Colchicine, a plant-derived alkaloid with relatively low toxicity on normal human epidermal keratinocytes (HEKn), has selective inhibitory effect on the growth of CaSki (HPV16-positive) and HeLa (HPV18-positive) human cervical cancer cell lines via the induction of apoptosis. Colchicine (2.5, 5.0 and 10.0 ng/ml) significantly reduced the expression of human papilloma virus (HPV) 16 E6/E7 mRNA and protein in CaSki and HeLa cells. Moreover, reduced expression of E6 and E7 induced by Colchicine resulted in the up-regulation of tumor suppressor proteins, p53 and Rb, as well as down-regulation of phospho Rb (pRb) protein. In addition, Bax, cytosolic cytochrome c and cleaved caspase-3 protein were increased while Bcl-2 protein was decreased significantly by 48 h of Colchicine treatment. These results implied that Colchicine could be explored as a potent candidate agent for the treatment and prevention of HPV-associated cervical cancer without deleterious effects.

Cervical cancer is one of the most common malignancies all over the world and ranked the second leading causes of cancer-related death in females [1,2], which is closely related with persistent infection with oncogenic or high-risk (HR) HPV. It is well established that nearly over 90% of cervical cancers are caused by HR-HPV [3]. In 2000, an estimated, 468,000 new cases were diagnosed annually, of which about 80% occurred in developing countries [4], and such extensive screening programs make them inappropriate for the developing world with limited resources [5]. Current common treatment strategies for cervical cancer include surgery, radiotherapy and chemotherapy alone or in combination. However, 5-year overall survival rates with this treatment are about only 50% [6]. Although the availability of two HPV vaccines, Cervarix® or Gardasil®, offer prophylactic protection against a minor fraction of HPV types-associated cervical lesions, they are not effective for existing post-HPV infection lesions [7,8]. What’s more, the cost of vaccination programs makes them inappropriate for developing countries. Despite advances in prevention and treatment of this disease, protocols for recurrence in a refractory form and available treatment options without undesirable side-effects are scanty. In recent years, more effective and less toxic anti-cancer agents from natural resources have become a research hot topic for cancer prevention and treatment based on their ability to attack multiple molecular targets [9]. These highlight an urgent need for development of efficacious plant-derived virus-specific inhibitors to overcome HPV-associated cervical cancer.

Colchicine is a very cheap natural alkaloid derived from either Colchicum autumnale (meadow saffron) or Gloriosa superba (glory lily) that has been utilized in the treatment of gout since 200 years ago [10,11]. It is an antimitotic compound, which has very strong binding capacity to β-tubulin heterodimers in solution to perturb microtubule dynamics, thus leading to cell cycle arrest, apoptosis and cell death [12]. Such inhibition of microtubule formation may contribute to the establishment of improved cancer therapies because cancer cells proliferate rapidly and uncontrollably [13,14]. Latter studies suggested that colchicine has notable effects in cancer cells including cervical cancer [15], which implies that colchicine may have potential as an anti-cancer drug for cervical cancer. Although colchicine exerts potent antimitotic properties, its mechanism of inducing cervical cancer cell apoptosis remains unclear. Understanding the anticancer mechanism of colchicine may lead to new clinical applications in cervical cancer treatment. Therefore, the purpose of the present study was to investigate whether colchicine also had anticancer effects on cervical cancer cells and its possible molecular mechanisms.

Materials and chemicals

Purified Colchicine (99.89%), MTT, and DMSO were purchased from Sigma–Aldrich Co. (St. Louis, MO, U.S.A.). Antibodies against Bcl-2, Bax, cleaved-caspase-3, p53, Rb, phospho Rb (pRb), β-actin and horseradish peroxidase (HRP) secondary conjugated antibodies were obtained from Santa Cruz Biotechnology, Inc., (Santa Cruz, CA, U.S.A.). Dulbecco’s Modified Eagle’s Medium (DMEM), RPMI 1640, fetal bovine serum (FBS), penicillin, and streptomycin were obtained from Cell Signaling Technology (Danvers, MA, U.S.A.). Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit and propidium iodide (PI) were purchased from BD Bioscience (San Jose, CA, U.S.A.).

Cell culture

The human cervical cancer cell lines CaSki (HPV 16 positive), HeLa (HPV 18 positive), C-33A (HPV negative) and primary human epidermal keratinocytes (HEKn, normal) were obtained from the Cell Bank of Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai, PR China) and grown according to their specifications. Cervical cancer cells were cultured in Dulbecco’s modified Eagle’s medium or RPMI 1640 media supplemented with 10% (v/v) heat-inactivated FBS, 100 U/ml penicillin G and 100 mg/ml streptomycin in a humidified atmosphere containing 95% air and 5% CO2 at 37°C. Primary HEKn cells were cultured in basal cell dermal media supplemented with keratinocyte growth kit components and antibiotics, as per conditioned described above.

