Open Access

CDK4 has the ability to regulate Aurora B and Cenpp expression in mouse keratinocytes

  • Authors:
    • Sung Hyun Lee
    • Liliana R.l. Rodriguez
    • Rima Majumdar
    • Paula L. Miliani De Marval
    • Marcelo L. Rodriguez‑Puebla
  • View Affiliations

  • Published online on: August 11, 2021     https://doi.org/10.3892/ol.2021.12993
  • Article Number: 732
  • Copyright: © Lee 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 4 (CDK4) is a critical molecule that regulates key aspects of cell proliferation through phosphorylation of the retinoblastoma (Rb) family of proteins. In the last few years, it has been suggested that CDK4 plays alternative roles in cell proliferation and tumorigenesis. The main aim of the present study was to define a novel CDK4 function as a transcriptional regulator of genes involved in chromosome segregation, contributing to the G2/M phase transition. Herein, chromatin‑immunoprecipitation reverse transcription‑quantitative PCR assays were performed to demonstrate that CDK4 could occupy the promoter region of genes associated with chromosomal segregation, such as Aurora‑B (Aurkb) and Centromere Protein P (CENP‑P). Moreover, gain‑ and loss‑of‑function experiments showed that CDK4 participated in the transcriptional regulation of Aurkb and CENP‑P. The finding that Aurkb may have a crucial role in chromosome bi‑orientation and the spindle assembly checkpoint, and that CENP‑P could be required for proper kinetochore function suggests that dysregulation of CDK4 expression induces chromosomal instability and, in some cases, cancer development.

Introduction

Progress from G0 to G1 and through the G1 phase of the mammalian cell cycle is mediated by the cyclin-dependent kinases 4 and 6 (CDK4, CDK6), which are activated through binding with their regulatory subunits D-type cyclins (D1, D2, and D3) (14). It is widely accepted that CDK4 regulates critical aspects of the cell cycle via phosphorylation of the retinoblastoma (Rb) family of proteins (5,6). Thus, the so-called CDK4/6-Rb axis is considered essential to cell-cycle entry and progression. Although this canonical role of CDK4 as a driver of cell proliferation has been firmly established, research carried out over the last few years has suggested cell cycle-independent functions of CDKs and D-type cyclins (7,8). For example, a novel role for CDK6 in hematopoietic cells that exceeds its function as a cell-cycle regulator has been recognized (9). Increasing evidence suggests that cyclin D1 carries out essential functions in other processes such as transcription and DNA damage (7,10,11).

Interestingly, a systematic screen has defined other potential substrates for CDK4, including the transcription factor forkhead box protein M1 (FOXM1) (12). Our skin carcinogenesis studies also suggested that CDK4 plays additional roles unrelated to its canonical function in the CDK-Rb axis. We reported that transgenic expression of CDK4 in mouse epidermis favors the malignant progression of skin tumors (13). However, forced expression of the other G1-CDKs, such as CDK6 and CDK2, in mouse keratinocytes resulted in elevated Rb phosphorylation but did not induce malignant progression such as observed in CDK4 transgenic mice (14,15). Moreover, a putative role of CDK4 in chromosome instability, and consequently in malignant progression, was reported by Adon et al, in which the absence of CDK4 or CDK2 prevents centrosome amplification (16).

Herein we report a novel function of CDK4 regulating the transcriptional expression of genes involved in chromosome segregation. Chromatin-immunoprecipitation (ChIP) analysis shows that CDK4 occupies the promoter of genes associated with chromosomal segregation, such as Aurkb (Aurora-B) and CENP-P (Centromere Protein P). Gain- and loss-of-function experiments showed that CDK4 participates in the transcriptional regulation of AurkB and CENP-P promoters. Importantly, Aurora-B is a subunit of the chromosome passenger complex controlling several aspects of chromosome segregation (17). Thus, deregulation of Aurora-B through CDK4 expression would result in a malfunction of the chromosome segregation events and potentially tumorigenesis (18). Our results suggest that CDK4 may contribute to G2/M regulation in addition to the prominent role in G0/G1- and G1/S-transitions. Aurora-B expression peaks during mitosis have a crucial role in chromosome bi-orientation and the spindle-assembly checkpoint, whereas CENP-P is required for proper kinetochore function (19,20), suggests that CDK4 plays a pivotal role in maintaining chromosomal stability.

Materials and methods

Cell lines and primary mouse keratinocytes

The 308-cell line was acquired through a previous research collaboration with Dr. Claudio Conti (MD Anderson Cancer Center, Texas). This immortalized cell line was derived from calcium-resistant foci of keratinocytes from adult Balb/c mouse initiated by 7,12-dimethylbenz[a]anthracene and has been extensively used as a model of cell proliferative (2123). NIH3T3 murine embryo fibroblasts cell line was obtained from the American Type Culture Collection (Catalog number CRL-1658; ATCC). Primary keratinocytes were isolated from newborn mice and cultured in a low Ca2+ medium (EMEM, 06-174 G; Cambrex-bioz) as described previously (24). Briefly, four newborns of 48 h of age were washed with ethanol and iodine solution and put in the refrigerator (4°C) for 30 min to induce hypothermic anesthesia. After anesthesia by refrigeration, the newborns were euthanized by decapitation, and skin was removed with forceps, rinsed, and continue with the cell culture process (24,25). The generation of mouse primary keratinocytes and protocols for animal use were approved for the North Carolina State University Institutional Animal Care and Use Committee (IACUC) protocol number 18-102-B, as required by federal regulations.

Cell extraction and immunofluorescence analysis

To visualize chromatin-bound proteins, unbound nuclear and cytosolic proteins from keratinocyte cell line 308 and primary culture of mouse keratinocytes were removed with cytoskeletal extraction buffer (CSK buffer). 70–80% confluent 308 cells were grown on coverslips coated with poly-L-lysine (Sigma-Aldrich; Merck KGaA). For CSK extraction, cells were washed with DPBS (Mediatech Inc.) twice and incubated with CSK buffer [100 mM NaCl, 300 mM sucrose, 3 mM MgCl2, 10 mM PIPES (pH.8), and 0.5% Triton X-100] containing 1 × protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA) for 2 min on ice. After extraction, cells were fixed in 10% formalin for 20 min at RT. For DNase extraction, cells permeabilized with CSK buffer were washed with DPBS twice and incubated with 10 units of Optizyme Recombinant DNase I (Thermo Fisher Scientific Inc.) 30 min at 37°C followed by fixation with 10% formalin.

