Expression of cyclin B1, D1 and K in non‑small cell lung cancer H1299 cells following treatment with sulforaphane

  • Authors:
    • Agnieszka Żuryń
    • Adrian Krajewski
    • Anna Klimaszewska‑Wiśniewska
    • Alina Grzanka
    • Dariusz Grzanka
  • View Affiliations

  • Published online on: December 7, 2018     https://doi.org/10.3892/or.2018.6919
  • Pages: 1313-1323
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sulforaphane (SFN) was first isolated from broccoli sprout and it is present at high concentrations in plants belonging to the Cruciferae family. The chemotherapeutic and anti‑cancerogenic capacities of SFN have been demonstrated by inhibition of cancer cell proliferation in several cancer cell lines. The aim of the present study was to evaluate the effect of SFN on apoptosis, cell cycle and expression of selected cell cycle‑associated proteins: Cyclin B1, cyclin D1 and cyclin K in the H1299 cell line. The non‑small cell lung cancer cell line H1299 was treated with increasing concentrations of SFN (5, 10 and 15 µM) for two days. After incubation, the percentage of cells in the individual cell cycle phases, as well as the percentage of necrotic and apoptotic cells, were estimated using flow cytometry. The expression of cyclins was examined by immunofluorescence staining, flow cytometry, western blot analysis and qRT‑PCR. Cyclin K was characterized by nuclear localization and increased expression after treatment with SFN. The expression data were confirmed by qRT‑PCR. SFN‑induced cell cycle arrest was associated with a decrease in cyclin B1 expression. Cells treated with SFN were also characterized by higher cyclin D1 and cyclin K expression. These data suggest the involvement of cyclin K in response to SFN. Moreover, we investigated the prognostic value of cyclin K, CDK12 and CDK13 in adenocarcinoma patients using ‘The Kaplan‑Meier plotter’ (KM plotter) database. It was shown that high expression of CDK12 and CDK13 but no cyclin K proteins is associated with worse overall survival among adenocarcinoma patients.

Introduction

Sulforaphane (SFN) was first isolated from broccoli sprout and is present at high concentrations in plants which belong to the Cruciferae family (1). SFN is a chemopreventive agent which displays functions, including inhibition of carcinogen-activating enzymes, such as the cytochrome p450 isoenzyme 2E1, induction of conjugating enzymes, such as glutathione S-transferases, and reduction of the DNA binding ability of nuclear factor-κB (NF-κB) (24). Furthermore, SFN was found to exert antiproliferative effects on various cancer cell lines in vitro and in vivo (57). According to Choi and Singh, SFN treatment induces Bax and Bak protein expression and conformational change and mitochondrial translocation of Bax to trigger the release of apoptogenic molecules from the mitochondria to the cytosol causing activation of caspases and cell death. Their research showed that both Bax and Bak are crucial for SFN-induced cell death (8). SFN acts as an indirect antioxidant and inducer of antioxidant response element (ARE) genes, and Moreover, the exposure to SFN results in a transient reactive oxygen species (ROS) burst, for which the duration and magnitude both depend on the SFN concentration and exposure period (9,10). Cyclin K plays a dual role by regulating CDKs and transcription.

Cyclin K was first discovered in a yeast screen, which was based on its ability to restore cell cycle progression and rescue Saccharomyces cerevisiae cells from lethality caused by deletion of G1 cyclin (11). Cyclin K triggers Cdk9 activity by creating a stable protein complex with Cdk9. The Cdk9/cyclin K complex phosphorylates the carboxyl-terminal domain (CTD) of RNA polymerase II (RNAP II). This reaction is said to be one of the most significant steps in the transcription of many genes (12,13). Moreover, the role of cyclin K and Cdk9 has been implied in the pathways that provide genomic stability in response to replication stress. Cyclin K also binds Cdk12 and Cdk13 to form two different complexes (cyclin K/Cdk12 or cyclin K/Cdk13) in human cells. Phosphorylation of Ser2 in the C-terminal domain of RNAP II and expression of a small subset of human genes are regulated by the cyclin K/Cdk12 complex as revealed in expression microarrays (14). Decreased expression in mainly long genes with a high number of exons is a result of depletion of cyclin K/Cdk12. Human cells without cyclin K/Cdk12 generate spontaneous DNA damage and are sensitive to a variety of DNA damage agents. Moreover, cyclin K/Cdk12 protects them from genomic instability (14). The regulation of Cdk13 activity is currently not well understood as the expression levels of the corresponding cyclin subunit, cyclin K, are rather stable. This is similar to other cyclins controlling transcriptional CDKs, e.g., cyclin T1, but different from cell cycle-regulating cyclins such as cyclin A (15). Cyclin D1 regulates the progression of cells from the G1 phase to the S phase of the cell cycle. It is involved in complexes with cyclin-dependent kinases 4 and 6 (Cdk4/6) (16).

Complexes of cyclin D1 with CDKs 4 or 6 are essential to preserve cell homeostasis when impairments in cyclin D1 expression can trigger tumorigenesis. Moreover, changes in cyclin D1 expression can significantly affect cellular response to drug treatment (1719). Cyclin B1 and Cdk1 form a complex called ‘mitotic promoting factor’ (MPF) that is crucial for G2/M transition (20). Therefore, insufficient levels of cyclin B/Cdk1 complexes, e.g. as a consequence of the attenuation of cyclin B1 promoter by p53, are related to cell cycle arrest at G2 phase (21). Cyclin B1 and Cdk1 are expressed in late S and G2 phases of the mammalian cell cycle and are active at late G2 phase. After cyclin B1 is produced in the cytoplasm during S phase it is transported to the nucleus at the late G2 phase and then finally removed during anaphase via a ubiquitin-related pathway (22). The redistribution of cyclin B1/Cdk1 to the mitochondrial matrix shows a unique mechanism which coordinates the mitotic events in other cellular compartment and mitochondrial activity for G2/M progression. In spite of the fact than cyclin B1/Cdk1 is detected in the mitochondria of asynchronous cells in different cell cycle phases, the timing of mitochondrial influx of cyclin B1/Cdk1 is consistent with the accumulation of G2/M cells. What is important, the kinase activity of Cdk1 is also maximized at the G2/M which indicates that cyclin B1/Cdk1-mediated mitochondrial bioenergetics is integrated into the overall process of G2/M transition (23). There are reports suggesting that changes in the regulation of cyclin expression may be connected not only with unrestrained cell growth and malignant transformation, but also in tumor suppressor mechanisms (24).

