Effects of combined phytochemicals on skin tumorigenesis in SENCAR mice

  • Authors:
    • Magdalena C. Kowalczyk
    • Jacob J. Junco
    • Piotr Kowalczyk
    • Olga Tolstykh
    • Margaret Hanausek
    • Thomas J. Slaga
    • Zbigniew Walaszek
  • View Affiliations

  • Published online on: July 5, 2013     https://doi.org/10.3892/ijo.2013.2005
  • Pages: 911-918
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The purpose of our study was to determine the effect of the combined action of phytochemicals on the early stages of skin tumorigenesis, i.e. initiation and promotion. We tested calcium D-glucarate (CG) given in the diet, while resveratrol (RES) and ursolic acid (UA) were applied topically. The 7,12-dimethylbenz[a]anthracene (DMBA)-initiated, 12-O-tetradecanoylphorbol-13-acetate (TPA)-promoted multistage skin carcinogenesis model in SENCAR mice was used. Mice received one topical dose of DMBA, then after one month, two weekly doses of TPA for 14 weeks until sacrifice. RES or UA were applied 20 min prior to DMBA or TPA treatment and 2% dietary CG was given from 2 weeks prior to 2 weeks after the DMBA dose or continually beginning 2 weeks prior to the first dose of TPA. UA applied alone and in combination with CG during the promotion stage was the only inhibitor of tumor multiplicity and tumor incidence. A number of combinations reduced epidermal proliferation, but only UA and the combination UA+CG applied during promotion significantly reduced epidermal hyperplasia. DMBA/TPA application resulted in significant increases in c-jun and p50, which were reversed by a number of different treatments. DMBA/TPA treatment also strongly increased mRNA levels of inflammation markers COX-2 and IL-6. All anti-promotion treatments caused a marked decrease in COX-2 and IL-6 expression compared to the DMBA/TPA control. These results show that UA is a potent inhibitor of skin tumor promotion and inflammatory signaling and it may be useful in the prevention of skin cancer and other epithelial cancers in humans.

Introduction

The induction of cancer is a multistage process and its stages have been defined experimentally as initiation, promotion, and progression. Carcinogenesis depends on inherited and acquired susceptibility factors, such as mutations in oncogenes and tumor suppressor genes (1,2), on exposure to initiation factors, i.e. exogenous and endogenous carcinogens and on promotion and progression factors. It is well known that a variety of chemical and physical agents can cause skin cancer in rodents and man. Repetitive treatment with known skin carcinogens will lead to skin damage followed by inflammation and regenerative hyperplasia, dysplasia, papillomas, basal and/or squamous cell carcinomas (35).

Papilloma formation can be chemically induced by the two-stage carcinogenesis protocol (reviewed in ref. 6). Application of the carcinogen 7,12-dimethylbenz[a]anthracene (DMBA) to the skin surface results in Ras mutations in long-lived keratinocytes, probably stem cells (7,8). In response to applications of the tumor promoter 12-O-tetradecanoylphorbol-13-acetate (TPA), oncogenic transcription factors NFκB and AP-1 are activated (911), leading to increased inflammation, proliferation of initiated cells and finally to clonal expansion and papilloma formation. The mouse skin carcinogenesis model has provided great understanding of the important cellular and molecular events involved in tumor initiation, promotion and progression (3,4).

The mouse skin cancer model has provided an important system not only for studying mechanisms involved in the various stages of carcinogenesis and for bioassay of tumor-promoting and carcinogenetic agents, but also for the study of inhibitors of tumor formation and malignant conversion (5). This model has been used to show that a variety of naturally occurring phytochemicals may be very useful for the prevention of skin cancer as well as for the prevention of other epithelial cancers in humans (12). The mouse skin cancer model relates very well to other models where squamous cell carcinomas are induced. The phytochemicals may modify carcinogen activation, enhance phase II enzymes detoxification, modify antioxidant enzymes, prevent oxidative damage to DNA bases and mutations, decrease inflammation and proliferation, modulate the immune response and induce apoptosis. Because of their diverse mechanisms of action, many combinations of phytochemicals can interact in a synergistic fashion. One phytochemical may impact the metabolism of the other (13,14), or change its ability to enter or leave the cell (15). Phytochemicals may synergize by acting along different points on cell regulatory systems (16,17). Synergistic interactions between phytochemicals allow for a decreased concentration of each drug to achieve the same effect. This may reduce cost of the therapy as well as reduce potential side effects.