Cytotoxicity assay

The cytotoxic effects of Colchicine on cell viability were determined with MTT assay [16]. Briefly, cells (10,000 per well) were seeded into 96-well culture plates and incubated with different concentrations of Colchicine for varying period. Subsequently, 20 μl MTT (5 mg/ml) reagents were added into each well containing the untreated and treated cells and incubated at 37°C for another 4 h, followed by the addition of 10 μl DMSO to dissolve the formazan. Then, the absorbance values were measured at 490 nm on an automatic microplate reader (Bio-Rad Benchmark, California, U.S.A.). The effect of Colchicine on growth inhibition was defined as percentage of cell viability, where cells with no treatment were considered 100% viable. Morphological changes in treated or untreated cells were observed using a phase-contrast inverse microscope (Olympus, Japan).

Annexin V/propidium iodide assay for apoptosis

Apoptosis was detected using an annexin V-FITC apoptosis detection kit according to the manufacturer’s instructions. In brief, treated or untreated cells were harvested, washed twice with cold phosphate-buffered saline, and then stained with 10 μl Annexin V- FITC and 10 μl of PI (20 μg/ml) in the dark at room temperature for 20 min. Apoptotic cells were analyzed immediately by BD FACScan flow cytometer (Becton Dickinson, San Jose, CA) with CellQuest 3.0 software. For each sample, the fluorescence of 10,000 cells was gated and counted. The degree of apoptosis was quantified as a percentage of the annexin V positive and PI-negative (annexin V+/PI) cells [17].

Quantitative real-time reverse transcription PCR (qRT-PCR) analysis of HPV16 E6 and E7 transcripts

Following different treatment, both adherent and floating cells were collected and frozen at −80°C until analysis. Total cellular RNAs were isolated with TRIzol reagent as per manufacturer’s protocol. RNA concentration and purity were evaluated on an ultraviolet spectrophotometer (Bio-Rad Inc., Hercules, CA, U.S.A.) by measuring the ratio of absorbance at 260 and 280 nm. qRT-PCR for the HPV E6, E7 mRNA and internal standard β-actin were performed on an ABI 7500 Fast RT-PCR system (Applied Biosystems, Foster City, CA, U.S.A.) using SuperScript III Platinum SYBR Green One-step RT-PCR Kit (Invitrogen, Carlsbad, California, U.S.A.) with the following procedures: 50°C for 4 min, 95°C for 14 min, followed by 40 cycles of 95°C for 15 s and 60°C for 31s. The primer sequences were as list below: HPV16 E6, forward: 5′-CTGCAATGTTTCAGGACCCA-3′ and reverse: 5′-TCATGTATAGTTGTGCAGCTCTGT-3′; HPV16 E7, forward: 5′-GAGGA GGAGGATGAAATAGATGGT-3′ and reverse: 5′-CACTTGCAACAAAACGTT ACAATATTG-3′; HPV18 E6, forward: 5′-AAGATTTATTTGTGGTGT-3′ and reverse: 5′-GGTGGATTG-3′; HPV18 E7, forward: 5′-CACGTAGAGAAACCCAGCTGTAA-3′ and reverse: 5′-GCAGGATCAGCCATGGTAGATT-3′; β-actin, forward: 5′- CCAACCGCGAGAAGATGA-3′ and reverse: 5′- CCAGAGGCGTACAGGGATAG-3′. Each measurement was performed in triplicate, and no-template controls were included for each assay. After completion of PCR, a dissociation curve analysis was done and Ct values were obtained from the ABI 7500 fast v2.0.1 software. Relative mRNA fold change was calculated using the 2−ΔΔCT method [18].

Protein extraction and Western blot analysis

Cells with indicated treatments were lysed with ice-cold lysis buffer (50 mM Tris-HCl, 150 mM NaCl, 1% NP40, 100 mM DTT, 0.1% SDS and 10% glycerol). The protein concentration was quantified using the BCA method. A total of 25 μg protein was separated using 10–15% SDS-PAGE and then transferred onto polyvinylidene fluoride membranes. The membrane was blocked with 5% nonfat milk in TBST solution and incubated overnight at 4°C with primary antibodies against Bcl-2 (1:1000), Bax (1:1000), cleaved-caspase-3 (1:500), p53 (1:1,000), Rb (1:1000), pRb (1:1000) and β-actin (1:2000), followed by incubation with HRP-conjugated second antibodies. Protein bands were visualized using the enhanced chemiluminescence detection system (ECL, Amersham Biosciences, Buckinghamshire, U.K.). The relative protein levels were calculated based on β-actin protein as a loading control.

