Open Access

Silencing cyclin-dependent kinase inhibitor 3 inhibits the migration of breast cancer cell lines

Retraction in: /10.3892/mmr.2023.13075

  • Authors:
    • Miao Deng
    • Jianguang Wang
    • Yanbin Chen
    • Like Zhang
    • Gangqiang Xie
    • Qipeng Liu
    • Ting Zhang
    • Pengfei Yuan
    • Dechun Liu
  • View Affiliations

  • Published online on: June 14, 2016     https://doi.org/10.3892/mmr.2016.5401
  • Pages: 1523-1530
  • Copyright: © Deng 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

Cyclin-dependent kinase inhibitor 3 (CDKN3) belongs to the dual-specificity protein phosphatase family, which is hypothesized to regulate cell cycle progression in tumor cells. However, whether CDKN3 is a potential therapeutic target for breast cancer remains to be elucidated. The present in vitro study aimed to investigate the potential roles of CDKN3 in breast cancer. Breast cancer cell lines were used to detect CDKN3 expression, and CDKN3 expression was silenced to investigate its role in cell apoptosis, cell cycle arrest and migration. The underlying mechanisms were screened by detecting proliferating cell nuclear antigen (PCNA), Ras homolog gene family, member A (RhoA), vimentin, B‑cell lymphoma 2 (Bcl‑2) and Bcl‑2‑associated X protein (Bax) expression. CDKN3 was highly expressed in MCF‑7 and BT474 cell lines. The silencing of CDKN3 in MCF‑7 and BT474 cell lines promoted cell apoptosis, induced G1 phase cell cycle arrest and inhibited cell migration. The expression levels of PCNA, RhoA, vimentin and Bcl‑2 were downregulated following CDKN3 silencing. Conversely, Bax expression was increased, as compared with the vehicle control. These results suggest that CDKN3 acts as an oncogene during breast cancer progression. The in vitro silencing of CDKN3 promoted apoptosis, induced G1 phase cell cycle arrest and inhibited cell migration. Possible mechanisms are associated with the regulation of PCNA, Bcl‑2, vimentin, RhoA and Bax expression. CDKN3 may therefore be considered a potential target for the treatment of breast cancer.

Introduction

Breast cancer is the most common type of malignancy in women, accounting for 29% of all female cancer cases and <1% of all male cancer cases (1). Each year, a large number of patients with breast cancer are diagnosed and the survival rate remains poor in developing countries. In certain cases, surgery, chemotherapy and radiotherapy are insufficient for successful treatment. Therefore, more efficient methods based on genetic factors should be developed to target breast cancer. In addition to benefiting treatment, identification of specific targets will be beneficial for disease diagnosis.

Cyclin-dependent kinases (CDKs) are a family of protein kinases first identified for their function in regulating the cell cycle (2,3). CDKs bind to cyclin proteins and form cyclin-CDK complexes that regulate cell cycle progression. CDK inhibitor 3 (CDKN3) is an enzyme encoded by the CDKN3 gene (2). The CDKN3 protein belongs to the dual-specificity protein phosphatase family. It was initially identified as a CDK inhibitor, which interacts with, and dephosphorylates CDK2, thus preventing its activation (4). Aberrant expression of CDKN3, including deletion, mutation, or overexpression has been reported in numerous types of cancer (46). However, the exact role of CDKN3 in the progression of breast cancer remains to be elucidated.

The association between CDKN3 and tumorigenesis has previously been reported in several studies (46). However, the molecular mechanisms underlying the effects of CDKN3 on cell cycle arrest, apoptosis and cell migration remain to be determined. In the present study, CDKN3 was observed to be highly expressed in MCF-7 and BT474 cell lines. An in vitro model produced by silencing CDKN3 was used to detect the potential roles of CDKN3 in tumor cell proliferation, apoptosis and migration. The present study hypothesized that CDKN3 is a potential target for the treatment of breast cancer.

Materials and methods

Cell lines and antibodies

The MCF-7, ZR-75-30, T47-D, MDA-MB-231 and BT474 breast cancer cell lines were obtained from the American Type Culture Collection (Manassas, VA, USA). The cell lines were cultured in Dulbecco's modified Eagle's medium (DMEM; Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.), 50 U/ml penicillin and 0.1 mg/ml streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C in a humidified atmosphere containing 5% CO2. Antibodies against CDKN3, proliferating cell nuclear antigen (PCNA), B-cell lymphoma 2 (Bcl-2), vimentin and Bcl-2-associated X protein (Bax) were purchased from Sigma-Aldrich (St. Louis, MO, USA).