Immunofluorescence staining cells were blocked with 10% goat serum diluted in 0.01% Triton X-100/PBS solution for 30 min at RT, incubated with primary antibodies against CDK4 (C-22), and HDAC1 (10E2) (Santa Cruz Biotechnology, Inc.) at 4°C overnight. Cells were washed with PBS four times and incubated with goat anti-rabbit-FITC conjugated antibody for CDK4 staining (Thermo Fisher Scientific Inc.; Pierce Biotechnology Inc.), and goat anti-mouse-Alexa Fluor 488 (Thermo Fisher Scientific Inc.; Molecular Probes) for HDAC1 staining. DAPI staining was utilized for counterstained (Vector Laboratories Inc.). Cells were examined under a Nikon Eclipse E400 fluorescence microscope (Nikon Corporation), and images were collected with Qcapture software (QImaging).

Retroviral infection and generation of stable cell lines

Murine CDK4 cDNA was subcloned into the pLPCX retroviral vector (Clontech Laboratories, Inc.) using primers containing NotI restriction sites. pLPCX-Cdk4 and pLPCX (empty plasmid) vectors were amplified in DH5α competent E. coli cells (Invitrogen; Thermo Fisher Scientific Inc.). The retroviral vector was transfected into the Platinum Retroviral Packaging Cell line (Plat-E cell) with psPAX2 packing vector (Addgene) using FuGENE® 6 Transfection Reagent (Promega Corp.). After 48 h of transfection, the virus-containing medium was filtered through a 0.45 µm of syringe filter (Corning Inc.). Harvested CDK4-retroviruses and control-retrovirus were utilized to infected NIH3T3 and 308 cells with 4 µg/ml of hexadimethrine bromide (polybrene; Sigma-Aldrich;) and incubated overnight. The pLPCX-Cdk4 and pLPCX-empty retrovirus infected cells (NIH3T3-CDK4 and 308-CDK4) were selected with 2 µg/ml of puromycin (Sigma-Aldrich Co. LLC, MO).

Western blot assays

NIH3T3 and 308 cell lines were lysed in RIPA buffer [150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, 0.1% SDS, 50 mM Tris-HCl (pH 8.0)] containing 1× protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA). Protein concentration was determined with the DC™ Protein Assay system (Bio-Rad Laboratories), 50 µg of protein were loaded on 10% SDS-PAGE gel and electrophoretically transferred onto nitrocellulose membranes. After being blocked with 5% nonfat powdered milk in Dulbecco phosphate-buffered saline, the membranes were incubated with 1 µg/ml of specific antibodies. The following antibodies were used: Polyclonal antibodies against CDK4 (C-22), β-actin (I-19) (Santa Cruz Biotechnology, Inc.), CENP-P (PA5-31186), and Aurora-B (MA5-17226) (Thermo Fisher Scientific Inc.; Pierce Biotechnology Inc.). Membranes were washed and incubated with goat anti-rabbit-HRP (sc-2004), donkey anti-goat-HRP, or goat anti-mouse-HRP secondary antibodies followed by enhanced chemiluminescence (ECL detection kit; GE Healthcare). Western blot bands were quantified using UN-SCAN-IT gel version 6.1 software. The experimental data are representative of three independent repetitions.

Chromatin immunoprecipitation (ChIP) assay

We utilized the SimpleChIP® Enzymatic chromatin IP kit (Cell Signaling Technology, Inc., MA) following the manufacturer's instructions. Briefly, 4×107 NIH3T3 semi-confluent cells were used for ChIP assay. Cell culture media was replaced with 10 ml of fresh media containing 1% formaldehyde to crosslink proteins/DNA and incubated for 10 min at RT. The reaction was stopped by the addition of glycine, a 0.125 M concentration in cell culture media. Cells were washed with PBS, scraped, collected into conical tubes, and centrifuged at 1,500 rpm. Chromatin was released by adding lysis buffer containing DTT, protease inhibitors, and PMSF and further fragmented by partial digestion with micrococcal nuclease to obtain chromatin fragments of 1 to 5 nucleosomes in size. Nuclei were pelleted by centrifugation at 13,000 rpm and resuspended in 1X ChIP buffer containing SDS, protease inhibitor, and PMSF. Pellets were sonicated (3 sets of 20-second pulses at setting 2 on a Branson sonifer 450) and clarified by centrifugation at 10,000 rpm for 10 min. The supernatant was kept at −80°C. 50 µl of chromatin sample were digested with 2 µl RNase A at 37°C for 30 min and 2 µl Proteinase K at 65°C for 2 h. DNAs were purified using spin columns, and 10 µl was used to electrophoresis on 1% agarose gel to check DNA fragment sizes. DNA concentration was determined with a NanoDrop 1000 (Thermo Fisher Scientific, Inc.). 10 µg chromatin was diluted in 500 µl of 1X ChIP buffer containing protease inhibitor cocktail and used for each immunoprecipitation. 2 µg of rabbit antibodies against CDK4 (C-22) (Santa Cruz Biotechnology, Inc.), normal IgG (New England Biolabs) or Histone H3 (D2B12) (New England Biolabs) were added to DNA samples and incubated overnight at 4°C. Thirty microliters of ChIP-grade protein G agarose beads (Cell Signaling Technology, Inc.) were added to each sample and incubated for 2 h at 4°C, followed by centrifugation 6,000 rpm for 1 min. The supernatant was transferred to a new tube and processed for immunoprecipitation 3 times. The agarose pellet was washed with ChIP buffer three times and ChIP buffer with high salt (350 mM NaCl). Chromatin was eluted from antibody/protein G bead through incubations at 65°C, 30 min with gentle vortex, separated by centrifugation at 6,000 rpm, and transferred to a new tube. RNA and proteins in Eluted DNA were removed through RNase A and proteinase K treatment, and DNA was purified through the spin column, as mentioned above.

