Assay

The cells were seeded in a 96-well plate (4 × 104 cell/well) and cultured at 37°C with 5% CO2. At 24, 48, and 72 h of culture, 10 μl of MTT solution (5 mg/ml, Sigma, USA) was added to each well and incubated at 37°C for 4 h. Dimethyl sulfoxide (DMSO) (150 μl/well) was added to dissolve the formazan crystals at 37°C for 30 min. Finally, a Multiskan™ FC Microplate Photometer (Multiskan FC, Thermo Fisher, USA) was used to measure the absorbance at 450 nm.

2.4. Transwell Invasion Assay

The serum-free cell suspension (5 × 105cells/ml; 200 μl) was added to the Transwell upper chamber, and 500 μl of DMEM medium containing 10% FBS was added to the lower chamber (Corning, USA). The Transwell chambers were cultured at 37°C and 5% CO2 for 24 h. Then, the cells were fixed with 4% paraformaldehyde for 15 min and stained with 0.1% crystal violet. Next, the cells were observed under an inverted fluorescence microscope (Axio Observer, Zeiss, Germany), and the migrated cells were counted.

2.5. Real-Time Quantitative PCR (RT-qPCR)

Total RNAs were extracted using TRIzol (10296010, Invitrogen, USA). The reverse transcription of RNA into cDNA was performed using a PrimeScript™ RT reagent kit (RR037A, Takara, Japan). We used a Mir-X miRNA first-strand synthesis kit (638315, Takara, Japan) to detect the expression of miR-19-3p, and U6 was the internal reference gene. Using the TB Green® Premix Ex Taq™ II kit (RR820 B, Takara, Japan), we detected the expression of MUC4 mRNA, with GAPDH being the internal reference gene. The primers were purchased from Sangon Biotech (Shanghai) Co., Ltd (Table 1). The PCR procedure was 95°C for 5 min followed by 40 cycles at 95°C for 15 s and 60°C for 30 s. The reaction was performed using a fluorescence quantitative PCR instrument (AFD4800, AGS, Hangzhou, China). The relative expression levels were calculated using the 2−ΔΔCt method [Reference Schmittgen, Lee and Jiang28].

TABLE 1: Primer sequences for real-time quantitative PCR.

2.6. Western Blot Analysis

RIPA Lysis Buffer (BL504A, Biosharp) was used to extract the total protein from the cervical cancer tissues and cells, and the protein concentration was determined by BCA (BL521A, Biosharp). The protein sample (40 μg) was subjected to 10% SDS-PAGE. The protein was then transferred to the PVDF membranes. After blocking with 5% BSA (4240GR025, Biofroxx) for 2 h at room temperature, the membranes were incubated with anti-PTEN (1 : 1000, ab32199, Abcam) and anti-GAPDH (1 : 1000. ab9485, Abcam) antibodies at 4°C overnight. After washing 3 times with TBST for 5 min each time, incubation with HRP-labelled secondary antibody (1 : 2000 dilution, ZB-2301, ASGB-BIO, China) was performed for 1 h at 37°C. Enhanced chemiluminescence (PE0010, Solarbio) was used for colour development. The protein bands were then analysed in Image J (V1.52s, Bharti Airtel Ltd.).

2.7. Acridine Orange and Propidium Iodide Staining

After staining with acridine orange (AO), red and yellow acid bodies were observed under a fluorescence microscope. SiHa and HeLa cells were collected and stained with AO and propidium iodide (PI) (5 μg/ml). An inverted fluorescence microscope was used to observe and analyse autophagy through the FITC/TRITC channel under 20x and 40x objective lenses [Reference Wu, Liu, Liu and Wu29, Reference Plemel, Caprariello and Keough30].