Construction of CDKN3-specific small interfering (si)RNA expression vectors

The siRNA-expressing constructs were generated by subcloning siRNA oligonucleotides (Shanghai Generay Biotech, Inc., Shanghai, China) into BamHI and EcoRI sites of pSIH-H1-GFP vector (System Biosciences, Mountain View, CA, USA), as previously described (7). The siRNA sequences were as follows: i) 5′-AAGAGCCUAUUGAAGAUGAUU-3′ for siRNA1; ii) 5′-UAGCUGCUUGUCUCCUACUUU-3′ for siRNA2; and iii) 5′-AAACCACCAGUGUUAUCAAUU-3′ for siRNA3. Negative control siRNA was also used, the sequence was 5′-UUCUCCGAACGUGUCACGUTT-3′. Briefly, cells were cultured on 6-well plates. Following a 24 h incubation, the original medium was replaced with serum and antibiotics-free medium. Transfection reagents were mixed with siRNAs; 10 µl Lipofectamine 2000 (Thermo Fisher Scientific, Inc.) was diluted with 250 µl Opti-MEM (Thermo Fisher Scientific, Inc.) in each well, and was incubated at room temperature for 5 min. In addition, 10 µl siRNAs were diluted with 250 µl Opti-MEM, and were gently agitated. The diluted Lipofectamine 2000 and diluted siRNAs were mixed and incubated at room temperature for 20 min. A total of 6 h post-transfection, the culture medium was replaced with medium containing antibiotics. The silencing effect was confirmed by detecting CDKN3 protein expression by western blotting. Breast cancer cells with Lipofectamine only were regarded as the control group, whereas breast cancer cells that were transfected with negative control siRNA were regarded as the vehicle group.

Apoptosis assay

Annexin V-fluorescein isothiocyanate (FITC) and propidium iodide (PI) were used to detect apoptotic cells following CDKN3 silencing. Subsequent to trypsinization, the cells were washed twice with phosphate-buffered saline (PBS) and resuspended in binding buffer containing Annexin-V-FITC and PI (BD Biosciences, Franklin Lakes, NJ, USA). The cells were incubated at room temperature for 15 min and were then analyzed using a flow cytometer (LSR Fortessa™; BD Biosciences).

Cell cycle assay

Cells were fixed in chilled 75% ethanol, and were stained with a solution containing 100 µg/ml RNase (Tiangen Biotech Co., Ltd., Beijing, China) and 50 µg/ml PI (BD Biosciences) in PBS for cell cycle analysis. The percentage of cells at each phase of the cell cycle was evaluated by flow cytometry (BD Biosciences).

Western blot analysis

Following the silencing of CDKN3, proteins were extracted using Cell Extraction Buffer (Thermo Fisher Scientific, Inc.) and protein concentration was measured using bicinchoninic acid protein assay kit (Thermo Fisher Scientific, Inc.). Equivalent quantities of protein from each group (10 µl) were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (120 V for 20 min in condensing gel; 180 V for 60 min in separating gel) and were transferred to polyvinylidene fluoride membranes (Thermo Fisher Scientific, Inc.) at 100 V for 120 min. After blocking with 5% non-fat milk powder for 1 h, the membranes were incubated with rabbit primary antibodies against CDKN3 (1:2,000; Abcam, Cambridge, MA, USA; ab118702), Bcl-2 (1:2,000; Abcam; cat. no. ab59348), Bax (1:3,000; Abcam; cat. no. ab32503), PCNA (1:2,000; Abcam; cat. no. ab18197), RhoA (1:2,000; Abcam; cat. no. ab187027), vimentin (1:2,000; Abcam; cat. no. ab92547) and GAPDH (1:5,000; Abcam; cat. no. ab9485) at 4°C overnight. Membranes were subsequently incubated with horseradish peroxidase-conjugated goat anti-rabbit secondary antibody (1:4,000; Abcam; cat. no. 6721) at room temperature for 1 h. Blots were visualized with enhanced chemiluminescence solution (Thermo Fisher Scientific, Inc.), and films were exposed in a dark room. Blot images were semi-quantified by ChemiDoc™ Touch Imaging System (Bio-Rad Laboratories, Hercules, CA, USA).