Standard PCR was performed using 50 ng DNAs with specific primers for CENP-P, Aurkb, Ckap2, Zw10, Top2a, and Mlf1ip genes as previously reported (7) with KAPA2G Fast PCR kit (KAPA Biosystems). PCR was executed using 34 cycles of amplification: Denaturation at 95°C for 30 sec, annealing at 50°C (CENP-P, Aurkb), 56.8°C (Mlf1ip) and 62°C (Zw10, Top2a) for 30 sec, extension at 72°C for 30 sec, and a final extension at 72°C for 5 min. PCR primers: CENP-P (CATGGAGATCCGCAGTACC; CATCCCTTCCTCATCGATTT) Aurkb (CCCAGAGAGTCCTACGGAAG; TGTTCTCAGCCAACTTCTGG) Ckap2 (ATTAAGCGATGGCAGAGTCC; TTTCTTTGTTCTCGGAAGGC) Zw10 (GAAGTGCCAGGATGTGATTG; AGCTTGTGATCAGCATCAGG) Top2A (ATCACCGACTCGCTCTCATT; GCACATGGACCTTCCTCATT).

Quantitative PCR (qRT-PCR)

Synthesis of cDNA with total RNA was performed using an iScript cDNA synthesis kit (Bio-Rad Laboratories). Two micrograms of total RNA and reverse transcriptase H was incubated in reaction buffer 5 min at 25°C, 30 min at 42°C, and 5 min at 85°C. The iQTM SYBR®-Green Supermix (Bio-Rad Laboratories) was used for quantitative real-time PCR. The same primers used for ChIP assays were utilized for this analysis, and mouse GAPDH as the reference gene. PCR amplification was performed with a 20 µl reaction mixture containing 2 µl of cDNA, 300 nM of each primer, and 1× iQTM SYBR®-Green supermix (Bio-Rad Laboratories). PCR condition as followed: Initial denaturation at 95°C for 3 min, followed by 40 cycles of denaturation at 95°C for 15 sec, annealing, and extension at 60°C for 30 sec. The transcriptional level of target genes was normalized by the transcriptional level of GAPDH (CATCACTGCCACCCAGAAGACTG; ATGCCAGTGAGCTTCCCGTTCAG). For downregulation assays, the transcriptional levels were compared with control siRNA-treated 308 cells or cell lines, not overexpressing CDK4 (NIH3T3 or 308) according to the algorithms 2-(ΔΔCt), respectively.

siRNA assay

Cell lines were transfected with CDK4 or control siRNAs with Lipofectamine® RNAiMAX Reagent (Invitrogen, Life Technologies) according to the manufacturer's instructions. 308 or 308-CDK4 cells were cultured in 60 mm Petri dishes at 70–80% of confluence. We utilized commercially available CDK4 specific siRNA (sc-29262) and control siRNA (sc-37007) (Santa Cruz Biotech). Briefly, 60 pmol of siRNA was diluted in 300 µl Opti-MEM® (Invitrogen; Life Technologies), mixed with 18 µl lipofectamine in 300 µl Opti-MEM®, and incubated 5 min at RT. The mixture was added to the cells in 4 ml of culture media without antibiotics. The media was replaced with fresh media after 24 h, and cells were harvested 96 h after the transfection. RNA was isolated using TRIzol® Reagent (Ambion, Life Technologies).

Statistical analysis

An unpaired Student's t-test was performed using GraphPad Prism 4 Software (GraphPad Software).

Results

CDK4 loaded onto the chromatin fraction of mouse keratinocytes

Various studies accomplished in the last decade have shown that cyclin D1 and CDK6 have additional functions non-related to their role in the cyclin-cdk-Rb axis (911). Based on these observations, we asked whether CDK4 has similar activities in transcriptional regulation (26). Thus, we first examined the CDK4 interaction with the chromatin fraction of mouse keratinocytes. We performed in situ cell extraction with the cytoskeleton extraction buffer (CSK), which removes the soluble proteins from the cytoplasm and nucleoplasm while leaving the chromatin-bound fraction intact (2729). We utilized asynchronous cell cultures of primary mouse keratinocytes and the keratinocyte cell line 308, a cell line derived from BALB/c mouse skin treated with 7,12-dimethylbenz[a]anthracene (2123,30). Immunofluorescence analysis of the extracted cells revealed the presence of CDK4 in the chromatin-bound fraction of both mouse primary keratinocytes and 308 keratinocyte cell lines (Fig. 1A-L). The CSK extraction buffer removes the soluble cell fractions, but the DNA and other insoluble material such as intermediate filament cytoskeleton remain in the so-called cell ghosts. Therefore, to verifies that CDK4 binds to DNA, we disrupt the cellular DNA and examine the presence of CDK4 and the positive control chromatin-bound histone deacetylase 1 (HDAC1). DNase treatment released the DNA-bound HDAC1 and CDK4 from the chromatin fraction (Fig. 1M-R), confirming that CDK4 is strongly associated with the DNA fraction of mouse keratinocytes.

CDK4 as a transcriptional regulator on the Aurkb and CENP-P promoters

In addition to their well-established role in the cell cycle, cyclin D1 and CDK6 have transcriptional functions (7,9,10). Whereas cyclin D1 plays a direct role in transcriptional regulation of genes governing chromosomal integrity (7), CDK6 is part of a complex that controls the transcription of p16Ink4 and the angiogenic factor VEGF-A (9). In view of the presence of the DNA-bound CDK4, its structural and functional similitudes with CDK6, and because cyclin D1 is one of the regulatory subunits of CDK4, we analyzed whether CDK4 may also act as a transcriptional regulator. We first examined a set of genes transcriptionally controlled by cyclin D1, which were previously reported that regulate chromosome segregation (7). Chromatin immunoprecipitation (ChIP) experiments revealed that CDK4 binds specifically to the Aurkb, CENP-P, and Zw10 promoters, while no binding to Ckap2, Top2a, and Mlf1ip was detected (Fig. 2A). These findings suggest that CDK4 may behave as a transcriptional regulator or form part of a transcriptional complex that controls genes associated with chromosomal segregation.

