Anti-proliferative and pro-apoptotic effects of 3,3'-diindolylmethane in human cervical cancer cells

  • Authors:
    • Junyong Zhu
    • Yuan Li
    • Chao Guan
    • Zuhua Chen
  • View Affiliations

  • Published online on: June 20, 2012     https://doi.org/10.3892/or.2012.1877
  • Pages: 1063-1068
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The antitumor effects of Indo-3-carbinol (I3C) have been proven in many human carcinoma cells. However, the roles of 3,3-diindolylmethane (DIM), an important polymer converted from I3C under pH 5.0-7.0, on the growth and proliferation of cervical cancer HeLa and SiHa cells still remain unrevealed. In the present study, we investigated the potential anti-proliferative and pro-apoptotic effects of DIM on HeLa and SiHa cells. Cell proliferation was detected by Cell Counting kit-8 and apoptosis was analyzed by flow cytometry. In addition, morphological changes accompanying cell apoptosis were observed using an inverted microscope after Hoechst 33258 staining. In addition, expression changes of proteins involved in the MAPK and PI3K pathways were determined by western blotting. DIM treatment inhibited the proliferation and induced apoptosis of HeLa and SiHa cells significantly in a time- and dose-dependent manner. Moreover, SiHa cells were more sensitive to DIM treatment than HeLa cells (P<0.05). In addition, the expression of ERK, p38 and p-p38, which are involved in the MAPK pathway, was downregulated by DIM treatment. Another protein involved in the MAPK pathway, JNK, was upregulated. Furthermore, DIM treatment significantly suppressed the expression of Akt, p-Akt, PI3K p110α, PI3K p110β, PI3K class III, GSK3-β, p-PDK1 and p-c-Raf which are involved in the PI3K pathway. These results demonstrate that DIM exerts antitumor effects on HeLa and SiHa cells through its anti-proliferative and pro-apoptotic roles, especially for SiHa cells. The molecular mechanism for these effects may be related to its regulatory effects on MAPK and PI3K pathway and apoptosis proteins. DIM may be a preventive and therapeutic agent against cervical cancer.

Introduction

Cervical carcinoma is the second most common malignancy of female reproductive tract and ranks second among female deaths caused by cancers. More than 50 million people worldwide suffer from this disease each year. Although early diagnosis of cervical cancer has made great progress, the incidence and mortality of this disease still remains high, and the onset age is becoming younger, seriously threatening women's health and lives. The treatments mainly depend on surgery and radiotherapy, supplemented with chemotherapy. Five-year survival rate of terminally-ill patients is only 50% although therapeutic techniques have been improved greatly in the past years (1). It was reported that the poor effects of chemotherapy on advanced stage of cervical cancer may be directly due to obstacles in apoptosis of tumor cells (2).

Indole-3-carbinol (I3C), a type of compound derived from cruciferous vegetables (3), was demonstrated to exhibit significant anticancer efficacy both in vivo and in vitro by inducing cell apoptosis and cell cycle arrest in G1 phase (4,5), affecting DNA damage repair (68), as well as resisting angiogenesis, metastasizing and invasion of cancer cells (9,10). 3,3′-diindolylmethane (DIM), an important polymer converted from I3C under pH 5.0–7.0 (11), was characterized with anti-proliferative and pro-apoptotic activities in cancer cells (12,13). Chinnakannu et al(13) reported that a formulated DIM (B-DIM) could inactivate NF-κB signaling and induce apoptosis through inhibiting proteasome activity in S phase of LNCaP and C4–2B cells, two types of prostate cancer cells, suggesting that B-DIM could be a potent agent for prevention and/or treatment of both hormone-sensitive and hormone-refractory prostate cancers. I3C-6, a derivative of I3C, was proven to significantly inhibit the growth and migration of HeLa cells (adenocarcinoma type) (14). However, there are no reports on anti-tumor effects of DIM in cervical carcinoma cells, or comparisons of its effects on two different types of cervical cancer cells, including HeLa cells (adenocarcinoma type) and SiHa cells (squamous carcinoma). The present study aimed to investigate the potential anticancer effects of DIM in cervical carcinoma cells in vitro and compare its different influence on HeLa cells and SiHa cells, thus providing useful information for clinical therapies of cervical carcinoma.