Migration assay

Equal numbers of cells (1×105) in DMEM with 5% FBS were added to the upper compartment of a Transwell plate and were maintained at 37°C for 15 h. The lower compartment was filled with DMEM supplemented with 10% FBS. Following incubation, the cells from the upper surface of the filter were removed with a cotton swab. The cells in the lower compartment were stained with crystal violet and visualized under light microscopy (CX31; Olympus Corporation, Tokyo, Japan). The number of cells that migrated to the lower chamber was counted in five randomly selected fields and the mean number of cells was calculated in each group.

Statistical analysis

All experiments were performed in triplicate, and the results were expressed as the mean ± standard deviation. A t-test was utilized to compare differences between two groups, and a one-way analysis of variance was used to compare differences between three or more groups. Statistical analyses were performed using IBM SPSS Statistics 20 software (IBM SPSS, Armonk, NY, USA).

Results

CDKN3 expression levels are increased in a number of breast cancer cell lines

To investigate the function of CDKN3 expression in tumor growth, CDKN3 expression levels were detected in a number of breast cancer cell lines, including MCF-7, ZR-75-30, T47-D, MDA-MB-231 and BT474. As demonstrated by western blot analysis, CDKN3 was highly expressed in the MCF-7 and BT474 cell lines (Fig. 1A). In subsequent experiments, siRNA was used to knockdown the expression of CDKN3 in MCF-7 and BT474 cell lines. Three siRNA sequences were designed and the optimal knockdown effect was observed using the siRNA3 sequence. As presented in Fig. 1B–D, siRNA significantly decreased the CDKN3 expression levels in the MCF-7 and BT474 cell lines compared with the control (P<0.001). However, the vehicle sequence did not affect CDKN3 expression. These results suggest that CDKN3 is overexpressed in certain types of breast cancer cell lines and that siRNA is an effective approach to knockdown the expression levels of CDKN3.

CDKN3 knockdown inhibits breast cancer cell migration

The present study also analyzed whether cancer cell migration was affected by CDKN3 silencing in breast cancer cell lines using a Transwell assay. As presented in Fig. 2, the cell number in the bottom chamber was significantly inhibited by CDKN3 deletion (P<0.01 in MCF-7 cells; P<0.001 in BT474 cells), but not by vehicle treatment.

CDKN3 knockdown induces apoptosis in breast cancer cell lines

The apoptotic rate in the breast cancer cell lines was analyzed following silencing of CDKN3. As presented in Fig. 3, 24 h after CDKN3 knockdown, the apoptotic rate was significantly increased in MCF-7 cells (P<0.001). Conversely, vehicle transfection did not affect the apoptotic rate compared with the control. Similarly, the apoptotic rate was significantly increased in BT474 cells following CDKN3 knockdown (P<0.001).

CDKN3 knockdown induces cell cycle arrest in breast cancer cell lines

CDKN3 is a cell cycle regulatory protein. In the present study, cell cycle distribution was detected following CDKN3 silencing in MCF-7 and BT474 cell lines. As presented in Fig. 4, the number of cells in G1 phase was significantly increased following CDKN3 silencing in MCF-7 and BT474 cell lines (P<0.01 in MCF-7 cells; P<0.05 in BT474 cells). These results suggest that CDKN3 knockdown results in G1 phase arrest in breast cancer cell lines.

CDKN3 knockdown decreases PCNA, Bcl-2 and RhoA expression levels, but increases Bax expression

The potential mechanisms underlying the effects of CDKN3 silencing were investigated by detection of PCNA, Bcl-2, Bax, vimentin and RhoA expression levels. As presented in Fig. 5, CDKN3 knockdown decreased PCNA, Bcl-2, vimentin and RhoA expression compared with in the control MCF-7 and BT474 cell lines (P<0.01 for PCNA, Bcl-2 and vimentin, and P<0.001 for RhoA in MCF-7 cells; P<0.01 for PCNA, P<0.001 for Bcl-2 and RhoA, and P<0.05 for vimentin in BT474 cells). Conversely, Bax expression was increased following CDKN3 silencing compared with in the control cells (P<0.001 in MCF-7 cells; P<0.01 in BT474 cells).

