Nutr Res Pract.  2018 Apr;12(2):101-109. 10.4162/nrp.2018.12.2.101.

Effect of vitamin C on azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced colitis-associated early colon cancer in mice

Affiliations
  • 1Department of Nutritional Science and Food Management, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea. yuri.kim@ewha.ac.kr
  • 2Department of Biomedical Laboratory Science, Eulji University, Gyeonggi 13135, Korea.
  • 3Kwang-Dong Pharmaceutical Co., Ltd., Seoul 06650, Korea.

Abstract

BACKGROUND/OBJECTIVES
The objective of this study was to investigate the effects of vitamin C on inflammation, tumor development, and dysbiosis of intestinal microbiota in an azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced inflammation-associated early colon cancer mouse model.
MATERIALS/METHODS
Male BALB/c mice were injected intraperitoneally with AOM [10 mg/kg body weight (b.w)] and given two 7-d cycles of 2% DSS drinking water with a 14 d inter-cycle interval. Vitamin C (60 mg/kg b.w. and 120 mg/kg b.w.) was supplemented by gavage for 5 weeks starting 2 d after the AOM injection.
RESULTS
The vitamin C treatment suppressed inflammatory morbidity, as reflected by disease activity index (DAI) in recovery phase and inhibited shortening of the colon, and reduced histological damage. In addition, vitamin C supplementation suppressed mRNA levels of pro-inflammatory mediators and cytokines, including cyclooxygenase-2, microsomal prostaglandin E synthase-2, tumor necrosis factor-α, Interleukin (IL)-1β, and IL-6, and reduced expression of the proliferation marker, proliferating cell nuclear antigen, compared to observations of AOM/DSS animals. Although the microbial composition did not differ significantly between the groups, administration of vitamin C improved the level of inflammation-related Lactococcus and JQ084893 to control levels.
CONCLUSION
Vitamin C treatment provided moderate suppression of inflammation, proliferation, and certain inflammation-related dysbiosis in a murine model of colitis associated-early colon cancer. These findings support that vitamin C supplementation can benefit colonic health. Long-term clinical studies with various doses of vitamin C are warranted.

Keyword

Vitamin C; colitis; inflammation; colonic neoplasm; microbiota

MeSH Terms

Animals
Ascorbic Acid*
Azoxymethane*
Body Weight
Colitis
Colon*
Colonic Neoplasms*
Cyclooxygenase 2
Cytokines
Drinking Water
Dysbiosis
Gastrointestinal Microbiome
Humans
Inflammation
Interleukin-6
Interleukins
Lactococcus
Male
Mice*
Microbiota
Necrosis
Proliferating Cell Nuclear Antigen
RNA, Messenger
Sodium*
Vitamins*
Ascorbic Acid
Azoxymethane
Cyclooxygenase 2
Cytokines
Drinking Water
Interleukin-6
Interleukins
Proliferating Cell Nuclear Antigen
RNA, Messenger
Sodium
Vitamins

Figure

  • Fig. 1 Effects of vitamin C supplementation on DAI score. DAI scores reflect weight loss, stool consistency, and fecal bleeding. (A) Bi-daily group DAI scores. (B) Analysis of day 16th, DAI scores among the Control, CC, V60, and V120 groups. Mean ± SEM are shown (Control, n = 9; CC, n = 12; V60 and V120, n = 11 per group). One-way ANOVAs and Newman-Keuls' post hoc tests were performed (P<0.05). CC, AOM/DSS-induced colon cancer; V60, AOM/DSS + 60 mg/kg body weight (b.w.) of vitamin C; V120, AOM/DSS + 120 mg/kg b.w. of vitamin C.

  • Fig. 2 Effects of vitamin C supplementation on colon length and histology in colitis-associated colon cancer. (A) Colon length was measured and compared among four groups. Mean ± SEM are shown (Control, n = 9; CC, n = 12; V60 and V120, n = 11 per group). One-way ANOVAs and Newman-Keuls' post hoc tests were performed (P<0.05). (B) Representative histologic sections of distal colon from among four groups. Glandular epithelium destruction (arrow head), neutrophil infiltration (arrow) or submucosa extension (S) were shown. HE stain, bar = 100 µm. Magnification, upper 100x, lower 200x CC, AOM/DSS-induced colon cancer; V60, AOM/DSS + 60 mg/kg body weight (b.w.) of vitamin C; V120, AOM/DSS + 120 mg/kg b.w. of vitamin C.

