Ocoxin® oral solution slows down tumor growth in an experimental model of colorectal cancer metastasis to the liver in Balb/c mice

Joana Márquez, Jorge Mena, Iera Hernandez-Unzueta, Aitor Benedicto, Eduardo Sanz, Beatriz Arteta, Elvira Olaso, Joana Márquez, Jorge Mena, Iera Hernandez-Unzueta, Aitor Benedicto, Eduardo Sanz, Beatriz Arteta, Elvira Olaso

Abstract

Liver metastatic disease is the main cause of death in colorectal cancer (CRC) patients. During metastatic spread of the disease an imbalance in the oxidative stress and inflammation plays a crucial role in tumor progression. In order to improve the efficacy of current therapies, new complementary therapeutic approaches are being analyzed including biologically active compounds with low side effects. The anti-inflammatory and anti-oxidant properties of Ocoxin® oral solution (OOS) prompt us to analyze its effect on the metastatic development of CRC to the liver. First, in vitro effect of OOS in tumor cell viability and migration was analyzed. Second, in vivo effect of different dosage patterns and concentrations in the development of hepatic metastasis was analyzed by intra-splenic inoculation of C26 colon carcinoma cells in Balb/c mice. Third, the expression of alpha smooth muscle actin, caspase-3 and Ki-67 expression was quantified by immunohistochemistry, then gene expression levels of inflammatory factors were measured by quantitative RT-PCR. According to our results, OOS reduced tumor cell viability and migration in vitro. Moreover, in vivo daily administration of OOS from the 7th day after tumor cell inoculation decreased the total area and size of metastatic foci in the liver. Furthermore, cell proliferation and fibroblast recruitment was decreased in tumor foci while a higher number of apoptotic cells were observed. Finally, RNA levels for the inflammatory mediators COX-2, IFNγ, IL1β, IL6 and TNFα were reduced in total liver. In conclusion, OOS reduced the metastatic development of colorectal cancer to the liver by increasing apoptosis, and decreasing tumor cell proliferation and fibroblast recruitment in the tumor foci, as well as the expression of inflammatory mediators in total liver. These results point out OOS as a potential supplement to be applied as complementary therapy for the treatment of liver metastasis from colorectal cancer.

Figures

Figure 1
Figure 1
Diagram of the experimental animal groups according to OOS dosage pattern. Animals were divided into 3 experimental groups as follows: group I, OOS untreated group; group II, mice treated with OOS 4 days prior to tumor cell inoculation; group III, mice treated with OOS from the 7th day after tumor cell inoculation. All treated groups were administered a daily dose during the period of treatment.
Figure 2
Figure 2
OOS reduces in vitro viability of C26 cells. (A) The viability of C26 cells was tested after 24-h incubation in the presence of OOS. C26 cells were treated with increasing concentrations of OOS ranging from 1:200 to 1:50 during 24 h before viability quantification and compared to the viability of initially cultures cells (light grey) and to that of untreated cells (black). Differences in the viability of treated cells vs. Untreated cells (*) and vs. initially cultured cells (**) were considered to be statistically significant at P<0.05. (B) The cell number in each cell phase respect to total gated cell number was analyzed after PI staining by flow cytometry after 72-h incubation with 1:100 OOS (V/Vf). Differences between treated and untreated cells were considered statistically significant at *P<0.05.
Figure 3
Figure 3
OOS reduces in vitro migration of C26 cells. C26 cells were cultured on top of 8 µm-pore membrane modified Boyden chambers. The number of migrated C26 cells were quantified after 18-h incubation in the presence of 1:100 OOS (V/Vf) Data are mean values ± SD from three different experiments. Differences were considered statistically significant at *P<0.05.
Figure 4
Figure 4
OOS slows down in vivo tumor growth. (A) C26 cells were i.s. inoculated and mice were treated with 100 µl of OOS under the administration patterns described in Materials and methods. Group I, untreated; group II, OOS administered four days prior to tumor cell inoculation; group III, OOS administered from day 7th after tumor cell inoculation until sacrifice. (B) Mice were treated with 50 and 100 µl of OOS under administration pattern of group III. (C) The amount of liver foci developed in the liver of mice untreated or treated with 50 and 100 µl of OOS was quantified in liver tissue based on tumor foci size. Image original magnification, ×20. Differences were considered statistically significant at *P<0.05.
Figure 5
Figure 5
OOS affects proliferation and apoptosis in vivo. The effect of OOS on Ki-67 and caspase-3 expression in livers from tumor-bearing mice was analyzed by immunohistochemistry. (A) Caspase-3 expression (brown) was quantified in livers collected from untreated, 50 and 100 µl OOS treated C26-bearing mice as the percentage of the area positive for caspase-3 expression respect to total liver section area. (B) Ki-67 expression (brown) was quantified in livers collected from untreated, 50 and 100 µl OOS treated C26-bearing mice as the percentage of the area positive for Ki-67 expression in respect to total liver section area. (C) The ratio between Ki-67 and caspase-3 was calculated as the expression of Ki-67 relative to caspase-3 expression. Image original magnification, ×20. Differences were considered statistically significant at *P<0.05.
Figure 6
Figure 6
In vivo HSC infiltration in the tumor is reduced by OOS. Expression levels of ASMA were analyzed in liver tissue by immunohistochemistry. ASMA was stained with specific antibodies in liver tissue collected from untreated, 50 and 100 µl OOS treated mice. Data are calculated as % of ASMA expression per tumor foci area. Image original magnification, ×20. Differences were considered statistically significant at *P<0.05.
Figure 7
Figure 7
OOS affects inflammatory gene expression in total liver. Gene expression of IFNγ, TNFα, IL1β, IL6 and COX-2 was analyzed by RT-PCR. Total RNA was extracted from paraffin-embedded liver tissue collected from untreated or 100 µl OOS treated C26-bearing mice. Data are expressed as the mean values ± SD from six livers from two independent experiments. Differences were considered statistically significant at *P<0.05.