The aim of the present study was to evaluate cyclin B1, cyclin D1 and cyclin K expression in H1299 cells treated with SFN as well as to investigate a potential involvement of these cyclins in the therapeutic outcome of SFN treatment.

Materials and methods

Cell culture and SFN treatment

The human non-small cell lung carcinoma cell line H1299 was purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were cultured in monolayers at 37°C in a humidified CO2 incubator (5% CO2) 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) and 50 µg/ml of gentamycin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany). Twenty-four hours after seeding, the cells were treated with SFN (Sigma-Aldrich; Merck KGaA) (5, 10 and 15 µM) for 48 h, and the following experimental procedures were performed.

MTT assay

Twenty-four hours prior to SFN treatment, the H1299 cells were seeded in 12-well plates. The cells were then treated with appropriate SFN concentrations (5, 10 and 15 µM) for 24 and 48 h. Following the treatment, cells were washed with phosphate-buffered saline (PBS) and 1 ml of DMEM without phenol red and 100 µl of the thiazolyl blue tetrazolium bromide (MTT) working solution (Sigma-Aldrich; Merck KGaA; 5 mg/ml in PBS) were added to each well and incubated for 3 h. Formazan crystals were diluted in isopropanol and the absorbance was read at 570 nm in a spectrophotometer (Spectra Academy, K-MAC, Daejeon, Korea).

Annexin V/propidium iodide (PI) binding assay

To assess the mode of cell death, the Alexa Fluor 488 Annexin V/Dead Cell Apoptosis kit (Invitrogen; Thermo Fisher Scientific, Inc.) was used according to the manufacturer's instructions. In short, after the SFN treatment, the cells were collected from 6-well plates using trypsin-EDTA solution, centrifuged at 300 × g for 8 min, resuspended in ABB (Annexin binding buffer) and incubated with Annexin V Alexa Fluor® 488 and propidium iodide (PI) at room temperature in the dark for 20 min. The cells were examined using a Guava 6HT-2L Cytometer (Merck KGaA). The data were analyzed by InCyte software (version 3.2; Merck KGaA) and expressed as the percentage of cells in each population (viable, Annexin V/PI; early apoptotic, Annexin V+/PI; late apoptotic, Annexin V+/PI+; necrotic, Annexin V/PI+).

DNA content analysis

For DNA content analysis, the Guava Cell Cycle reagent (Merck KGaA) was used according to the manufacturer's instructions. Briefly, the cells were harvested from 6-well plates by trypsinization, rinsed with PBS, fixed in ice-cold 70% ethanol at 4°C and maintained at −25°C overnight. The cells were then centrifuged at 650 × g for 5 min at room temperature (RT) and washed with PBS. After centrifugation at 500 × g for 7 min, the cells were resuspended in Guava Cell Cycle reagent. Following a 30-min incubation at RT in the dark, the cells were analyzed using Guava 6HT-2L cytometer (Merck KGaA), and the percentage of cells in each phase of the cell cycle was determined using InCyte software (version 4.03; De Novo Software, Piscataway, NJ, USA).

Flow cytometric analysis of cyclins B1, D1 and K

Cells grown in 6-well plates were harvested, washed with PBS, centrifuged (for 5 min, at 300 × g) and incubated with eBioscience Fixable Viability Dye eFluor 660 (Thermo Fisher Scientific, Inc.) for 30 min at 4°C to exclude dead cells. Then, the cells were fixed with Cytofix/Cytoperm solution (BD Biosciences, San Jose, CA, USA). After incubation on ice (for 15 min in the dark) and subsequent centrifugation (for 5 min, at 300 × g), the cells in pellets were permeabilized by the addition of 1 ml of ice-cold 80% (v/v) methanol (POCH) overnight at −20°C, washed with cold Perm/Wash solution (BD Biosciences) and resuspended in 3% bovine serum albumin (BSA; Sigma-Aldrich; Merck KGaA). For intracellular staining, cells were incubated with the appropriate antibody diluted in Perm/Wash solution. Following a 30-min incubation at room temperature (RT) in the dark and washing with Perm/Wash solution, the cells were centrifuged for 5 min, at 500 × g to wash off excess antibody. Cells stained with cyclin B1 and cyclin K antibody were incubated with Alexa Fluor 488 for 30 min at RT. Cells were resuspended in 300 µl of PBS for flow cytometric analysis on Guava 6HT-2L Cytometer. InCyte software was used to calculate the mean fluorescence intensity. The antibodies used in the experiment were the following: Mouse monoclonal anti-cyclin B1 antibody (dilution 1:100; cat. no. MA5-14319; Thermo Fisher Scientific, Inc.), FITC conjugated mouse cyclin D1 antibody (cat. no. 554109; BD Biosciences), or mouse monoclonal anti-cyclin K antibody (dilution 1:50; cat. no. sc-376371; Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA).