Materials and methods

Reagents

Cell Counting kit-8 (CCK-8) was purchased from Dongji (Japan). All antibodies were purchased from Cell Signaling Technology (USA), including p-p38, t-p38, p-ERK, t-ERK, p-JNK, t-JNK, PI3K p85, PI3K p110α, PI3K p110β, p-Akt, t-Akt, p-PDK1, p-c-Raf and GAPDH. Annexin V FITC/PI Apoptosis Detection kit was from Lianke (China).

Cell cultures

The HeLa cells and SiHa cells, kindly provided by the Type Culture Collection of Chinese Academy of Sciences, were maintained in RPMI-1640 medium (Sigma, USA) supplemented with 10% fetal bovine serum (FBS) (Hyclone, USA) and 20 μg/ml antibiotics (ampicillin and kanamycin). The cultivations were performed in a humidified atmosphere of 5% CO2 at 37°C. The medium was changed at 48-h intervals and culture transfers were performed every 4 days when the culture almost reached confluence.

Measurement of cell proliferation

Cells were harvested by trypsinization at their logarithmic growth phases and plated in 96-well microwell plates in 100 μl RPMI-1640 medium with 10% FBS at a density of 1×103 cells/well. Cells were treated with DIM at different concentrations (25, 50 and 100 μM) after 24 h of incubation and continuously cultured for 0, 24, 48 and 72 h before proliferation analysis. Cell proliferation assays were performed using the WST-8 based Colorimetric Assay Cell Counting kit-8 (CCK-8). In addition, proliferation was also measured in parallel negative groups without DIM treatment and the blank groups without cells in the wells. Six repeats were prepared for each group. Quantification of viable cells was performed by measuring the absorbance at 450 nm (A450) using a microplate reader (Beckman, USA). The following formula was used to calculate the inhibitory rate of DIM on cell proliferation: inhibitory rate (%) = [A450 (negative group) − A450 (treated group)] / [A450 (negative group) − A450 (blank group)] × 100%.

Apoptosis analysis

After DIM treatment for 48 h, HeLa and SiHa cells were harvested by trypsinization (≥1×104) and washed twice with cold phosphate-buffered saline (PBS). The cells resuspended in 200 μl annexin-V binding buffer were incubated in 5 μl of annexin-V fluorescein isothiocyanate (FITC) solution and 10 μl of propidium iodide (PI) solution for 15 min in the dark at room temperature. The fluorescences of each group were analyzed on a FACSCanto™II spectrometer (BD Biosciences, San Jose, CA, USA) within 1 h and the cells stained with FITC/PI were counted as apoptotic cells.

Western blot analysis

Proteins were extracted from the cultured cells and loaded onto a polyacrylamide gel (10%). Then electrophoresis was performed at room temperature at 50 V for 30 min and then 200 V for 1.5 h, followed by transferring to nitrocellulose membranes. The membranes were incubated with blocking solution for 2 h at room temperature, and then incubated with different primary antibodies including p-p38, t-p38, p-ERK, t-ERK, p-JNK, t-JNK, PI3K p85, PI3K p110α, PI3K p110β, p-Akt, t-Akt, p-PDK1, p-c-Raf and GAPDH (Cell Signaling) for 12 h at 4°C. After washing for 1 h with 20 mM Tris·HCl and 0.5 M NaCl (TBS) with 0.05% Tween-20 (TTBS), membranes were incubated with the secondary antibody conjugated to horseradish peroxidase for 30 min at room temperature and washed again for 1 h. GAPDH was used as an internal reference. Protein bands were finally colored with enhanced chemiluminescence (ECL) reagent (Thermo, USA) and exposed to X-ray.

