Open Access

DDX46 silencing inhibits cell proliferation by activating apoptosis and autophagy in cutaneous squamous cell carcinoma

  • Authors:
    • Quan Lin
    • Hong-Juan Jin
    • Duo Zhang
    • Ling Gao
  • View Affiliations

  • Published online on: September 14, 2020     https://doi.org/10.3892/mmr.2020.11509
  • Pages: 4236-4242
  • Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DEAD‑Box Helicase 46 (DDX46) is an ATP‑dependent RNA helicase that plays a central role in transcription splicing and ribosome assembly. However, the role of DDX46 in cutaneous squamous cell carcinoma (CSCC) remains to be elucidated. The aim of the present study was to investigate the role of DDX46 in CSCC by assessing DDX46 expression levels in CSCC tissues and cell lines. The effect of DDX46 silencing on CSCC cell proliferation, apoptosis and autophagy were also analyzed. It was demonstrated that DDX46 was significantly overexpressed in CSCC tissues and cells (P<0.05). Furthermore, it was found that DDX46 silencing could dramatically inhibit cell proliferation (P<0.05). Moreover, cell apoptosis and autophagy were activated in DDX46 silencing groups (P<0.05). Therefore, the present results suggested that DDX46 was overexpressed in CSCC and that DDX46 silencing can inhibit cell proliferation by inducing apoptosis and activating autophagy. Thus, DDX46 may serve as a novel potential therapeutic target for CSCC.

Introduction

Cutaneous squamous cell carcinoma (CSCC) is one of the most common cancer types, accounting for 25–35% of cutaneous malignancies (1). CSCC develops from epidermal keratinocytes or adnexal squamous epithelial cells (2). Currently, immunosuppression factors that are sensitive to sunlight or UV radiation and genetic factors are thought to be closely associated with CSCC (3). Treatments for cutaneous squamous cell carcinoma include excision, radiotherapy, photodynamic therapy and topical drug treatment (4).

DEAD-box (DDX) RNA helicases are a large ATP-dependent RNA helicases family, which originates from a wide number of organisms ranging from prokaryotes, including viruses, to eukaryotes such as yeast, plants and animals (5). DDX is involved in almost every cellular process of RNA metabolism, from RNA synthesis to RNA degradation and plays important roles during cell development and growth in almost all organisms (6). DDX46 is a member of the DDX RNA helicase family located on chromosome 5q31.1 and plays a critical role in transcript splicing and ribosome assembly (7). Previous studies (7,8) have shown that DDX46 is abnormally expressed and plays an oncogenic role in various tumors, and is also involved in tumorigenesis and cancer progression. However, the role of DDX46 in CSCC is still not fully understood.

Therefore, in the present study, three pairs of CSCC tissues and corresponding adjacent tissues were collected, and the expression level of DDX46 was assessed. Furthermore, the effect of DDX46 silencing on CSCC cell proliferation, apoptosis and autophagy was analyzed. To our best knowledge, the present study is the first to identify the possible role of DDX46 in CSCC.

Materials and methods

Clinical specimens and cell lines

Human CSCC A431 and SCL-1 cells, and human keratinocyte HaCaT cells were purchased from Shanghai Enzyme Biological Technology Co., Ltd. In addition, three pairs of CSCC tissues and corresponding adjacent tissues were collected from the Department of Plastic Surgery, The First Hospital of Jilin University from September 2015 to November 2018. All the specimens were confirmed for CSCC by clinical and pathological diagnosis. Consent was obtained from the patients and their families. The present study was informed and approved by the Ethics Committee of The First Hospital of Jilin University.

Reagents and instruments

DMEM and FBS were purchased from Santa Co., Ltd. DMSO, crystal violet and tryptase were purchased from Sigma-Aldrich (Merck KGaA). Cell Counting Kit-8 (CCK-8) kits, Annexin V/PI cell apoptosis detection kits and TaqMan™ reverse transcription reagents (cat. no. 4304134) were obtained from Santa Cruz Biotechnology, Inc. TRIzol® kit, the transfection reagent Lipofectamine 3000® and 3,3′-diaminobenzidine (DAB) reagent kit were provided by Thermo Fisher Scientific, Inc. Rabbit anti-DDX46 (1:500; cat. no. ab72083) antibody was purchased from Abcam. The forward (F) and reverse (R) primers sequences for RT-qPCR of DDX46 were designed and synthesized by Arraystar, Inc., and were as follows: DDX46 F, 5′-AAAATGGCGAGAAGAGCAACG-3′ and R, 5′-CATCATCGTCCTCTAAACTCCAC-3′; GAPDH F, 5′-TGACTTCAACAGCGACACCCA-3′ and R, CACCCTGTTGCTGTAGCCAAA-3′. The preparation and packaging of DDX46 gene RNAi target sequence and shDX46 lentivirus (shDDX46) were designed and synthesized by Arraystar, Inc.