Statistical analysis

Results are expressed as mean of three individual experiments ± standard deviation (SD), and data were analyzed using the GraphPad Prism version 6.01 statistical program (GraphPad Software, Inc.). Statistical differences between groups were evaluated using one-way analysis of variance (ANOVA) followed by the Student’s t-tests. P < 0.05 was considered to indicate a statistically significant result.

Cytotoxicity of Colchicine on CaSki, HeLa, C-33A and HEKn cells

To determine the cytotoxic effect of Colchicine on cell growth of different HPV cervical cancer cell lines, CaSki (HPV 16 positive), HeLa (HPV 18 positive), C-33A (HPV negative) and normal HEKn cells exposed to increasing doses of Colchicine for 24, 48 and 72 h, and cell viability was examied by the MTT assay. The results showed that the cell viabilities of CaSki and HeLa were markedly decreased after exposure to Colchicine in dose- and time-responsive manners. It was observed that Colchicine decreased the growth of CaSki cells by 12.21% (2.5 ng/ml), 23.02% (5.0 ng/ml, P < 0.05) and 32.25% (10.0 ng/ml, P < 0.05) at 24 h, by 28.21% (2.5 ng/ml, P < 0.05), 42.62% (5.0 ng/ml, P < 0.01) and 54.12% (10.0 ng/ml, P < 0.01) at 48 h, and by 32.50% (2.5 ng/ml, P < 0.05), 47.14% (5.0 ng/ml, P < 0.01) and 58.25% (10.0 ng/ml, P < 0.01) at 72 h, respectively, compared with the untreated control cells (Figure 1A). Similarly, HeLa cells exhibited, 17.34% (2.5 ng/ml, P < 0.05), 25.05% (5.0 ng/ml, P < 0.05) and 34.98% (10.0 ng/ml, P < 0.01) decrease in the cell growth at 24 h, 30.25% (2.5 ng/ml, P < 0.05), 43.21% (5.0 ng/ml, P < 0.01) and 57.02% (10.0 ng/ml, P < 0.01) decrease at 48 h, and 32.68% (2.5 ng/ml, P < 0.05), 40.37% (5.0 ng/ml, P < 0.01) and 59.67% (10.0 ng/ml, P < 0.01) decrease at 72 h, respectively, compared with the untreated control cells (Figure 1B). We observed that the inhibitory effect of Colchicine on cell proliferation of CaSki or HeLa cells at 48 h reached the same potent efficiency at 72 h, thus we choose the 48-h incubation time as following assay. Nevertheless, no cell viability inhibitory effect was observed in C-33A cervical cancer cells and HEKn normal cells following Colchicine treatment in the same condition (Figure 1C,D). This result indicated that Colchicine exhibited selective toxicity for HPV positive CaSki and HeLa cervical cancer cells.

Cytotoxicity of Colchicine on CaSki, HeLa, C-33A and HEKn cells

Figure 1
Cytotoxicity of Colchicine on CaSki, HeLa, C-33A and HEKn cells

(A) The effect of Colchicine on cell viability of CaSki cells. (B) The effect of Colchicine on cell viability of HeLa cells. (C) The effect of Colchicine on cell viability of C-33A cells. (D) The effect of Colchicine on cell viability of HEKn cells. Values are expressed as mean ± S.D. of three independent experiments. *P <0.05 and **P <0.01 compared with control.

Figure 1
Cytotoxicity of Colchicine on CaSki, HeLa, C-33A and HEKn cells

(A) The effect of Colchicine on cell viability of CaSki cells. (B) The effect of Colchicine on cell viability of HeLa cells. (C) The effect of Colchicine on cell viability of C-33A cells. (D) The effect of Colchicine on cell viability of HEKn cells. Values are expressed as mean ± S.D. of three independent experiments. *P <0.05 and **P <0.01 compared with control.

Close modal

Effects of Colchicine on the apoptosis in CaSki and HeLa cells

To determin whether Colchicine induced inhibition of cell proliferation is related to the induction of apoptosis, an annexin V-FITC apoptosis detection kit was used to quantify the rate of apoptosis in both cervical cancer cells. Flow cytometry revealed that the number of apoptotic cells was increased in response to treatment with Colchicine by 25.22% (2.5 ng/ml), 40.12% (5.0 ng/ml) and 53.92% (10.0 ng/ml) for CaSki cells, and 28.95% (2.5 ng/ml), 42.85% (5.0 ng/ml) and 56.35% (10.0 ng/ml) for HeLa cells, as compared with the control group, respectively (Figure 2A,B). Therefore, the antiproliferation activity of Colchicine on CaSki or HeLa cells was related to apoptosis.