To delineate whether variation in the CDK4 protein level affects the interaction to the regulatory sites of these genes, we performed ChIP analysis of NIH3T3 cells overexpressing murine CDK4 (NIH3T3-CDK4). We carry out ChIP assay in three sequential immunoprecipitations and quantify the association of CDK4 with Aurkb and CENP-P promoters in NIH3T3-CDK4 cells and the parental cell line NIH3T3. CDK4 overexpressing cells showed a 3-, 5- and 9-fold increase binding to the CENP-P promoter in the three sequential immunoprecipitations, respectively, compared to NIH3T3 cells (Fig. 2B). Similarly, NIH3T3-CDK4 cells showed 2- and 7-fold increase binding to Aurkb promoter in the first and second immunoprecipitation, respectively, compared to NIH3T3 cells (Fig. 2B). ChIP analysis of the Zw10 promoter showed no differences between CDK4 overexpressing and parental cell lines. These results support the specificity of CDK4 binding to Aurkb and CENP-P promoters and suggest that variation in the CDK4 level might affect the transcription of Aurkb and CENP-P genes.

To examine whether CDK4 regulates the transcription of these genes, we quantified the transcription of Aurkb B and CENP-P genes upon overexpression and downregulation of CDK4. We performed quantitative PCR (qRT-PCR) of CENP-P, Aurkb, Zw10, Ckap2, and Top2a on NIH3T3 cells and the keratinocyte cell line 308 overexpressing CDK4 (NIH3T3-CDK4, 308-CDK4) and the parental cells lines NIH3T3 and 308. The transcriptional levels were normalized to the Gapdh, and a transcriptional ratio calculated between CDK4 overexpressing and the parental cell lines. NIH3T3-CDK4 cells showed a 2-fold increase in transcription of Aurkb (P<0.005, t-test) and 1.5-fold elevate transcription of CENP-P (P<0.05, t-test) compared to NIH3T3 cells. Similarly, we observed 3-(P<0.05, t-test) and 2-fold (P<0.005, t-test) elevated transcription of Aurkb and CENP-P, respectively, in the keratinocyte cell line 308-CDK4 (Fig. 3A). The enhanced expression of CDK4 did not significantly change the transcriptional levels of Ckap2 gene, although a 2- and 1.3-fold reduction of Zw10 (P<0.05, t-test) and Top2a (P<0.0005, t-test) genes. Recently reports showed that Top2a and Zw10 proteins are involved in chromosome segregation and mitotic checkpoint proteins (3135); therefore, their potential role downstream of CDK4 expression in cell proliferation and tumor development warrants further investigation. We also studied the protein levels of Aurora-B and CENP-P in both NIH3T3-CDK4 and 308-CDK4 cell lines. Increased protein levels of both CENP-P and Aurora-B were observed upon overexpression of CDK4 in the 308-cell line (Fig. 3B). 308-CDK4 cells showed a 20-fold increase of CENP-P (P<0.05, t-test) and a 4-fold increase of Aurora-B (P<0.05, t-test) compared to 308 cells. Expression of CENP-P protein was elevated 2-fold (P<0.05, t-test) in NIH3T3-CDK4 cells, although the protein level of Aurora-B showed a non-statistically significant change compared to NIH3T3 cells (Fig. 3B). Altogether, these results demonstrated that the CDK4 protein indeed regulates the transcription of Aurkb and CENP-P.

To validate our conclusions, we determined the effect of the reduction level of CDK4 on Aurkb and CENP-P transcription. We evaluated the inhibitory effect of CDK4-specific siRNA on the levels of Aurkb and CENP-P. The keratinocyte cell line 308 overexpressing CDK4 and the parental cell line 308 were transfected with CDK4-specific siRNA and a control scramble-siRNA. The transcription levels of Aurkb and CENP-P, normalized to the transcriptional level of Gapdh, were quantified by qRT-PCR and compared between CDK4-siRNA and control-siRNA transfected cells. We observed a reduction in Aurkb and CENP-P RNA levels in both 308-CDK4 and 308 cells 72 h after transfection with CDK4-siRNA (Fig. 4A). CDK4-siRNA lead to a 24% (P=0.02) and 51% (P=0.01) decrease of CENP-P RNA expression in 308 and 308-CDK4 cells respectively (Fig. 4A). Aurkb RNA levels were reduced by 46% (P=0.01, t-test) and 78% (P=0.0005, t-test) in 308 and 308-CDK4 cells respectively (Fig. 4A). The effect of CDK4-siRNA was monitored by Western blot analysis 72 h after transfection showing a 50- and 10-times reduction of CDK4, correlating with the 10- and 2-fold decreased level of Aurora-B protein in 308 and 308-CDK4 cells, respectively (Fig. 4B). Taken together, these analyses revealed that the binding of CDK4 to the regulatory site of Aurkb and CENP-P genes leads to positive transcriptional regulation.

Discussion

The canonical role of CDK4 and D-type cyclins as drivers of cell proliferation and tumorigenesis via phosphorylation of the retinoblastoma (Rb) family of proteins has been firmly established. However, over the last few years, it has been suggested that CDK4 plays alternative functions in proliferation and tumorigenesis (8). For example, CDK4 can also phosphorylate the transcription factor FOXM1, which in turn induces the transcription of other genes involved in the G2/M phases (12). Likewise, additional functions have been identified in different cell cycle regulators. For instance, the CDK4-related kinase, CDK6, performs transcriptional functions regulating the expression of VEGF-A and p16INK4a (9). Cyclin D1, a regulatory subunit of CDK4 and CDK6, participates in activities other than cell-cycle regulation, such as interaction with the androgen and estrogen receptors and DNA repair (8,10,11).