Statistical analysis

Data are expressed as mean ± SD. Difference in the effects of DIM on HeLa and SiHa cells were compared with one-way ANOVA (SPSS statistical software, SPSS Inc., Chicago, IL, USA). P<0.05 (two-tailed) was considered statistically significant.

Results

Effects of DIM on the proliferations of HeLa and SiHa cells

HeLa and SiHa cells were treated with various concentrations (0, 25, 50 and 100 μM) of DIM for 0–72 h. Cell proliferation was assessed by the CCK-8 method and represented by absorbance value at 450 nm. DIM showed a dose- and time-dependent anti-proliferative effect on the growth of HeLa and SiHa cells (Fig. 1).

DIM showed a profound dose-dependent inhibition of the growth of HeLa and SiHa cells with the inhibitory rates ranging from 14.61 to 93.03 (Table I). It was noticeable that the inhibitory effects of DIM on SiHa cells were much stronger than that on HeLa cells after 48 h of treatment in each concentration group (except for 50 μM group after 48 h of treatment). In addition, after DIM treatment for 48 h, the calculated IC50 of DIM for HeLa and SiHa cells were 46.8 and 44.44 μM respectively, suggesting a much more sensitive response of SiHa cells to DIM than that of HeLa cells.

Table I

Inhibitory rates of DIM on the proliferation of HeLa and SiHa cells.

Table I

Inhibitory rates of DIM on the proliferation of HeLa and SiHa cells.

24 h48 h72 h



DIM (μM)HeLaSiHaHeLaSiHaHeLaSiHa
2514.6110.1823.3831.5826.1940.04
5029.2128.7747.5442.1148.0656.30
10057.0256.1484.0084.9492.6693.03

[i] Cells were treated with different concentrations (25–100 μM) of DIM for 0–72 h. Cell proliferation was assessed by the CCK-8 method and represented by absorbance value at 450 nm (A450). Inhibitory rate (%) = [A450 (negative group) − A450 (treated group)] / [A450 (negative group) − A450 (blank group)] × 100%. DIM, 3,3′-diindolylmethane.

Effects of DIM on the apoptosis of HeLa and SiHa cells

Flow cytometry was performed to detect the apoptosis of HeLa cells and SiHa cells treated with various concentrations of DIM for 48 h. As DIM concentrations increased from 0 μM (negative control group) to 100 μM, both HeLa cells and SiHa cells showed the following tendencies (Fig. 2): i) The cells distributed in the 3rd region (normal zone) gradually decreased, indicating that the number of living cancer cells were reduced by DIM treatment. ii) The cells distributed in the 4th region gradually increased, suggesting the number of cells at early stage of apoptosis were increased after DIM treatment. iii) The number of cells with broken nuclei, which would be distributed in the 2nd region (advanced apoptosis cells), also increased with the increment of DIM concentrations.

DIM showed a significantly stronger pro-apoptotic effect on SiHa cells compared with HeLa cells (Fig. 3). Apoptosis was induced in (4.45±0.56), (7.11±0.67) and (25.16±2.12)% of HeLa cells at 25, 50 and 100 μM DIM respectively, while the apoptotic ratio in SiHa cells were (10.09±1.32), (21.11±3.36) and (55.46±6.33)% at each concentration (all P<0.01).

Effects of DIM on the expressions of proteins involved in apoptosis

Among many apoptosis pathways, the activation of mitogen-activated protein kinases (MAPK) and phosphoinositide 3-kinases (PI3K) signaling pathways drive a significant percentage of human cancer and serve as the targets for drug development. Western blottting was performed to determine the expression changes of proteins involved in these two pathways. The results showed that expression levels of different proteins involved were more or less influenced by DIM treatment. As shown in Fig. 4, GAPDH, the internal reference, remained stable in the two types of cells before and after DIM treatment. For MAPK pathway (Fig. 4A), the expression levels of extracellular signal regulated kinase (ERK), as well as the levels of phosphorylated ERK (p-ERK), were all decreased in HeLa and SiHa cells in accordance with the increase of DIM concentration. p38 and p-p38, also proteins involved in MAPK pathway, were downregulated with relatively lower DIM concentrations (25 and 50 μM), but then upregulated when the concentration reached 100 μM. Moreover, the expression levels of p38 and p-p38 in SiHa cells treated with 100 μM DIM were lower than those in HeLa cells. Another protein involved in MAPK pathway, c-Jun N-terminal kinase (JNK), was upregulated by DIM treatment in a dose-dependent manner, accompanied with the increase of phosphorylated JNK (p-JNK) and there were no evident differences between HeLa and SiHa cells.

