DOI QR코드

DOI QR Code

Effect of Luteolin on the Levels of Glycoproteins During Azoxymethane-induced Colon Carcinogenesis in Mice

  • Published : 2012.04.30

Abstract

Luteolin (LUT), a bioflavonoid has been used as a chemopreventive agent world-wide against chemically induced cancer. Hence we designed an experiment to assess chemopreventive action of LUT on lipid peroxidation (LPO) and glycoconjugates in azoxymethane (AOM)-induced colon carcinogenesis. Colon cancer was induced by 15 mg/body kg. body weight of AOM and administration of LUT (at the dose of 1.2 mg/kg. body weight) was till end of the study. Analysis of lipid peroxidative end products such as protein carbonyl (PC), malonadehyde (MDA) and conjucated dienes (CD) demonstrated significant increase in in AOM-induced animals with reduction by LUT (p<0.05). Increased levels of glycoconjugates such as hexose, hexosamine, sialic acid, fucose and mucoprotein were analyzed in serum and colon tissues examined histopathologically by periodic acid Schiff's (PAS) staining were also reversed by LUT l(p<0.05). The secondary marker of colon cancer mucin depleted foci (MDF) was assessed in control and experimental group of animals. A characteristic increase of MDF was observed in AOM-induced colon cancer animals. Treatment with LUT decreased the incidence of MDF. These results suggest that LUT alters the expression of glycoconjugates and suppress colon cancer. Hence, we speculate that LUT can be used as a chemopreventive agent to treat colon cancer.