Notably, it was recently reported a transcriptional role of cyclin D1 regulating chromosome segregation genes such as Aurkb, CENP-P, Zw10, Ckap2, Top2a and Mlf1ip (7). Our present findings indicate that CDK4 also regulates the transcription of Aurkb and CENP-P, two genes involved in chromosome segregation. Aurora B expression has a key role in chromosome bi-orientation and spindle-assembly checkpoint, whereas Cenpp is required for proper kinetochore function (19,20,36). Our studies have also established a major difference between the transcriptional activities of CDK4 and cyclin D1 (7). Both CDK4 and cyclin D1 localizes on the regulatory sites of Aurkb, CENP-P, and Zw10 genes, but only cyclin D1 bind to Ckap2, Top2a, and Mlf1ip promoters (7). These results led us to hypothesize that CDK4 and cyclin D1 may act as a complex at the regulatory sites of Aurkb and CENP-P genes, whereas cyclin D1 may act independently of CDK4 in other contexts. Interestingly, the CDK4 binding to the Zw10 promoter does not result in changes in the transcription level of this gene. It is worth mentioning that the transcriptional role of cyclin D1 was determined in Ccnd1−/− mouse embryo fibroblasts (MEF) transfected with an epitope-tagged cyclin D1. In contrast, we studied the effect of CDK4 gain- and lost-of-function in keratinocytes and embryo fibroblast cell lines. Therefore, whether the differences observed in the transcription of Ckap2, Top2a, Mlf1ip, and Zw10 genes represent cell-specific regulation or technical discrepancies between these experiments merit further analysis.

Notably, it has been suggested that CDK4 activity is necessary for regulation of phase others than G0 and G1 (8,37). Inhibition of CDK4 activity results in delayed progression from G2 to mitosis due to a failure of chromosomes to migrate to the metaphase plate, implying that CDK4 is necessary for entry into mitosis (38,39). Consistent with these findings, our studies also showed that the transcriptional and protein levels of Aurkb and CENP-P correlate well with CDK4 expression. Notably, Aurora-B mRNA and protein levels are tightly regulated and peak at the G2-M phases (40,41), correlating well with the putative activity of CDK4 in the G2/M phase. Although the mechanisms regulated by CDK4 in the G2-M phase have not been clearly defined, it is known that CDK4 inhibition reduces mitosis's fidelity, implying that CDK4 indeed takes part in the G2/M phase by regulating Aurora B (38).

Given that CDK4 inhibitors are in active clinical development (4245), it is crucial to understand the role of CDK4 regulating Aurkb and CENP-P in tumor development. In this regard, we demonstrated that transgenic expression of CDK4 induces keratinocyte proliferation and accelerates the malignant progression of mouse skin tumors (1315). In contrast, lack of CDK4 expression inhibits skin and oral tumor development (46,47). We have also determined that CDK4, but not the related kinases CDK6 and CDK2, induce skin tumor malignant progression (1315). Thus, the effect of CDK4 inducing tumor progression might be related to its role in the transcriptional activity of Aurkb and CENP-P. Aurora-B is a subunit of the chromosomal passenger complex (CPC) controlling chromosome segregation (41,48,49) and potentially contribute to the Spindle-Assembly-Checkpoint (SAC), which malfunction leads to aneuploidy and tumorigenesis (18). In fact, long-term overexpression of Aurora-B in vivo results in defective chromosome segregation, aneuploidy, and the development of multiple tumors in mice (5054). Similarly, CENP-P is a subunit of the centromeric complex required for proper kinetochore function contributing to chromosome segregation (20). Thus, the potential effect of CDK4 dysregulation in Aurkb and CENP-P expression leading to CIN and tumorigenesis warrant further investigation.

Studies demonstrating that inactivation of CDK4 and D-type cyclins can prevent tumor development in murine models reinforced the view that CDK4 is suitable for cancer-specific targets (46,47,55,56). Based on these results, in the last decade, CDK inhibitors were designed, which are in clinical development or have already been approved by the US Food and Drug Administration (4245,5759). For example, the observed preclinical and clinical effects of palbociclib are consistent with the notion that inhibition of CDK4/6 is a crucial mechanism underlying tumor growth activity (6063). However, some of these drugs have been met with variable degrees of success in preclinical and clinical studies. Thus, the CDK4 binding to the promoter regions should be confirmed with assays in which specific CDK4-inhibitors, such as Abemaciclib and Palbociclib, are administered to keratinocytes. Those experiments will be fundamentals to determine the effect of the CDK4 kinase activity in the transcriptional role of CDK4. If the function of CDK4 regulating Aurkb and CENP-P levels is not inhibited by the current drugs, then this new activity might represent an important therapeutic target to disrupt cell cycle progression in cancer cells. Such a scenario could help to explain the reduced efficacy of the existing CDK4 drugs in some cancers and open new research avenues for future studies directed to provide new CDK4-related targets for combined therapeutic interventions.

Acknowledgements

The authors would like to thank the Laboratory Animal Resources and the Histology Core at the College of Veterinary Medicine, North Carolina State University (Raleigh, USA) for helping with the processing and staining of skin samples.

Funding

Research reported in this publication was supported by grants from the National Cancer Institute (grant no. RO1CA116328) and the National Institute of Environmental Health Sciences (grant no. P30ES025128; Center for Human Health and the Environment).