RT-qPCR

Total RNAs of CSCC tissues and cells were extracted with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Total RNA was reverse transcribed into cDNA at room temperature using TaqMan™ reverse transcription reagents (cat. no. 4304134; Thermo Fisher Scientific, Inc.). qPCR was subsequently performed using the iScript™ cDNA Synthesis kit (cat. no. 1708890; Bio-Rad Laboratories, Inc.). The PCR conditions were as follows: 95°C for 3 min, followed by 40 cycles of 95°C for 10 sec, 60°C for 30 sec and 72°C for 30 sec, according to the manufacturer's protocol of the PCR kit. The relative levels were assessed by 2−ΔΔCq method (9); ΔCq=Cqtarget-CqGAPDH.

Western blotting

Total protein of CSCC tissues and cells were collected with RIPA buffer (Sigma-Aldrich; Merck KGaA). Protein concentrations were determined with a BCA assay (Bio-Rad Laboratories, Inc.). Then, a total of 20 µg protein per lane was transferred to the nitrocellulose membrane after 10% SDS-PAGE and blocked by 5% TBST (0.1% Tween-20) for 1 h at room temperature. The membranes were incubated with rabbit anti-DDX46 (1:1,000; cat. no. ab72083; Abcam), rabbit anti-Bcl-2 antibody (1:1500; cat. no. ab59348; Abcam), rabbit anti-Survivin antibody (1:1,000; cat. no. ab469; Abcam), rabbit anti-Bax antibody (1:1,000; cat. no. ab53154; Abcam), rabbit anti-Cleaved Caspase-3 antibody (1:1,000; cat. no. ab2302; Abcam), rabbit anti-Beclin 1 antibody (ab62557) (1:1,000; cat. no. ab62557; Abcam), rabbit anti-microtubule-associated protein 1A/1B-light chain 3 (LC3) antibody (1:1,000; cat. no. ab128025; Abcam) and anti-β-actin (1:2,000; cat. no. ab8227; Abcam) at 4°C overnight. The PVDF membrane was washed three times with TBST and incubated at room temperature for 2 h with horseradish peroxidase-conjugated secondary antibody (1:2,000; cat. no. ab97051; Abcam). Blot bands were visualized by ECL systems (Pierce; Thermo Fisher Scientific, Inc.). Densitometry was performed by using ImageJ v1.47 software (National Institutes of Health) (10).

Cell transfection

The preparation and packaging of DDX46 gene RNAi target sequence (shDDX46) and negative control sequence (shNC) were designed and synthesized by Arraystar, Inc. short hairpin (sh)RNA sequence of DDX46 was 5′-AGAAATCACCAGGCTCATA-3′, and the negative control shRNA (shNC) sequence was 5′-TTCTCCGAACGTGTCACGT-3′. The quantity of siRNA transfected was 50 nM. Cells (1×105/well) were transfected using Lipofectamine 3000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Then, 48 h after transfection, the transfection efficiency was evaluated by labeling vectors with green fluorescence protein (GFP) to ensure that cells were successfully transfected, and evaluated by RT-qPCR using the same steps as mentioned above).

CCK-8 assay

Transfected cells in the logarithmic growth stage were collected and inoculated into 96-well plates with density of 2×103 cells per pore. Each group was assessed with three multiple pores. After adherence, the cells were replaced with 100 ml RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.).containing 10 ml CCK-8 reagent and incubated for 2 h at 37°C according to the manufacturer's instructions. The absorbance (D) of each pore were measured at 450 nm by enzyme label. The D values of the cells in each group were measured at 24, 48, 72 and 96 h. The growth curves of each group cells were drawn according to the measured D value.