The phytochemicals selected for this study occur in many medicinal herbs and plants. Resveratrol (RES) is a naturally occurring phytoalexin associated with many health benefits, most notably the mitigation of age-related diseases, including neurodegeneration, carcinogenesis and atherosclerosis (1820). RES has also shown promise as an antidiabetic (21) and anticancer (22,23) agent in recent clinical trials. Calcium D-glucarate (CG) is the salt and the commercial form of D-glucaric acid, which occurs naturally in a variety of foods, including broccoli, oranges and apples. Following oral administration, CG is converted to D-glucaro-1,4-lactone, which inhibits the enzyme β-glucuronidase and enhances phase II detoxification (24). Some in vitro and animal data suggest that inhibition of β-glucuronidase may suppress carcinogenesis (25), as well as inhibit the initiation and promotion/progression stages of tumorigenesis (12,26,27). Ursolic acid (UA) is a pentacyclic triterpenoid which has been shown to suppress skin (28) and breast (29) tumorigenesis. UA has also been found to induce apoptosis in a wide variety of cancer cells (3032).

The overall goal of the present study was to determine the effect of combined action of phytochemicals on early stages of skin tumorigenesis, i.e. initiation and promotion. Our hypothesis was that concurrent topical and dietary treatment with selected compounds would lead to more efficient synergistic prevention of chemically-induced murine skin tumorigenesis. Tumors per mouse, tumor incidence, epidermal thickness, epidermal proliferation, and a number of inflammatory biomarkers were measured to determine the effects of these combinations of phytochemicals on the initiation and promotion stages of tumorigenesis.

Materials and methods

Scheme of DMBA-initiated, TPA-promoted skin carcinogenesis

Female SENCAR mice, 5 weeks old, were purchased from the National Cancer Institute, Frederick Cancer Research and Development Center (Frederick, MD, USA). At 6–7 weeks of age the backs of mice were shaved and 20 nmol of DMBA in 0.2 ml acetone (ACT) was applied topically, then after one month, two weekly doses of 2 μg TPA in 0.2 ml of ACT were applied for up to 14 weeks until sacrifice. The phytochemicals RES (2.5 μmol in 0.2 ml ACT) or UA (1 μmol in 0.2 ml DMSO) were applied topically to the dorsal surface of mice 20 min prior to DMBA or TPA treatment and 2% dietary CG was given in the AIN-93G diet from 2 weeks prior until 2 weeks after the DMBA dose or continually beginning 2 weeks prior to the first dose of TPA. The compounds were used in these same concentrations for the combination studies. After the 14th week of treatment, mice were euthanized, tumors were removed and two 1-cm2 skin samples were removed for histology and RNA extraction.

Tumor counting

Upon the appearance of papillomas (7th week of TPA treatment) tumors on the backs of each mouse were counted weekly. Tumor multiplicity and tumor incidence were calculated for each group.

Histological evaluation

The tissues were prepared for histological evaluation by using conventional paraffin sections and hematoxylin-eosin staining. The average epithelial thickness was determined from at least 12 randomly selected sites in formalin-fixed skin samples, with 3 measurements per image.

For proliferative analysis, mice were given an i.p. injection of BrdU (Sigma Chemical Co., St. Louis, MO, USA) (1.5 mg in saline per mouse) 60 min prior to sacrifice. The tissue sections were immunostained with anti-BrdU antibody (Lab Vision Corp., Fremont, CA, USA). The percentage of stained cells in the basal layer of the epidermis of 12 randomly selected sites was determined. For analysis of NFκB and AP-1 activities, slides were stained with anti-p50 (Lab Vision Co.) or anti-c-jun antibodies (BD Biosciences, Franklin Lakes, NJ, USA). Positive cells were counted as with BrdU samples, however, 3 mice per group were used in the analyses.