Keywords

References

  1. Asano N, Kuno T, Hirose Y, et al (2007). Preventive effects of a flavonoid myricitrin on the formation of azoxymethaneinduced premalignant lesions in colons of rats. Asian Pac J Cancer Prev, 8, 73-6.
  2. Ashokkumar P, Sudhandiran G (2008). Protective role of Luteolin on the status of lipid peroxidation and antioxidant defense against Azoxymethane-induced experimental colon carcinogenesis. Biomed Pharmacother, 62, 590-7. https://doi.org/10.1016/j.biopha.2008.06.031
  3. Ashokkumar P, Sudhandiran G (2011). Luteolin inhibits cell proliferation during Azoxymethane-induced experimental colon carcinogenesis via Wnt/$\beta$-catenin pathway. Invest New Drugs, 29, 273-84. https://doi.org/10.1007/s10637-009-9359-9
  4. Burnstein MJ (1993). Dietary factors related to colorectal neoplasms. Surg Clin North America, 73, 13-29.
  5. Caderni G, Femia AP, Giannini A, et al (2003). Identification of mucin-depleted foci in the unsectioned colon of azoxymethane-treated rats: correlation with carcinogenesis. Cancer Res, 63, 2388-92.
  6. Chihara T, Shimpo K, Kaneko T, Beppu H (2010). Inhibition of 1, 2-dimethylhydrazine-induced mucin-depleted foci and O6-methylguanine DNA adducts in the ratolorectum by boiled garlic powder. Asian Pac J Cancer Prev, 11, 1301-4
  7. Dische Z, Shuttle S (1960). Fucose and sialic acid in glycoproteins of the mucus of the digestive tract. Fed Proc, 19, 904-10.
  8. Femia AP, Dolara P, Caderni G (2004). Mucin-depleted foci (MDF) in the colon of rats treated with azoxymethane (AOM) are useful biomarkers for colon carcinogenesis. Carcinogenesis, 25, 277-81.
  9. Feron VJ, Tie HP, DeVrjer F, et al (1991). Aldehydes occurrence, carcinogenic potential mechanism of action and risk assessment. Mutat Res, 259, 362-6.
  10. Greenlee R, Bolden SM, Wingo PA (2000). Cancer statistics, 2000. CA Cancer J Clin, 50, 7-33. https://doi.org/10.3322/canjclin.50.1.7
  11. Hakomori S (1996). Tumor malignancy defined by aberrant glycosylation and sphingo (glyco) lipid metabolism. Cancer Res, 56, 5309-18.
  12. Hicky R, Clark RL, Cumbley RW (1986). Year book of cancer. Chicago: Year book, Med Publishers, 427.
  13. Hynes RO (1978). Cell surface proteins and malignant transformation. Biochem Biophys Acta, 458, 73-107.
  14. Kelin RA (1970). The detection of oxidation in liposome preparations. Biochem Biophys Acta, 210, 486-92. https://doi.org/10.1016/0005-2760(70)90046-9
  15. Kierman JA (1990). Histological & histochemical methods - Theory and practice, second ed. Pergamon Press, Frankfurt, p 433.
  16. Kimata M, Inagaki N, Nagai H (2000a). Effects of luteolin and other flavonoids on IgE-mediated allergic reactions. Planta Med, 66, 25-9. https://doi.org/10.1055/s-2000-11107
  17. Kimata M, Shichijo M, Miura T, et al (2000b). Effects of luteolin, quercetin and baicalein on immunoglobulin E-mediated mediator release from human cultured mast cells. Clin Exp Allergy, 30, 501-8. https://doi.org/10.1046/j.1365-2222.2000.00768.x
  18. HK, Cheon BS, Kim SY, et al (1999). Effects of naturally occurring flavonoids on nitric oxide production in the macrophage cell line RAW 264.7 and their structure and activity relationships. Biochem Pharmacol, 58, 759-65 https://doi.org/10.1016/S0006-2952(99)00160-4
  19. Levine M, Dharwal KR, Welch RW, et al (1995). Determination of optimal vitamin C requirements in humans. Am J Clin Nutr, 62, 1347-56.
  20. Macbeth RA, Bskesi JG (1964). Plasma glycoproteins of malignant disease. Adv Carbohydr Chem Biochem, 88, 635-7.
  21. Montreuil J (1980). Primary structure of glycoprotein glycans basis for the molecular biology of glycoproteins. Adv Carbohydr Chem Biochem, 37, 157-223. https://doi.org/10.1016/S0065-2318(08)60021-9
  22. Niebes P (1972). Determination of enzymes and degradation products of glucosamine glycan metabolism in the serum of health and various subjects. Clin Chim Acta, 42, 399-408. https://doi.org/10.1016/0009-8981(72)90105-2
  23. Ohkawa H, Ohishi N, Yagi K (1979). Assay for lipid peroxides in animal tissues by thiobarbutaric acid reaction. Anal Biochem, 95, 351-8. https://doi.org/10.1016/0003-2697(79)90738-3
  24. Olden K, Parent JB, White SL (1982). Carbohydrate moieties of glycoproteins a re-evaluation of their functions. Biochem Biophys Acta, 650, 209-32. https://doi.org/10.1016/0304-4157(82)90017-X
  25. Patel PS, Baxi BR, Balar DP (1989). Significance of serum sialoglycoproteins in patients with lung cancer. Neoplasma, 36, 53-9.
  26. Patel PS, Baxi BR, Adhavava SG, et al (1990). Individual and combined usefulness of lipid associated sialic acid and hexoses as tumour marker in breast cancers. Cancer Lett, 51, 203-8. https://doi.org/10.1016/0304-3835(90)90103-5