References

    1. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic Biol Med. 2010;49:1603–1616. doi: 10.1016/j.freeradbiomed.2010.09.006.
    1. Mamlouk S, Wielockx B. Hypoxia-inducible factors as key regulators of tumor inflammation. Int J Cancer. 2013;132:2721–2729. doi: 10.1002/ijc.27901.
    1. Zhang Y, Tang H, Cai J, Zhang T, Guo J, Feng D, Wang Z. Ovarian cancer-associated fibroblasts contribute to epithelial ovarian carcinoma metastasis by promoting angiogenesis, lymphangiogenesis and tumor cell invasion. Cancer Lett. 2011;303:47–55. doi: 10.1016/j.canlet.2011.01.011.
    1. Shankar S, Suthakar G, Srivastava RK. Epigallocatechin-3-gallate inhibits cell cycle and induces apoptosis in pancreatic cancer. Front Biosci. 2007;12:5039–5051. doi: 10.2741/2446.
    1. Goldberg ED, Amosova EN, Zueva EP, Razina TG, Krylova SG, Zorikov PS. Licorice preparations improve efficiency of chemotherapy and surgical treatment of transplanted tumors. Bull Exp Biol Med. 2008;145:252–255. doi: 10.1007/s10517-008-0063-0.
    1. Ma YC, Li C, Gao F, Xu Y, Jiang ZB, Liu JX, Jin LY. Epigallocatechin gallate inhibits the growth of human lung cancer by directly targeting the EGFR signaling pathway. Oncol Rep. 2014;31:1343–1349.
    1. Maruyama T, Murata S, Nakayama K, Sano N, Ogawa K, Nowatari T, Tamura T, Nozaki R, Fukunaga K, Ohkohchi N. (−)-Epigallocatechin-3-gallate suppresses liver metastasis of human colorectal cancer. Oncol Rep. 2014;31:625–633.
    1. Roomi MW, Roomi N, Ivanov V, Kalinovsky T, Niedzwiecki A, Rath M. Inhibition of pulmonary metastasis of melanoma b16fo cells in C57BL/6 mice by a nutrient mixture consisting of ascorbic Acid, lysine, proline, arginine, and green tea extract. Exp Lung Res. 2006;32:517–530. doi: 10.1080/01902140601098552.
    1. Roomi MW, Kalinovsky T, Niedzwiecki A, Rath M. Modulation of uPA, MMPs and their inhibitors by a novel nutrient mixture in human colorectal, pancreatic and hepatic carcinoma cell lines. Int J Oncol. 2015;47:370–376.
    1. Gomez EV, Perez YM, Sanchez HV, Forment GR, Soler EA, Bertot LC, Garcia AY, del Rosario Abreu vazquez M, Fabian LG. Antioxidant and immunomodulatory effects of viusid in patients with chronic hepatitis C. World J Gastroenterol. 2010;16:2638–2647. doi: 10.3748/wjg.v16.i21.2638.
    1. Vilar Gomez E, Sanchez Rodriguez Y, Torres Gonzalez A, Calzadilla Bertot L, Arus Soler E, Martinez Perez Y, Yasells Garcia A, Abreu Vazquez MR. Viusid, a nutritional supplement, increases survival and reduces disease progression in HCV-related decompensated cirrhosis: A randomised and controlled trial. BMJ Open. 2011;1:e000140. doi: 10.1136/bmjopen-2011-000140.
    1. Hernández-García S, González V, Sanz E, Pandiella A. Effect of oncoxin oral solution in HER2-overexpressing breast cancer. Nutr Cancer. 2015;67:1159–1169. doi: 10.1080/01635581.2015.1068819.
    1. Tommelein J, Verset L, Boterberg T, Demetter P, Bracke M, De Wever O. Cancer-associated fibroblasts connect metastasis-promoting communication in colorectal cancer. Front Oncol. 2015;5(63) doi: 10.3389/fonc.2015.00063.