Discussion

Dysregulation of CDKN3 has been demonstrated to be involved in tumorigenesis; however, the exact function of CDKN3 in breast cancer remains to be elucidated. In the present study, CDKN3 was highly expressed in breast cancer cell lines. Knockdown of this specific gene was able to inhibit migratory ability and cell cycle progression, and induce apoptosis. Possible mechanisms underlying these effects are associated with PCNA, RhoA and Bcl-2 downregulation.

Aberrant expression of CDKN3 has been reported to exert various effects on human tumorigenesis. Either overexpression or mutation may result in the proliferation of tumor cells; therefore, a dual function of CDKN3 as an oncogene and tumor suppressor gene has been detected in various types of tumor (810). However, the functional relevance of CDKN3 in breast cancer remains inconclusive, and more research is required. In the present study, CDKN3 was highly expressed in breast cancer cell lines, which is consistent with a previous report demonstrating CDKN3 as an important oncogene in breast cancer tumorigenesis (11). Although a previous study demonstrated that overexpression of CDKN3 may markedly sensitize imatinib-induced apoptosis in K562 leukemic cells, CDKN3 functions differently in various types of cancer (12). CDKN3 has been suggested to function as a tumor suppressor, and the effect of a loss of function of this protein has been observed in various cancers (8,9), including glioblastoma (8) and hepatocellular carcinoma (13). In addition, elevated levels of CDKN3 have been detected in renal cell carcinoma, which enhanced xenograft tumor growth, thus suggesting an oncogenic function of CDKN3 (14). Notably, numerous spliced transcript variants encoding different isoforms of CDKN3 have been observed in diverse types of cancer, indicating that these isoforms may be associated with specific tumor formation (13,15). Numerous breast cancer cell lines are available for use in therapeutic agent screening or mechanistic investigation (16). However, the characteristics of these cell lines are not the same. In the present study, CDKN3 expression was measured in MCF-7, ZR-75-30, T47-D, MDA-MA-231 and BT474 cell lines. CDKN3 exhibited the highest expression in the MCF-7 and BT474 cell lines. Therefore, in the subsequent mechanistic investigations, these two cell lines were used to investigate the function of CDKN3.

The CDKN3 gene is located on chromosome 14 at 14q22 (8). CDKN3 dephosphorylates CDK2 and inhibits G1/S-phase cell cycle progression (4). A further function of CDKN3 is interaction with CDK1 to facilitate mitosis by dephosphorylating CDK1 at Thr161. CDKN3 belongs to the protein phosphatase family and has a dual function in regulating the cell cycle and proliferation. In the present study, the knockdown of CDKN3 in MCF-7 and BT474 cells contributed to G1 phase arrest. Conversely, in a previous study, CDKN3 expression was upregulated following Y-box binding protein 1 silencing, resulting in G1 phase arrest (17). This may be due to the dual function of CDKN3 in tumorigenesis. In addition, the type of cancer may also affect the function of CDKN3, for example, in prostate cancer cells and hepatocellular carcinoma, downregulation of CDKN3 results in G1 phase arrest (18,19).

In addition to cell cycle arrest, CDKN3 silencing also triggered apoptosis in the two breast cancer cell lines. Cell cycle arrest contributes to the inhibition of proliferation, and induction of apoptosis is also considered an important strategy to inhibit tumor cell proliferation.

In addition to proliferation, migration is a key factor in determining tumorigenesis. Although CDKN3 predominantly functions via regulation of the cell cycle, CDKN3 inhibits migration by decreasing CDK1 mRNA and protein expression (15). In the present study, CDKN3 silencing inhibited cell migration in the breast cancer cell lines.

The present study also demonstrated that CDKN3 silencing inhibits PCNA, Bcl-2 and RhoA expression. The potential mechanisms underlying the effects of CDKN3 on apoptosis induction and cell cycle arrest were investigated in the present study. PCNA is a DNA clamp, the function of which is essential for DNA replication (20). The downregulation of PCNA was observed in breast cancer cell lines following silencing of CDKN3, thus suggesting that CDKN3 is important for PCNA expression. However, the mechanism underlying the CDKN3-induced regulation of PCNA remains to be elucidated. RhoA is important for actin-polymerization, which is required for cell proliferation (21). Decreased RhoA expression increases the turnover of actin, blocking proliferation. In the present study, RhoA expression levels were markedly decreased, which may be associated with inhibition of the cell cycle or cell migration.