  27. Perez-Garcia F, Adzet T, Canigueral S (2000). Activity of artichoke leaf extract on reactive oxygen species in human leukocytes. Free Rad Res, 33, 661-5. https://doi.org/10.1080/10715760000301171
  28. Prabhu PN, Ashokkumar P, Sudhandiran G (2009). Antioxidative and antiproliferative effects of astaxanthin during the initiation stages of 1,2-dimethyl hydrazine-induced experimental colon carcinogenesis. Fund Clin Pharmacol, 23, 225-34. https://doi.org/10.1111/j.1472-8206.2009.00669.x
  29. Samy RP, Gopalakrishnan P, Ignacimuthu S (2006). Anti-tumor promoting potential of luteolin against 7, 12-dimethylbenz(a) anthracene-induced mammary tumors in rats. Chem Biol Interact, 164, 1-13. https://doi.org/10.1016/j.cbi.2006.08.018
  30. Scholz D, Horpacsy G, Mebel M (1979). Late prognosis in acute post-transplant renal failure in 102 patients. Eur Urol, 5, 14-7.
  31. Selvam S, Nagini S (1995). Administration of the plasticizer di(engl hexyl)phthalate alters glycoconjugate profile. Ind J Physiol Pharmacol, 39, 252-4.
  32. Sengupta A, Ghosh S, Das S (2002). Inhibition of Cell Proliferation and Induction of Apoptosis During Azoxymethane Induced Colon Carcinogenesis by Black Tea. Asian Pac J Cancer Prev, 3, 41-6.
  33. Sen U, Guha S, Chowdhury JR (1983). Serum fucosyl transferase activity and serum fucose levels as diagnostic tools in malignancy. Acta Med Okayama, 37, 457-62.
  34. Shetlar MR, Erwir CP, Evereft MR (1950). Serum polysaccharide levels in rats bearing the walker 256 tumor. Cancer Res, 10, 445-7.
  35. Shimoi K, Masuda S, Furugori M, et al (1994). Radioprotective effect of antioxidative flavonoids in g-ray irradiated mice. Carcinogenesis, 15, 2669-72. https://doi.org/10.1093/carcin/15.11.2669
  36. Skrzydlewska E, Kouszko B, Sulkowska M, et al (2003). Antioxidant potential in esophageal, stomach and colorectal cancers. Hepatogasteroenterol, 50, 126-31.
  37. Slattery ML, Edwards SL, Boucher KM, et al (1999). Life style and colon cancer: an assessment of factors associated with risk. Am J Epidemiol, 150, 869-77. https://doi.org/10.1093/oxfordjournals.aje.a010092
  38. Srinivasan P, Sabitha KE, Shyamaladevi CS (2006). Modulatory efficacy of Green tea polyphenols on glycoconjugates and immunological markers in 4-Nitroquinoline 1-oxide-induced oral carcinogenesis-A therapeutic approach. Chem Biol Inter, 162, 149-56. https://doi.org/10.1016/j.cbi.2006.05.021
  39. Sriram N, Kalayarasan S, Ashokkumar P, et al (2008). Diallyl sulfide induces apoptosis in Colo 320 DM human colon cancer cells: involvement of caspase-3, NF-$\kappa B$, and ERK-2. Mol Cell Biochem, 311, 157-63. https://doi.org/10.1007/s11010-008-9706-8
  40. Swan DK, Ford B (1997). Chemoprevention of cancer: review of the literature. Oncol. Nursing Forum, 24, 719-27.
  41. Tanwar L, Vaish V, Sanyal SN (2009). Chemoprevention of 1,2-Dimethylhydrazine-induced colon carcinogenesis by a Non-steroidal anti-inflammatory drug, etoricoxib, in rats: Inhibition of nuclear factor $\kappa B$. Asian Pac J Cancer Prev, 10, 1141-6.
  42. Upsani CD, Khera A, Balaraman R (2001). Effect of Lead and vitamin E, C or spiruline on malondialdehyde, conjucated dienes and hydroperoxides in rats. Ind J Exp Biol, 39, 70-4.
  43. Vaca CE, Wilhelm J, Harms-Rihsdash M (1998). Interaction of lipid peroxidation product with DNA. Mutat Res Rev Genet Toxicol, 195, 137.
  44. Veda H, Yamazaki C, Yamazaki M (2002). Luteolin as an anti-inflammatory and anti-allergic constituent of Perilla frutescens. Biol Pharm Bull, 25, 1197-202. https://doi.org/10.1248/bpb.25.1197
  45. Wagner WD (1979). A more sensitive assay discriminating galactosamine and glucosamine in mixture. Anal Biochem, 94, 394-6. https://doi.org/10.1016/0003-2697(79)90379-8
  46. Williams CS, Mann M, DuBois RN (1999). The role of cyclooxygenases in inflammation, cancer, and development. Oncogene, 18, 7908-16.
  47. Winzler RJ (1955). Determination of serum glycoproteins. Methods Biochem Anal, 2, 279-311. https://doi.org/10.1002/9780470110188.ch10
  48. Yasukawa K, Takido M, Takeuchi M, et al (1989). Effect of chemical constituents from plants on 12-O-tetradecanoylphorbol-13- acetate induced inflammation in mice. Chem Pharm Bull, 37, 1071-3. https://doi.org/10.1248/cpb.37.1071
  49. Yoshimi N, Morioka T, Kinjo T, et al (2004). Histological and immunohistochemical observations of mucin-depleted foci (MDF) stained with Alcian blue, in rat colon carcinogenesis induced with 1, 2-dimethylhydrazine dihydrochloride. Cancer Sci, 95, 792-7. https://doi.org/10.1111/j.1349-7006.2004.tb02183.x
  50. Zieba M, Nowak D, Suwalski M, et al (2001). Enhanced lipid peroxidation in cancer tissue homogenate in non-small cell lung cancer. Monaldi Arch Chest Dis, 56, 110-4.