RT PCR analysis of cyclin B1, D1 and cyclin K expression

Total RNA from H1299 cells was isolated using a Total RNA kit (A&A Biotechnology, Gdynia, Poland) according to the manufacturer's instruction. The concentration and purity of RNA were determined spectrophotometrically (BioSpectrometer Basic; Eppendorf, Hamburg, Germany). The reverse transcription and quantitative PCR reactions were performed in a single 20-µl LightCycler capillary (Roche Applied Science, Mannheim, Germany) as a one-step real-time qRT-PCR with using LightCycler RNA Master SYBR-Green I (Roche Applied Science). For each target gene, the reactions were carried out in a 20-µl volume containing 100 ng of RNA and 0.2 µM of each primer in addition to LightCycler RNA Master SYBR-Green I kit components. The sequences of the primers were as follows: CCNK forward, 5′-ACCCAAAGGAGGAAGTAATGG-3′ and CCNK reverse, 5′-GAACTGGTATGGATGTTCTACCT-3′; CCND1 forward, 5′-TGAGGCGGTAGTAGGACAGG-3′ and CCND1 reverse, 5′-GACCTTCGTTGCCCTCTGT-3; CCNB1 forward, 5′-TTTCGCCTGAGCCTATTTTG-3′ and CCNB1 reverse, 5′-GCACATCCAGATGTTTCCATT-3′. The thermocycling conditions used for the qRT-PCR were as follows: One cycle of reverse transcription for 20 min at 61°C, one cycle of denaturation for 1 min at 95°C, and 45 cycles of denaturation for 5 sec at 95°C, followed by annealing and extension for 20 sec at 57–61°C (depending on the melting temperature of the primers) and 5 sec at 72°C, respectively. The samples were run in at least triplicate on the LightCycler 2.0 Instrument (Roche Applied Science) and evaluated with LightCycler Software (version 4.0; Roche Applied Science). The expression of the target gene was normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) internal control and assessed using the ΔΔCq method (2−ΔΔCq method) (25).

Cyclin B1, cyclin D1 and cyclin K immunofluorescence

H1299 cells growing on coverslips were briefly washed with PBS, fixed in 4% paraformaldehyde (for 15 min at RT) and then washed with PBS (3×5 min). After that, the cells were incubated in permeabilization solution (0.1% Triton X-100 in PBS) and blocked with 3% BSA. After permeabilization, the cells were incubated with mouse monoclonal anti-cyclin B1 antibody (dilution 1:100; cat. no. MA5-14319; Thermo Fisher Scientific, Inc.) or mouse monoclonal anti-cyclin D1 antibody (cat. no. C7464; Sigma-Aldrich; Merck KGaA) or mouse monoclonal anti-cyclin K antibody (dilution 1:50; cat. no. sc-376371; Santa Cruz Biotechnology, Inc.), respectively (60 min at RT), washed three times with PBS and incubated with Alexa Fluor 488 goat anti-mouse IgG (Invitrogen; Molecular Probes) (60 min, in the dark). Nuclear staining was performed with DAPI (Sigma-Aldrich; Merck KGaA). Contract labeling with Alexa Fluor 594 phalloidin (Thermo Fisher Scientific, Inc.) was used additionally. After incubation, the cells were washed with PBS and then mounted on microscope slides in Aqua-Poly/Mount (Polysciences, Inc., Warrington, PA, USA). The cells were examined using a C1 laser-scanning confocal microscopy system (Nikon, Tokyo, Japan) with a 100× oil immersion objective. Fluorescence images were obtained and analyzed with Nikon EZ-C1 software (ver3.80; Nikon Instruments, Melville, NY, USA).

Western blot assay

For semi-quantitative protein expression measurement, western blot analysis was conducted. Whole-cell lysates were obtained from lysis in RIPA buffer (Merck KGaA). Following normalization of the protein concentration using the BCA protein assay kit (Thermo Fisher Scientific, Inc.), equal amounts of protein (25 µg of total protein per lane) were separated using 4–12 or 16% NuPAGE Bis-Tris Gel (Novex/Life Technologies; Thermo Fisher Scientific, Inc.) and transferred onto nitrocellulose membranes using the iBlot dry western blotting system (Invitrogen; Thermo Fisher Scientific, Inc.). The membrane was processed in iBind Flex Western Blot system (Thermo Fisher Scientific, Inc.) following the manufacturer's protocol. Bands were detected using 1-Step™ Ultra TMB-Blotting solution (Thermo Fisher Scientific, Inc.).

Gene expression analysis (KM plotter)

Kaplan-Meier database (http://kmplot.com/analysis/) was conducted to evaluate the impact of CCNK, CDK12 and the CDK13 expression on the outcome of adenocarcinoma patients (26). The following probeset was used (ID Affymetrix): CCNK (219273_at), CDK12 (225690_at), CDK13 (228991_at).

Statistical analysis

The analysis was performed using statistical software (ver. 6.0; GraphPad Prism, San Diego, CA, USA). The data were compared with the non-parametric Mann-Whitney U test or non-parametric Kruskal-Wallis test with Dunn's multiple comparisons test, and the changes were considered statistically significant at the level of P<0.05. To analyze Kaplan-Meier survival curves, hazard ratio and log-rank test were calculated on the KM plotter web page.

Results

Annexin V and MTT assay

Following treatment with 5, 10 and 15 µM of SFN used in this study, the percentage of proliferating cells was decreased in a dose-dependent manner at both 24 and 48 h and the difference was significant at 10 and 15 µM of SFN (P<0.05) when compared with the control as determined by the MTT assay (Fig. 1A). In addition, as shown by the Annexin V/propidium iodide (PI) binding assay, the percentage of living cells was significantly decreased at 10 and 15 µM of SFN treatment (P<0.05). A dose-dependent increase in the percentage of apoptotic cells and a slight increase in the percentage of necrotic cells were observed and the differences were significant at 10 and 15 µM (Mann-Whitney U; P<0.05) (Fig. 1B). The results indicated that SFN induces both apoptosis and necrosis in a similar extent.

DNA content analysis

Flow cytometry was conducted to assess whether SFN treatment induced changes in cell cycle distribution in the H1299 cell line. Cell cycle analysis showed that along with the increase in the concentration of SFN, the percentage of cells arrested at the G2/M phase of the cell cycle was increased which was correlated with a decrease in cells in the G0/G1 phase. These results indicated that SFN suppressed cell cycle progression in the H1299 cell line (Fig. 2).

Expression of cyclin B1

Following SFN treatment, expression of cyclin B1 was decreased. qRT-PCR analysis showed that treatment with 5, 10 and 15 µM concentrations of SFN induced a significant decrease in cyclin B1 mRNA (Mann-Whitney U; P<0.05) (Fig. 3A), similarly to cytometric analysis which revealed a slight decrease at all doses of SFN (Fig. 4A). Immunofluorescent labeling of cyclin B1 in SFN-treated cells showed a trend similar to that noted in the flow cytometric and qRT-PCR results (Fig. 5). Western blot analysis showed a decrease in cyclin B1 expression (Fig. 6).