    1. Roomi MW, Kalinovsky T, Cha J, Roomi NW, Niedzwiecki A, Rath M. Effects of a nutrient mixture on immunohistochemical localization of cancer markers in human cervical cancer Hela cell tumor xenografts in female nude mice. Exp Ther Med. 2015;9:294–302.
    1. Lee SK, Park KK, Park JH, Lim SS, Chung WY. The inhibitory effect of roasted licorice extract on human metastatic breast cancer cell-induced bone destruction. Phytother Res. 2013;27:1776–1783. doi: 10.1002/ptr.4930.
    1. Liu Y, Yu QY, Zhu ZL, Tang PY, LI K. Vitamin B2 intake and the risk of colorectal cancer: A meta-analysis of observational studies. Asian Pac J Cancer Prev. 2015;16:909–913. doi: 10.7314/APJCP.2015.16.3.909.
    1. Lode HN, Huebener N, Strandsby A, Gaedicke G. Nutrient mixture including vitamin C, L-lysine, L-proline, and epigal-locatechin is ineffective against tumor growth and metastasis in a syngeneic neuroblastoma model. Pediatr Blood Cancer. 2008;50:284–288. doi: 10.1002/pbc.21172.
    1. Niedzwiecki A. Micronutrient mixture in tumor growth and metastasis. Pediatr Blood Cancer. 2008;50:422–423. 424–425. doi: 10.1002/pbc.21467. author reply.
    1. Srividhya R, Jyothilakshmi V, Arulmathi K, Senthilkumaran V, Kalaiselvi P. Attenuation of senescence-induced oxidative exacerbations in aged rat brain by (−)-epigallocatechin-3-gallate. Int J Dev Neurosci. 2008;26:217–223. doi: 10.1016/j.ijdevneu.2007.12.003.
    1. Eferl R, Wagner EF. AP-1: A double-edged sword in tumori-genesis. Nat Rev Cancer. 2003;3:859–868. doi: 10.1038/nrc1209.
    1. Takada Y, Aggarwal BB. Flavopiridol inhibits NF-kappaB activation induced by various carcinogens and inflammatory agents through inhibition of IkappaBalpha kinase and p65 phosphorylation: Abrogation of cyclin D1, cyclooxygenase-2, and matrix metalloprotease-9. J Biol Chem. 2004;279:4750–4759. doi: 10.1074/jbc.M304546200.
    1. Gupta S, Hussain T, Mukhtar H. Molecular pathway for (−)-epigallocatechin-3-gallate-induced cell cycle arrest and apoptosis of human prostate carcinoma cells. Arch Biochem Biophys. 2003;410:177–185. doi: 10.1016/S0003-9861(02)00668-9.
    1. Hsia SM, Yu CC, Shih YH, Yuanchien Chen M, Wang TH, Huang YT, Shieh TM. Head Neck. Jan 10, 2015. Isoliquiritigenin as a cause of DNA damage and inhibitor of ataxia-telangiectasia mutated expression leading to G2/M phase arrest and apoptosis in oral squamous cell carcinoma. Epub ahead of print.
    1. He Y, Zhang X, Zeng X, Huang Y, Wei JA, Han L, Li CX, Zhang GW. HuR-mediated posttranscriptional regulation of p21 is involved in the effect of Glycyrrhiza uralensis licorice aqueous extract on polyamine-depleted intestinal crypt cells proliferation. J Nutr Biochem. 2012;23:1285–1293. doi: 10.1016/j.jnutbio.2011.07.009.
    1. Lee JS, Ahn JH, Cho YJ, Kim HY, Yang YI, Lee KT, Jang DS, Choi JH. α-Terthienylmethanol, isolated from Eclipta prostrata, induces apoptosis by generating reactive oxygen species via NADPH oxidase in human endometrial cancer cells. J Ethnopharmacol. 2015;169:426–434. doi: 10.1016/j.jep.2015.04.029.