2.8. Dual-Luciferase Reporter Gene Assay

The target genes of miR-19-3p were predicted using TargetScan7.2 (https://www.targetscan.org/vert_71/). The 3′-UTR of PTEN mRNA containing the putative miR-19-3p binding site was amplified by PCR, and the resulting products were cloned into the pmirGLO dual-luciferase vector (E1330, Promega, USA). Site-directed mutagenesis on the predicted target site of miR-19-3p in wild-type (WT) PTEN 3′-UTR was conducted using a QuickChange II site-directed mutagenesis kit (200521, Agilent Technologies, Inc., Santa Clara, CA, USA). A PTEN 3′-UTR mutation (MUT-PTEN 3′-UTR) reporter gene plasmid was constructed. A Lipofectamine 2000 kit (Invitrogen, 11668019, USA) was used to cotransfect the reporter gene plasmid and miR-19-3p mimics or miR-19-3p inhibitor into cells. At 48 h after transfection, luciferase activity was determined using a dual-luciferase reporter assay system (E2920, Promega, USA) according to the manufacturer’s instructions.

2.9. Statistical Analysis

SPSS 18.0 (IBM Corp., Armonk, NY, USA) was used for the statistical analysis. The data were expressed as the mean ± standard deviation (mean ± SD). A two-tailed t-test was used to compare the differences between the two groups. A one-way ANOVA was used to compare differences among multiple groups. The correlation between two variables was analysed using Pearson correlation. P < 0.05 was considered to be significant [Reference Mishra, Singh, Pandey, Mishra and Pandey31].

3. Results

3.1. Upregulation of miR-19-3p and downregulation of PTEN in Cervical Cancer

The relative expressions of miR-19-3p and PTEN mRNA were detected using RT-qPCR. Our results showed that the relative expression of miR-19-3p in the tumour samples was significantly higher than in the normal samples (P < 0.01) (Figure 1(a)), while the relative expression of PTEN mRNA in the tumour samples was significantly lower than in the normal samples (P < 0.01) (Figure 1(b)). For the expression of miR-19-3p and PTEN in SiHA, HeLa, and NC cells, the results showed that the relative expression of miR-19-3p in SiHa and HeLa cells was significantly higher than in the NC cells (P < 0.01) (Figure 1(c)). However, the relative expression of PTEN mRNA in the SiHa and HeLa cells was significantly lower than that in the NC cells (P < 0.01) (Figure 1(d)). The expression of PTEN protein in tumour and normal samples was detected by western blot. The results showed that the expression of PTEN protein in tumour samples was significantly lower than in normal samples (P < 0.01) (Figures 1(e) and 1(f)). These results suggest that miR-19-3p is upregulated and PTEN is downregulated in cervical cancer.

FIGURE 1: Expression of miR-19-3p and PTEN in cervical cancer tissues and cells. (a) Relative expression of miR-19-3p in tumour tissues (tumour) and adjacent tissues (normal). (b) Relative expression of PTEN in the tumour and normal tissues. (c) Relative expression of miR-19-3p in SiHa, HeLa, and normal cervical exfoliated cells (NC). (d) Relative expression of PTEN mRNA in SiHa, HeLa, and NC. (e, f) Expression of PTEN protein in the tumour and normal tissues. *P < 0.05, **P < 0.01.

3.2. The Proliferation and Invasion of Cervical Cancer Cells are Regulated by miR-19-3p

The mimics and inhibitors were transfected into SiHa and HeLa cells. The relative expression of miR-19-3p was detected using RT-qPCR. Cell proliferation was detected with an MTT assay, and cell invasion was detected with a Transwell assay. The results showed that the miR-19-3p expression, cell proliferation, and invasion after transfection showed the same trend in the SiHa and HeLa cells. The relative expression of miR-19-3p in the mimic group was significantly higher than that of the mimic-NC group (P < 0.01), while the relative expression of miR-19-3p in the inhibitor group was significantly lower than in the inhibitor-NC group (P < 0.01) (Figures 2(a) and 2(b)). The proliferation of SiHa and Hela was significantly upregulated at 48 h after mimic transfection (P < 0.01) but was significantly downregulated at 48 h and 72 h after transfection with inhibitor (P < 0.01) (Figures 2(c) and 2(d)). The invasion of the SiHa and HeLa cells was significantly inhibited at 48 h after transfection with the inhibitor (P < 0.05) (Figures 2(e) and 2(f)). These results suggest that miR-19-3p regulates the proliferation and invasion of cervical cancer cells.