For proteins involved in PI3K pathway (p110α, p110β, class III, Akt, GSK3-β, p-PDK1 and p-c-Raf) (Fig. 4B), their expressions and phosphorylation levels were all downregulated in both HeLa and SiHa cells by DIM treatment with decreasing the drug concentration. In addition, the expression levels of Akt, PI3K class III, GSK3-β, and p-PDK1 of SiHa cells treated with 100 μM DIM were significantly lower than those of HeLa cells.

Disscussion

There is well documented evidence that I3C, a natural anticancer compound extracted from cruciferous vegetables, exerted anti-proliferative effects on many cancers in vitro, including breast, colon and prostate cancers (12,13). While DIM, the polymer converted from I3C under acidic conditions, was suggested to be the real actor during anticancer process induced by I3C (15). In the present study we investigated the effects of DIM on the proliferation and apoptosis of HeLa and SiHa cells, as they are two different typical types of cervical cancer cells.

In the present study it was demonstrated that DIM inhibited proliferation and induced apoptosis in HeLa and SiHa cells in a time- and dose-dependent manner, consistent with other reports concerning the anti-proliferative and pro-apoptotic ablities of DIM (12,13). Interestingly, SiHa cells, a type of squamous carcinoma cells, seemed to be more sensitive to DIM treatment than the adenocarcinoma cells (HeLa cells), contrary to the effects of many other anti-cancer compounds. According to Zhou et al(16), artesunate inhibited the growth of HeLa cells more efficiently than SiHa cells, and it also increased the radio-sensitivity of HeLa cells but not SiHa cells. Cisplatin, a widely-used drug in cancer chemotherapy, was demonstrated to be more effective in HeLa cells compared with SiHa cells (17). However, Han et al(18) found that extracts from shiitake (Lentinula edodes) mycelial culture broth, by an organic solvent ethyl acetate, inhibited the proliferation of cultured tumor cells, including Caski, SiHa, HeLa, HP-1 and A375 cells. Among them, HeLa cells showed lower sensitivity to the extracts than SiHa cells, not only in cell growth, but also in fragmentation of DNA, as well as the activity of caspase-3 in cancer cell extracts. In the present study a similar phenomenon for DIM treatment was observed.

Mitogen-activated protein kinases (MAPK), including ERK, JNK, and p38 MAPK (19), are ubiquitous serine/threonine protein kinases that have been implicated in many cellular processes such as proliferation, differentiation, and apoptosis (20,21). Human MAPK pathway was often deregulated in cancer cells (22) and thus considered as potential therapeutic targets (23). Elevated ERK activity was frequently observed in human tumors and suggested to be a good indicator of tumor progression and evaluation of the efficacy of therapeutic agents (2426). Studies have shown that targeting ERK1/2 using siRNAs effectively reduced lung metastasis and sensitized tumor cells to chemotherapeutic agents such as cisplatin (25,27). The activation of p38 was also reported to be elevated in human non-small cell lung cancer (28) and the inhibition and reduction of p38 by inhibitor or RNAi could restrain the invasion and movement of cancer cells (2931), suggesting the important roles of p38 in oncogenesis, tumor development, tumor invasion and metastasis. Several reports also provided evidence on JNK's function as proapoptotic kinase in response to various stimuli (3237). The inhibitory effect of DIM on the proliferation of cervical cancer cells, as well as the induction of apoptosis in the present study, were demonstrated to be accompanied by downregulation of ERK, P38, and upregulation of JNK, indicating that MAPK pathway was involved in the pro-apoptotic effects of DIM treatment on HeLa and SiHa cells, which was consistent to reports on other cancer cells. However, it remained unclear why expression of p38 and p-p38 was reduced at relatively lower concentration of DIM treatment, but then increased at the concentration of 100 μM. This might be explained by a compensatory mechanism triggered at the advanced apotosis stage since the main function of p38 was to protect cells from apoptosis (38).