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

MLRP was responsible for the design and conception of the experiments, and was the guarantor of this work and, as such, takes responsibility for the integrity of the data and the accuracy of the data analysis. SHL carried out the experiments and contributed to data analysis and interpretation. LRLR and PLMDM conceived and designed part of the experiments, and wrote part of the results section. RM designed the primers utilized in part of this paper, and performed the immunofluorescence staining and semi-quantification of the western blots. All authors provided critical feedback and helped shape the research, analysis and manuscript. SHL and MLRP wrote the manuscript and confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The generation of mouse primary keratinocytes and protocols for animal use were approved by the Institutional Animal Care and Use Committee of North Carolina State University (approval no. 18-102-B; Raleigh, USA).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CDK4

cyclin-dependent kinase 4

CIN

chromosome instability

ChIP

chromatin immunoprecipitation

References

1 

Fischer PM, Endicott J and Meijer L: Cyclin-dependent kinase inhibitors. Prog Cell Cycle Res. 5:235–248. 2003.PubMed/NCBI

2 

Lukasik P, Zaluski M and Gutowska I: Cyclin-dependent kinases (CDK) and their role in diseases development-review. Int J Mol Sci. 22:29352021. View Article : Google Scholar : PubMed/NCBI

3 

Suryadinata R, Sadowski M and Sarcevic B: Control of cell cycle progression by phosphorylation of cyclin-dependent kinase (CDK) substrates. Biosci Rep. 30:243–255. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Gitig DM and Koff A: Cdk pathway: Cyclin-dependent kinases and cyclin-dependent kinase inhibitors. Methods Mol Biol. 142:109–123. 2000.PubMed/NCBI

5 

Weinberg RA: The molecular basis of carcinogenesis: Understanding the cell cycle clock. Cytokines Mol Ther. 2:105–110. 1996.PubMed/NCBI

6 

Ewen ME, Sluss HK, Sherr CJ, Matsushime H, Kato J and Livingston DM: Functional interactions of the retinoblastoma protein with mammalian D-type cyclins. Cell. 73:487–497. 1993. View Article : Google Scholar : PubMed/NCBI

7 

Casimiro MC, Crosariol M, Loro E, Ertel A, Yu Z, Dampier W, Saria EA, Papanikolaou A, Stanek TJ, Li Z, et al: ChIP sequencing of cyclin D1 reveals a transcriptional role in chromosomal instability in mice. J Clin Invest. 122:833–843. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Hydbring P, Malumbres M and Sicinski P: Non-canonical functions of cell cycle cyclins and cyclin-dependent kinases. Nat Rev Mol Cell Biol. 17:280–292. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Kollmann K, Heller G, Schneckenleithner C, Warsch W, Scheicher R, Ott RG, Schäfer M, Fajmann S, Schlederer M, Schiefer AI, et al: A Kinase-Independent function of CDK6 links the cell cycle to tumor angiogenesis. Cancer Cell. 24:167–181. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Bienvenu F, Jirawatnotai S, Elias JE, Meyer CA, Mizeracka K, Marson A, Frampton GM, Cole MF, Odom DT, Odajima J, et al: Transcriptional role of cyclin D1 in development revealed by a genetic-proteomic screen. Nature. 463:374–378. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Jirawatnotai S, Hu Y, Michowski W, Elias JE, Becks L, Bienvenu F, Zagozdzon A, Goswami T, Wang YE, Clark AB, et al: A function for cyclin D1 in DNA repair uncovered by protein interactome analyses in human cancers. Nature. 474:230–234. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Anders L, Ke N, Hydbring P, Choi YJ, Widlund HR, Chick JM, Zhai H, Vidal M, Gygi SP, Braun P and Sicinski P: A systematic screen for CDK4/6 substrates links FOXM1 phosphorylation to senescence suppression in cancer cells. Cancer Cell. 20:620–634. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Miliani de Marval PL, Macias E, Conti CJ and Rodriguez-Puebla ML: Enhanced malignant tumorigenesis in Cdk4 transgenic mice. Oncogene. 23:1863–1873. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Macias E, Miliani de Marval PL, De Siervi A, Conti CJ, Senderowicz AM and Rodriguez-Puebla ML: CDK2 activation in mouse epidermis induces keratinocyte proliferation but does not affect skin tumor development. Am J Pathol. 173:526–535. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Wang X, Sistrunk C and Rodriguez-Puebla ML: Unexpected reduction of skin tumorigenesis on expression of cyclin-dependent kinase 6 in mouse epidermis. Am J Pathol. 178:345–354. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Adon AM, Zeng X, Harrison MK, Sannem S, Kiyokawa H, Kaldis P and Saavedra HI: Cdk2 and Cdk4 regulate the centrosome cycle and are critical mediators of centrosome amplification in p53-null cells. Mol Cell Biol. 30:694–710. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Carmena M, Wheelock M, Funabiki H and Earnshaw WC: The chromosomal passenger complex (CPC): From easy rider to the godfather of mitosis. Nat Rev Mol Cell Biol. 13:789–803. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Musacchio A and Salmon ED: The spindle-assembly checkpoint in space and time. Nat Rev Mol Cell Biol. 8:379–393. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Peters AH, Kubicek S, Mechtler K, O'Sullivan RJ, Derijck AA, Perez-Burgos L, Kohlmaier A, Opravil S, Tachibana M, Shinkai Y, et al: Partitioning and plasticity of repressive histone methylation states in mammalian chromatin. Mol Cell. 12:1577–1589. 2003. View Article : Google Scholar : PubMed/NCBI

20 

D'Avino PP and Capalbo L: New auroras on the roles of the chromosomal passenger complex in cytokinesis: Implications for cancer therapies. Front Oncol. 5:2212015.

21 

Strickland JE, Greenhalgh DA, Koceva-Chyla A, Hennings H, Restrepo C, Balaschak M and Yuspa SH: Development of murine epidermal cell lines which contain an activated rasHa oncogene and form papillomas in skin grafts on athymic nude mouse hosts. Cancer Res. 48:165–169. 1988.PubMed/NCBI

22 

Hennings H, Michael D, Lichti U and Yuspa SH: Response of carcinogen-altered mouse epidermal cells to phorbol ester tumor promoters and calcium. J Invest Dermatol. 88:60–65. 1987. View Article : Google Scholar : PubMed/NCBI

23 

Yuspa SH and Morgan DL: Mouse skin cells resistant to terminal differentiation associated with initiation of carcinogenesis. Nature. 293:72–74. 1981. View Article : Google Scholar : PubMed/NCBI