Real-time PCR analysis

Total RNA was extracted using TRI reagent (MRC Inc., Cincinnati, OH, USA). RNA (1 mg) was reverse transcribed with oligo(dT), using cMaster RT kit (Eppendorf North America, Westbury, NY, USA) according to the manufacturer’s protocol. Primers used were: 5′-ATCCTGC CAGCTCCACCG-3′, 5′-TGGTCAAATCCTGTGCTCATA CAT-3′ for COX-2, 5′-GATGTGGAGCAACTTGGAAT-3′, 5′-AGCTCTCCACTTGCAGAAAA-3′ for IL-6 and 5′-CAT CCTGGCCTCGCTGTC-3′, 5′-CTCGTCGTACTCCTGC TTGGT-3′ for β-actin. Standard quantitative RT-PCR was performed in triplicate using SYBR Green RealMasterMix (Eppendorf North America) on the Realplex MasterCycler (Eppendorf). RT-PCR cycle thresholds (Ct) of candidate genes were normalized to control gene β-actin. The formula 2Ct(Candidate)/2Ct(Control) was used to calculate the normalized ratios.

Statistical analysis

The results for Figs. 28 are expressed as means ± standard deviation (SD). For visual clarity, SD is not shown in Fig. 1. For comparison of the differences between the groups, a two-tailed, unpaired, Student’s t-test was used. p<0.05 was considered to be statistically significant.

Results

Effect of phytochemicals on tumor multiplicity and tumor incidence

Tumor multiplicity (average papillomas per mouse) and tumor incidence (% of mice with at least one papilloma) for each treatment group were determined by counting papillomas on the backs of mice once per week for the 7th through the 14th week of TPA treatment. No tested compound or combination inhibited carcinogenesis when applied during the initiation (DMBA) phase. In fact, UA significantly increased DMBA/TPA-mediated tumor formation when applied with DMBA in weeks 7 and 9–14 of TPA treatment (p<0.05). However, when applied with TPA, UA inhibited tumor multiplicity by 90% by itself and by 78.9% in CG-fed mice, and both treatments inhibited tumor incidence by 75% at the 14th week of TPA treatment. UA and UA+CG significantly p<0.05 inhibited tumor multiplicity when applied with TPA in each of weeks 7–14 of TPA treatment (Fig. 1).

Effect of phytochemicals on epidermal hyperplasia and cell proliferation determined by incorporation of BrdU

The level of epidermal proliferation for each treatment group was determined by counting BrdU-positive cells at 12 random locations per sample, then expressing the data as a percentage of BrdU-positive cells in the basal layer. The proliferation rate of keratinocytes located in the basal layer of epidermis and overall epidermal thickness were greatly increased in the DMBA/TPA treated mice compared to negative control. Despite not having an effect on tumor formation when applied with DMBA, UA alone and in combination with CG was able to inhibit proliferation when applied during the initiation phase, while no compound significantly reduced epidermal hyperplasia when applied with DMBA (Fig. 2). All treatments except CG or UA alone had a statistically significant impact on proliferation when applied during promotion, however, only UA and UA+CG significantly affected epidermal hyperplasia when applied with TPA (Fig. 3). In similar studies, there is typically a strong concordance between levels of epidermal proliferation (measured by BrdU staining) and epidermal hyperplasia. However, in our experiments, we observed some disagreement between these two metrics of tumor promotion. For example, we observed that UA was the strongest inhibitor of epidermal hyperplasia when applied with TPA, however, it had no effect on epidermal proliferation (Fig. 3). This effect may be mediated by a UA-induced upregulation of apoptosis, which would decrease epidermal thickness without specifically affecting proliferation.

