Journal List > J Nutr Health > v.51(1) > 1081549

Park and Kim: Effects of luteolin on chemical induced colon carcinogenesis in high fat diet-fed obese mouse

Abstract

Purpose

Colorectal cancer, which is one of the most commonly diagnosed cancers in developing and developed countries, is highly associated with obesity. The association is largely attributed to changes to western style diets in those countries containing high-fat and high-energy. Luteolin (LUT) is a known potent inhibitor of inflammation, obesity, and cancer. In this study, we investigated the effects of LUT on chemical-induced colon carcinogenesis in high fat diet (HFD)-fed obese mice.

Methods

Five-week-old male C57BL/6 mice received a single intraperitoneal injection of azoxymethane (AOM) at a dose of 12.5 mg/kg body weight. Mice were then divided into four groups (n = 10) that received one of the following diets for 11 weeks after the AOM injection: normal diet (ND); HFD; HFD with 0.0025% LUT (HFD LL); HFD with 0.005% LUT (HFD HL). One week after AOM injection, animals received 1~2% dextran sodium sulfate in their drinking water over three cycles consisting of five consecutive days each that were separated by 16 days.

Results

Body weight, ratio of colon weight/length, and tumor multiplicity increased significantly in the HFD group compared to the ND group. Luteolin supplementation of the HFD significantly reduced the ratio of colon weight/length and colon tumors, but not body weight. The levels of plasma TNF-α and colonic expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 protein increased in response to HFD, but were suppressed by LUT supplementation. Immunohistochemistry analysis also showed that iNOS expression was decreased by LUT.

Conclusion

Consumption of LUT may reduce the risk of obesity-associated colorectal cancer by suppression of colonic inflammation.

Figures and Tables

Fig. 1

Weight and length of large intestine. The entire large intestine from cecum to rectum was taken out and the length of large intestine was measured with a ruler. The large intestine was weighed after flushing out luminal contents with phosphate buffered saline. Values are presented as the mean ± SE. Means with different letters are significantly different at p < 0.05 by Duncan's multiple range test. The alphabet a in the figure was given to the largest number. ND, normal diet; HFD, high fat diet; HFD LL, HFD + 0.0025% luteolin; HFD HL, HFD + 0.005% luteolin

jnh-51-14-g001
Fig. 2

Chemopreventive effect of luteolin in high fat diet-fed C57BL/6 mouse. After colon carcinogenesis experiment, the entire large intestine was taken out, open longitudinally and the number of tumors was counted macroscopically. Values are presented as the mean ± SE. Means with different letters are significantly different at p < 0.05 by Duncan's multiple range test. The alphabet a in the figure was given to the largest number. ND, normal diet; HFD, high fat diet; HFD LL, HFD + 0.0025% luteolin; HFD HL, HFD + 0.005% luteolin

jnh-51-14-g002
Fig. 3

Anti-inflammatory effects of luteolin in high fat diet-fed C57BL/6 mouse. (A) Plasma TNF-α was measured as described in the methods. (B) The distal part of large intestine was immunoblotted with relevant antibodies. Photograph of chemiluminescent detection of the representative blots (left). The relative abundance of each band to its own β-actin was quantified (right). (C) Rectum was removed and fixed in 10% formalin. Tissue sections were stained with antibody against iNOS and photographed at × 200. (a) ND (b) HFD (c) HFD LL (d) HFD HL. Values are presented as the mean ± SE. Means with different letters are significantly different at p < 0.05 by Duncan's multiple range test. The alphabet a in the figure was given to the largest number. ND, normal diet; HFD, high fat diet; HFD LL, HFD + 0.0025% luteolin; HFD HL, HFD + 0.005% luteolin

jnh-51-14-g003
Table 1

Composition of experimental diets

jnh-51-14-i001

1) ND, normal diet; HFD, high fat diet; HFD LL, HFD+0.0025% luteolin; HFD HL, HFD+0.005% luteolin. 2) Composition of AIN-76A mineral Mix (g/kg): Calcium phosphate, dibasic 500; Sodium chloride 74; Potassium citrate, monohydrate 220; Potassium sulfate 52; Magnesium oxide 24; Manganous carbonate (43–48% Mn) 3.5; Ferric citrate (16–17% Fe) 6; Zinc carbonate (70% ZnO) 1.6; Cupric carbonate (53–55% Cu) 0.3; Potassium iodate 0.01; Sodium selenite 0.01; Chromium potassium sulfate 0.55; Sucrose, finely powdered 118.03. 3) Composition of AIN-76A vitamin Mix (g/kg): Thiamin hydrochloride 0.6; Riboflavin 0.6; Pyridoxine hydrochloride 0.7; Nicotinic acid 3.0; D-calcium pantothenate 1.6; Folic acid 0.2; D-biotin 0.02; Cyanocobalamine 0.001; Cholecalciferol (400,000 IU/g) 0.25; Manaquinone 0.005; Ascorbic acid 0.2; Sucrose, finely powdered 992.824. 4) t-BHQ: tert-butylhydroquinone.