FIGURE 2: Proliferation and invasion of SiHa and HeLa cells after miR-19-3p mimic and inhibitor transfection. (a, b) Relative expression of miR-19-3p after miR-19-3p mimic and inhibitor transfection in SiHa and HeLa cells. (c, d) Cell proliferation of SiHa and HeLa cells after transfection. (e, f) Cell invasion of SiHa and HeLa after transfection. *P < 0.05, **P < 0.01.

3.3. Cervical Cancer Cell Autophagy is Regulated by miR-19-3p

The degree of autophagy was observed using fluorescence microscopy after miR-19-3p mimic transfection. The results showed that SiHa and HeLa in the mimic, mimic-NC, and inhibitor-NC groups mainly exhibited green fluorescence, and there was little red fluorescence in the cytoplasm and nucleus, suggesting that there was virtually no autophagy. The cells in the inhibitor group showed obvious red fluorescence, indicating that a large number of acidic autophagic lysosomal vacuoles had formed and that early apoptosis was induced (Figure 3).

FIGURE 3: The effect of miR-19-3p on the autophagy of SiHa and HeLa. The intensity of red fluorescence reflects the degree of autophagy.

3.4. miR-19b-3p Targets PTEN

TargetScan7.2 was used to predict the target genes of miR-19b-3p. PTEN was identified as a potential target of miR-19-3p. The putative targeting sites of miR-19-3p in PTEN 3′-UTR are shown in Figure 4(a). The dual-luciferase reporter gene assay showed that the miR-19-3p mimic attenuated the luciferase activity of WT-PTEN 3′-UTR in SiHa and HeLa but had no significant effect on MUT-PTEN 3′-UTR (P < 0.01) (Figures 4(b) and 4(c)). Western blot was used to detect the regulation of PTEN by miR-19-3p mimics in SiHa and HeLa. The results showed that the miR-19-3p mimic significantly downregulated PTEN protein expression in SiHa and HeLa (P < 0.01) (Figures 4(d) and 4(e)). These results suggest that PTEN is the direct target of miR-19-3p.

FIGURE 4: PTEN is the direct target of miR-19-3p. (a) The target site of miR-19-3p in PTEN 3′-UTR. The box indicates the mutation site; (b, c) luciferase reporter analysis of cells transfected with the WT or MUT-PTEN 3′-UTR reporter vector; (d, e) western blot was used to detect the expression of PTEN protein in SiHa and HeLa after miR-19-3p mimic transfection. *P < 0.05, **P < 0.01.

4. Discussion

There has been increasing evidence that dysregulated miRNAs are involved in the regulation of a variety of physiological processes in tumours, including cell proliferation, drug resistance, apoptosis, metastasis, and angiogenesis [Reference Wang, Zhang, Zhang and Xu10, Reference Ohtsuka, Ling, Doki, Mori and Calin32, Reference Zhang, Chen and Khan33]. Research on miRNAs focuses on cancer-specific up- or downregulated miRNAs and their target genes in order to clarify the pathogenesis of cancer [Reference Guzman, Agarwal and Asthagiri34, Reference Kim, Won and Ban35]. Numerous reports have shown that miR-19 positively regulates tumorigenesis, EMT, cancer cell proliferation and invasion, and metastasis by targeting different targets in rectal cancer [Reference Su, Zang, Wang and Ding24], breast cancer [Reference Zhou, Zhang, Shi and Fan25], pancreatic cancer [Reference Qiu, Gu, Li, Nie and Yu26], osteosarcoma [Reference Xie, Liu and Wu36], and lung cancer [Reference Song, Sun, Xia, Chen and Yang27, Reference Peng, Guan and Gao37], indicating that miR-19 plays a carcinogenic role in cancer development. Our research confirmed that miR-19-3p is upregulated in cervical cancer and that miR-19-3p upregulation in vitro promotes the proliferation and invasion of SiHa and HeLa cells. These results indicate that miR-19-3p exerts a carcinogenic effect in cervical cancer. The target of miR-19-3p was further predicted and analysed to determine the potential mechanism of miR-19-3p in the occurrence and development of cervical cancer.