Effect of phytochemicals on COX-2 and IL-6 expression

DMBA/TPA treatments resulted in increased expression of the inflammation markers COX-2 and IL-6, compared to control. These genes play important roles in such inflammatory responses as edema formation and hyperplasia, as well as in papilloma development in mouse skin (33,34). While only UA and RES+CG significantly reversed COX-2 mRNA levels when applied with DMBA in the two-stage model, every compound or combination decreased COX-2 mRNA levels when applied alongside TPA during the promotion stage. All treatments strongly decreased IL-6 expression; moreover, anti-promotion treatments reduced IL-6 expression to the negative control level (Fig. 4).

Effect of phytochemicals on c-jun-positive cells

AP-1 activation levels were measured by immunostaining for c-jun-positive cells within the interfollicular epidermis. An average of 200 cells per tissue section were counted at 12 random locations per sample, with the data expressed as the mean ± SD of three animals per treatment group. As expected, DMBA/TPA treatment strongly increased the number of c-jun-positive cells. All treatments resulted in a slight decrease of c-jun-positive cells when applied with DMBA (Fig. 5). All treatments except for CG alone resulted in a significant decrease in c-jun-positive cells when applied alongside TPA. In case of mice treated with UA+CG during the TPA stage, the percentage of c-jun-positive cells was decreased to the negative control level revealing at least an additive effect of this combination (Fig. 6).

Effect of phytochemicals on p50-positive cells

NFκB expression for each treatment group was determined by counting p50 protein-positive cells at 12 random locations per sample, then expressing the data as a percentage of positive cells within the interfollicular epidermis. The DMBA/TPA-treated skin presented strong nuclear staining. UA+CG applied during the DMBA initiation stage was the only treatment to significantly suppress the percentage of p50-positive cells (Fig. 7). UA alone, its combination with CG and additionally dietary CG with RES during the promotion stage significantly reduced the percentage of p50-stained cells. UA+CG treatment applied during the promotion phase was the greatest inhibitor of the increase in p50-positive cells among used treatments, showing some additive effect in comparison to single treatments (Fig. 8).

Discussion

DMBA/TPA treatments resulted in epidermal hyperplasia, characterized by a significant increase in cells positive for AP-1 and NFκB components with a simultaneously increased expression of the inflammation markers COX-2 and IL-6, compared to untreated controls. Despite our initial hypothesis, we observed no definitive synergistic effect between any of the tested phytochemicals.

UA applied alone and in combination with CG during the promotion stage was a strong inhibitor of tumor multiplicity and tumor incidence. While different treatments reduced epidermal proliferation when applied with either DMBA or TPA, only UA and the combination UA+CG applied during promotion significantly reduced hyperplasia. All anti-promotion treatments (either single compounds or their combinations), a majority of single treatments and all combinations during anti-initiation stage caused a marked decrease of inflammation related genes levels compared to the DMBA/TPA control. On the other hand, only UA+CG significantly diminished both c-jun-positive cells and p50-positive cells. These results show that UA alone or in combination with CG is a potent inhibitor of skin tumor promotion and it may be useful in the prevention of skin cancer and other epithelial cancers in humans. In addition, UA+CG is a strong inhibitor of AP-1 and NFκB, and may be useful for the prevention or treatment of maladies associated with activation of these factors.

In our study, we observed UA to be a much stronger inhibitor of tumor formation than RES or CG. This may be because UA and UA+CG were the most potent inhibitors of DMBA/TPA-induced c-jun positive cells and p50-positive cells of the treatments tested. This agrees with studies in other systems, which also show UA is a more potent inhibitor of inflammation-mediated processes such as monocyte recruitment than RES (35). Our other in vivo studies have demonstrated that UA more strongly inhibits TPA-induced phosphorylation and activation of NFκB subunit p65 in vivo (unpublished data). As AP-1 (36,37) and NFκB (9) activities have been shown to be necessary for chemically-induced skin cancer formation, we suggest that UA and UA+CG-mediated decreases in these transcription factors resulted in the observed antitumor effect.