Cited by

  1. Dietary Non-nutritive Factors in Targeting of Regulatory Molecules in Colorectal Cancer: An Update vol.14, pp.10, 2013, https://doi.org/10.7314/APJCP.2013.14.10.5543
  2. Luteolin, a Bioflavonoid, Attenuates Azoxymethane-Induced Effects on Mitochondrial Enzymes in Balb/c Mice vol.14, pp.11, 2013, https://doi.org/10.7314/APJCP.2013.14.11.6669
  3. Potential Targets for Prevention of Colorectal Cancer: a Focus on PI3K/Akt/mTOR and Wnt Pathways vol.14, pp.4, 2013, https://doi.org/10.7314/APJCP.2013.14.4.2201
  4. Luteolin-loaded Phytosomes Sensitize Human Breast Carcinoma MDA-MB 231 Cells to Doxorubicin by Suppressing Nrf2 Mediated Signalling vol.15, pp.13, 2014, https://doi.org/10.7314/APJCP.2014.15.13.5311
  5. Luteolin, a Bioflavonoid Inhibits Colorectal Cancer through Modulation of Multiple Signaling Pathways: A Review vol.15, pp.14, 2014, https://doi.org/10.7314/APJCP.2014.15.14.5501
  6. Signal Transducer and Activator of Transcription 3 - A Promising Target in Colitis-Associated Cancer vol.15, pp.2, 2014, https://doi.org/10.7314/APJCP.2014.15.2.551
  7. Luteolin, a bioflavonoid inhibits Azoxymethane-induced colorectal cancer through activation of Nrf2 signaling vol.24, pp.1, 2014, https://doi.org/10.3109/15376516.2013.843111
  8. S-allyl cysteine alleviates nonsteroidal anti-inflammatory drug-induced gastric mucosal damages by increasing cyclooxygenase-2 inhibition, heme oxygenase-1 induction, and histone deacetylation inhibition vol.29, pp.08159319, 2014, https://doi.org/10.1111/jgh.12730
  9. Luteolin inhibits matrix metalloproteinase 9 and 2 in azoxymethane-induced colon carcinogenesis vol.33, pp.11, 2014, https://doi.org/10.1177/0960327114522502
  10. The Chemopotential Effect of Annona muricata Leaves against Azoxymethane-Induced Colonic Aberrant Crypt Foci in Rats and the Apoptotic Effect of Acetogenin Annomuricin E in HT-29 Cells: A Bioassay-Guided Approach vol.10, pp.4, 2015, https://doi.org/10.1371/journal.pone.0122288
  11. Luteolin-induced protection of H2O2-induced apoptosis in PC12 cells and the associated pathway vol.12, pp.5, 2015, https://doi.org/10.3892/mmr.2015.4400
  12. Exogenous Natural Glycoprotein Multiple Mechanisms of Anti-tumor Activity vol.16, pp.4, 2015, https://doi.org/10.7314/APJCP.2015.16.4.1331
  13. -Associated Chronic Atrophic Gastritis and to Prevent Tumorigenesis vol.21, pp.1, 2016, https://doi.org/10.1111/hel.12229
  14. Modulatory Effect of Taurine on 7,12-Dimethylbenz(a)Anthracene-Induced Alterations in Detoxification Enzyme System, Membrane Bound Enzymes, Glycoprotein Profile and Proliferative Cell Nuclear Antigen in Rat Breast Tissue vol.30, pp.8, 2016, https://doi.org/10.1002/jbt.21805
  15. Preparation and Characterization of Copolymer Micelles for the Solubilization and In Vitro Release of Luteolin and Luteoloside vol.18, pp.6, 2017, https://doi.org/10.1208/s12249-016-0692-y
  16. Pro-Apoptotic Effect of Rice Bran Inositol Hexaphosphate (IP6) on HT-29 Colorectal Cancer Cells vol.14, pp.12, 2013, https://doi.org/10.3390/ijms141223545
  17. Luteolin induces apoptosis in vitro through suppressing the MAPK and PI3K signaling pathways in gastric cancer vol.14, pp.2, 2017, https://doi.org/10.3892/ol.2017.6380
  18. Polyphenols in Colorectal Cancer: Current State of Knowledge including Clinical Trials and Molecular Mechanism of Action vol.2018, pp.2314-6141, 2018, https://doi.org/10.1155/2018/4154185
  19. Protective effect of Luteolin against methyl methanesulfonate-induced toxicity pp.1556-9551, 2021, https://doi.org/10.1080/15569543.2018.1564142