Apoptosis is a type of programmed cell death. Various signaling pathways are associated with apoptosis, particularly the mitochondria-dependent and mitochondria-independent signaling pathways (22). A major checkpoint in the mitochondria-dependent signaling pathway is the ratio of proapoptotic (Bax) to anti-apoptotic (Bcl-2) proteins. In the present study, Bcl-2 and Bax expression levels were detected following CDKN3 silencing. The results of the present study demonstrated that CDKN3 silencing increased Bax expression levels and decreased Bcl-2 expression levels. Consistently, the ratio of Bax/Bcl-2 was increased following CDKN3 silencing, which was consistent with apoptosis. In addition, vimentin expression levels were detected following CDKN3 silencing. As a key marker of cancer (23), vimentin expression was significantly decreased following CDKN3 silencing.

In conclusion, results from the present study suggest that CDKN3 acts as a tumor oncogene during breast tumorigenesis. The in vitro silencing of CDKN3 promoted apoptosis, induced G1 phase cell cycle arrest and inhibited cell migration. Possible underlying mechanisms are associated with regulation of PCNA, Bcl-2, RhoA and Bax expression. Therefore, the present study suggested that CDKN3 silencing may be considered an effective method for the inhibition of breast cancer tumorigenesis.

References

1 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Baghdassarian N and Ffrench M: Cyclin-dependent kinase inhibitors (CKIs) and hematological malignancies. Hematol Cell Ther. 38:313–323. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Malumbres M: Cyclin-dependent kinases. Genome Biol. 15:1222014. View Article : Google Scholar : PubMed/NCBI

4 

Berumen J, Espinosa AM and Medina I: Targeting CDKN3 in cervical cancer. Expert Opin Ther Targets. 18:1149–1162. 2014. View Article : Google Scholar : PubMed/NCBI

5 

García-Escudero R, Martínez-Cruz AB, Santos M, Lorz C, Segrelles C, Garaulet G, Saiz-Ladera C, Costa C, Buitrago-Pérez A, Dueñas M and Paramio JM: Gene expression profiling of mouse p53-deficient epidermal carcinoma defines molecular determinants of human cancer malignancy. Mol Cancer. 9:1932010. View Article : Google Scholar : PubMed/NCBI

6 

Gerald WL: Gene expression analysis of prostate carcinoma. Expression Profiling of Human Tumors. Ladanyi M and Gerald WL: Humana Press; New York: pp. 173–197. 2003, View Article : Google Scholar

7 

Chen CF, Feng X, Liao HY, Jin WJ, Zhang J, Wang Y, Gong LL, Liu JJ, Yuan XH, Zhao BB, et al: Regulation of T cell proliferation by JMJD6 and PDGF-BB during chronic hepatitis B infection. Sci Rep. 4:63592014. View Article : Google Scholar : PubMed/NCBI

8 

Nalepa G, Barnholtz-Sloan J, Enzor R, Dey D, He Y, Gehlhausen JR, Lehmann AS, Park SJ, Yang Y, Yang X, et al: The tumor suppressor CDKN3 controls mitosis. J Cell Biol. 201:997–1012. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Yeh CT, Lu SC, Chao CH and Chao ML: Abolishment of the interaction between cyclin-dependent kinase 2 and Cdk-associated protein phosphatase by a truncated KAP mutant. Biochem Biophys Res Commun. 305:311–314. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Lee SW, Reimer CL, Fang L, Iruela-Arispe ML and Aaronson SA: Overexpression of kinase-associated phosphatase (KAP) in breast and prostate cancer and inhibition of the transformed phenotype by antisense KAP expression. Mol Cell Biol. 20:1723–1732. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Li H, Jiang X, Yu Y, Huang W, Xing H, Agar NY, Yang HW, Yang B, Carroll RS and Johnson MD: KAP regulates ROCK2 and Cdk2 in an RNA-activated glioblastoma invasion pathway. Oncogene. 34:1432–1441. 2015. View Article : Google Scholar