The PI3K/AKT pathway plays a vital role in cancer development. As a tumour suppressor gene, PTEN is a key negative regulator of the PI3K/AKT pathway, which is involved in regulating cell biological processes such as cell proliferation, invasion, and cycle arrest [Reference Pan, Li and Jiang38, Reference Pérez-Ramírez, Cañadas-Garre, Molina, Faus-Dáder and Calleja-Hernández39]. Downregulation of PTEN expression in a variety of cancers has been reported [Reference Li, Shen and Wang40Reference Huse, Brennan and Hambardzumyan42]. PTEN is the downstream target of many miRNAs. For example, Chai et al. predicted that the potential target of miR-498 is PTEN and identified that miR-498 is overexpressed in triple-negative breast cancer cells and downregulated PTEN by directly binding to the 3′-UTR of PTEN mRNA [Reference Chai, Wu, Wang, Eisenstat and Leng43]. Wang et al. reported that miR-301 regulates cell proliferation and invasion by regulating the expression of PTEN in oesophageal carcinoma [Reference Wang, Hua and Wang44]. Cao et al. reported that inhibition of miR-144-3p expression can upregulate PTEN expression and affect cell growth and metastasis [Reference Cao, Gu, Sun and Chen45]. In this study, it was predicted that using TargetScan7.2 that PTEN is a target gene of miR-19-3p, and a dual-luciferase reporter gene assay further confirmed that miR-19-3p could bind specifically to the 3′-UTR of PTEN. We further found that the miR-19-3p mimics upregulated miR-19-3p in SiHa and HeLa cells and inhibited the expression of PTEN protein. However, the miR-19-3p inhibitor reversed the inhibitory effect on PTEN. These results indicate that miR-19-3p can negatively regulate the expression of the target gene PTEN.

In summary, miR-19-3p can regulate the biological functions of cervical cancer cells by targeting PTEN. miR-19-3p is expected to become a potential therapeutic target for cervical cancer.

5. Conclusions

miR-19-3p downregulates the expression of PTEN by directly targeting the 3′-UTR of PTEN mRNA, thereby regulating the biological behaviours of cervical cancer cells, such as cell proliferation, invasion, and autophagy. This research provides new insights into the diagnosis and treatment strategies for cervical cancer.

Data Availability

The data used to support the study are available from the corresponding author upon request.

Ethical Approval

All works were conducted in accordance with the Declaration of Helsinki (1964). The study was approved by the Institutional Ethics Board of Fuyang City People’s Hospital (no.[2020]0026).

Consent

Informed consent was obtained from all individual participants.

Disclosure

The authors have completed the MDAR reporting checklist. Liu Lu is co-first author.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

Wei Wang and Lu Liu equally contributed to this study.

