DOI QR코드

DOI QR Code

Luteolin, a Bioflavonoid, Attenuates Azoxymethane-Induced Effects on Mitochondrial Enzymes in Balb/c Mice

  • Published : 2013.11.30

Abstract

Colon cancer (CRC) is a serious health problem throughout the world. Development of novel drugs without side effects for this cancer is crucial. Luteolin (LUT), a bioflavonoid, has many beneficial effects such as antioxidant, anti-inflammatory and anti-proliferative potential. was a potent chemical carcinogen used for the induction of colon cancer. Colon carcinogenesis was initiated by intraperitoneal injection of azoxymethane (AOM) to mice at the dose of 15 mg/body kg weight in Balb/C mice for 3 weeks. Mice were treated with LUT at the dose of 1.2 mg/body kg weight orally. Mitochondrial enzymes such as isocitrate dehydrogenase (ICDH), ${\alpha}$-keto dehydrogenase (${\alpha}$-KDH), succinate dehydrogenase (SDH) and the activities of respiratory chain enzymes NADH dehydrogenase and cytochrome c oxidase were found to be elevated in AOM-treated animals. Treatment with LUT decreased the activities of all the parameters significantly. Hence, LUT might be a potent anticancer agent against colorectal cancer.

Keywords

References

  1. 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
  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. Dalton TP, Shertzer HG (1999). Puga A. Regulation of gene expression by reactive oxygen. Annu Rev Pharmacol Toxicol, 39, 67-101. https://doi.org/10.1146/annurev.pharmtox.39.1.67
  4. Hagen TM, Wehr CM, Ames BN (1998). Mitochondrial decay in aging. Reversal through supplementation of acetyl-L-carnitine and N-tert-butyl-alpha-phenyl-nitrone. Ann NY Acad Sci, 854, 214-23. https://doi.org/10.1111/j.1749-6632.1998.tb09904.x
  5. Johnson D, Lardy H (1967). Isolation of liver and kidney mitochondria. Methods Enzymol, 10, 94-6. https://doi.org/10.1016/0076-6879(67)10018-9
  6. King J (1965). Practical clinical enzymology. London: D. van. Nostrand Company, 83-93.
  7. Kumar A, Pandurangan AK, Lu F, et al (2012). Chemopreventive sphingadienes downregulate wnt signaling via a PP2A/Akt/GSK3$\beta$ pathway in colon cancer. Carcinogenesis, 33, 1726-35. https://doi.org/10.1093/carcin/bgs174
  8. Lenaz G (1998). Role of mitochondria in oxidative stress and ageing. Biochim Biophys Acta, 1366, 53-67. https://doi.org/10.1016/S0005-2728(98)00120-0
  9. Lowry OH, Rusebrough NJ, Farr AL, Randall RJ (1951). Protein measurement with folin-phenol reagent. J Biol Chem, 193, 265-75.
  10. Melov S, Coskun P, Patel M, et al (1999). Mitochondrial disease in superoxide dismutase 2 mutant mice. Proc Natl Acad Sci USA, 96, 846-51. https://doi.org/10.1073/pnas.96.3.846
  11. Minakami S, Ringler RL, Singer TP (1962). Studies on the respiratory-chain linked dihydrodiphosphopyridine nucleotide dehydrogenase. Assay of the enzyme in particulate and in soluble preparations. J Biol Chem, 237, 569-76.
  12. Pandurangan AK (2013). Potential targets for the prevention of colorectal cancer: A focus on PI3K/Akt/mTOR and Wnt pathways. Asian Pac J Cancer Prev, 14, 3007-11.
  13. Pandurangan AK, Anandasadagopan SK, Dharmalingam P, et al (2013c). Luteolin, a bioflavonoid inhibits Azoxymethane-induced colorectal cancer through Nrf2 signaling. Toxicol Mech and Methods, [Epub Ahead of Print].
  14. Pandurangan AK, Dharmalingam P, Ananda Sadagopan SK, et al (2013). Luteolin induces growth arrest in colon cancer cells through involvement of Wnt/$\beta$-catenin/GSK-3$\beta$. J Environ Pathol Toxicol Oncol, 32, 131-9. https://doi.org/10.1615/JEnvironPatholToxicolOncol.2013007522