Expression of cyclin D1

The qRT-PCR experiment showed a significant increase in cyclin D1 mRNA after treatment with 10 µM SFN. Slightly increased levels resulted from the treatment with other doses, i.e. 5 and 15 µM SFN, when compared with the control (Fig. 3B). Flow cytometric measurements indicated that the fluorescence intensity of cyclin D1 protein was increased at all doses of SFN treatment with significant increases at 10 and 15 µM doses (Fig. 4B). Fluorescence microscopic examination of cyclin D1 in H1299 control and SFN-treated cells revealed the highest induction of this protein at 10 and 15 µM SFN (Fig. 7). Western blot analysis showed a slight decrease at the protein level following treatment with SFN, which suggests post-transcriptional inhibition of cyclin D1 (Fig. 6).

Expression of cyclin K

qRT-PCR analysis displayed that treatment of SFN at a 15 µM concentration induced a significant increase in cyclin K mRNA (Mann-Whitney U, P<0.05) (Fig. 3C). Flow cytometric measurements demonstrated that the fluorescence intensity of cyclin K protein was significantly higher comparing to the control cells (Fig. 4C). Immunofluorescent labeling of cyclin K in SFN-treated cells showed a trend similar to that noted in the qRT-PCR results (Fig. 8). Importantly, we observed downregulation of cyclin K after treatment with the lowest dose of SFN. However, to date, not enough data exist to explain this change in expression profile. We will try to investigate this phenomenon in the future and we hope that we can provide a more detailed mechanism which is responsible for changes in cyclin K expression after the treatment with a variety of drug and compounds. Additionally, due to the very low cyclin K expression in the H1299 cell line, we failed to perform western blot analysis.

Prognostic value of cyclin K, CDK12 and CDK13

Overall survival analysis of the patients with adenocarcinoma revealed that high expression of CDK12 and CDK13 but no cyclin K expression (CCNK) is associated with a worse prognosis (Fig. 9).

Discussion

Despite the significant advance in diagnosis and treatment, cancer remains a major cause of death worldwide. One of the deadliest cancers, which leads to a high death rate for both women and men, is lung cancer. With a 5-year survival rate below 20%, it is still a challenge for all those involved in cancer research (27). Phytochemicals are a potent source of anticancer agents, which can act unassisted or synergize with traditional drugs. Sulforaphane (SFN) is one of the most promising natural compound which exerts anticancer activities. This broccoli sprout-derived isothiocyanate acts through different pathways, which are not yet fully understood. Tumor cells are often characterized by imbalanced processes responsible for maintaining homeostasis, regulation of cell cycle and cell death. Uncontrolled divisions hold the danger of genetic impairments which directly lead to malignant transformation. In the present study, we demonstrated that SFN induces apoptosis, cell cycle arrest and changes in the expression of proliferation markers. What is important, the chosen doses can be attained by a normal consumption of broccoli. Approximately 50 grams of this vegetable delivers a sufficient amount of SFN to achieve a 20-µM concentration in urine (28). The ability of SFN to induce apoptosis has been demonstrated in several models in vivo and in vitro. In the highly metastatic melanoma cell line B16F-10 apoptosis was associated with morphological changes involving membrane blebbing and presence of apoptotic bodies. On the molecular level, activation of caspases-3 and −9, Bax and p53 with a simultaneous decrease in Bcl-2, caspase-8, Bid and NF-κB was observed (29). In U251MG glioblastoma cells, SFN induced apoptosis associated with expression of Bad, Bax and cytochrome c and decreased levels of survivin and Bcl-2. Moreover, cells treated with SFN were characterized by reduced invasion capabilities connected with increased expression of E-cadherin and lower expression of MMP-2, MMP-9 and galectin-3 (30). Mutations in the TP53 gene have been observed in most human cancers and p53 protein responses to low or constitutive stress through cell cycle arrest. When the stress drives to severe DNA damage, p53 triggers apoptosis. The H1299 cell line is characterized by p53 deficiency and only a few studies have shown details for the p53-independent response following SFN treatment. Ferreira de Oliveira et al (31) found that SFN induced DNA damage and increased the number of nuclear and mitotic abnormalities. In another study by Ferreira de Oliveira et al (32), the authors presented a model of SFN involvement in ROS generation and cytotoxicity in the p53 null osteosarcoma MG-63 cell line. SFN treatment was found to induce a significant increase in ROS generation and subsequent changes in mitochondrial membrane potential and eventually execution of apoptosis in a p53-independent manner. Higher concentrations of SFN were found to lead to inhibition of ROS-scavenging enzymes such as SOD, Cat and GPx and contribute to lowered glutathione regeneration and impaired antioxidative potential. Cancer cells have a lower ratio of oxidized glutathione to reduced glutathione which confers drug resistance (33,34). Targeting the glutathione system can be utilized to increase the effectiveness of chemotherapy. We showed that SFN alone has the potential to inhibit proliferation of the H1299 cell line, thus it is possible to use SFN as a sensitizer in the combinational therapy of tumors with dysfunctional p53 protein.