Colony formation assay

Transfected cells in the logarithmic growth stage were digested with 0.5% trypsin (Gibco; Thermo Fisher Scientific, Inc.) at room temperature for 5 sec and seeded in 6-well plates with 1×105/well. After cultured for 2 weeks, 6-well plates were washed twice with PBS and cells were fixed with 2% paraformaldehyde (PFA) at 37°C for 30 min. Then, cells were stained with 0.5% crystal violet for 15 min at 37°C. Cell colony forming units were imaged under a confocal microscope (Nikon Corporation; magnification, ×400) and analyzed using ImageJ v1.47 software (National Institutes of Health) (10).

5-Ethynyl-2′-deoxyuridine (EdU) assay

Transfected cells in the logarithmic growth stage were digested with 0.5% trypsin (Gibco; Thermo Fisher Scientific, Inc.) at room temperature for 5 sec and seeded in 24-well plates with 1×104/well. After cultured for 24 h, 5 µl EdU reagent (Sigma-Aldrich; Merck KGaA)was added to each dish and incubated for 2 h at 37°C. Then, plates were washed twice with PBS and cell were fixed with 2% PFA for 30 min at 37°C. EdU positive cells were visualized and photographed under fluorescence microscope (BD Biosciences; magnification, ×400), and analyzed using ImageJ v1.47 software (National Institute of Health).

Flow cytometry

Transfected cells in the logarithmic growth stage were digested with 0.5% trypsin (Gibco; Thermo Fisher Scientific, Inc.) at room temperature for 5 sec and seeded in 6-well plates with 1×104/well. After cultured for 24 h, the cells were collected and washed twice with pre-cooled PBS. Then, cells were re-suspended with 1X combined buffer (Sigma-Aldrich; Merck KGaA) and the cell density was adjusted to ~1×106/ml. Then, 100 ml cell fluid was added to the flow tube, 5 µl Annexin V-FITC and 5 µl PI solution were added into flow tube to stain cells for 20 min in the dark at 4°C. Then, 400 ml combined buffer was added and the apoptotic rate was detected by a FACSCalibur flow cytometer (Becton-Dickinson and Company) for cell-cycle distribution analysis using Cell Quest Software (v5.1; Becton-Dickinson and Company).

Statistical analysis

SPSS v17.0 (SPSS, Inc.) software were used for statistical analyses. Data are presented as the mean ± SD of three experimental repeats. Student's t-test and χ2 test were performed among two groups, one-way ANOVA followed by Tukey's multiple comparison tests were performed among three groups. P<0.05 was considered to indicate a statistically significant difference.

Results

DDX46 is overexpressed in CSCC tissues

In the present study, the expression level of DDX46 in three pairs of CSCC tissues and corresponding adjacent tissues was assessed using RT-qPCR and Western blot assay. The RT-qPCR and Western blot results demonstrated that DDX46 mRNA (P<0.01; Fig. 1) and protein (P<0.05; Figs. 2 and 3) expression levels were significantly overexpressed in CSCC cells, and also in CSCC tissues (n=4) compared with corresponding adjacent tissues.

shRNA is successfully transfected into CSCC cells

shDDX46 and shNC were transfected into A431 and SCL-1 cells. Then, 48 h after transfection using a fluorescence microscope GFP was observed in A431 and SCL-1 cells (Fig. 4A). RT-qPCR results indicated that DDX46 mRNA expression levels were significantly decreased in the shDDX46 group compared with the shNC group (P<0.05; Fig. 4B).

DDX46 silencing inhibits cell proliferation

CCK-8, colony formation and EdU assay were used to analyze the effect of DDX46 on cell proliferation. CCK-8 assay results demonstrated that the proliferative ability of cells was significantly decreased in the shDDX46 group compared with the control group (P<0.05; Fig. 5). Furthermore, colony formation assay results suggested that the number of colony-forming units were significantly reduced in the shDDX46 group (P<0.05; Fig. 6) compared with the shNC group. In addition, it was found that the number of EdU positive cells was significantly decreased in the shDDX46 group compared with the shNC group (P<0.05; Fig. 7).