24 

Lichti U, Anders J and Yuspa SH: Isolation and short-term culture of primary keratinocytes, hair follicle populations and dermal cells from newborn mice and keratinocytes from adult mice for in vitro analysis and for grafting to immunodeficient mice. Nat Protoc. 3:799–810. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Miliani de Marval PL, Kim SH and Rodriguez-Puebla ML: Isolation and characterization of a stem cell side-population from mouse hair follicles. Methods Mol Biol. 1195:259–268. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Kitagawa M and Lee SH: The chromosomal passenger complex (CPC) as a key orchestrator of orderly mitotic exit and cytokinesis. Front Cell Dev Biol. 3:142015. View Article : Google Scholar : PubMed/NCBI

27 

Madine MA, Swietlik M, Pelizon C, Romanowski P, Mills AD and Laskey RA: The roles of the MCM, ORC, and Cdc6 proteins in determining the replication competence of chromatin in quiescent cells. J Struct Biol. 129:198–210. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Martini E, Roche DM, Marheineke K, Verreault A and Almouzni G: Recruitment of phosphorylated chromatin assembly factor 1 to chromatin after UV irradiation of human cells. J Cell Biol. 143:563–575. 1998. View Article : Google Scholar : PubMed/NCBI

29 

Geng Y, Whoriskey W, Park MY, Bronson RT, Medema RH, Li T, Weinberg RA and Sicinski P: Rescue of cyclin D1 deficiency by knockin cyclin E. Cell. 97:767–777. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Hennings H, Robinson VA, Michael DM, Petit GR, Jung R and Yuspa SH: Development of an in vitro analogue of initiated mouse epidermis to study tumor promoters and antipromoters. Cancer Res. 50:4794–4800. 1990.PubMed/NCBI

31 

Li HN, Zheng WH, Du YY, Wang G, Dong ML, Yang ZF and Li XR: ZW10 interacting kinetochore protein may serve as a prognostic biomarker for human breast cancer: An integrated bioinformatics analysis. Oncol Lett. 19:2163–2174. 2020.PubMed/NCBI

32 

Pauleau AL, Bergner A, Kajtez J and Erhardt S: The checkpoint protein Zw10 connects CAL1-dependent CENP-A centromeric loading and mitosis duration in Drosophila cells. PLoS Genet. 15:e10083802019. View Article : Google Scholar : PubMed/NCBI

33 

Park Y, Kim JS and Oh JS: Zw10 is a spindle assembly checkpoint protein that regulates meiotic maturation in mouse oocytes. Histochem Cell Biol. 152:207–215. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Leonard J, Sen N, Torres R, Sutani T, Jarmuz A, Shirahige K and Aragón L: Condensin relocalization from centromeres to chromosome arms promotes Top2 recruitment during anaphase. Cell Rep. 13:2336–2344. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Bermejo R, Capra T, Gonzalez-Huici V, Fachinetti D, Cocito A, Natoli G, Katou Y, Mori H, Kurokawa K, Shirahige K and Foiani M: Genome-organizing factors Top2 and Hmo1 prevent chromosome fragility at sites of S phase transcription. Cell. 138:870–884. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Gruneberg U, Neef R, Honda R, Nigg EA and Barr FA: Relocation of Aurora B from centromeres to the central spindle at the metaphase to anaphase transition requires MKlp2. J Cell Biol. 166:167–172. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Neuman E, Ladha MH, Lin N, Upton TM, Miller SJ, DiRenzo J, Pestell RG, Hinds PW, Dowdy SF, Brown M and Ewen ME: Cyclin D1 stimulation of estrogen receptor transcriptional activity independent of cdk4. Mol Cell Biol. 17:5338–5347. 1997. View Article : Google Scholar : PubMed/NCBI

38 

Burgess A, Wigan M, Giles N, Depinto W, Gillespie P, Stevens F and Gabrielli B: Inhibition of S/G2 phase CDK4 reduces mitotic fidelity. J Biol Chem. 281:9987–9995. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Gabrielli BG, Sarcevic B, Sinnamon J, Walker G, Castellano M, Wang XQ and Ellem KA: A cyclin D-Cdk4 activity required for G2 phase cell cycle progression is inhibited in ultraviolet radiation-induced G2 phase delay. J Biol Chem. 274:13961–13969. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Kimura M, Uchida C, Takano Y, Kitagawa M and Okano Y: Cell cycle-dependent regulation of the human aurora B promoter. Biochem Biophys Res Commun. 316:930–936. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Kimura M, Kotani S, Hattori T, Sumi N, Yoshioka T, Todokoro K and kano Y: Cell cycle-dependent expression and spindle pole localization of a novel human protein kinase, Aik, related to Aurora of Drosophila and yeast Ipl1. J Biol Chem. 272:13766–13771. 1997. View Article : Google Scholar : PubMed/NCBI

42 

Fry DW, Harvey PJ, Keller PR, Elliott WL, Meade M, Trachet E, Albassam M, Zheng X, Leopold WR, Pryer NK and Toogood PL: Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol Cancer Ther. 3:1427–1438. 2004.PubMed/NCBI

43 

Toogood PL, Harvey PJ, Repine JT, Sheehan DJ, VanderWel SN, Zhou H, Keller PR, McNamara DJ, Sherry D, Zhu T, et al: Discovery of a potent and selective inhibitor of cyclin-dependent kinase 4/6. J Med Chem. 48:2388–2406. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Infante JR, Cassier PA, Gerecitano JF, Witteveen PO, Chugh R, Ribrag V, Chakraborty A, Matano A, Dobson JR, Crystal AS, et al: A Phase I study of the cyclin-dependent kinase 4/6 inhibitor ribociclib (LEE011) in patients with advanced solid tumors and lymphomas. Clin Cancer Res. 22:5696–5705. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Patnaik A, Rosen LS, Tolaney SM, Tolcher AW, Goldman JW, Gandhi L, Papadopoulos KP, Beeram M, Rasco DW, Hilton JF, et al: Efficacy and safety of abemaciclib, an inhibitor of CDK4 and CDK6, for patients with breast cancer, non-small cell lung cancer, and other solid tumors. Cancer Discov. 6:740–753. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Rodriguez-Puebla ML, Miliani de Marval PL, LaCava M, Moons DS, Kiyokawa H and Conti CJ: Cdk4 deficiency inhibits skin tumor development but does not affect normal keratinocyte proliferation. Am J Pathol. 161:405–411. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Miliani de Marval PL, Macias E, Rounbehler R, Sicinski P, Kiyokawa H, Johnson DG, Conti CJ and Rodriguez-Puebla ML: Lack of cyclin-dependent kinase 4 inhibits c-myc tumorigenic activities in epithelial tissues. Mol Cell Biol. 24:7538–7547. 2004. View Article : Google Scholar : PubMed/NCBI

