
Cervical cancer is the second leading cause of cancer mortality among women worldwide (Łuczak and Jagodzinski, 2008; Chang
High-risk HPV encodes two transforming genes, E6 and E7, both of which play important roles in malignant transformation (Zhou
E7 is a major transforming protein of HPV and its transforming activity is due to its interaction with retinoblastoma tumor suppressor (Rb) (Thierry, 2009; Moody and Laimins, 2010; Xiong
In this study, the Elk-1 transcription factor was identified as an interaction partner for the E7 oncoprotein. We demonstrated
We suggest that Elk-1 transcriptional activity should be increased for the formation of a novel oncogenic action of E7 in cervical cancer through its interaction with E7. Our findings may provide a new logic for inhibiting E7-induced carcinogenesis.
The pCMV-GAL4/Elk-1 plasmid was constructed by cloning
Table 1 . List of primer sequences used for the deletion mutants or site-directed mutagenesis
Gene | Primer sequences |
---|---|
E7 (1~20) | Forward: 5’-GATC |
Reverse: 5’-GATC | |
E7 (21~40) | Forward: 5’-GATC |
Reverse: 5’-GATC | |
E7 (41~106) | Forward: 5’-GATC |
Reverse: 5’-GATC | |
Elk-1 (1~117) | Forward : 5'-ATGC |
Reverse: 5'-ATGC | |
Elk-1 (118~199) | Forward : 5'-ATGC |
Reverse : 5'-ATGC | |
Elk-1 (200~286) | Forward : 5'-ATGC |
Reverse : 5'-ATGC | |
Elk-1 (287~339) | Forward : 5'-ATGC |
Reverse : 5'-ATGC | |
Elk-1 (340~428) | Forward : 5'-ATGCGGATCCCGATGGGAAGTGGCTCCGGC-3' |
Reverse : 5'-TAATCTCGAGTCATGGCTTCTGGGG-3' | |
E7 (∆25~29) | Forward: 5’-ATGATTCCGGTTGACCAATT -AAGCGACTCA-3’ |
Reverse: 5’-TGAGTCGCTT-AATTGGTCAACCGGAATCAT-3’ | |
E7 (L25A) | Forward: 5’-TCCGGTTGACGCTCTA -TGTCACGAGCAATTAAGCG-3’ |
Reverse: 5’-CGCTTAA-TTGCTCGTGACATAG-AGCGTCAACCGGA-3’ |
Human cervical cancer cell lines were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA) and maintained in recommended medium supplemented with 10% fetal bovine serum (JBI, Seoul, Korea) and 1% penicillin/streptomycin in a humidified 5% CO2 incubator at 37°C. Transfections were performed using FuGENE6 (Promega, Madison, WI, USA) with indicated amounts of expression plasmids.
Yeast two-hybrid screening was performed using the Matchmaker LexA two-hybrid system (Clontech, Mountain View, CA, USA) according to the manufacturer’s instructions (Rho
Equal amounts of GST and GST fusion proteins bound to glutathione-linked agarose beads (GE Healthcare, Little Chalfont, Buckinghamshire, UK) were incubated
The luciferase assays were performed using the Dual-Luciferase® Reporter Assay System (Promega) (Rho
At 48 h post-transfection, cells were harvested and lysed on ice in RIPA buffer [50 mM Tris-HCl (pH 8.0), 150 mM NaCl, 1% NP-40, 0.1% SDS, and 0.5% Na-deoxycholate] (Rho
Cells were lysed and equal amounts of cell extracts (20-40 µg) or the samples from the co-immunoprecipitations were electrophoresed on 10% SDS polyacrylamide gel, electro transferred onto a nitrocellulose membrane (Yoon
For the colony formation assay, 1,000 cells transfected with E7-, Elk-1-, and En-expressing vectors and a control vector plasmid were seeded on 100-mm plates and allowed to grow for 14 days in culture medium (Choi
Values are presented as the mean ± SD or ± SE. Statistical comparisons between groups were performed using the Student’s t-test.
E7-binding proteins were screened using the Y2H system to identify novel interaction proteins of the E7. The pGilda-E7 was used as bait to screen a human cDNA library. Based on the results, Elk-1 was identified as an interacting partner. Growth and β-galactosidase assays were conducted to verify the result of the Y2H screening (Fig. 1).
Strains transformed with pGilda/Elk-1 and pB42AD/E7 were unable to grow in normal culture media, but those grown in a Leu-deficient culture medium were able to grow (Fig. 1A). The culture medium containing X-Gal (5-bromo-4-chloro-3-indolyl-β-D-galactopyranoside) turned blue after X-Gal had been decomposed by β-galactosidase (Fig. 1B), demonstrating a strong interaction between the E7 and Elk-1.