In the present study, the antiproliferative effect of SFN was manifested by G2/M phase arrest associated with downregulation of cyclin B1 which is a potent cell cycle regulator. Its overexpression is observed in many different cancer types, including lung, breast and gastric cancer (3537). Cyclin B1 silencing can exert antiproliferative effects on cancer cells, thus cyclin B1 can be considered as a potential therapeutic target. Kedinger et al downregulated cyclin B1 expression using modified siRNAs and achieved a reduction in the growth of melanoma xenografts and inhibition of formation and dissemination of melanoma lung metastases (38). In cervical cancer, G2/M arrest induced by SFN was also associated with downregulation of cyclin B1. Moreover, the cellular response involved upregulation of GADD45β, known as cyclin B1/Cdk1 inhibitor. The results suggest that SFN has the potential to inhibit cancer growth via the cyclin B/GADD45β pathway (39). It is noteworthy that downregulation of cyclin B1 is not unequivocally beneficial for cancer patients. In colorectal cancer patients, negative or low cyclin B1 staining was inversely correlated with lymph node metastatic potential. Tumors with low cyclin B1 expression more frequently present with lymphatic permeation or vessel invasion (40). Fang et al showed that cyclin B inhibits lymph node metastasis via E-cadherin both in vivo and in vitro. Cyclin B1 silencing resulted in decreased E-cadherin expression, which is one of the molecular hallmarks associated with EMT. Downregulation of cyclin B1 suppressed p53+/+ HCT116 and p53−/− HCT116 colorectal cancer cell lines. The same significant effect was observed in the SW480 cell line and in a xenograft model (41). Correspondingly with a previously described model, Wang et al showed that SFN suppressed EMT and metastasis in human lung cancer cell lines through miR-616-5p-mediated GSK3β/β-catenin signaling pathways. SFN treatment was found to alter the expression of EMT-related proteins such as β-catenin, N-cadherin, vimentin and E-cadherin (42). These data show that the impact of cyclin B1 depends on the cellular environment and it is important to determine the factors which promote the antimetastatic effect of cyclin B1 and when cyclin B1 enhances EMT events.

Cyclin D1 is frequently overexpressed in many cancer types. After treatment with SFN, we observe an increase in cyclin D1 mRNA and protein expression. It is possible that cyclin D1 overexpression in the H1299 cell line is responsible for moderate response to drug intervention. Our previous study showed that cyclin D1 overexpression in the A549 cell line prevented SFN-induced apoptosis (43). Moreover, we also observed an increase in cyclin D1 after treatment with the lowest dose of SFN (30 µM). It is possible that at lower doses, SFN inhibits cyclin D1 association with cyclin-dependent kinases and this is the cause of the increase in cyclin fluorescence. Hagemann et al showed that SFN can upregulate MDM-2 protein expression which is a p53 suppressor but also can act independently of p53. MDM-2 as an oncoprotein promotes the survival of cancer cells and contributes to drug resistance. The protective effect of SFN can be blocked with MDM-2 inhibitor, Nutlin-3 (44). Moreover, silencing of cyclin D1 enhances the cytostatic effect of Nutlin-3 and makes different cancer cell lines more susceptible to cell death (45). It is feasible that the axis cyclin D1-MDM-2 significantly contributes to the cellular response to SFN treatment. Treating the H1299 cell line with another cytostatic drug, actinomycin D induced an increase in MDM2 expression. Silencing of MDM2 sensitized cells to actinomycin D and increase the number of cells undergoing apoptosis (46). The interaction between cyclin D1 and MDM-2 in the H1299 cell line warrants further investigations.

Cyclin-dependent kinases 12 and 13 with its regulatory subunit cyclin K, take part in gene transcription regulation through interaction with RNA polymerase II and phosphorylation of C-terminal domain which is an important step in the generation of mature mRNA (47,48). The activity and oncogenic status of cyclin K/Cdk12 and cyclin K/Cdk13 warrant further elucidation. To investigate how the expression of CDK12 and CDK13 mRNA affects adenocarcinoma and the squamous cell carcinoma outcome we employed ‘The Kaplan-Meier plotter’ (KM plotter) database and discovered that high expression of Cdk12 and Cdk13 mRNA is correlated with worse overall survival. To date, limited and inconsistent data have been published, showing both a suppressive and a promoting effect on different cancer types. The loss of function mutations in high-grade serous ovarian cancer probably result in deregulated expression of DDR genes and contribute to genomic instability (49,50). Moreover, patients bearing mutations in the CCNK gene also present with abnormalities in BRCA1 and BRCA2 genes, well-known cancer suppressors (51). Amplification of the CDK12 mRNA increased protein levels, and elevated phosphorylation was observed in HER-2 driven breast cancer samples (52). Involvement in DNA repair leads to believe that targeting of Cdk12/cyclin K complex can significantly alter the drug response and can be considered as a new strategy in therapeutic development. The cyclin K gene was found to sensitize cancer cells to camptothecin which is a topoisomerase inhibitor inducing DNA damage (53). The loss of function mutations in the CDK12 gene leads to increased cisplatin sensitivity and downregulation of CDK12 induces spontaneous cell death and decreases resistance to different DNA-damaging agents such as etoposide, mitomycin C and cisplatin (14,54).

In turn, the impact of CDK13 activity on cancer development and disease outcome is still an uncharted territory. The CKD13 gene is frequently amplified in many different cancer cell lines. Clonogenic and invasion assays on the NIH3T3 cell line revealed the high oncogenic potential of the CDK13 product and showed that stable expression of CDK13 resulted in the stronger colony forming ability and an intermediate degree of migration activity. Additionally, cells overexpressing CDK13 exhibited increased resistance to 5-fluorouracil and doxorubicin and a simultaneous increase in tamoxifen susceptibility (55). These data suggest a significant role of CDK12 and CDK13 in human cancer and response to a treatment. The role of cyclin K in cancer development is unclear. Cyclin K plays important roles in transcriptional regulation and cell cycle control, but we know little about how cyclin K expression affects drug response. Schecher et al demonstrated the potential role of cyclin K in prostate cancer. Depletion of cyclin K in prostate cancer cell lines led to an increase in the number of multinucleated cells, mitotic catastrophe and eventually reduction of proliferation or apoptotic cell death. Moreover, prostate cancer patients with high expression of cyclin K, treated with an adjuvant therapy had worse biochemical recurrence-free survival rates compared to patients with low cyclin K expression. Silencing of cyclin K resulted in a decrease in Aurora B mRNA and protein expression. Aurora B is crucial for the formation of chromosomes and spindle assembly and its depletion is probably caused by the reduction of CDK12 activity rather than cyclin K itself (56). In this study, the increase in cyclin K expression could be a part of drug response. SFN induces DNA damage and activates various genes associated with DNA repair. The results suggest that high expression of cyclin K and CDK12 and CDK13 kinases should be considered as a potential obstacle in the successful therapy of tumors. It was shown that inhibitors of Cdk12 and Cdk13 can exert an antiproliferative effect on cancer cells thus it is reasonable to conduct further studies to investigate CDK12, CDK13 and cyclin K as targets for cancer treatment (57,58).