48 

Glover DM, Leibowitz MH, McLean DA and Parry H: Mutations in aurora prevent centrosome separation leading to the formation of monopolar spindles. Cell. 81:95–105. 1995. View Article : Google Scholar : PubMed/NCBI

49 

Gopalan G, Chan CS and Donovan PJ: A novel mammalian, mitotic spindle-associated kinase is related to yeast and fly chromosome segregation regulators. J Cell Biol. 138:643–656. 1997. View Article : Google Scholar : PubMed/NCBI

50 

Gonzalez-Loyola A, Fernandez-Miranda G, Trakala M, Partida D, Samejima K, Ogawa H, Cañamero M, de Martino A, Martínez-Ramírez Á, de Cárcer G, et al: Aurora B overexpression causes aneuploidy and p21Cip1 repression during tumor development. Mol Cell Biol. 35:3566–3578. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Nguyen HG, Makitalo M, Yang D, Chinnappan D, St Hilaire C and Ravid K: Deregulated Aurora-B induced tetraploidy promotes tumorigenesis. FASEB J. 23:2741–2748. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Nguyen HG and Ravid K: Tetraploidy/aneuploidy and stem cells in cancer promotion: The role of chromosome passenger proteins. J Cell Physiol. 208:12–22. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Giet R, Petretti C and Prigent C: Aurora kinases, aneuploidy and cancer, a coincidence or a real link? Trends Cell Biol. 15:241–250. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Katayama H, Brinkley WR and Sen S: The Aurora kinases: Role in cell transformation and tumorigenesis. Cancer Metastasis Rev. 22:451–464. 2003. View Article : Google Scholar : PubMed/NCBI

55 

Yu Q, Geng Y and Sicinski P: Specific protection against breast cancers by cyclin D1 ablation. Nature. 411:1017–1021. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Yu Q, Sicinska E, Geng Y, Ahnström M, Zagozdzon A, Kong Y, Gardner H, Kiyokawa H, Harris LN, Stål O and Sicinski P: Requirement for CDK4 kinase function in breast cancer. Cancer Cell. 9:23–32. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Finn RS, Martin M, Rugo HS, Jones S, Im SA, Gelmon K, Harbeck N, Lipatov ON, Walshe JM, Moulder S, et al: Palbociclib and letrozole in advanced breast cancer. N Engl J Med. 375:1925–1936. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Long F, He Y, Fu H, Li Y, Bao X, Wang Q, Wang Y, Xie C and Lou L: Preclinical characterization of SHR6390, a novel CDK 4/6 inhibitor, in vitro and in human tumor xenograft models. Cancer Sci. 110:1420–1430. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Bisi JE, Sorrentino JA, Jordan JL, Darr DD, Roberts PJ, Tavares FX and Strum JC: Preclinical development of G1T38: A novel, potent and selective inhibitor of cyclin dependent kinases 4/6 for use as an oral antineoplastic in patients with CDK4/6 sensitive tumors. Oncotarget. 8:42343–42358. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Dean JL, Thangavel C, McClendon AK, Reed CA and Knudsen ES: Therapeutic CDK4/6 inhibition in breast cancer: Key mechanisms of response and failure. Oncogene. 29:4018–4032. 2010. View Article : Google Scholar : PubMed/NCBI

61 

Dean JL, McClendon AK, Hickey TE, Butler LM, Tilley WD, Witkiewicz AK and Knudsen ES: Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors. Cell Cycle. 11:2756–2761. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Schwartz GK, LoRusso PM, Dickson MA, Randolph SS, Shaik MN, Wilner KD, Courtney R and O'Dwyer PJ: Phase I study of PD 0332991, a cyclin-dependent kinase inhibitor, administered in 3-week cycles (Schedule 2/1). Br J Cancer. 104:1862–1868. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Zhang YX, Sicinska E, Czaplinski JT, Remillard SP, Moss S, Wang Y, Brain C, Loo A, Snyder EL, Demetri GD, et al: Antiproliferative effects of CDK4/6 inhibition in CDK4-amplified human liposarcoma in vitro and in vivo. Mol Cancer Ther. 13:2184–2193. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2021
Volume 22 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee SH, Rodriguez LR, Majumdar R, De Marval PL and Rodriguez‑Puebla ML: CDK4 has the ability to regulate Aurora B and Cenpp expression in mouse keratinocytes. Oncol Lett 22: 732, 2021
APA
Lee, S.H., Rodriguez, L.R., Majumdar, R., De Marval, P.L., & Rodriguez‑Puebla, M.L. (2021). CDK4 has the ability to regulate Aurora B and Cenpp expression in mouse keratinocytes. Oncology Letters, 22, 732. https://doi.org/10.3892/ol.2021.12993
MLA
Lee, S. H., Rodriguez, L. R., Majumdar, R., De Marval, P. L., Rodriguez‑Puebla, M. L."CDK4 has the ability to regulate Aurora B and Cenpp expression in mouse keratinocytes". Oncology Letters 22.4 (2021): 732.
Chicago
Lee, S. H., Rodriguez, L. R., Majumdar, R., De Marval, P. L., Rodriguez‑Puebla, M. L."CDK4 has the ability to regulate Aurora B and Cenpp expression in mouse keratinocytes". Oncology Letters 22, no. 4 (2021): 732. https://doi.org/10.3892/ol.2021.12993