DDX46 silencing inhibits cell apoptosis

Flow cytometry was used to assess cell apoptosis and Western blotting was used to measure the expression levels of the apoptotic-related proteins Bcl-2, Survivin, Bax and cleaved caspase-3. It was demonstrated that the apoptotic rate was significantly increased in the shDDX46 group, compared with shNC group (P<0.05; Fig. 8). Furthermore, Western blotting results indicated that the protein expression levels of Bcl-2 and Survivin were significantly decreased in the shDDX46 group, and that the protein expression levels of Bax and cleaved caspase-3 protein were significantly increased in shDDX46 group (P<0.05; Fig. 9).

DDX46 silencing can activate cell autophagy

Western blotting was used to detect the protein expression levels of Beclin1 and LC3 II/I. It was demonstrated that the protein expression levels of Beclin1 protein and the ratio of LC3 II/I were significantly increased in the shDDX46 group compared with the shNC group (P<0.05; Fig. 10).

Discussion

RNA helicase is essential for RNA metabolism, including RNA splicing, transcription, transport, translation and degradation (7). DDX is an important ATP-dependent RNA helicase family, which exists in almost all prokaryotes and eukaryotes (11). Furthermore, DDX plays an important role in RNA metabolism, it is required for RNA functions, including gene transcription, RNA precursor mothering, ribosome synthesis, RNA output, translation initiation and RNA degradation (12,13). Several DDX RNA helicase subtypes have been reported to be abnormally expressed in various cancer types (14). Previous studies have shown that DDX1 is overexpressed in retinoblastoma, transitional blastoma, breast cancer and testicular cancer (15,16). Moreover, DDX5 is upregulated in human melanoma and hepatocellular carcinoma (17,18), and DDX6 is increased in glioblastoma, rhabdomyosarcoma, lung cancer, colon cancer and liver cancer (19). Thus, the DDX family may be involved in the occurrence and progression cancer.

DDX46 is a member of the DDX RNA helicase family with ATP-dependent RNA helicase, that is located on chromosome 5q31.1 and plays a critical role in transcript splicing and ribosome assembly (20). Previous studies (2123) in zebrafish have shown that DDX46 is required in the multilineage differentiation of hematopoietic stem cells and in the development of digestive organs and the brain. Li et al (24) reported that DDX46 is significantly overexpressed in colorectal cancer and that lentiviral DDX46 knockdown inhibits growth and induces apoptosis in human colorectal cancer cells. Liu et al (25) showed that DDX46 is upregulated in human bladder cancer 5637 and T24 cells. Li et al (26) found that DDX46 is significantly upregulated in esophageal squamous cell carcinoma tissues and cells, and that DDX46 knockdown decreases proliferation and increased apoptosis in TE-1 and Eca-109 cells. Thus, DDX46 may be involved in tumorigenesis and cancer progression. However, the role of DDX46 in CSCC still remains unknown.

The present results suggested that DDX46 was overexpressed in CSCC tissues, as indicated by results from IHC, RT-qPCR and Western blotting. The present study used DDX46 RNAi lentivirus to silence DDX46 expression in CSCC cells. It was found that DDX46 silencing can inhibit cell proliferation. Furthermore, DDX46 silencing can also induce cell apoptosis and activate autophagy by regulating the protein expression levels of Bcl-2, Survivin, Bax, cleaved caspase-3, Beclin1 and LC3 II/I. Thus, the present results suggested that DDX46 was overexpressed in CSCC and that DDX46 silencing can inhibit cell proliferation, the mechanism of which may be associated with apoptosis and autophagy. Previous studies have found that autophagy and apoptotic activity changes occur in a variety of human tumors (26,27). In some cases, autophagy inhibits cell apoptosis, which promotes cell survival (28). In other instances, autophagy can induce cell death, or function alongside apoptosis to induce cell death in the case of apoptotic defect (29,30). Thus, there is a close relationship between autophagy and apoptosis during cell death, and these pathways are interrelated and regulate each other. The present results indicated that apoptosis and autophagy worked together to inhibit CSCC cell proliferation.