Table 2

Daily food intake and body weight change

jnh-51-14-i002

1) Fresh food was provided to the mice in each group and daily food intake was measured everyday. 2) Body weight was measured once a week. 3) Body weight gain was calculated by (final body weight-initial body weight)/experimental period (days). 4) FER was calculated by body weight gain/daily food intake.

Values are means ± SE. Significant differences are indicated by different letters in a row at p < 0.05 as determined by Duncan's multiple range test. The alphabet a in the table was given to the largest number. FER; food efficiency ratio. ND, nomal diet; HFD, high fat diet; HFD LL, HFD + 0.0025% luteolin; HFD HL, HFD + 0.005% luteolin; NS, not significant.

Notes

This work was supported by research grants from Daegu Catholic University in 2012.

References

1. World Cancer Research Fund. American Institute for Cancer Research. Food, nutrition, physical activity, and the prevention of cancer: a global perspective. Washington, D.C.: WCRF/AICR;2007.
2. Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009; 22(4):191–197.
crossref
3. Boyle P, Langman JS. ABC of colorectal cancer: epidemiology. BMJ. 2000; 321(7264):805–808.
crossref
4. Young GP, Le Leu RK. Preventing cancer: dietary lifestyle or clinical intervention? Asia Pac J Clin Nutr. 2002; 11:Suppl 3. S618–S631.
crossref
5. Na SY, Myung SJ. Obesity and colorectal cancer. Korean J Gastroenterol. 2012; 59(1):16–26.
crossref
6. Padidar S, Farquharson AJ, Williams LM, Kearney R, Arthur JR, Drew JE. High-fat diet alters gene expression in the liver and colon: links to increased development of aberrant crypt foci. Dig Dis Sci. 2012; 57(7):1866–1874.
crossref
7. Sung MK, Yeon JY, Park SY, Park JH, Choi MS. Obesity-induced metabolic stresses in breast and colon cancer. Ann N Y Acad Sci. 2011; 1229:61–68.
crossref
8. Schlesinger S, Lieb W, Koch M, Fedirko V, Dahm CC, Pischon T, Nöthlings U, Boeing H, Aleksandrova K. Body weight gain and risk of colorectal cancer: a systematic review and meta-analysis of observational studies. Obes Rev. 2015; 16(7):607–619.
crossref
9. Liu Z, Brooks RS, Ciappio ED, Kim SJ, Crott JW, Bennett G, Greenberg AS, Mason JB. Diet-induced obesity elevates colonic TNF-α in mice and is accompanied by an activation of Wnt signaling: a mechanism for obesity-associated colorectal cancer. J Nutr Biochem. 2012; 23(10):1207–1213.
crossref
10. Jochem C, Leitzmann M. Obesity and colorectal cancer. Recent Results Cancer Res. 2016; 208:17–41.
crossref
11. López-Lázaro M. Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem. 2009; 9(1):31–59.
12. Ashokkumar P, Sudhandiran G. Protective role of luteolin on the status of lipid peroxidation and antioxidant defense against azoxymethane-induced experimental colon carcinogenesis. Biomed Pharmacother. 2008; 62(9):590–597.
crossref
13. Nishitani Y, Yamamoto K, Yoshida M, Azuma T, Kanazawa K, Hashimoto T, Mizuno M. Intestinal anti-inflammatory activity of luteolin: role of the aglycone in NF-κB inactivation in macrophages co-cultured with intestinal epithelial cells. Biofactors. 2013; 39(5):522–533.
crossref
14. Salib JY, Michael HN, Eskande EF. Anti-diabetic properties of flavonoid compounds isolated from Hyphaene thebaica epicarp on alloxan induced diabetic rats. Pharmacognosy Res. 2013; 5(1):22–29.
crossref
15. Pandurangan AK, Esa NM. Luteolin, a bioflavonoid inhibits colorectal cancer through modulation of multiple signaling pathways: a review. Asian Pac J Cancer Prev. 2014; 15(14):5501–5508.
crossref