  15. Pandurangan AK, Dharmalingam P, Anandasadagopan SK, Ganapasam S (2012a). Effect of luteolin on the levels of glycoproteins during azoxymethane-induced colon carcinogenesis in mice. Asian Pac J Cancer Prev, 13, 1569-73. https://doi.org/10.7314/APJCP.2012.13.4.1569
  16. Pandurangan AK, Dharmalingam P, Anandasadagopan SK, Ganapasam S (2013a). Inhibitory effect of Luteolin on the status of membrane bound ATPases against Azoxymethane-induced colorectal cancer. J Chem Pharm Res, 5, 123-127.
  17. Pandurangan AK, Ganapasam S (2013a). Luteolin modulates cellular thiols on azoxymenthane-induced colon carcinogenesis. Asian J Exp Biol Sci, 4, 245-50.
  18. Pandurangan AK, Ganapsam S (2013b). Luteolin induces apoptosis in azoxymethane-induced colon carcinogenesis through involvement of Bcl-2, Bax, and Caspase-3. J Chem Pharm Res, 5, 143-8.
  19. Pandurangan AK, Periasamy S, Anandasadagopan SK, et al (2012). Green tea polyphenol protection against 4-nitroquinoline 1-oxide-induced bone marrow lipid peroxidation and genotoxicity in Wistar rats. Asian Pac J Cancer Prev, 13, 4107-12. https://doi.org/10.7314/APJCP.2012.13.8.4107
  20. Pearl N, Cancrcao J, Zweifach BW (1963). Micro determination of cytochrome oxidase in rat tissues by the oxidation of N-phenyl-p-phenylenediamine or ascorbic acid. J Histochem Cytochem, 11, 102-4. https://doi.org/10.1177/11.1.102
  21. Pederson PL (1978). Tumor mitochondria and the bioenergetics of cancer cells. Progress in Exp Tumor Res, 22, 190-274.
  22. Reed LJ, Mukherjee BB (1969). Alpha-Ketoglutarate dehydrogenase complex from Escherichia coli. Methods Enzymol, 13, 55-61. https://doi.org/10.1016/0076-6879(69)13016-5
  23. Ross JA, Kasum CM (2002). Dietary flavonoids: bio availability, metabolic effects and safety. Annu Rev Nutr, 22, 19-34. https://doi.org/10.1146/annurev.nutr.22.111401.144957
  24. 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, 1-14. https://doi.org/10.1016/j.cbi.2006.08.018
  25. Shafie NH, Mohd Esa NM, Ithnin H, et al (2013). Prophylactic Inositol Hexaphosphate (IP6) inhibits colon cancer through involvement of Wnt/$\beta$-catenin and COX-2 pathway. BioMed Res Int, 2013, 681027.
  26. 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
  27. Slater EC, Bonner WD (1952). Effect of fluoride on succinate oxidase system. Biochem J, 52, 185-96. https://doi.org/10.1042/bj0520185
  28. Sohal RS (1993). Aging, cytochrome oxidase activity and hydrogen peroxide release by mitochondria. Free Radic Biol Med, 14, 583-8. https://doi.org/10.1016/0891-5849(93)90139-L
  29. Sriram N, Kalayrasan 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. Mol Cell Biochem, 311, 157-65. https://doi.org/10.1007/s11010-008-9706-8
  30. Ueda 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
  31. Yamashita N, Kawanishi S (2000). Distinct mechanisms of DNA damage in apoptosis induced by quercetin and luteolin. Free Radic Res, 33, 623-33. https://doi.org/10.1080/10715760000301141

Cited by

  1. 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
  2. The dietary flavone luteolin epigenetically activates the Nrf2 pathway and blocks cell transformation in human colorectal cancer HCT116 cells vol.119, pp.11, 2018, https://doi.org/10.1002/jcb.27275
  3. Selective Cytotoxicity of Luteolin and Kaempferol on Cancerous Hepatocytes Obtained from Rat Model of Hepatocellular Carcinoma: Involvement of ROS-Mediated Mitochondrial Targeting vol.70, pp.4, 2018, https://doi.org/10.1080/01635581.2018.1460679
  4. Effects of luteolin on chemical induced colon carcinogenesis in high fat diet-fed obese mouse vol.51, pp.1, 2018, https://doi.org/10.4163/jnh.2018.51.1.14