In conclusion, to the best of our knowledge, the present study provides the first evidence that DDX46 was significantly overexpressed in CSCC. Furthermore, it was found that DDX46 silencing inhibited cell proliferation by inducing apoptosis and activating autophagy. Thus, DDX46 may serve as a novel potential therapeutic target for CSCC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

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

Authors' contributions

LG and QL designed the study and performed the experiments. HJJ and DZ analyzed the data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of the First Hospital of Jilin University (Jilin, China), and written informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Que SKT, Zwald FO and Schmults CD: Cutaneous squamous cell carcinoma: Incidence, risk factors, diagnosis, and staging. J Am Acad Dermatol. 78:237–247. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Ratushny V, Gober MD, Hick R, Ridky TW and Seykora JT: From keratinocyte to cancer: The pathogenesis and modeling of cutaneous squamous cell carcinoma. J Clin Invest. 122:464–472. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Umezono Y, Sato Y, Noto M, Yamada K, Noguchi N, Hasunuma N, Osada SI and Manabe M: Incidence rate of cutaneous squamous cell carcinoma is rapidly increasing in Akita Prefecture: Urgent alert for super-aged society. J Dermatol. 46:259–262. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Peters FS, Peeters AMA, Mandaviya PR, van Meurs JB, Hofland LJ, van de Wetering J, Betjes MG, Baan CC and Boer K: Differentially methylated regions in T cells identify kidney transplant patients at risk for de novo skin cancer. Clin Epigenetics. 10:812018. View Article : Google Scholar : PubMed/NCBI

5 

Jiang F, Zhang D, Li G and Wang X: Knockdown of DDX46 inhibits the invasion and tumorigenesis in osteosarcoma cells. Oncol Res. 25:417–425. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Nawaz G and Kang H: Rice OsRH58, a chloroplast DEAD-box RNA helicase, improves salt or drought stress tolerance in Arabidopsis by affecting chloroplast translation. BMC Plant Biol. 19:172019. View Article : Google Scholar : PubMed/NCBI

7 

Baek W, Lim CW and Lee SC: A DEAD-box RNA helicase, RH8, is critical for regulation of ABA signaling and the drought stress response via inhibition of PP2CA activity. Plant Cell Environ. 41:1593–1604. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Zheng Q, Hou J, Zhou Y, Li Z and Cao X: The RNA helicase DDX46 inhibits innate immunity by entrapping m6A-demethylated antiviral transcripts in the nucleus. Nat Immunol. 18:1094–1103. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

10 

Schindelin J, Rueden CT, Hiner MC and Eliceiri KW: The ImageJ ecosystem: An open platform for biomedical image analysis. Mol Reprod Dev. 82:518–529. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Pauszek R, Lamichhane R, Rajkarnikar Singh A, van der Schans E and Millar D: The 5 nuclease domain of DNA polymerase I mediates a novel DNA transfer pathway during proofreading. Biophys J. 114:82a2018. View Article : Google Scholar

12 

Sohn SO and Chay KO: The ATP-dependent RNA helicase, DDX42 interacts with paxillin and regulates apoptosis and polarization of Ba/F3 cells. Anim Cells Syst Seoul. 23:1–9. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Gao J, Byrd AK, Zybailov BL, Marecki JC, Guderyon MJ, Edwards AD, Chib S, West KL, Waldrip ZJ, Mackintosh SG, et al: DEAD-box RNA helicases Dbp2, Ded1 and Mss116 bind to G-quadruplex nucleic acids and destabilize G-quadruplex RNA. Chem Commun (Camb). 55:4467–4470. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Curmi F and Cauchi RJ: The multiple lives of DEAD-box RNA helicase DP103/DDX20/Gemin3. Biochem Soc Trans. 46:329–341. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Tang J, Chen H, Wong CC, Liu D, Li T, Wang X, Ji J, Sung JJ, Fang JY and Yu J: DEAD-box helicase 27 promotes colorectal cancer growth and metastasis and predicts poor survival in CRC patients. Oncogene. 37:3006–3021. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Ribeiro de Almeida C, Dhir S, Dhir A, Moghaddam AE, Sattentau Q, Meinhart A and Proudfoot NJ: RNA helicase DDX1 converts RNA G-Quadruplex structures into R-Loops to promote IgH class switch recombination. Mol Cell. 70:650–662.e8. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Zhong W, Li Z, Zhou M, Xu T and Wang Y: DDX1 regulates alternative splicing and insulin secretion in pancreatic β cells. Biochem Biophys Res Commun. 500:751–757. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Wu N, Jiang M, Han Y, Liu H, Chu Y, Liu H, Cao J, Hou Q, Zhao Y, Xu B, et al: O-GlcNAcylation promotes colorectal cancer progression by regulating protein stability and potential catcinogenic function of DDX5. J Cell Mol Med. 23:1354–1362. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Huang W, Thomas B, Flynn RA, Gavzy SJ, Wu L, Kim SV, Hall JA, Miraldi ER, Ng CP, Rigo F, et al: Retraction note: DDX5 and its associated lncRNA Rmrp modulate TH17 cell effector functions. Nature. 562:1502018. View Article : Google Scholar : PubMed/NCBI