None of the tested treatment groups significantly decreased tumor formation when applied during the DMBA stage. These compounds may only affect traits associated with skin tumor promotion, such as proliferation/clonal expansion, resistance to apoptosis and inflammation. In fact, UA added during the DMBA stage increased tumor formation. This may have been due to UA increasing the reactive concentration of DMBA. UA shows relatively little inhibition of CYP1A1 and CYP1B1, which metabolize polycyclic aromatic hydrocarbons like DMBA to their more reactive form (38). However, RES inhibits CYP1A1 and CYP1B1, but still showed no decrease in tumor multiplicity when applied with DMBA in our study. Another potential explanation is that the glucocorticoid-like structure of UA may cause an increase in nuclear envelope permeability (39), allowing for more DMBA intake and an increase in tumor formation. However, the classical glucocorticoid dexamethasone has been shown to inhibit skin tumor formation when applied with different initiators, including DMBA, in the two-stage skin carcinogenesis model (40). Finally, in our study, UA was applied topically in DMSO, as we could not solubilize the desired dose in ACT. Relative to ACT, DMSO has been shown previously to enhance solute penetration in skin explants of various animals (41) and enhance penetration of the carcinogen benzidine into porcine skin explants (42). The penetration-enhancing effects of DMSO may be mediated by a number of processes, including changing keratin structure and interactions with lipid bilayers (43), as well as the hygroscopic nature of DMSO (42). These effects of DMSO may have allowed for more DMBA epidermal penetration in the UA-treated mice in our study. However, we did not observe the same effect in the UA+CG group, which was also treated with UA in DMSO 20 min prior to DMBA application. Further studies are needed to determine how UA may increase chemically-mediated skin tumor formation when applied with DMBA.

Other studies have shown a potent antitumor effect of RES (4446), including in the DMBA/TPA-mediated skin cancer model in Balb/c and ICR mouse strains (47,48). However, in our studies we observed no antitumor effect of RES, even at doses which significantly inhibited the transcription of inflammatory genes and the levels of c-jun protein. This lack of effect may be explained by the strain of mouse used. Our previous studies revealed RES has limited effect on epidermal hyperplasia and proliferation when applied with DMBA or TPA in short-term, DMBA- or TPA-only models in SENCAR mice (49) (unpublished data). However, in the same TPA-only model equimolar doses of UA reduced epidermal hyperplasia to the level of negative control. The lack of sensitivity to RES relative to UA observed in SENCAR mice may be explained by different levels of metabolizing enzymes or RES targets in the skin of different strains. Future studies, perhaps in a simplified system with keratinocytes isolated from each mouse strain, are needed to confirm this hypothesis.

These results indicate UA, alone or in combination with CG, functions as a potent preventer of chemically-induced skin cancer formation. Our previous experiments also identified combinations of natural compounds which synergistically inhibited DMBA-mediated mutation frequency as well as inflammation in the epidermis (49). Future studies may reveal these treatments to have an effect on full skin tumor formation or on other cancers.

Acknowledgements

This study was supported by NIH grants R01 CA 102747 and P30 CA 54174-1651.

Abbreviations:

RES

resveratrol

GC

calcium D-glucarate

UA

ursolic acid

DMBA

7,12-dimethylbenz[a]anthracene

TPA

12-O-tetradecanoylphorbol-13-acetate

References

1 

Bishop JM: Molecular themes in oncogenesis. Cell. 64:235–248. 1991. View Article : Google Scholar : PubMed/NCBI

2 

Knudson AG: Hereditary predisposition to cancer. Ann NY Acad Sci. 833:58–67. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Boutwell RK: Some biological aspects of skin carcinogenisis. Prog Exp Tumor Res. 4:207–250. 1964.PubMed/NCBI

4 

Slaga TJ: Cancer: etiology, mechanisms, and prevention - a summary. Carcinog Compr Surv. 5:243–262. 1980.PubMed/NCBI

5 

DiGiovanni J: Multistage carcinogenesis in mouse skin. Pharmacol Ther. 54:63–128. 1992. View Article : Google Scholar : PubMed/NCBI