PI3K, a major signaling hub downstream of HER2 (human epidermal growth factor receptor 2) and other receptor tyrosine kinases (RTKs), activates Akt, SGK, PDK1, mTOR, and several other molecules involved in cell cycle progression and survival (39). Three classes of PI3K enzymes have been defined and class I is the most intensely studied which includes p110α, β, γ, and δ catalytic isoforms (40). As a direct downstream target of PI3K, Akt is also a key oncogenic survival factor and can phosphorylate and inactivate a panel of critical proapoptotic molecules, including Bad, caspase 9, GSK3-β, cell cycle inhibitors p21 and p27, and tumor suppressor TSC2 (4145). The PI3K pathway is an important regulator in cell survival, proliferation, and apoptosis. Moreover, it is one of the most frequently altered networks in cancer (46). Overexpression of PI3K/AKT was observed recently in various malignant tumors, such as cervical carcinoma (47), prostate cancer (48), as well as breast cancer (49). Furthermore, PI3K p110 expression was detected to be positive in all stages of cervical lesions except normal cervical tissue (50). The inhibitors of PI3K or Akt, curcumin (51), celecoxib (52), perifosine (5355) and CMEP (56), were all reported to attenuate the growth and proliferation of cancer cells and induce cell apoptosis. In the present study, downregulation of proteins associated with PI3K/Akt signaling pathway in HeLa and SiHa cells suggested that this pathway was also involved in the apoptosis of cervical cancer cells due to DIM treatment. In addition, different expression changes of p38, p-p38, Akt, PI3K classIII, GSK3-β, and p-PDK1 after DIM treatment between SiHa cells and HeLa cells might explain their different sensitivities to DIM treatment.

In conclusion, according to our results in the present study, DIM showed evident anti-proliferative and pro-apoptotic effects on cervical carcinoma cells in time and dose-dependent manner, and SiHa cells presented a stronger sensitivity than HeLa cells. MAPK and PI3K signaling pathways were proven to be involved in the pro-apoptotic effects of DIM on cervical cancer cells. Given that DIM exerted the above anti-tumor effects, especially on SiHa cells, it might be helpful for the development of novel therapeutic compounds for cervical cancers. Specific therapies for different types of cervical carcinoma are necessary since they react differently to different drugs.

References

1 

Newall AT, Beutels P, Wood JG, Edmunds WJ and MacIntyre CR: Cost-effectiveness analyses of human papillomavirus vaccination. Lancet Infect Dis. 7:289–296. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Tanaka T, Bai T, Yukawa K and Umesaki N: Optimal combination chemotherapy and chemoradiotherapy with etoposide for advanced cervical squamous cancer cells in vitro. Oncol Rep. 15:939–947. 2006.PubMed/NCBI

3 

Sarkar FH, Li Y, Wang Z and Kong D: Cellular signaling perturbation by natural products. Cell Signal. 21:1541–1547. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Brew CT, Aronchik I, Hsu JC, et al: Indole-3-carbinol activates the ATM signaling pathway independent of DNA damage to stabilize p53 and induce G1 arrest of human mammary epithelial cells. Int J Cancer. 118:857–868. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Chinni SR, Li Y, Upadhyay S, Koppolu PK and Sarkar FH: Indole-3-carbinol (I3C) induced cell growth inhibition, G1 cell cycle arrest and apoptosis in prostate cancer cells. Oncogene. 20:2927–2936. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Rahman KM, Aranha O, Glazyrin A, Chinni SR and Sarkar FH: Translocation of Bax to mitochondria induces apoptotic cell death in indole-3-carbinol (I3C) treated breast cancer cells. Oncogene. 19:5764–5771. 2000. View Article : Google Scholar : PubMed/NCBI