Effects of Colchicine on the apoptosis in CaSki and HeLa cells

Figure 2
Effects of Colchicine on the apoptosis in CaSki and HeLa cells

(A) The effect of Colchicine on apoptosis of CaSki cells. (B) The effect of Colchicine on apoptosis of HeLa cells. Values are expressed as mean ± S.D. of three independent experiments.

Figure 2
Effects of Colchicine on the apoptosis in CaSki and HeLa cells

(A) The effect of Colchicine on apoptosis of CaSki cells. (B) The effect of Colchicine on apoptosis of HeLa cells. Values are expressed as mean ± S.D. of three independent experiments.

Close modal

Effect of Colchicine on the expression of HPV E6 and E7 in CaSki and HeLa cells

Colchicine exhibited significant sensitive toxicity to both HPV16 (CaSki) and HPV18 (HeLa) positive cell lines via induction of apoptosis, so next we aimed to investigate the expression of the viral E6 and E7 gene or proteins in two cell lines by qRT-PCR or Western blot, respectively. There was a significant dose-dependent reduction of E6 and E7 mRNA or oncoproteins in both the cervical cancer cell lines, CaSki (Figure 3A,C) and HeLa (Figure 3B,D), compared with those in the untreated control cells (P < 0.05, P < 0.01 or P < 0.001). All these results suggested decreased the expression of the HPV E6 and E7 were involved in the antitumor effect of Colchicine toward CaSki and HeLa cells.

Effect of Colchicine on the expression of HPV E6 and E7 in CaSki and HeLa cells

Figure 3
Effect of Colchicine on the expression of HPV E6 and E7 in CaSki and HeLa cells

(A) The effect of Colchicine on the mRNA expression of E6 and E7 in CaSki cells. (B) The effect of Colchicine on the mRNA expression of E6 and E7 in HeLa cells. (C) The effect of Colchicine on the protein expression of E6 and E7 in CaSki cells. (D) The effect of Colchicine on the protein expression of E6 and E7 in HeLa cells. Values are expressed as mean ± S.D. of three independent experiments. *P <0.05, **P <0.01 and ***P <0.001 compared with control.

Figure 3
Effect of Colchicine on the expression of HPV E6 and E7 in CaSki and HeLa cells

(A) The effect of Colchicine on the mRNA expression of E6 and E7 in CaSki cells. (B) The effect of Colchicine on the mRNA expression of E6 and E7 in HeLa cells. (C) The effect of Colchicine on the protein expression of E6 and E7 in CaSki cells. (D) The effect of Colchicine on the protein expression of E6 and E7 in HeLa cells. Values are expressed as mean ± S.D. of three independent experiments. *P <0.05, **P <0.01 and ***P <0.001 compared with control.

Close modal

Effects of Colchicine on the expression of p53, Rb and pRb proteins

Considering the repression of oncoproteins E6 and E7, we thus examined the effect of Colchicine on the expression of p53, Rb and pRb protiens in both cervical cancer cells. The results showed that Colchicine dose-dependently up-regulated p53 and Rb protein expressions in HPV-positive CaSki cells or HeLa cells (Figure 4A,B). In contrast, the expression of pRb was decreased dose dependently in both cancer cells. Interestingly, no change was observed on p53 and Rb proteins level in HPV-negtive C-33A cells (data not shown). These results confirmed that reduced expression of E6 and E7 is the key contributor to Colchicine-mediated up-regulation of p53 and Rb.

Effects of Colchicine on the expression of p53, Rb and pRb proteins in CaSki and HeLa cells

Figure 4
Effects of Colchicine on the expression of p53, Rb and pRb proteins in CaSki and HeLa cells

(A) The effect of Colchicine on the protein expression of p53, Rb and pRb in CaSki cells. (B) The effect of Colchicine on the protein expression of p53, Rb and pRb in HeLa cells. Values are expressed as mean ± S.D. of three independent experiments. *P <0.05, **P <0.01 and ***P <0.001 compared with control.

Figure 4
Effects of Colchicine on the expression of p53, Rb and pRb proteins in CaSki and HeLa cells

(A) The effect of Colchicine on the protein expression of p53, Rb and pRb in CaSki cells. (B) The effect of Colchicine on the protein expression of p53, Rb and pRb in HeLa cells. Values are expressed as mean ± S.D. of three independent experiments. *P <0.05, **P <0.01 and ***P <0.001 compared with control.