    1. Olaso E, Salado C, Egilegor E, Gutierrez V, Santisteban A, Sancho-Bru P, Friedman SL, Vidal-Vanaclocha F. Proangiogenic role of tumor-activated hepatic stellate cells in experimental melanoma metastasis. Hepatology. 2003;37:674–685. doi: 10.1053/jhep.2003.50068.
    1. Berdiel-Acer M, Sanz-Pamplona R, Calon A, Cuadras D, Berenguer A, Sanjuan X, Paules MJ, Salazar R, Moreno V, Batlle E, et al. Differences between CAFs and their paired NCF from adjacent colonic mucosa reveal functional heterogeneity of CAFs, providing prognostic information. Mol Oncol. 2014;8:1290–1305. doi: 10.1016/j.molonc.2014.04.006.
    1. Hassan LE, Ahamed MB, Majid AS, Baharetha HM, Muslim NS, Nassar ZD, Majid AM. Correlation of antiangiogenic, antioxidant and cytotoxic activities of some Sudanese medicinal plants with phenolic and flavonoid contents. BMC Complement Altern Med. 2014;14(406)
    1. Fassina G, Venè R, Morini M, Minghelli S, Benelli R, Noonan DM, Albini A. Mechanisms of inhibition of tumor angiogenesis and vascular tumor growth by epigallocatechin-3-gallate. Clin Cancer Res. 2004;10:4865–4873. doi: 10.1158/1078-0432.CCR-03-0672.
    1. Leibovich SJ, Polverini PJ, Shepard HM, Wiseman DM, Shively V, Nuseir N. Macrophage-induced angiogenesis is mediated by tumour necrosis factor-alpha. Nature. 1987;329:630–632. doi: 10.1038/329630a0.
    1. Saijo Y, Tanaka M, Miki M, Usui K, Suzuki T, Maemondo M, Hong X, Tazawa R, Kikuchi T, Matsushima K, et al. Proinflammatory cytokine IL-1 beta promotes tumor growth of lewis lung carcinoma by induction of angiogenic factors: In vivo analysis of tumor-stromal interaction. J Immunol. 2002;169:469–475. doi: 10.4049/jimmunol.169.1.469.
    1. Tsujii M, Kawano S, DuBois RN. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci USA. 1997;94:3336–3340. doi: 10.1073/pnas.94.7.3336.
    1. Tsujii M, Kawano S, Tsuji S, Sawaoka H, Hori M, DuBois RN. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell. 1998;93:705–716. doi: 10.1016/S0092-8674(00)81433-6.
    1. Sun Y, Tang XM, Half E, Kuo MT, Sinicrope FA. Cyclooxygenase-2 overexpression reduces apoptotic susceptibility by inhibiting the cytochrome c-dependent apoptotic pathway in human colon cancer cells. Cancer Res. 2002;62:6323–6328.
    1. Han YD, Hong YK, Kang JG, Choi YJ, Park CH. Relation of the expression of cyclooxygenase-2 in colorectal adenomas and adenocarcinomas to angiogenesis and prognosis. J Korean Soc Coloproctol. 2010;26:339–346. doi: 10.3393/jksc.2010.26.5.339.
    1. Valcárcel M, Arteta B, Jaureguibeitia A, Lopategi A, Martínez I, Mendoza L, Muruzabal FJ, Salado C, Vidal-Vanaclocha F. Three-dimensional growth as multicellular spheroid activates the proangiogenic phenotype of colorectal carcinoma cells via LFA-1-dependent VEGF: Implications on hepatic micrometastasis. J Transl Med. 2008;6(57) doi: 10.1186/1479-5876-6-57.
    1. Arteta B, Lasuen N, Lopategi A, Sveinbjörnsson B, Smedsrød B, Vidal-Vanaclocha F. Colon carcinoma cell interaction with liver sinusoidal endothelium inhibits organ-specific antitumor immunity through interleukin-1-induced mannose receptor in mice. Hepatology. 2010;51:2172–2182. doi: 10.1002/hep.23590.

Source: PubMed

3
Prenumerera