16. Zhang X, Zhang QX, Wang X, Zhang L, Qu W, Bao B, Liu CA, Liu J. Dietary luteolin activates browning and thermogenesis in mice through an AMPK/PGC1α pathway-mediated mechanism. Int J Obes (Lond). 2016; 40(12):1841–1849.
crossref
17. Kwon EY, Jung UJ, Park T, Yun JW, Choi MS. Luteolin attenuates hepatic steatosis and insulin resistance through the interplay between the liver and adipose tissue in mice with diet-induced obesity. Diabetes. 2015; 64(5):1658–1669.
crossref
18. Zhang L, Han YJ, Zhang X, Wang X, Bao B, Qu W, Liu J. Luteolin reduces obesity-associated insulin resistance in mice by activating AMPKα1 signalling in adipose tissue macrophages. Diabetologia. 2016; 59(10):2219–2228.
crossref
19. Ding L, Jin D, Chen X. Luteolin enhances insulin sensitivity via activation of PPARγ transcriptional activity in adipocytes. J Nutr Biochem. 2010; 21(10):941–947.
crossref
20. Nepali S, Son JS, Poudel B, Lee JH, Lee YM, Kim DK. Luteolin is a bioflavonoid that attenuates adipocyte-derived inflammatory responses via suppression of nuclear factor-κ B/mitogen-activated protein kinases pathway. Pharmacogn Mag. 2015; 11(43):627–635.
21. Cooper HS, Murthy SN, Shah RS, Sedergran DJ. Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 1993; 69(2):238–249.
22. Cooper HS, Murthy S, Kido K, Yoshitake H, Flanigan A. Dysplasia and cancer in the dextran sulfate sodium mouse colitis model. Relevance to colitis-associated neoplasia in the human: a study of histopathology, B-catenin and p53 expression and the role of inflammation. Carcinogenesis. 2000; 21(4):757–768.
crossref
23. Tanaka T, Kohno H, Suzuki R, Hata K, Sugie S, Niho N, Sakano K, Takahashi M, Wakabayashi K. Dextran sodium sulfate strongly promotes colorectal carcinogenesis in ApcMin/+ mice: inflammatory stimuli by dextran sodium sulfate results in development of multiple colonic neoplasms. Int J Cancer. 2006; 118(1):25–34.
24. Fiocchi C. Inflammatory bowel disease: etiology and pathogenesis. Gastroenterology. 1998; 115(1):182–205.
crossref
25. Hendrickson BA, Gokhale R, Cho JH. Clinical aspects and pathophysiology of inflammatory bowel disease. Clin Microbiol Rev. 2002; 15(1):79–94.
crossref
26. Renehan AG, Soerjomataram I, Tyson M, Egger M, Zwahlen M, Coebergh JW, Buchan I. Incident cancer burden attributable to excess body mass index in 30 European countries. Int J Cancer. 2010; 126(3):692–702.
crossref
27. Moghaddam AA, Woodward M, Huxley R. Obesity and risk of colorectal cancer: a meta-analysis of 31 studies with 70,000 events. Cancer Epidemiol Biomarkers Prev. 2007; 16(12):2533–2547.
crossref
28. Martínez ME, Giovannucci E, Spiegelman D, Hunter DJ, Willett WC, Colditz GA. Leisure-time physical activity, body size, and colon cancer in women. Nurses' Health Study Research Group. J Natl Cancer Inst. 1997; 89(13):948–955.
29. Larsson SC, Wolk A. Obesity and colon and rectal cancer risk: a meta-analysis of prospective studies. Am J Clin Nutr. 2007; 86(3):556–565.
crossref
30. Baltgalvis KA, Berger FG, Peña MM, Davis JM, Carson JA. The interaction of a high-fat diet and regular moderate intensity exercise on intestinal polyp development in Apc Min/+ mice. Cancer Prev Res (Phila). 2009; 2(7):641–649.
31. Tang FY, Pai MH, Chiang EP. Consumption of high-fat diet induces tumor progression and epithelial-mesenchymal transition of colorectal cancer in a mouse xenograft model. J Nutr Biochem. 2012; 23(10):1302–1313.
crossref
32. Reddy BS. Types and amount of dietary fat and colon cancer risk: Prevention by omega-3 fatty acid-rich diets. Environ Health Prev Med. 2002; 7(3):95–102.
crossref