6 

Kemp CJ: Multistep skin cancer in mice as a model to study the evolution of cancer cells. Semin Cancer Biol. 15:460–473. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Perez-Losada J and Balmain A: Stem-cell hierarchy in skin cancer. Nat Rev Cancer. 3:434–443. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Hanausek M, Ganesh P, Walaszek Z, Arntzen CJ, Slaga TJ and Gutterman JU: Avicins, a family of triterpenoid saponins from Acacia victoriae(Bentham), suppress H-ras mutations and aneuploidy in a murine skin carcinogenesis model. Proc Natl Acad Sci USA. 98:11551–11556. 2001.PubMed/NCBI

9 

Kim EJ, Park H, Kim J and Park JH: 3,3′-diindolylmethane suppresses 12-O-tetradecanoylphorbol-13-acetate-induced inflammation and tumor promotion in mouse skin via the downregulation of inflammatory mediators. Mol Carcinog. 49:672–683. 2010.

10 

Budunova IV, Perez P, Vaden VR, Spiegelman VS, Slaga TJ and Jorcano JL: Increased expression of p50-NF-kappaB and constitutive activation of NF-kappaB transcription factors during mouse skin carcinogenesis. Oncogene. 18:7423–7431. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Przybyszewski J, Yaktine AL, Duysen E, et al: Inhibition of phorbol ester-induced AP-1-DNA binding, c-Jun protein and c-jun mRNA by dietary energy restriction is reversed by adrenalectomy in SENCAR mouse epidermis. Carcinogenesis. 22:1421–1427. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Walaszek Z, Hanausek M and Slaga TJ: Mechanisms of chemoprevention. Chest. 125:S128–S133. 2004. View Article : Google Scholar

13 

Taesotikul T, Dumrongsakulchai W, Wattanachai N, Navinpipat V, Somanabandhu A and Tassaneeyakul W: Inhibitory effects of Phyllanthus amarus and its major lignans on human microsomal cytochrome P450 activities: evidence for CYP3A4 mechanism-based inhibition. Drug Metab Pharmacokinet. 26:154–161. 2011. View Article : Google Scholar

14 

Kimura Y, Ito H, Ohnishi R and Hatano T: Inhibitory effects of polyphenols on human cytochrome P450 3A4 and 2C9 activity. Food Chem Toxicol. 48:429–435. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Suganuma M, Okabe S, Kai Y, Sueoka N, Sueoka E and Fujiki H: Synergistic effects of (−)-epigallocatechin gallate with (−)-epicatechin, sulindac, or tamoxifen on cancer-preventive activity in the human lung cancer cell line PC-9. Cancer Res. 59:44–47. 1999.

16 

Saw CL, Cintron M, Wu TY, et al: Pharmacodynamics of dietary phytochemical indoles I3C and DIM: induction of Nrf2-mediated phase II drug metabolizing and antioxidant genes and synergism with isothiocyanates. Biopharm Drug Dispos. 32:289–300. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Khafif A, Schantz SP, Chou TC, Edelstein D and Sacks PG: Quantitation of chemopreventive synergism between (−)-epigallocatechin-3-gallate and curcumin in normal, premalignant and malignant human oral epithelial cells. Carcinogenesis. 19:419–424. 1998.

18 

Vingtdeux V, Dreses-Werringloer U, Zhao H, Davies P and Marambaud P: Therapeutic potential of resveratrol in Alzheimer’s disease. BMC Neurosci. 9(Suppl 2): S62008.