In conclusion, this study revealed that SFN induced cell cycle arrest and apoptosis in the H1299 cell line. Cell cycle arrest was associated with downregulation of cyclin B1. Significant changes in the expression of cyclin D1 and K suggest the role of these proteins in response to SFN treatment. Moreover, our in silico analysis showed that upregulated mRNA expression of the cyclin K catalytic partners CDK12 and CDK13 is associated with lower overall survival rates in adenocarcinoma patients. The present study shows the urgent need for elucidating the role of cyclin K/CDK12/CDK13 complexes in development and progression of lung cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Nicolaus Copernicus University in Toruń, Collegium Medicum, Faculty of Medicine (grant no. 114).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

AŻ conceived and designed the study. AK and AŻ were responsible for the DNA content analysis and the DNA content analysis for cytometric analysis, the immunofluorescent labeling and the western blot analysis, they jointly designed the figures and they wrote the study. AK was responsible for the Annexin V and MTT assay and for the prognostic value of cyclin K, CDK12 and CDK13. AKW was responsible for the qRT-PCR analysis. DG and AG performed the statistical analysis; AZ, AK, AG, AKW and DG reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All experiments have been approved by the Bioethics Committee of the Nicolaus Copernicus University in Toruń functioning at Collegium Medicum in Bydgoszcz.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Clarke JD, Dashwood RH and Ho E: Multi-targeted prevention of cancer by sulforaphane. Cancer Lett. 269:291–304. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Barcelo S, Gardiner JM, Gescher A and Chipman JK: CYP2E1-mediated mechanism of anti-genotoxicity of the broccoli constituent sulforaphane. Carcinogenesis. 17:277–282. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Langouët S, Furge LL, Kerriguy N, Nakamura K, Guillouzo A and Guengerich FP: Inhibition of human cytochrome P450 enzymes by 1,2-dithiole-3-thione, oltipraz and its derivatives, and sulforaphane. Chem Res Toxicol. 13:245–252. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Heiss E, Herhaus C, Klimo K, Bartsch H and Gerhäuser C: Nuclear factor kappa B is a molecular target for sulforaphane-mediated anti-inflammatory mechanisms. J Biol Chem. 276:32008–32015. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Parnaud G, Li P, Cassar G, Rouimi P, Tulliez J, Combaret L and Gamet-Payrastre L: Mechanism of sulforaphane-induced cell cycle arrest and apoptosis in human colon cancer cells. Nutr Cancer. 48:198–206. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Singh SV, Herman-Antosiewicz A, Singh AV, Lew KL, Srivastava SK, Kamath R, Brown KD, Zhang L and Baskaran R: Sulforaphane-induced G2/M phase cell cycle arrest involves checkpoint kinase 2-mediated phosphorylation of cell division cycle 25C. J Biol Chem. 279:25813–25822. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Herman-Antosiewicz A, Johnson DE and Singh SV: Sulforaphane causes autophagy to inhibit release of cytochrome c and apoptosis in human prostate cancer cells. Cancer Res. 66:5828–5835. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Choi S and Singh SV: Bax and Bak Are required for apoptosis induction by sulforaphane, a cruciferous vegetable-derived cancer chemopreventive agent. Cancer Res. 65:2035–2043. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Shankar S, Ganapathy S and Srivastava RK: Sulforaphane enhances the therapeutic potential of TRAIL in prostate cancer orthotopic model through regulation of apoptosis, metastasis, and angiogenesis. Clin Cancer Res. 14:6855–6866. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Moon DO, Kim MO, Kang SH, Choi YH and Kim GY: Sulforaphane suppresses TNF-alpha-mediated activation of NF-kappaB and induces apoptosis through activation of reactive oxygen species-dependent caspase-3. Cancer Lett. 274:132–142. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Edwards MC, Wong C and Elledge SJ: Human cyclin K, a novel RNA polymerase II-associated cyclin possessing both carboxy-terminal domain kinase and Cdk-activating kinase activity. Mol Cell Biol. 18:4291–4300. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Fu TJ, Peng J, Lee G, Price DH and Flores O: Cyclin K functions as a CDK9 regulatory subunit and participates in RNA polymerase II transcription. J Biol Chem. 274:34527–34530. 1999. View Article : Google Scholar : PubMed/NCBI

13 

Napolitano G, Majello B, Licciardo P, Giordano A and Lania L: Transcriptional activity of positive transcription elongation factor b kinase in vivo requires the C-terminal domain of RNA polymerase II. Gene. 254:139–145. 2000. View Article : Google Scholar : PubMed/NCBI

14 

Blazek D, Kohoutek J, Bartholomeeusen K, Johansen E, Hulinkova P, Luo Z, Cimermancic P, Ule J and Peterlin BM: The Cyclin K/Cdk12 complex maintains genomic stability via regulation of expression of DNA damage response genes. Genes Dev. 25:2158–2172. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Zhou Z and Fu XD: Regulation of splicing by SR proteins and SR protein-specific kinases. Chromosoma. 122:191–207. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Baldin V, Lukas J, Marcote MJ, Pagano M and Draetta G: Cyclin D1 is a nuclear protein required for cell cycle progression in G1. Genes Dev. 7:812–821. 1993. View Article : Google Scholar : PubMed/NCBI

17 

Akiyama N, Sasaki H, Katoh O, Sato T, Hirai H, Yazaki Y, Sugimura T and Terada M: Increment of the cyclin D1 mRNA level in TPA-treated three human myeloid leukemia cell lines: HEL, CMK and HL-60 cells. Biochem Biophys Res Commun. 195:1041–1049. 1993. View Article : Google Scholar : PubMed/NCBI