33. Dai W, Liu T, Wang Q, Rao CV, Reddy BS. Down-regulation of PLK3 gene expression by types and amount of dietary fat in rat colon tumors. Int J Oncol. 2002; 20(1):121–126.
crossref
34. van Beelen VA, Spenkelink B, Mooibroek H, Sijtsma L, Bosch D, Rietjens IM, Alink GM. An n-3 PUFA-rich microalgal oil diet protects to a similar extent as a fish oil-rich diet against AOM-induced colonic aberrant crypt foci in F344 rats. Food Chem Toxicol. 2009; 47(2):316–320.
crossref
35. Kang KA, Piao MJ, Ryu YS, Hyun YJ, Park JE, Shilnikova K, Zhen AX, Kang HK, Koh YS, Jeong YJ, Hyun JW. Luteolin induces apoptotic cell death via antioxidant activity in human colon cancer cells. Int J Oncol. 2017; 51(4):1169–1178.
crossref
36. Pandurangan AK, Dharmalingam P, Sadagopan SK, Ramar M, Munusamy A, Ganapasam S. Luteolin induces growth arrest in colon cancer cells through involvement of Wnt/β-catenin/GSK-3β signaling. J Environ Pathol Toxicol Oncol. 2013; 32(2):131–139.
crossref
37. Lim DY, Jeong Y, Tyner AL, Park JH. Induction of cell cycle arrest and apoptosis in HT-29 human colon cancer cells by the dietary compound luteolin. Am J Physiol Gastrointest Liver Physiol. 2007; 292(1):G66–G75.
crossref
38. Ramos AA, Pereira-Wilson C, Collins AR. Protective effects of ursolic acid and luteolin against oxidative DNA damage include enhancement of DNA repair in Caco-2 cells. Mutat Res. 2010; 692(1-2):6–11.
crossref
39. Pandurangan AK, Ananda Sadagopan SK, Dharmalingam P, Ganapasam S. Luteolin, a bioflavonoid, attenuates azoxymethane-induced effects on mitochondrial enzymes in BALB/c mice. Asian Pac J Cancer Prev. 2014; 14(11):6669–6672.
crossref
40. Pandurangan AK, Dharmalingam P, Sadagopan SK, Ganapasam S. Luteolin inhibits matrix metalloproteinase 9 and 2 in azoxymethane-induced colon carcinogenesis. Hum Exp Toxicol. 2014; 33(11):1176–1185.
crossref
41. Rankin JW, Turpyn AD. Low carbohydrate, high fat diet increases C-reactive protein during weight loss. J Am Coll Nutr. 2007; 26(2):163–169.
crossref
42. Kim IW, Myung SJ, Do MY, Ryu YM, Kim MJ, Do EJ, Park S, Yoon SM, Ye BD, Byeon JS, Yang SK, Kim JH. Western-style diets induce macrophage infiltration and contribute to colitis-associated carcinogenesis. J Gastroenterol Hepatol. 2010; 25(11):1785–1794.
crossref
43. Clapper ML, Cooper HS, Chang WC. Dextran sulfate sodium-induced colitis-associated neoplasia: a promising model for the development of chemopreventive interventions. Acta Pharmacol Sin. 2007; 28(9):1450–1459.
crossref
44. Park YH, Kim N, Shim YK, Choi YJ, Nam RH, Choi YJ, Ham MH, Suh JH, Lee SM, Lee CM, Yoon H, Lee HS, Lee DH. Adequate dextran sodium sulfate-induced colitis model in mice and effective outcome measurement method. J Cancer Prev. 2015; 20(4):260–267.
crossref
45. Randhawa PK, Singh K, Singh N, Jaggi AS. A review on chemical-induced inflammatory bowel disease models in rodents. Korean J Physiol Pharmacol. 2014; 18(4):279–288.
crossref
46. Takahashi M, Mutoh M, Kawamori T, Sugimura T, Wakabayashi K. Altered expression of beta-catenin, inducible nitric oxide synthase and cyclooxygenase-2 in azoxymethane-induced rat colon carcinogenesis. Carcinogenesis. 2000; 21(7):1319–1327.
47. Ahn B, Ohshima H. Suppression of intestinal polyposis in Apc(Min/+) mice by inhibiting nitric oxide production. Cancer Res. 2001; 61(23):8357–8360.
48. Yagihashi N, Kasajima H, Sugai S, Matsumoto K, Ebina Y, Morita T, Murakami T, Yagihashi S. Increased in situ expression of nitric oxide synthase in human colorectal cancer. Virchows Arch. 2000; 436(2):109–114.
crossref
49. Turini ME, DuBois RN. Cyclooxygenase-2: a therapeutic target. Annu Rev Med. 2002; 53:35–57.
crossref
TOOLS
Similar articles