7 

Cover CM, Hsieh SJ, Tran SH, et al: Indole-3-carbinol inhibits the expression of cyclin-dependent kinase-6 and induces a G1 cell cycle arrest of human breast cancer cells independent of estrogen receptor signaling. J Biol Chem. 273:3838–3847. 1998. View Article : Google Scholar

8 

Fan S, Wang J, Yuan R, et al: BRCA1 inhibition of estrogen receptor signaling in transfected cells. Science. 284:1354–1356. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Meng Q, Qi M, Chen DZ, et al: Suppression of breast cancer invasion and migration by indole-3-carbinol: associated with up-regulation of BRCA1 and E-cadherin/catenin complexes. J Mol Med. 78:155–165. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Chang X, Tou JC, Hong C, et al: 3,3′-Diindolylmethane inhibits angiogenesis and the growth of transplantable human breast carcinoma in athymic mice. Carcinogenesis. 26:771–778. 2005.

11 

Safe S, Papineni S and Chintharlapalli S: Cancer chemotherapy with indole-3-carbinol, bis(3′-indolyl)methane and synthetic analogs. Cancer Lett. 269:326–338. 2008.

12 

Khwaja FS, Wynne S, Posey I and Djakiew D: 3,3′-diindolylmethane induction of p75NTR-dependent cell death via the p38 mitogen-activated protein kinase pathway in prostate cancer cells. Cancer Prev Res. 2:566–571. 2009.

13 

Chinnakannu K, Chen D, Li Y, et al: Cell cycle-dependent effects of 3,3′-diindolylmethane on proliferation and apoptosis of prostate cancer cells. J Cell Physiol. 219:94–99. 2009.

14 

Ge C, Xu J and Jiao Y: New derivatives I3C-6 of indole-3-carbinol in cervical cancer HeLa cells. Suzhou University J Med Sci. 29:838–834. 2009.

15 

Chang YC, Riby J, Chang GH, Peng BC, Firestone G and Bjeldanes LF: Cytostatic and antiestrogenic effects of 2-(indol-3-ylmethyl)-3,3′-diindolylmethane, a major in vivo product of dietary indole-3-carbinol. Biochem Pharmacol. 58:825–834. 1999.PubMed/NCBI

16 

Zhou Y, Feng Y and Zhang X: Study on the effect of artesunate combined with irradiation on DNA damage of HeLa and SiHa cells of human cervical cancer. J Radiat Res Radiat Process. 29:33–36. 2011.

17 

Wang H, Zhang Y and Jiang K: Enhancement of photodynamic therapy sensitivity by cisplatin in human cervical carcinoma cell lines. J Xian Jiaotong University (Med Sci ). 31:625–630. 2010.

18 

Han B, Toyomasu T and Shinozawa T: Induction of apoptosis in cultured cells by extracts from shiitake (Lentinula edodes) mycelial culture broth. Mycoscience. 41:623–631. 2000. View Article : Google Scholar

19 

Fecher LA, Amaravadi RK and Flaherty KT: The MAPK pathway in melanoma. Curr Opin Oncol. 20:183–189. 2008. View Article : Google Scholar

20 

Seger R and Krebs EG: The MAPK signaling cascade. FASEB J. 9:726–735. 1995.PubMed/NCBI

21 

Cobb MH: MAP kinase pathways. Prog Biophys Mol Biol. 71:479–500. 1999. View Article : Google Scholar