19 

Bishayee A: Cancer prevention and treatment with resveratrol: from rodent studies to clinical trials. Cancer Prev Res. 2:409–418. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Das M and Das DK: Resveratrol and cardiovascular health. Mol Aspects Med. 31:503–512. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Brasnyo P, Molnar GA, Mohas M, et al: Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the Akt pathway in type 2 diabetic patients. Br J Nutr. 106:383–389. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Howells LM, Berry DP, Elliott PJ, et al: Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases - safety, pharmacokinetics, and pharmacodynamics. Cancer Prev Res. 4:1419–1425. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Patel KR, Brown VA, Jones DJ, et al: Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 70:7392–7399. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Walaszek Z, Szemraj J, Narog M, et al: Metabolism, uptake, and excretion of a D-glucaric acid salt and its potential use in cancer prevention. Cancer Detect Prev. 21:178–190. 1997.PubMed/NCBI

25 

Yoshimi N, Walaszek Z, Mori H, Hanausek M, Szemraj J and Slaga TJ: Inhibition of azoxymethane-induced rat colon carcinogenesis by potassium hydrogen D-glucarate. Int J Oncol. 16:43–48. 2000.PubMed/NCBI

26 

Hanausek M, Walaszek Z and Slaga TJ: Detoxifying cancer causing agents to prevent cancer. Integr Cancer Ther. 2:139–144. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Abou-Issa H, Moeschberger M, el-Masry W, Tejwani S, Curley RW Jr and Webb TE: Relative efficacy of glucarate on the initiation and promotion phases of rat mammary carcinogenesis. Anticancer Res. 15:805–810. 1995.PubMed/NCBI

28 

Huang MT, Ho CT, Wang ZY, et al: Inhibition of skin tumorigenesis by rosemary and its constituents carnosol and ursolic acid. Cancer Res. 54:701–708. 1994.PubMed/NCBI

29 

De Angel RE, Smith SM, Glickman RD, Perkins SN and Hursting SD: Antitumor effects of ursolic acid in a mouse model of postmenopausal breast cancer. Nutr Cancer. 62:1074–1086. 2010.PubMed/NCBI

30 

Shan JZ, Xuan YY, Zheng S, Dong Q and Zhang SZ: Ursolic acid inhibits proliferation and induces apoptosis of HT-29 colon cancer cells by inhibiting the EGFR/MAPK pathway. J Zhejiang Univ Sci B. 10:668–674. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Kassi E, Papoutsi Z, Pratsinis H, Aligiannis N, Manoussakis M and Moutsatsou P: Ursolic acid, a naturally occurring triterpenoid, demonstrates anticancer activity on human prostate cancer cells. J Cancer Res Clin Oncol. 133:493–500. 2007. View Article : Google Scholar

32 

Kassi E, Sourlingas TG, Spiliotaki M, et al: Ursolic acid triggers apoptosis and Bcl-2 downregulation in MCF-7 breast cancer cells. Cancer Invest. 27:723–733. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Fischer SM, Pavone A, Mikulec C, Langenbach R and Rundhaug JE: Cyclooxygenase-2 expression is critical for chronic UV-induced murine skin carcinogenesis. Mol Carcinog. 46:363–371. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Lederle W, Depner S, Schnur S, et al: IL-6 promotes malignant growth of skin SCCs by regulating a network of autocrine and paracrine cytokines. Int J Cancer. 128:2803–2814. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Ullevig SL, Zhao Q, Zamora D and Asmis R: Ursolic acid protects diabetic mice against monocyte dysfunction and accelerated atherosclerosis. Atherosclerosis. 219:409–416. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Young MR, Li JJ, Rincon M, et al: Transgenic mice demonstrate AP-1 (activator protein-1) transactivation is required for tumor promotion. Proc Natl Acad Sci USA. 96:9827–9832. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Thompson EJ, MacGowan J, Young MR, Colburn N and Bowden GT: A dominant negative c-jun specifically blocks okadaic acid-induced skin tumor promotion. Cancer Res. 62:3044–3047. 2002.PubMed/NCBI

38 

Kowalczyk MC, Walaszek Z, Kowalczyk P, Kinjo T, Hanausek M and Slaga TJ: Differential effects of several phytochemicals and their derivatives on murine keratinocytes in vitro and in vivo: implications for skin cancer prevention. Carcinogenesis. 30:1008–1015. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Shahin V, Ludwig Y, Schafer C, Nikova D and Oberleithner H: Glucocorticoids remodel nuclear envelope structure and permeability. J Cell Sci. 118:2881–2889. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Thompson S and Slaga TJ: The effects of dexamethasone on mouse skin initiation and aryl hydrocarbon hydroxylase. Eur J Cancer. 12:363–370. 1976. View Article : Google Scholar : PubMed/NCBI