18 

Tiemann K, Alluin JV, Honegger A, Chomchan P, Gaur S, Yun Y, Forman SJ, Rossi JJ and Chen RW: Small interfering RNAs targeting cyclin D1 and cyclin D2 enhance the cytotoxicity of chemotherapeutic agents in mantle cell lymphoma cell lines. Leuk Lymphoma. 52:2148–2154. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Danos AM, Liao Y, Li X and Du W: Functional inactivation of Rb sensitizes cancer cells to TSC2 inactivation induced cell death. Cancer Lett. 328:36–43. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Pines J and Hunter T: Human cyclin A is adenovirus E1A-associated protein p60 and behaves differently from cyclin B. Nature. 346:760–763. 1990. View Article : Google Scholar : PubMed/NCBI

21 

Innocente SA, Abrahamson JL, Cogswell JP and Lee JM: p53 regulates a G2 checkpoint through cyclin B1. Proc Natl Acad Sci USA. 96:2147–2152. 1999. View Article : Google Scholar : PubMed/NCBI

22 

Holloway SL, Glotzer M, King RW and Murray AW: Anaphase is initiated by proteolysis rather than by the inactivation of maturation-promoting factor. Cell. 73:1393–1402. 1993. View Article : Google Scholar : PubMed/NCBI

23 

Wang Z, Fan M, Candas D, Zhang TQ, Qin L, Eldridge A, Wachsmann-Hogiu S, Ahmed KM, Chromy BA, Nantajit D, et al: Cyclin B1/Cdk1 coordinates mitochondrial respiration for cell-cycle G2/M progression. Dev Cell. 29:217–232. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Sugrue MM, Shin DY, Lee SW and Aaronson SA: Wild-type p53 triggers a rapid senescence program in human tumor cells lacking functional p53. Proc Natl Acad Sci USA. 94:9648–9653. 1997. View Article : Google Scholar : PubMed/NCBI

25 

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

26 

Lánczky A, Nagy Á, Bottai G, Munkácsy G, Szabó A, Santarpia L and Győrffy B: miRpower: A web-tool to validate survival-associated miRNAs utilizing expression data from 2178 breast cancer patients. Breast Cancer Res Treat. 160:439–446. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Zhang Y and Callaway EC: High cellular accumulation of sulphoraphane, a dietary anticarcinogen, is followed by rapid transporter-mediated export as a glutathione conjugate. Biochem J. 364:301–307. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Hamsa TP, Thejass P and Kuttan G: Induction of apoptosis by sulforaphane in highly metastatic B16F-10 melanoma cells. Drug Chem Toxicol. 34:332–340. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Zhang Z, Li C, Shang L, Zhang Y, Zou R, Zhan Y and Bi B: Sulforaphane induces apoptosis and inhibits invasion in U251MG glioblastoma cells. Springerplus. 5:2352016. View Article : Google Scholar : PubMed/NCBI

31 

Ferreira de Oliveira JM, Remédios C, Oliveira H, Pinto P, Pinho F, Pinho S, Costa M and Santos C: Sulforaphane induces DNA damage and mitotic abnormalities in human osteosarcoma MG-63 cells: Correlation with cell cycle arrest and apoptosis. Nutr Cancer. 66:325–334. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Ferreira de Oliveira JM, Costa M, Pedrosa T, Pinto P, Remédios C, Oliveira H, Pimentel F, Almeida L and Santos C: Sulforaphane induces oxidative stress and death by p53-independent mechanism: Implication of impaired glutathione recycling. PLoS One. 9:e929802014. View Article : Google Scholar : PubMed/NCBI

33 

Chen HH and Kuo MT: Role of glutathione in the regulation of cisplatin resistance in cancer chemotherapy. Met Based Drugs. 2010(pii): 4309392010.PubMed/NCBI

34 

Backos DS, Franklin CC and Reigan P: The role of glutathione in brain tumor drug resistance. Biochem Pharmacol. 83:1005–1012. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Cooper WA, Kohonen-Corish MR, McCaughan B, Kennedy C, Sutherland RL and Lee CS: Expression and prognostic significance of cyclin B1 and cyclin A in non-small cell lung cancer. Histopathology. 55:28–36. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Aaltonen K, Amini RM, Heikkilä P, Aittomäki K, Tamminen A, Nevanlinna H and Blomqvist C: High cyclin B1 expression is associated with poor survival in breast cancer. Br J Cancer. 100:1055–1060. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Begnami MD, Fregnani JH, Nonogaki S and Soares FA: Evaluation of cell cycle protein expression in gastric cancer: Cyclin B1 expression and its prognostic implication. Hum Pathol. 41:1120–1127. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Kedinger V, Meulle A, Zounib O, Bonnet ME, Gossart JB, Benoit E, Messmer M, Shankaranarayanan P, Behr JP, Erbacher P, et al: Sticky siRNAs targeting survivin and cyclin B1 exert an antitumoral effect on melanoma subcutaneous xenografts and lung metastases. BMC Cancer. 13:3382013. View Article : Google Scholar : PubMed/NCBI

39 

Cheng YM, Tsai CC and Hsu YC: Sulforaphane, a dietary isothiocyanate, induces G2/M arrest in cervical cancer cells through cyclinB1 downregulation and GADD45β/CDC2 association. Int J Mol Sci. 17(pii): E15302016. View Article : Google Scholar : PubMed/NCBI

40 

Korenaga D, Takesue F, Yasuda M, Honda M, Nozoe T and Inutsuka S: The relationship between cyclin B1 overexpression and lymph node metastasis in human colorectal cancer. Surgery. 131 Suppl 1:S114–S120. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Fang Y, Liang X, Jiang W, Li J, Xu J and Cai X: Cyclin B1 suppresses colorectal cancer invasion and metastasis by regulating E-cadherin. PLoS One. 10:e01268752015. View Article : Google Scholar : PubMed/NCBI