22 

Meira DD, de Almeida VH, Mororo JS, et al: Combination of cetuximab with chemoradiation, trastuzumab or MAPK inhibitors: mechanisms of sensitisation of cervical cancer cells. Br J Cancer. 101:782–791. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Inamdar GS, Madhunapantula SV and Robertson GP: Targeting the MAPK pathway in melanoma: why some approaches succeed and other fail. Biochem Pharmacol. 80:624–637. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Lopez-Bergami P, Huang C, Goydos JS, et al: Rewired ERK-JNK signaling pathways in melanoma. Cancer Cell. 11:447–460. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Mirmohammadsadegh A, Mota R, Gustrau A, et al: ERK1/2 is highly phosphorylated in melanoma metastases and protects melanoma cells from cisplatin-mediated apoptosis. J Invest Dermatol. 127:2207–2215. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Flaherty KT: Sorafenib: delivering a targeted drug to the right targets. Expert Rev Anticancer Ther. 7:617–626. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Sharma A, Tran MA, Liang S, et al: Targeting mitogen-activated protein kinase/extracellular signal-regulated kinase kinase in the mutant (V600E) B-Raf signaling cascade effectively inhibits melanoma lung metastases. Cancer Res. 66:8200–8209. 2006. View Article : Google Scholar

28 

Greenberg AK, Basu S, Hu J, et al: Selective p38 activation in human non-small cell lung cancer. Am J Respir Cell Mol Biol. 26:558–564. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Kim MS, Lee EJ, Kim HR and Moon A: p38 kinase is a key signaling molecule for H-Ras-induced cell motility and invasive phenotype in human breast epithelial cells. Cancer Res. 63:5454–5461. 2003.PubMed/NCBI

30 

Sossey-Alaoui K, Ranalli TA, Li X, Bakin AV and Cowell JK: WAVE3 promotes cell motility and invasion through the regulation of MMP-1, MMP-3, and MMP-9 expression. Exp Cell Res. 308:135–145. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Huang X, Chen S, Xu L, et al: Genistein inhibits p38 map kinase activation, matrix metalloproteinase type 2, and cell invasion in human prostate epithelial cells. Cancer Res. 65:3470–3478. 2005.PubMed/NCBI

32 

Zwang Y and Yarden Y: p38 MAP kinase mediates stress-induced internalization of EGFR: implications for cancer chemotherapy. EMBO J. 25:4195–4206. 2006. View Article : Google Scholar

33 

Lin A: A five-year itch in TNF-alpha cytotoxicity: the time factor determines JNK action. Dev Cell. 10:277–278. 2006.PubMed/NCBI

34 

Bowen C, Birrer M and Gelmann EP: Retinoblastoma protein-mediated apoptosis after gamma-irradiation. J Biol Chem. 277:44969–44979. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Li T, Dai W and Lu L: Ultraviolet-induced junD activation and apoptosis in myeloblastic leukemia ML-1 cells. J Biol Chem. 277:32668–32676. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Ouwens DM, Gomes de Mesquita DS, Dekker J and Maassen JA: Hyperosmotic stress activates the insulin receptor in CHO cells. Biochim Biophys Acta. 1540:97–106. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Stadheim TA and Kucera GL: c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) is required for mitoxantrone- and anisomycin-induced apoptosis in HL-60 cells. Leuk Res. 26:55–65. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Liao Y and Hung MC: Regulation of the activity of p38 mitogen-activated protein kinase by Akt in cancer and adenoviral protein E1A-mediated sensitization to apoptosis. Mol Cell Biol. 23:6836–6848. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Garrett JT, Chakrabarty A and Arteaga CL: Will PI3K pathway inhibitors be effective as single agents in patients with cancer? Oncotarget. 2:1314–1321. 2011.PubMed/NCBI

40 

Cantley LC: The phosphoinositide 3-kinase pathway. Science. 296:1655–1657. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Blain SW and Massague J: Breast cancer banishes p27 from nucleus. Nat Med. 8:1076–1078. 2002. View Article : Google Scholar : PubMed/NCBI