References

Sung, H., Ferlay, J., Siegel, R. L. et al., “Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries,CA: A Cancer Journal for Clinicians, vol. 71, no. 3, pp. 209249, 2021.Google ScholarPubMed
Overmeer, R. M., “Altered DNA Methylation during HPV-Induced Cervical Carcinogenesis: Basic Aspects and Diagnostic Implications,” PhD-Thesis - Research and graduation internal, Vrije Universiteit Amsterdam, Amsterdam, Netherlands, 2011.Google Scholar
Hausen, H. Z., “Papillomavirus infections--a major cause of human cancers,Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, vol. 1288, no. 2, pp. F55F78, 1996.10.1016/0304-419X(96)00020-0CrossRefGoogle ScholarPubMed
Syrjänen, K., Mäntyjärvi, R., Väyrynen, M. et al., “Human papillomavirus (HPV) infections involved in the neoplastic process of the uterine cervix as established by prospective follow-up of 513 women for two years,European Journal of Gynaecological Oncology, vol. 8, no. 1, pp. 516, 1987.Google ScholarPubMed
Walboomers, J. M. M., Jacobs, M. V., Manos, M. M. et al., “Human papillomavirus is a necessary cause of invasive cervical cancer worldwide,The Journal of Pathology, vol. 189, Article ID 199909, pp. 1219, 1999.10.1002/(SICI)1096-9896(199909)189:1<12::AID-PATH431>3.0.CO;2-F3.0.CO;2-F>CrossRefGoogle ScholarPubMed
De Vuyst, H., Clifford, G. M., Nascimento, M. C., Madeleine, M. M., and Franceschi, S., “Prevalence and type distribution of human papillomavirus in carcinoma and intraepithelial neoplasia of the vulva, vagina and anus: a meta-analysis,International Journal of Cancer, vol. 124, no. 7, pp. 16261636, 2009 Apr 1.10.1002/ijc.24116CrossRefGoogle ScholarPubMed
Crow, J. M., “HPV: the global burden,Nature, vol. 488, no. 7413, pp. S2S3, 2012 Aug 30.10.1038/488S2aCrossRefGoogle ScholarPubMed
Bracken, C. P., Scott, H. S., and Goodall, G. J., “A network-biology perspective of microRNA function and dysfunction in cancer,Nature Reviews Genetics, vol. 17, no. 12, pp. 719732, 2016.10.1038/nrg.2016.134CrossRefGoogle ScholarPubMed
Dhawan, A., Scott, J. G., Harris, A. L., and Buffa, F. M., “Pan-cancer characterisation of microRNA across cancer hallmarks reveals microRNA-mediated downregulation of tumour suppressors,Nature Communications, vol. 9, no. 1, p. 5228, 2018.10.1038/s41467-018-07657-1CrossRefGoogle ScholarPubMed
Wang, P., Zhang, L., Zhang, J., and Xu, G., “[Retracted] MicroRNA‑124‑3p inhibits cell growth and metastasis in cervical cancer by targeting IGF2BP1,Experimental and Therapeutic Medicine, vol. 15, no. 2, pp. 13851393, 2018.Google ScholarPubMed
Rotondo, J. C., Mazziotta, C., Lanzillotti, C., Tognon, M., and Martini, F., “Epigenetic dysregulations in merkel cell polyomavirus-driven merkel cell carcinoma,International Journal of Molecular Sciences, vol. 22, no. 21, Article ID 11464, 2021.10.3390/ijms222111464CrossRefGoogle ScholarPubMed
Wang, Y. and Lin, C., “Exosomes miR-22-3p derived from mesenchymal stem cells suppress colorectal cancer cell proliferation and invasion by regulating RAP2B and PI3K/AKT pathway,JAMA Oncology, vol. 2021, Article ID 3874478, 10 pages, 2021.Google ScholarPubMed
Wang, R., Sun, Y., Yu, W. et al., “Downregulation of miRNA-214 in cancer-associated fibroblasts contributes to migration and invasion of gastric cancer cells through targeting FGF9 and inducing EMT,Journal of Experimental & Clinical Cancer Research, vol. 38, no. 1, p. 20, 2019.10.1186/s13046-018-0995-9CrossRefGoogle ScholarPubMed
Li, T. K., Yin, K., Chen, Z., Bao, Y., and Zhang, S. X., “MiR-214 regulates oral cancer KB cell apoptosis through targeting RASSF5,Genetics and Molecular Research, vol. 16, no. 1, p. 16, 2017.10.4238/gmr16019327CrossRefGoogle ScholarPubMed
Li, J., Wang, Q., Wen, R. et al., “MiR-138 inhibits cell proliferation and reverses epithelial-mesenchymal transition in non-small cell lung cancer cells by targeting GIT1 and SEMA4C,Journal of Cellular and Molecular Medicine, vol. 19, no. 12, pp. 27932805, 2015.10.1111/jcmm.12666CrossRefGoogle ScholarPubMed