42 

Wang DX, Zou YJ, Zhuang XB, Chen SX, Lin Y, Li WL, Lin JJ and Lin ZQ: Sulforaphane suppresses EMT and metastasis in human lung cancer through miR-616-5p-mediated GSK3β/β-catenin signaling pathways. Acta Pharmacol Sin. 38:241–251. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Żuryń A, Litwiniec A, Safiejko-Mroczka B, Klimaszewska-Wiśniewska A, Gagat M, Krajewski A, Gackowska L and Grzanka D: The effect of sulforaphane on the cell cycle, apoptosis and expression of cyclin D1 and p21 in the A549 non-small cell lung cancer cell line. Int J Oncol. 48:2521–2533. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Hagemann JH, Thomasova D, Mulay SR and Anders HJ: Nrf2 signalling promotes ex vivo tubular epithelial cell survival and regeneration via murine double minute (MDM)-2. Nephrol Dial Transplant. 28:2028–2037. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Yang P, Chen W, Li X, Eilers G, He Q, Liu L, Wu Y, Wu Y, Yu W, Fletcher JA and Ou WB: Downregulation of cyclin D1 sensitizes cancer cells to MDM2 antagonist Nutlin-3. Oncotarget. 7:32652–32663. 2016.PubMed/NCBI

46 

Li L, Cui D, Zheng SJ, Lou H and Tang J: Regulation of Actinomycin D induced upregulation of Mdm2 in H1299 cells. DNA Repair. 11:112–119. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Sansó M and Fisher RP: Pause, play, repeat: CDKs push RNAP II's buttons. Transcription. 4:146–152. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Bartkowiak B, Liu P, Phatnani HP, Fuda NJ, Cooper JJ, Price DH, Adelman K, Lis JT and Greenleaf AL: CDK12 is a transcription elongation-associated CTD kinase, the metazoan ortholog of yeast Ctk1. Genes Dev. 24:2303–2316. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Bell D, Berchuck A, Birrer M, Chien J, Cramer D, Dao F, Dhir R, DiSaia P, Gabra H, Glenn P, et al: Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Ekumi KM, Paculova H, Lenasi T, Pospichalova V, Bösken CA, Rybarikova J, Bryja V, Geyer M, Blazek D and Barboric M: Ovarian carcinoma CDK12 mutations misregulate expression of DNA repair genes via deficient formation and function of the Cdk12/CycK complex. Nucleic Acids Res. 43:2575–2589. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Carter SL, Cibulskis K, Helman E, McKenna A, Shen H, Zack T, Laird PW, Onofrio RC, Winckler W, Weir BA, et al: Absolute quantification of somatic DNA alterations in human cancer. Nat Biotechnol. 30:413–421. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Mertins P, Mani DR, Ruggles KV, Gillette MA, Clauser KR, Wang P, Wang X, Qiao JW, Cao S, Petralia F, et al: Proteogenomics connects somatic mutations to signalling in breast cancer. Nature. 534:55–62. 2016. View Article : Google Scholar : PubMed/NCBI

53 

O'Connell BC, Adamson B, Lydeard JR, Sowa ME, Ciccia A, Bredemeyer AL, Schlabach M, Gygi SP, Elledge SJ and Harper JW: A genome-wide camptothecin sensitivity screen identifies a mammalian MMS22L-NFKBIL2 complex required for genomic stability. Mol Cell. 40:645–657. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Joshi PM, Sutor SL, Huntoon CJ and Karnitz LM: Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly(ADP-ribose) polymerase inhibitors. J Biol Chem. 289:9247–9253. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Kim HE, Kim DG, Lee KJ, Son JG, Song MY, Park YM, Kim JJ, Cho SW, Chi SG, Cheong HS, et al: Frequent amplification of CENPF, GMNN and CDK13 genes in hepatocellular carcinomas. PLoS One. 7:e432232012. View Article : Google Scholar : PubMed/NCBI

56 

Schecher S, Walter B, Falkenstein M, Macher-Goeppinger S, Stenzel P, Krümpelmann K, Hadaschik B, Perner S, Kristiansen G, Duensing S, et al: Cyclin K dependent regulation of Aurora B affects apoptosis and proliferation by induction of mitotic catastrophe in prostate cancer: Cyclin K in prostate cancer. Int J Cancer. 141:1643–1653. 2017. View Article : Google Scholar : PubMed/NCBI

57 

Johannes JW, Denz CR, Su N, Wu A, Impastato AC, Mlynarski S, Varnes JG, Prince DB, Cidado J, Gao N, et al: Structure-based design of selective noncovalent CDK12 inhibitors. ChemMedChem. 13:231–235. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Zhang T, Kwiatkowski N, Olson CM, Dixon-Clarke SE, Abraham BJ, Greifenberg AK, Ficarro SB, Elkins JM, Liang Y, Hannett NM, et al: Covalent targeting of remote cysteine residues to develop CDK12 and CDK13 inhibitors. Nat Chem Biol. 12:876–884. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2019
Volume 41 Issue 2

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Żuryń A, Krajewski A, Klimaszewska‑Wiśniewska A, Grzanka A and Grzanka D: Expression of cyclin B1, D1 and K in non‑small cell lung cancer H1299 cells following treatment with sulforaphane. Oncol Rep 41: 1313-1323, 2019
APA
Żuryń, A., Krajewski, A., Klimaszewska‑Wiśniewska, A., Grzanka, A., & Grzanka, D. (2019). Expression of cyclin B1, D1 and K in non‑small cell lung cancer H1299 cells following treatment with sulforaphane. Oncology Reports, 41, 1313-1323. https://doi.org/10.3892/or.2018.6919
MLA
Żuryń, A., Krajewski, A., Klimaszewska‑Wiśniewska, A., Grzanka, A., Grzanka, D."Expression of cyclin B1, D1 and K in non‑small cell lung cancer H1299 cells following treatment with sulforaphane". Oncology Reports 41.2 (2019): 1313-1323.
Chicago
Żuryń, A., Krajewski, A., Klimaszewska‑Wiśniewska, A., Grzanka, A., Grzanka, D."Expression of cyclin B1, D1 and K in non‑small cell lung cancer H1299 cells following treatment with sulforaphane". Oncology Reports 41, no. 2 (2019): 1313-1323. https://doi.org/10.3892/or.2018.6919