Close modal

Effects of Colchicine on apoptosis-related protein in CaSki and HeLa cells

To further explore the effects of Colchicine on the apoptosis of CaSki and HeLa cells, the present study determined the altered expression levels of apoptosis-associated proteins by Western blotting. When both cervical cancer cells were treated with 2.5, 5.0 and 10.0 ng/ml of Colchicine for 48 h, the protein expression levels of Bax, cleaved caspase 3 and cytochrome c in cytosolic fraction were increased in a dose-dependent manner, whereas the Bcl-2 protein expression was in the opposite manner (Figure 5A,B).

Effects of Colchicine on apoptosis-related protein in CaSki and HeLa cells

Figure 5
Effects of Colchicine on apoptosis-related protein in CaSki and HeLa cells

(A) The effect of Colchicine on the protein expression of Bax, Bcl-2, cleaved caspase 3 and cytoplasmic cytochrome c in CaSki cells. (B) The effect of Colchicine on the protein expression of Bax, Bcl-2, cleaved caspase 3 and cytoplasmic cytochrome c in HeLa cells.

Figure 5
Effects of Colchicine on apoptosis-related protein in CaSki and HeLa cells

(A) The effect of Colchicine on the protein expression of Bax, Bcl-2, cleaved caspase 3 and cytoplasmic cytochrome c in CaSki cells. (B) The effect of Colchicine on the protein expression of Bax, Bcl-2, cleaved caspase 3 and cytoplasmic cytochrome c in HeLa cells.

Close modal

During cervical carcinogenesis, an increasing imbalance between proliferation and apoptosis (programmed cell death) has been observed, when compared with normal cervical epithelium. HPV is the major cause of development of cervical cancer. As the prevalence of HPV infection increased, the incidence of cervical cancer increased rapidly [19,20]. HPVs included many subtype, such as HPV 16, 18, 31 and 33, out of which HPV-16 and HPV-18 are the most common genotypes in malignant biopsies with little geographic variation and account for almost 70% of the cancers [21,22]. Two HPV encoded viral oncoproteins, E6 and E7, are essential for cervical carcinogenesis by their potential to inactivate the tumor suppressor proteins p53 and retinoblastoma protein (pRb), respectively [23]. These tumor suppressor proteins are involved in many signaling pathways that regulate the cell cycle and monitor and protect the integrity of the genome [24,25]. The anti-oncogene p53 has long been involved in tumor necrosis factor (TNF)-α-mediated apoptosis, which is achieved by induction of cell cycle G1 arrest and inhibition of cell proliferation, thus leading eventually to apoptosis and cell death [26,27]. Despite the critical role of p53 in cell survival, characterization of the downstream signaling events remains largely undefined at present. As of now, there is a cross-talk between the p53 and TNF-α mediated pathway. Once transcription factor NF-κB-induced threshold level of p53 is reached as a result of TNF-α stimulation, the cells will be definitely committed to apoptosis [27–29]. Moreover, continual cell stress induced by TNF-α may actually lead to up-regulation of p53 and the inactivation of IκB kinase 2 (IKK2 or IKKβ), which is responsible for triggering the nuclear import of the NF-κB [26,30]. Thereof, the resulting NF-κB suppression is able to inactivate the transcription of genes that regulate a variety of fundamental biological processes, including apoptosis [30]. Unlike other kind of cancers, where the p53 gene is mutated irreversibly, cervical cancer cell lines uniquely harbor wild-type p53 and Rb genes [31]. In this case, the growth-regulatory machinery of normal cells is masked by the expression of HPV E6 and E7 proteins [32]. To this end, HPV E6 and E7 oncoproteins have become a target for cervical cancer prevention and treatment. Although HPV vaccines against two main stains of HPV (16 and 18) by large pharmaceutical companies block the development of cervical cancer in a great degree, an estimated cost at $300 for a person in vaccination programs will prohibit the access of most women in developing countries to obtain these vaccines [33]. Down-regulation of viral HPV E6 and E7 represents a viable strategy to induce functional p53 for cervical cancer therapies, which would in turn further induce downstream target genes involved in cell circle arrest and apoptosis and thus restore growth control in tumor cells or sensitize cells to cancer therapies [34]. Actually, consistent with this notion, there has been a growing body of evidence to suggest that the possibility of reactivating the p53 pathway has been extensively studied in several cancers [35,36].