To further investigate this interaction, an
To determine the feasibility of Elk-1 and E7 association in mammalian cells, Elk-1 and His-E7 were transiently co-transfected into human HEK293a cells for immunoprecipitation analysis.
After 48 h of transfection, cell lysates were prepared and co-immunoprecipitation assays were performed using an anti-His (His-E7) antibody. As seen in Fig. 2, Elk-1 co-precipitated with the His-E7 antibody. However, Ras overexpression slightly increased the interaction between Elk-1 and E7 in the HEK293a cells (Fig. 2A, lane 4).
The co-immunoprecipitation assays were repeated in HeLa cells to express the endogenous E7 protein. The HeLa cells were transfected with Elk-1 and treated with 5 mM DTBP before cell lysis to preserve the intracellular protein complexes in the lysates. Cell lysates from the HeLa cells underwent co-immunoprecipitation with the E7 antibody, and the immune complexes were subsequently analysed by Western blotting using the Elk-1 antibody. As shown in Fig. 2B, Elk-1 co-precipitated with the E7 antibody, but not with a control IgG antibody (Fig. 2B, lane 1).
To further confirm the interaction between Elk-1 and E7
These results revealed the
To investigate the specific Elk-1 binding domain of E7, three E7 truncation fragments were designed, as shown in Fig. 3A. In the Y2H system, full-length human Elk-1 cDNA and a plasmid containing either the full-length E7 cDNA (1–106) or three deletion fragments (CR1 (1–20), CR2 (21–40), or CR3 (41–106)) were co-transformed into EGY48 yeast cells. Cells containing the full-length and deletion fragments of E7 grew on plates without Ura, His, Trp, and Leu. To confirm these results, the binding activity of these constructs was determined by measuring the relative expression of β-galactosidase.
As shown in Fig. 3B (lower panel), the β-galactosidase assay results confirmed that all the fragments were able to bind to Elk-1. For further confirmation, GST pull-down assays were conducted using various GST-E7 deletion proteins bound to glutathione-linked agarose beads and
The E7 domains that interacted with Elk-1 were identified, but the potential Elk-1 motifs involved in E7 interaction were yet to be identified. To this end, a series of GST-Elk-1 derivative proteins were constructed (Fig. 4A). First, to confirm that full-length Elk-1 interacted with E7, (His6)-Elk-1 (immobilised on Ni2+-NTA agarose beads) was used in the pull-down assays. The bound E7 was examined by autoradiography (Fig. 4B).
GST pull-down assays were then performed using GST-Elk-1 deletion proteins and
Together, these results suggest that the N-terminal domain of Elk-1 including the ETS DNA-binding domain and serum response factor (SRF) binding domain was required for E7 association.
To clarify the functional consequences of the interaction between E7 and Elk-1, the role of E7 in controlling the transactivation capacity of Elk-1 was investigated. First, the promoter activity of the Elk-1 target genes, egr-1 and c-fos, were evaluated. The egr-1 or c-fos promoter-reporter gene construct was co-transfected into cells together with plasmid vectors that express E7, Elk-1, and Elk-En (a dominant-negative form of Elk-1) (Fig. 5).
In HeLa cells, transient transfection of Elk-1 stimulated the promoter activity of both egr-1 and c-fos, as expected (Fig. 5A, 5B). However, E7 also stimulated the promoter activity of the two Elk-1 target genes. The intensity of activation of the egr-1 promoter by Elk-1 and/or E7 was much stronger than that of the c-fos promoter (Fig. 5A, 5B). Moreover, when E7 and Elk-1 were co-transfected, a synergistic effect was observed on the activation of the Elk-1 target gene promoter. Egr-1 promoter activity was increased by ~35-fold, and c-fos promoter activity was increased by ~4-fold. These results suggested that E7 enhanced Elk-1 transactivation activity. To confirm that E7 binding to Elk-1 enhanced transactivation, Elk-En was transiently transfected with E7. The results showed that upregulation of Egr-1 and c-fos promoter activity by E7 was significantly downregulated by Elk-En, suggesting that E7 interacted with Elk-1 to enhance transcriptional activity. In contrast, the expression of the E2F transcription factor activated by E7 (not the target gene of Elk1) was not increased by co-transfection with Elk-1 and E7 or decreased by co-transfection with Elk1-EN and E7 (Fig. 5C).
After transiently transfecting E7, the expression of the egr-1 and c-fos proteins was investigated since E7 increased the transcriptional activity of Elk-1. Cells were co-transfected with 2 µg of the Elk-1-expressing vector and various amounts of the E7-expressing vector (0, 2, 5, and 8 µg), and the protein expression levels of egr-1 and c-fos were examined using a Western hybridisation assay. In both HeLa (HPV positive) and C33A (HPV negative) cell lines, cells transiently transfected with E7 exhibited increased protein expression levels of egr-1 and c-fos in a dose-dependent manner (Fig. 5D, 5E). These results suggest that E7 upregulated the expression of target gene products egr-1 and c-fos while activating the transcriptional activity of Elk-1.