41 

Baynes RE, Halling KB and Riviere JE: The influence of diethyl-m-toluamide (DEET) on the percutaneous absorption of permethrin and carbaryl. Toxicol Appl Pharmacol. 144:332–339. 1997. View Article : Google Scholar : PubMed/NCBI

42 

Baynes RE, Brownie C, Freeman H and Riviere JE: In vitro percutaneous absorption of benzidine in complex mechanistically defined chemical mixtures. Toxicol Appl Pharmacol. 141:497–506. 1996. View Article : Google Scholar : PubMed/NCBI

43 

Williams AC and Barry BW: Penetration enhancers. Adv Drug Deliv Rev. 56:603–618. 2004. View Article : Google Scholar

44 

Banerjee S, Bueso-Ramos C and Aggarwal BB: Suppression of 7,12-dimethylbenz(a)anthracene-induced mammary carcinogenesis in rats by resveratrol: role of nuclear factor-kappaB, cyclooxygenase 2, and matrix metalloprotease 9. Cancer Res. 62:4945–4954. 2002.PubMed/NCBI

45 

Cui X, Jin Y, Hofseth AB, et al: Resveratrol suppresses colitis and colon cancer associated with colitis. Cancer Prev Res. 3:549–559. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Harper CE, Patel BB, Wang J, Arabshahi A, Eltoum IA and Lamartiniere CA: Resveratrol suppresses prostate cancer progression in transgenic mice. Carcinogenesis. 28:1946–1953. 2007. View Article : Google Scholar : PubMed/NCBI

47 

George J, Singh M, Srivastava AK, et al: Resveratrol and black tea polyphenol combination synergistically suppress mouse skin tumors growth by inhibition of activated MAPKs and p53. PLoS One. 6:e233952011. View Article : Google Scholar : PubMed/NCBI

48 

Kapadia GJ, Azuine MA, Tokuda H, et al: Chemopreventive effect of resveratrol, sesamol, sesame oil and sunflower oil in the Epstein-Barr virus early antigen activation assay and the mouse skin two-stage carcinogenesis. Pharmacol Res. 45:499–505. 2002. View Article : Google Scholar

49 

Kowalczyk MC, Kowalczyk P, Tolstykh O, Hanausek M, Walaszek Z and Slaga TJ: Synergistic effects of combined phytochemicals and skin cancer prevention in SENCAR mice. Cancer Prev Res. 3:170–178. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September 2013
Volume 43 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kowalczyk MC, Junco JJ, Kowalczyk P, Tolstykh O, Hanausek M, Slaga T and Walaszek Z: Effects of combined phytochemicals on skin tumorigenesis in SENCAR mice. Int J Oncol 43: 911-918, 2013
APA
Kowalczyk, M.C., Junco, J.J., Kowalczyk, P., Tolstykh, O., Hanausek, M., Slaga, T. ., & Walaszek, Z. (2013). Effects of combined phytochemicals on skin tumorigenesis in SENCAR mice. International Journal of Oncology, 43, 911-918. https://doi.org/10.3892/ijo.2013.2005
MLA
Kowalczyk, M. C., Junco, J. J., Kowalczyk, P., Tolstykh, O., Hanausek, M., Slaga, T. ., Walaszek, Z."Effects of combined phytochemicals on skin tumorigenesis in SENCAR mice". International Journal of Oncology 43.3 (2013): 911-918.
Chicago
Kowalczyk, M. C., Junco, J. J., Kowalczyk, P., Tolstykh, O., Hanausek, M., Slaga, T. ., Walaszek, Z."Effects of combined phytochemicals on skin tumorigenesis in SENCAR mice". International Journal of Oncology 43, no. 3 (2013): 911-918. https://doi.org/10.3892/ijo.2013.2005