Nascimento, N. P. G., Gally, T. B., Borges, G. F., Campos, L. C. G., and Kaneto, C. M., “Systematic review of circulating MICRORNAS as biomarkers of cervical carcinogenesis,BMC Cancer, vol. 22, no. 1, p. 862, 2022.10.1186/s12885-022-09936-zCrossRefGoogle ScholarPubMed
Aftab, M., Poojary, S. S., Seshan, V. et al., “Urine miRNA signature as a potential non-invasive diagnostic and prognostic biomarker in cervical cancer,Scientific Reports, vol. 11, no. 1, Article ID 10323, 2021 May 14.10.1038/s41598-021-89388-wCrossRefGoogle ScholarPubMed
Hu, X., Schwarz, J. K., Lewis, J. S. et al., “A microRNA expression signature for cervical cancer prognosis,Cancer Research, vol. 70, no. 4, pp. 14411448, 2010.10.1158/0008-5472.CAN-09-3289CrossRefGoogle ScholarPubMed
Park, S., Kim, J., Eom, K. et al., “microRNA-944 overexpression is a biomarker for poor prognosis of advanced cervical cancer,BMC Cancer, vol. 19, no. 1, p. 419, 2019.10.1186/s12885-019-5620-6CrossRefGoogle ScholarPubMed
Sun, P., Shen, Y., Gong, J. M., Zhou, L. L., Sheng, J. H., and Duan, F. J., “A new MicroRNA expression signature for cervical cancer,International Journal of Gynecological Cancer, vol. 27, no. 2, pp. 339343, 2017 Feb.10.1097/IGC.0000000000000863CrossRefGoogle ScholarPubMed
Liu, M., Wang, W., Chen, H. et al., “miR-9, miR-21, miR-27b, and miR-34a Expression in HPV16/58/52-Infected Cervical Cancer,BioMed Research International, vol. 2020, Article ID 2474235, 2020.Google Scholar
Yao, Q., Xu, H., Zhang, Q. Q., Zhou, H., and Qu, L. H., “MicroRNA-21 promotes cell proliferation and down-regulates the expression of programmed cell death 4 (PDCD4) in HeLa cervical carcinoma cells,Biochemical and Biophysical Research Communications, vol. 388, no. 3, pp. 539542, 2009.10.1016/j.bbrc.2009.08.044CrossRefGoogle ScholarPubMed
Xu, D., Dong, P., Xiong, Y. et al., “MicroRNA-361-Mediated inhibition of HSP90 expression and EMT in cervical cancer is counteracted by oncogenic lncRNA NEAT1,Cells, vol. 9, no. 3, p. 632, 2020 Mar 5.10.3390/cells9030632CrossRefGoogle ScholarPubMed
Su, Y. F., Zang, Y. F., Wang, Y. H., and Ding, Y. L., “MiR-19-3p induces tumor cell apoptosis via targeting FAS in rectal cancer cells,Technology in Cancer Research and Treatment, vol. 19, Article ID 1533033820917978, 2020.10.1177/1533033820917978CrossRefGoogle ScholarPubMed
Zhou, J., Zhang, X., Shi, H., and Fan, C., “MiR-19 regulates breast cancer cell aggressiveness by targeting profilin 1,FEBS Letters, vol. 591, no. 11, p. 1623, 2017.10.1002/1873-3468.12657CrossRefGoogle ScholarPubMed
Qiu, F., Gu, W. G., Li, C., Nie, S. L., and Yu, F., “Analysis on expression level and diagnostic value of miR-19 and miR-21 in peripheral blood of patients with undifferentiated lung cancer,European Review for Medical and Pharmacological Sciences, vol. 22, no. 23, pp. 83678373, 2018.Google ScholarPubMed
Song, M., Sun, M., Xia, L., Chen, W., and Yang, C., “miR-19b-3p promotes human pancreatic cancer Capan-2 cells proliferation by targeting phosphatase and tension homolog,Annals of Translational Medicine, vol. 7, no. 11, p. 236, 2019.10.21037/atm.2019.04.61CrossRefGoogle ScholarPubMed
Schmittgen, T. D., Lee, E. J., Jiang, J. et al., “Real-time PCR quantification of precursor and mature microRNA,Methods, vol. 44, no. 1, pp. 3138, 2008.10.1016/j.ymeth.2007.09.006CrossRefGoogle ScholarPubMed
Wu, X., Liu, P. C., Liu, R., and Wu, X., “Dual AO/EB staining to detect apoptosis in osteosarcoma cells compared with flow cytometry,Med Sci Monit Basic Res, vol. 21, pp. 1520, 2015.Google Scholar
Plemel, J. R., Caprariello, A. V., Keough, M. B. et al., “Unique spectral signatures of the nucleic acid dye acridine orange can distinguish cell death by apoptosis and necroptosis,Journal of Cell Biology, vol. 216, no. 4, pp. 11631181, 2017.10.1083/jcb.201602028CrossRefGoogle ScholarPubMed