In our screening for drug candidates exhibiting inhibitory activity against HPV, we found that Colchicine displayed dramatic growth inhibitory effect on HPV-16 (CaSki) or -18 (HeLa) positive cervical cancer cell lines, but not on HPV negative (C-33A) cell line. The antiproliferative effect of Colchicine was clearly associated with induction of apoptosis, as evidenced by the increasing percentage of apoptotic CaSki or HeLa cells after Annexin/PI staining on flow cytometer. These results showed that CaSki and HeLa cells treated with Colchicine undergo typical apoptosis. However, these changes did not occur in HPV negative C-33A and normal HEKn cells. Furthermore, RT-PCR and Western blotting indicated that Colchicine dose-dependently inhibited both mRNA and proteins expression of HPV E6 and E7 in CaSki and HeLa cells, respectively, confirming that Colchicine might have potential inhibitory activity against cervical cancer associated with HPV. We next examined if repression of HPV E6 and E7 oncogenes resulted in reactivation of dormant tumor suppressor p53 and Rb. It was evident that Colchicine dose-dependently increased the expression of p53 and Rb tumor suppressor proteins in CaSki and HeLa cells, but decreased pRb protein expression. At the same time, the tumor suppressor protein levels in the HPV negative cell lines C-33A largely remained unaltered. Hence, we presumed that Colchicine did not directly change p53 and Rb expression, and the resulting up-regulation in both cancer cells was caused by the down-regulation of E6 and E7.

The restoration of tumor suppressor functions of p53 proteins might lead to the activation of downstream signaling molecules, such as Bax, Bcl-2, cytochrome c and cleaved caspase-3, thus finally causing either cell cycle arrest or apoptosis [37]. The change of these apoptosis-related proteins is regulated by p53 via the protein–protein interaction and it is known as the intrinsic apoptotic pathway or mitochondrial-mediated pathway [38–40]. Western blotting indicated that the addition of Colchicine stimulated the protein expression of Bax, cytochrome c and cleaved caspase-3, but inhibited the expression of Bcl-2 protein in CaSki and HeLa cells. These results suggested that the antitumor effect of Colchicine on CaSki and HeLa cells was associated with the p53-dependent intrinsic apoptotic pathway.

In summary, the present study demonstrated that Colchicine has profound in vitro antiproliferative activity against HPV-16 or -18 positive human cervical cancer cell lines, which was achieved by HPV E6/E7 inhibition and subsequently p53-dependent intrinsic apoptosis. Our findings suggest Colchicine may be exploited as a promising candidate agent for HPV associated cervical cancer prevention and treatment. Further more work including in vivo studies are required to further elucidate the exact mechanism and therapeutic effects.

The authors declare that there are no competing interests associated with the manuscript.

The authors declare that there are no sources of funding to be acknowledged.

L.C.Y. and X.P.L. designed the study and reviewed the draft. L.C.Y. and H.H. conducted the analyses and writed the original draft. Y.Z. and X.R.Y. completed the methodology of study. Z.H.Y. and C.Y.C. collected the data and done the work of statistics.