20 

Taniguchi K, Iwatsuki A, Sugito N, Shinohara H, Kuranaga Y, Oshikawa Y, Tajirika T, Futamura M, Yoshida K, Uchiyama K, et al: Oncogene RNA helicase DDX6 promotes the process of c-Myc expression in gastric cancer cells. Mol Carcinog. 57:579–589. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Lumb JH, Li Q, Popov LM, Ding S, Keith MT, Merrill BD, Greenberg HB, Li JB and Carette JE: DDX6 Represses Aberrant Activation of Interferon-Stimulated Genes. Cell Rep. 20:819–831. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Hirabayashi R, Hozumi S, Higashijima S and Kikuchi Y: Ddx46 is required for multi-lineage differentiation of hematopoietic stem cells in zebrafish. Stem Cells Dev. 22:2532–2542. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Hozumi S, Hirabayashi R, Yoshizawa A, Ogata M, Ishitani T, Tsutsumi M, Kuroiwa A, Itoh M and Kikuchi Y: DEAD-box protein Ddx46 is required for the development of the digestive organs and brain in zebrafish. PLoS One. 7:e336752012. View Article : Google Scholar : PubMed/NCBI

24 

Li M, Ma Y, Huang P, Du A, Yang X, Zhang S, Xing C, Liu F and Cao J: Lentiviral DDX46 knockdown inhibits growth and induces apoptosis in human colorectal cancer cells. Gene. 560:237–244. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Liu X, Zhang C, Huang D, et al: Construction of DDX46 lentiviral vector and its expression identification in human bladder cancer cells. Anhui Medical Journal. 2018.(In Chinese).

26 

Li B, Li YM, He WT, Chen H, Zhu HW, Liu T, Zhang JH, Song TN and Zhou YL: Knockdown of DDX46 inhibits proliferation and induces apoptosis in esophageal squamous cell carcinoma cells. Oncol Rep. 36:223–230. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Mane SD and Kamatham AN: Ascorbyl stearate stimulates cell death by oxidative stress-mediated apoptosis and autophagy in HeLa cervical cancer cell line in vitro. 3 Biotech. 9:1152019. View Article : Google Scholar : PubMed/NCBI

28 

Chen Q, Kang J and Fu C: The independence of and associations among apoptosis, autophagy, and necrosis. Signal Transduct Target Ther. 3:18–23. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Song S, Tan J, Miao Y, Li M and Zhang Q: Crosstalk of autophagy and apoptosis: Involvement of the dual role of autophagy under ER stress. J Cell Physiol. 232:2977–2984. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Yan X, Zhou R and Ma Z: Autophagy-cell survival and death. Adv Exp Med Biol. 1206:667–696. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 22 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin Q, Jin H, Zhang D and Gao L: DDX46 silencing inhibits cell proliferation by activating apoptosis and autophagy in cutaneous squamous cell carcinoma. Mol Med Rep 22: 4236-4242, 2020
APA
Lin, Q., Jin, H., Zhang, D., & Gao, L. (2020). DDX46 silencing inhibits cell proliferation by activating apoptosis and autophagy in cutaneous squamous cell carcinoma. Molecular Medicine Reports, 22, 4236-4242. https://doi.org/10.3892/mmr.2020.11509
MLA
Lin, Q., Jin, H., Zhang, D., Gao, L."DDX46 silencing inhibits cell proliferation by activating apoptosis and autophagy in cutaneous squamous cell carcinoma". Molecular Medicine Reports 22.5 (2020): 4236-4242.
Chicago
Lin, Q., Jin, H., Zhang, D., Gao, L."DDX46 silencing inhibits cell proliferation by activating apoptosis and autophagy in cutaneous squamous cell carcinoma". Molecular Medicine Reports 22, no. 5 (2020): 4236-4242. https://doi.org/10.3892/mmr.2020.11509