Mishra, P., Singh, U., Pandey, C. M., Mishra, P., and Pandey, G., “Application of student’s t-test, analysis of variance, and covariance,Annals of Cardiac Anaesthesia, vol. 22, no. 4, pp. 407411, 2019.10.4103/aca.ACA_94_19CrossRefGoogle ScholarPubMed
Ohtsuka, M., Ling, H., Doki, Y., Mori, M., and Calin, G. A., “MicroRNA processing and human cancer,Journal of Clinical Medicine, vol. 4, no. 8, pp. 16511667, 2015.10.3390/jcm4081651CrossRefGoogle ScholarPubMed
Zhang, G., Chen, L., Khan, A. A. et al., “miRNA-124-3p/neuropilin-1(NRP-1) axis plays an important role in mediating glioblastoma growth and angiogenesis,International Journal of Cancer, vol. 143, no. 3, pp. 635644, 2018.10.1002/ijc.31329CrossRefGoogle ScholarPubMed
Guzman, N., Agarwal, K., Asthagiri, D. et al., “Breast cancer-specific miR signature unique to extracellular vesicles includes “microRNA-like” tRNA fragments,Molecular Cancer Research, vol. 13, no. 5, pp. 891901, 2015.10.1158/1541-7786.MCR-14-0533CrossRefGoogle ScholarPubMed
Kim, J., Won, R., Ban, G. et al., “Targeted regression of hepatocellular carcinoma by cancer-specific RNA replacement through MicroRNA regulation,Scientific Reports, vol. 5, Article ID 12315, 2015.Google ScholarPubMed
Xie, C., Liu, S., Wu, B. et al., “miR-19 promotes cell proliferation, invasion, migration, and EMT by inhibiting SPRED2-mediated autophagy in osteosarcoma cells,Cell Transplantation, vol. 29, Article ID 963689720962460, 2020.10.1177/0963689720962460CrossRefGoogle ScholarPubMed
Peng, X., Guan, L., and Gao, B., “miRNA-19 promotes non-small-cell lung cancer cell proliferation via inhibiting CBX7 expression,OncoTargets and Therapy, vol. 11, pp. 88658874, 2018.10.2147/OTT.S181433CrossRefGoogle ScholarPubMed
Pan, H., Li, T., Jiang, Y. et al., “Overexpression of circular RNA ciRS-7 abrogates the tumor suppressive effect of miR-7 on gastric cancer via PTEN/PI3K/AKT signaling pathway,Journal of Cellular Biochemistry, vol. 119, no. 1, pp. 440446, 2018.10.1002/jcb.26201CrossRefGoogle ScholarPubMed
Pérez-Ramírez, C., Cañadas-Garre, M., Molina, M. Á., Faus-Dáder, M. J., and Calleja-Hernández, M. Á., “PTEN and PI3K/AKT in non-small-cell lung cancer,Pharmacogenomics, vol. 16, no. 16, pp. 18431862, 2015.10.2217/pgs.15.122CrossRefGoogle ScholarPubMed
Li, S., Shen, Y., Wang, M. et al., “Loss of PTEN expression in breast cancer: association with clinicopathological characteristics and prognosis,Oncotarget, vol. 8, no. 19, pp. 3204332054, 2017.10.18632/oncotarget.16761CrossRefGoogle ScholarPubMed
Garofalo, M., Di Leva, G., Romano, G. et al., “miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation,Cancer Cell, vol. 16, no. 6, pp. 498509, 2009.10.1016/j.ccr.2009.10.014CrossRefGoogle ScholarPubMed
Huse, J. T., Brennan, C., Hambardzumyan, D. et al., “The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliomagenesis in vivo,Genes & Development, vol. 23, no. 11, pp. 13271337, 2009.10.1101/gad.1777409CrossRefGoogle ScholarPubMed
Chai, C., Wu, H., Wang, B., Eisenstat, D. D., and Leng, R. P., “MicroRNA-498 promotes proliferation and migration by targeting the tumor suppressor PTEN in breast cancer cells,Carcinogenesis, vol. 39, no. 9, pp. 11851196, 2018.10.1093/carcin/bgy092CrossRefGoogle ScholarPubMed
Wang, B., Hua, P., Wang, R. et al., “Inhibited MicroRNA-301 restrains angiogenesis and cell growth in esophageal squamous cell carcinoma by elevating PTEN,Nanoscale Research Letters, vol. 16, no. 1, p. 3, 2021.10.1186/s11671-020-03452-4CrossRefGoogle ScholarPubMed
Cao, H. L., Gu, M. Q., Sun, Z., and Chen, Z. J., “<p>miR-144-3p Contributes to the Development of Thyroid Tumors through the PTEN/PI3K/AKT Pathway</p&gt,Cancer Management and Research, vol. 12, pp. 98459855, 2020.10.2147/CMAR.S265196CrossRefGoogle Scholar
Figure 0