42 

Inoki K, Li Y, Zhu T, Wu J and Guan KL: TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 4:648–657. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Nicholson KM and Anderson NG: The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 14:381–395. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Testa JR and Bellacosa A: AKT plays a central role in tumorigenesis. Proc Natl Acad Sci USA. 98:10983–10985. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Zhou BP, Liao Y, Xia W, Spohn B, Lee MH and Hung MC: Cytoplasmic localization of p21Cip1/WAF1 by Akt-induced phosphorylation in HER-2/neu-overexpressing cells. Nat Cell Biol. 3:245–252. 2001. View Article : Google Scholar : PubMed/NCBI

46 

Brugge J, Hung MC and Mills GB: A new mutational AKTivation in the PI3K pathway. Cancer Cell. 12:104–107. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Ma JL, Lee SJ, Duong JK and Stern DF: Activation of the checkpoint kinase Rad53 by the phosphatidyl inositol kinase-like kinase Mec1. J Biol Chem. 281:3954–3963. 2006. View Article : Google Scholar : PubMed/NCBI

48 

Xu J, Gao M, Fan S, et al: Effect of Akt inhibition on scatter factor-regulated gene expression in DU-145 human prostate cancer cells. Oncogene. 26:2925–2938. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Junttila TT, Akita RW, Parsons K, et al: Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell. 15:429–440. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Xie L, Chen X and Liu Q: Study of PI3K p110α expression in cervical lesions. Pract Prev Med. 18:2011.

51 

Shankar S and Srivastava RK: Involvement of Bcl-2 family members, phosphatidylinositol 3′-kinase/AKT and mitochondrial p53 in curcumin (diferulolylmethane)-induced apoptosis in prostate cancer. Int J Oncol. 30:905–918. 2007.

52 

Barnes NL, Warnberg F, Farnie G, et al: Cyclooxygenase-2 inhibition: effects on tumour growth, cell cycling and lymphangiogenesis in a xenograft model of breast cancer. Br J Cancer. 96:575–582. 2007. View Article : Google Scholar : PubMed/NCBI

53 

de Marsh RW, Rocha Lima CM, Levy DE, Mitchell EP, Rowland KM Jr and Benson AB III: A phase II trial of perifosine in locally advanced, unresectable, or metastatic pancreatic adenocarcinoma. Am J Clin Oncol. 30:26–31. 2007.

54 

Cirstea D, Hideshima T, Rodig S, et al: Dual inhibition of akt/mammalian target of rapamycin pathway by nanoparticle albumin-bound-rapamycin and perifosine induces antitumor activity in multiple myeloma. Mol Cancer Ther. 9:963–975. 2010. View Article : Google Scholar

55 

Floryk D and Thompson TC: Perifosine induces differentiation and cell death in prostate cancer cells. Cancer Lett. 266:216–226. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Zhang M, Fang X, Liu H, Wang S and Yang D: Blockade of AKT activation in prostate cancer cells with a small molecule inhibitor, 9-chloro-2-methylellipticinium acetate (CMEP). Biochem Pharmacol. 73:15–24. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September 2012
Volume 28 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu J, Li Y, Guan C and Chen Z: Anti-proliferative and pro-apoptotic effects of 3,3'-diindolylmethane in human cervical cancer cells. Oncol Rep 28: 1063-1068, 2012
APA
Zhu, J., Li, Y., Guan, C., & Chen, Z. (2012). Anti-proliferative and pro-apoptotic effects of 3,3'-diindolylmethane in human cervical cancer cells. Oncology Reports, 28, 1063-1068. https://doi.org/10.3892/or.2012.1877
MLA
Zhu, J., Li, Y., Guan, C., Chen, Z."Anti-proliferative and pro-apoptotic effects of 3,3'-diindolylmethane in human cervical cancer cells". Oncology Reports 28.3 (2012): 1063-1068.
Chicago
Zhu, J., Li, Y., Guan, C., Chen, Z."Anti-proliferative and pro-apoptotic effects of 3,3'-diindolylmethane in human cervical cancer cells". Oncology Reports 28, no. 3 (2012): 1063-1068. https://doi.org/10.3892/or.2012.1877