HEKn

human epidermal keratinocytes

HPV

human papilloma virus

pRb

phospho Rb

TNF

tumor necrosis factor

1.
Bruni
L.
,
Barrionuevo-Rosas
L.
,
Albero
G.
,
Aldea
M.
,
Serrano
B.
and
Valencia
S.
(
2014
)
ICO Information Centre on HPV and Cancer (HPV Information Centre)
.
Human Papillomavirus Related Diseases World
Summary Report 2015 04 08
2.
Schiffman
M.
,
Wentzensen
N.
,
Wacholder
S.
,
Kinney
W.
,
Gage
J.C.
and
Castle
P.E.
(
2011
)
Human papillomavirus testing in the prevention of cervical cancer
.
J. Natl. Cancer Inst.
103
,
368
383
[PubMed]
3.
Jemal
A.
,
Bray
F.
,
Center
M.M.
,
Ferlay
J.
,
Ward
E.
and
Forman
D.
(
2011
)
Global cancer statistics
.
CA Cancer J. Clin.
61
,
69
90
[PubMed]
4.
Parkin
D.M.
(
2001
)
Global cancer statistics in the year 2000
.
Lancet Oncol.
2
,
533
543
[PubMed]
5.
Fowler
R.S.
(
2000
)
Vulvar vestibulitis: response to hypocontactant vulvar therapy
.
J. Lower Genital Tract Disease
4
,
200
203
[PubMed]
6.
Hougardy
B.M.
,
Maduro
J.H.
,
van der Zee
A.G.
,
Willemse
P.H.
,
de Jong
S.
and
de Vries
E.G.
(
2005
)
Clinical potential of inhibitors of survival pathways and activators of apoptotic pathways in treatment of cervical cancer: changing the apoptotic balance
.
Lancet Oncol.
6
,
589
598
[PubMed]
7.
Stanley
M.
(
2010
)
Potential mechanisms for HPV vaccine-induced long-term protection
.
Gynecol. Oncol.
118
,
S2
S7
[PubMed]
8.
Pogoda
C.S.
,
Roden
R.B.
and
Garcea
R.L.
(
2016
)
Immunizing against anogenital cancer: HPV vaccines
.
PLoS Pathog.
12
,
e1005587
[PubMed]
9.
Gordaliza
M.
(
2007
)
Natural products as leads to anticancer drugs
.
Clin. Transl. Oncol.
9
,
767
776
[PubMed]
10.
Sivakumar
G.
(
2013
)
Colchicine semisynthetics: chemotherapeutics for cancer?
Curr. Med. Chem.
20
,
892
898
[PubMed]
11.
Schlesinger
N.
,
Schumacher
R.
,
Catton
M.
and
Maxwell
L.
(
2006
)
Colchicine for acute gout
.
Cochrane Database Syst. Rev.
4
,
CD006190
[PubMed]
12.
Bhattacharyya
B.
,
Panda
D.
,
Gupta
S.
and
Banerjee
M.
(
2008
)
Anti‐mitotic activity of colchicine and the structural basis for its interaction with tubulin
.
Med. Res. Rev.
28
,
155
183
[PubMed]
13.
Risinger
A.L.
,
Giles
F.J.
and
Mooberry
S.L.
(
2009
)
Microtubule dynamics as a target in oncology
.
Cancer Treat. Rev.
35
,
255
261
[PubMed]
14.
Jordan
M.A.
and
Wilson
L.
(
2004
)
Microtubules as a target for anticancer drugs
.
Nat. Rev. Cancer
4
,
253
[PubMed]
15.
Feletig
P.
(
1955
)
Karyological study on the antimitotic action of colchicine in cervical cancer
.
Tumori
41
,
467
480
[PubMed]
16.
Zhai
Q.
,
Li
X.
,
Yang
Y.
,
Yu
L.
and
Yao
Y.
(
2014
)
Antitumor activity of a polysaccharide fraction from Laminaria japonica on U14 cervical carcinoma-bearing mice
.
Tumor Biol.
35
,
117
122
[PubMed]
17.
Lee
P.Y.
,
Park
B.C.
,
Chi
S.W.
,
Bae
K.H.
,
Kim
S.
,
Cho
S.
et al.
(
2016
)
Histone H4 is cleaved by granzyme A during staurosporine-induced cell death in B-lymphoid Raji cells
.
B.M.B. Rep.
49
,
560
[PubMed]
18.
Livak
K.J.
and
Schmittgen
T.D.
(
2001
)
Analysis of relative gene expression data using real-time quantitative PCR and the 2− ΔΔCT method
.
Methods
25
,
402
408
[PubMed]
19.
Munoz
N.
(
2000
)
Human papillomavirus and cancer: the epidemiological evidence
.
J. Clin. Virol.
19
,
1
5
[PubMed]
20.
Bosch
F.X.
and
de Sanjosé
S.
(
2007
)
The epidemiology of human papillomavirus infection and cervical cancer
.
Dis. Markers
23
,
213
227
[PubMed]
21.
Roa
J.C.
,
Garcia
P.
,
Gomez
J.
,
Fernández
W.
,
Gaete
F.
,
Espinoza
A.
et al.
(
2009
)
HPV genotyping from invasive cervical cancer in Chile
.
Int. J. Gynecol. Obst.
105
,
150
153
22.
Bruni
L.
,
Barrionuevo-Rosas
L.
,
Albero
G.
,
Aldea
M.
,
Serrano
B.
and
Valencia
S.
(
2014
)
ICO Information Centre on HPV and Cancer (HPV Information Centre)
.
Human Papillomavirus Related Diseases World.
Summary Report 2015 04 08
23.