TABLE 1: Primer sequences for real-time quantitative PCR.

Figure 1

FIGURE 1: Expression of miR-19-3p and PTEN in cervical cancer tissues and cells. (a) Relative expression of miR-19-3p in tumour tissues (tumour) and adjacent tissues (normal). (b) Relative expression of PTEN in the tumour and normal tissues. (c) Relative expression of miR-19-3p in SiHa, HeLa, and normal cervical exfoliated cells (NC). (d) Relative expression of PTEN mRNA in SiHa, HeLa, and NC. (e, f) Expression of PTEN protein in the tumour and normal tissues. *P < 0.05, **P < 0.01.

Figure 2

FIGURE 2: Proliferation and invasion of SiHa and HeLa cells after miR-19-3p mimic and inhibitor transfection. (a, b) Relative expression of miR-19-3p after miR-19-3p mimic and inhibitor transfection in SiHa and HeLa cells. (c, d) Cell proliferation of SiHa and HeLa cells after transfection. (e, f) Cell invasion of SiHa and HeLa after transfection. *P < 0.05, **P < 0.01.

Figure 3

FIGURE 3: The effect of miR-19-3p on the autophagy of SiHa and HeLa. The intensity of red fluorescence reflects the degree of autophagy.

Figure 4

FIGURE 4: PTEN is the direct target of miR-19-3p. (a) The target site of miR-19-3p in PTEN 3′-UTR. The box indicates the mutation site; (b, c) luciferase reporter analysis of cells transfected with the WT or MUT-PTEN 3′-UTR reporter vector; (d, e) western blot was used to detect the expression of PTEN protein in SiHa and HeLa after miR-19-3p mimic transfection. *P < 0.05, **P < 0.01.