To determine the synergistic effects of the E7 and Elk-1 on cervical cancer cell proliferation, a colony formation assay was performed in the C33A cells. Transfection of E7 or Elk-1 induced colony formation of C33A cells. More importantly, the co-transfection of E7 and Elk-1 resulted in increased colony formation (Fig. 6A, 6B). However, compared to the untreated control and E7-treated cells, Elk-En significantly reduced colony formation in C33A cells. These results suggest that E7 significantly increased cell proliferation.
High-risk HPV encodes oncogenes E6 and E7, which have transforming properties. According to several studies, E6 and E7 are potent oncogenes that promote cervical cancer development (Yim and Park, 2005; Pal and Kundu, 2020).
As previously stated, one of the major oncogenic functions of E7 is the inactivation of pRb through proteasome degradation induction. However, some mutants with changes to the CK2 site or C-terminal zinc finger domain inhibit the transforming activity of E7 while maintaining their ability to bind to pRb (McINTYRE
In this case, a Y2H method was used to examine a partner molecule that binds to E7 to discover a new E7 mechanism in the carcinogenesis of human cervical cancer. Elk-1 was identified as a binding partner capable of interacting with E7. Elk-1, a transcriptional activator of a proto-oncogene from the ETS family of transcription factors, binds to the serum response element (SRE) in the promoter region and mediates transcriptional activity in response to growth factors. Elk-1 has a significant role in regulating ras-induced tumorigenesis in lung cancer cells (De Luca
Elk-1 functions as a cofactor to enable the activation of the c-myc promoter by the nuclear factor of the activated T cells (NFAT) and pancreatic cancer growth
We demonstrated the direct binding of E7 to Elk-1 (Fig. 1, 2). Deletion constructs were used to locate the Elk-1-interacting region of E7. The E7 protein is divided into three domains [conserved regions (CR) 1–3]. CR1 at the N-terminus was required for the cellular transformation and pRb degradation but did not directly bind to pRb; CR2, which contained an LXCXE motif sufficient for binding to pRb, blocked the association of E2F and pRb; and CR3, which contained two Cys-X-X-Cys motifs that bind directly to a C-terminal region of pRb, was also required to inhibit the binding of E2F to pRb. The CR3 region of E7 includes a nuclear export signal (NES) and type 1 PDZ binding motifs (Chemes
All E7 deletion constructs (CR1, CR2, and the CR3 domain) bonded to Elk-1, but Elk-1 interacted more strongly with the CR2 and CR3 domains than with the CR1 domain (Fig. 3, 4). Additionally, the LXCXE pRb-binding motif of the E7 mutants (deletion or deficient in the pRb-binding motif) did not bind to Elk-1, suggesting that the LXCXE motif was required for binding Elk-1 to E7 (McINTYRE
Next, the E7-interacting region in Elk-1 was identified. E7 binds to the ETS DNA-binding and SRF-binding domains of Elk-1. Elk-1 was recruited to the SRE via protein–DNA (via ETS DNA binding) and protein–protein (via the SRF-binding domain) interactions. The ETS DNA-binding domain, which is located in the N-terminal region, was required for DNA binding and ternary complex formation with SRF. The SRF-binding domain was located downstream of the ETS DNA-binding domain, which was necessary for binding to SRF. Since E7 interacted with two different domains of Elk-1, the E7/Elk-1 binding may affect the protein–DNA and protein–protein interactions. These results suggest that E7 enhanced the DNA binding affinity of Elk-1 and increased the interaction between Elk-1 and SRF through the ETS DNA-binding and the SRF-binding domains, respectively. As a result, E7 may be crucial in Elk-1-SRF-SRE ternary complex formation.
Elk-1 is a member of a subfamily of ETS-domain proteins known as TCFs (ternary complex factors), which associate with the SRF to regulate c-fos proto-oncogene transcription (Gille
Interestingly, Elk-1 controls various functional groups of target genes and different modes of transcription factor binding. This activates the SRF function, which acts like actin when the SRF binds to the Elk-1-SRF binding domain. SRF also controls the actin cytoskeleton and cell migration (Odrowaz and Sharrocks, 2012; Azam
In conclusion, E7 promotes cervical cancer in addition to deactivating pRB by binding to Elk-1, a new target of E7, and activating the function of Elk-1. We expect that blocking this accelerating process can be a good treatment strategy for suppressing cervical cancer.
The work is supported by grants from the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2020R1F1A107633713, BK21 FOUR program), National Cancer Center, Korea (NCC-2210450-1) and the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Korea (HP20C0131).
The authors have declared that no competing interest exists.
![]() |
![]() |