Zur Hausen
H.
(
1996
)
Papillomavirus infections—a major cause of human cancers
.
Biochim. Biophys. Acta
1288
,
F55
F78
24.
Yeung
C.A.
,
Tsang
T.Y.
,
Yau
P.L.
and
Kwok
T.T.
(
2011
)
Human papillomavirus type 16 E6 induces cervical cancer cell migration through the p53/microRNA-23b/urokinase-type plasminogen activator pathway
.
Oncogene
30
,
2401
[PubMed]
25.
Nevins
J.R.
(
2001
)
The Rb/E2F pathway and cancer
.
Hum. Mol. Genet.
10
,
699
703
[PubMed]
26.
Rokhlin
O.W.
,
Gudkov
A.V.
,
Kwek
S.
,
Glover
R.A.
,
Gewies
A.S.
and
Cohen
M.B.
(
2000
)
p53 is involved in tumor necrosis factor-α-induced apoptosis in the human prostatic carcinoma cell line LNCaP
.
Oncogene
19
,
1959
1968
[PubMed]
27.
Yeung
M.C.
and
Lau
A.S.
(
1998
)
Tumor suppressor p53 as a component of the tumor necrosis factor-induced, protein kinase PKR-mediated apoptotic pathway in human promonocytic U937 cells
.
J. Biol. Chem.
273
,
25198
25202
[PubMed]
28.
Schneider
G.
,
Henrich
A.
,
Greiner
G.
,
Wolf
V.
,
Lovas
A.
,
Wieczorek
M.
et al.
(
2010
)
Cross talk between stimulated NF-κB and the tumor suppressor p53
.
Oncogene
29
,
2795
2806
[PubMed]
29.
Liu
Y.
,
Tergaonkar
V.
,
Krishna
S.
and
Androphy
E.J.
(
1999
)
Human papillomavirus type 16 E6-enhanced susceptibility of L929 cells to tumor necrosis factor α correlates with increased accumulation of reactive oxygen species
.
J. Biol. Chem.
274
,
24819
24827
[PubMed]
30.
Irelan
J.T.
,
Murphy
T.J.
,
DeJesus
P.D.
,
Teo
H.
,
Xu
D.
,
Gomez-Ferreria
M.A.
et al.
(
2007
)
A role for IκB kinase 2 in bipolar spindle assembly
.
Proc. Natl. Acad. Sci. U.S.A.
104
,
16940
16945
31.
Abdulkarim
B.
,
Sabri
S.
,
Deutsch
E.
,
Chagraoui
H.
,
Maggiorella
L.
,
Thierry
J.
et al.
(
2002
)
Antiviral agent Cidofovir restores p53 function and enhances the radiosensitivity in HPV-associated cancers
.
Oncogene
21
,
2334
[PubMed]
32.
Scheffner
M.
,
Münger
K.
,
Byrne
J.C.
and
Howley
P.M.
(
1991
)
The state of the p53 and retinoblastoma genes in human cervical carcinoma cell lines
.
Proc. Natl. Acad. Sci. U.S.A.
88
,
5523
[PubMed]
33.
Capell
K.
and
Barrett
A.
(
2004
)
A vaccine every women should take
.
Business Week Online
34.
Vousden
K.H.
and
Lu
X.
(
2002
)
Live or let die: the cell's response to p53
.
Nat. Rev. Cancer
2
,
594
[PubMed]
35.
Bykov
V.J.
and
Wiman
K.G.
(
2003
)
Novel cancer therapy by reactivation of the p53 apoptosis pathway
.
Ann. Med.
35
,
458
465
[PubMed]
36.
Goodwin
E.C.
and
DiMaio
D.
(
2000
)
Repression of human papillomavirus oncogenes in HeLa cervical carcinoma cells causes the orderly reactivation of dormant tumor suppressor pathways
.
Proc. Natl. Acad. Sci. U.S.A.
97
,
12513
12518
37.
Choudhari
A.S.
,
Suryavanshi
S.A.
and
Kaul-Ghanekar
R.
(
2013
)
The aqueous extract of Ficus religiosa induces cell cycle arrest in human cervical cancer cell lines SiHa (HPV-16 Positive) and apoptosis in HeLa (HPV-18 Positive)
.
PLoS ONE
8
,
e70127
[PubMed]
38.
Sangthong
S.
,
Sangphech
N.
,
Palaga
T.
,
Ngamrojanavanich
N.
,
Puthong
S.
,
Vilaivan
T.
et al.
(
2014
)
Anthracene-9, 10-dione derivatives induced apoptosis in human cervical cancer cell line (CaSki) by interfering with HPV E6 expression
.
Eur. J. Med. Chem.
77
,
334
342
[PubMed]
39.
Bava
S.V.
,
Puliappadamba
V.T.
,
Deepti
A.
,
Nair
A.
,
Karunagaran
D.
and
Anto
R.J.
(
2005
)
Sensitization of taxol-induced apoptosis by curcumin involves down-regulation of nuclear factor-κB and the serine/threonine kinase Akt and is independent of tubulin polymerization
.
J. Biol. Chem.
280
,
6301
6308
[PubMed]
40.
Teissier
S.
,
Khalifa
Y.B.
,
Mori
M.
,
Pautier
P.
,
Desaintes
C.
and
Thierry
F.
(
2007
)
A new E6/P63 pathway, together with a strong E7/E2F mitotic pathway, modulates the transcriptome in cervical cancer cells
.
J. Virol.
81
,
9368
9376
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).