12 

Drullion C, Trégoat C, Lagarde V, Tan S, Gioia R, Priault M, Djavaheri-Mergny M, Brisson A, Auberger P, Mahon FX and Pasquet JM: Apoptosis and autophagy have opposite roles on imatinib-induced K562 leukemia cell senescence. Cell Death Dis. 3:e3732012. View Article : Google Scholar : PubMed/NCBI

13 

Yeh CT, Lu SC, Chen TC, Peng CY and Liaw YF: Aberrant transcripts of the cyclin-dependent kinase-associated protein phosphatase in hepatocellular carcinoma. Cancer Res. 60:4697–4700. 2000.PubMed/NCBI

14 

Lai MW, Chen TC, Pang ST and Yeh CT: Overexpression of cyclin-dependent kinase-associated protein phosphatase enhances cell proliferation in renal cancer cells. Urol Oncol. 30:871–878. 2012. View Article : Google Scholar

15 

Yu Y, Jiang X, Schoch BS, Carroll RS, Black PM and Johnson MD: Aberrant splicing of cyclin-dependent kinase-associated protein phosphatase KAP increases proliferation and migration in glioblastoma. Cancer Res. 67:130–138. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, Clark L, Bayani N, Coppe JP, Tong F, et al: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 10:515–527. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Yu YN, Yip GW, Tan PH, Thike AA, Matsumoto K, Tsujimoto M and Bay BH: Y-box binding protein 1 is up-regulated in proliferative breast cancer and its inhibition deregulates the cell cycle. Int J Oncol. 37:483–492. 2010.PubMed/NCBI

18 

Padua MB and Hansen PJ: Changes in expression of cell-cycle-related genes in PC-3 prostate cancer cells caused by ovine uterine serpin. J Cell Biochem. 107:1182–1188. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Xing C, Xie H, Zhou L, Zhou W, Zhang W, Ding S, Wei B, Yu X, Su R and Zheng S: Cyclin-dependent kinase inhibitor 3 is overexpressed in hepatocellular carcinoma and promotes tumor cell proliferation. Biochem Biophys Res Commun. 420:29–35. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Shibahara K and Stillman B: Replication-dependent marking of DNA by PCNA facilitates CAF-1-coupled inheritance of chromatin. Cell. 96:575–585. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Pillé JY, Denoyelle C, Varet J, Bertrand JR, Soria J, Opolon P, Lu H, Pritchard LL, Vannier JP, Malvy C, et al: Anti-RhoA and anti-RhoC siRNAs inhibit the proliferation and invasiveness of MDA-MB-231 breast cancer cells in vitro and in vivo. Mol Ther. 11:267–274. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Fulda S and Debatin KM: Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 25:4798–4811. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Satelli A and Li S: Vimentin in cancer and its potential as a molecular target for cancer therapy. Cell Mol Life Sci. 68:3033–3046. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2016
Volume 14 Issue 2

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
Deng M, Wang J, Chen Y, Zhang L, Xie G, Liu Q, Zhang T, Yuan P and Liu D: Silencing cyclin-dependent kinase inhibitor 3 inhibits the migration of breast cancer cell lines Retraction in /10.3892/mmr.2023.13075. Mol Med Rep 14: 1523-1530, 2016
APA
Deng, M., Wang, J., Chen, Y., Zhang, L., Xie, G., Liu, Q. ... Liu, D. (2016). Silencing cyclin-dependent kinase inhibitor 3 inhibits the migration of breast cancer cell lines Retraction in /10.3892/mmr.2023.13075. Molecular Medicine Reports, 14, 1523-1530. https://doi.org/10.3892/mmr.2016.5401
MLA
Deng, M., Wang, J., Chen, Y., Zhang, L., Xie, G., Liu, Q., Zhang, T., Yuan, P., Liu, D."Silencing cyclin-dependent kinase inhibitor 3 inhibits the migration of breast cancer cell lines Retraction in /10.3892/mmr.2023.13075". Molecular Medicine Reports 14.2 (2016): 1523-1530.
Chicago
Deng, M., Wang, J., Chen, Y., Zhang, L., Xie, G., Liu, Q., Zhang, T., Yuan, P., Liu, D."Silencing cyclin-dependent kinase inhibitor 3 inhibits the migration of breast cancer cell lines Retraction in /10.3892/mmr.2023.13075". Molecular Medicine Reports 14, no. 2 (2016): 1523-1530. https://doi.org/10.3892/mmr.2016.5401