  • Fig. 3 Effect of vitamin C supplementation on proinflammatory mediators and cytokines in middle and distal colon. (A) mRNA expressions of COX-2 and mPGES-2 (B) TNF-α, IL-1β, and IL-6 in middle colon (a) and in the distal colon (b) were analyzed using real-time PCR. β-actin and GAPDH were used as the loading control. Mean ± SEM are shown (Control, n = 9; CC, n = 12; V60 and V120, n = 11 per group). One-way ANOVAs and Newman-Keuls' post hoc tests were performed (P<0.05). CC, AOM/DSS-induced colon cancer; V60, AOM/DSS + 60 mg/kg body weight (b.w.) of vitamin C; V120, AOM/DSS + 120 mg/kg b.w. of vitamin C.

  • Fig. 4 The effects of vitamin C on colon tumor number and mRNA expression of PCNA. (A) Number of colon tumor were counted with naked eye when sacrificing the mice. (B) mRNA expressions of PCNA was analyzed in the middle colon (a) and in the distal colon (b) using real-time PCR. β-actin and GAPDH were used as the loading control. Mean ± SEM are shown (Control, n = 9; CC, n = 12; V60 and V120, n = 11 per group). One-way ANOVAs and Newman-Keuls' post hoc tests were performed (P<0.05). CC, AOM/DSS-induced colon cancer; V60, AOM/DSS + 60 mg/kg body weight (b.w.) of vitamin C; V120, AOM/DSS + 120 mg/kg b.w. of vitamin C.

  • Fig. 5 Effects of vitamin C on gut microbiome taxonomy. (A) Composition of gut microbacteria at the phylum level. (a) Changes in the proportion of Bacteroidetes and Firmicutes phyla. (b) Relative abundance of Bacteridetes/Firmicutes across the Control, CC, and V60 groups. (B) Relative abundance of genus Lactoccocus. (C) Relative abundance of JQ084893 species. Mean ± SEM are shown (n = 6 per group). One-way ANOVAs and Newman-Keuls' post hoc tests were used (P<0.05). CC, AOM/DSS-induced colon cancer; V60, AOM/DSS + 60 mg/kg body weight (b.w.) of vitamin C.


Reference

1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016; 66:7–30.
Article
2. Boyle P, Langman JS. ABC of colorectal cancer: Epidemiology. BMJ. 2000; 321:805–808.
Article
3. Bernstein CN, Blanchard JF, Kliewer E, Wajda A. Cancer risk in patients with inflammatory bowel disease: a population-based study. Cancer. 2001; 91:854–862.
Article
4. Itzkowitz SH, Yio X. Inflammation and cancer IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol. 2004; 287:G7–G17.
Article
5. Tanaka T. Development of an inflammation-associated colorectal cancer model and its application for research on carcinogenesis and chemoprevention. Int J Inflam. 2012; 2012:658786.
Article
6. Meira LB, Bugni JM, Green SL, Lee CW, Pang B, Borenshtein D, Rickman BH, Rogers AB, Moroski-Erkul CA, McFaline JL, Schauer DB, Dedon PC, Fox JG, Samson LD. DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J Clin Invest. 2008; 118:2516–2525.
Article
7. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011; 140:1756–1767.
Article
8. Szkaradkiewicz A, Marciniak R, Chudzicka-Strugala I, Wasilewska A, Drews M, Majewski P, Karpiński T, Zwoździak B. Proinflammatory cytokines and IL-10 in inflammatory bowel disease and colorectal cancer patients. Arch Immunol Ther Exp (Warsz). 2009; 57:291–294.
Article
9. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007; 449:804–810.
Article
10. Laukens D, Brinkman BM, Raes J, De Vos M, Vandenabeele P. Heterogeneity of the gut microbiome in mice: guidelines for optimizing experimental design. FEMS Microbiol Rev. 2016; 40:117–132.
Article
11. Kamada N, Seo SU, Chen GY, Nunez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013; 13:321–335.
Article
12. Holmes E, Li JV, Athanasiou T, Ashrafian H, Nicholson JK. Understanding the role of gut microbiome-host metabolic signal disruption in health and disease. Trends Microbiol. 2011; 19:349–359.
Article
13. Mai V, Morris JG Jr. Colonic bacterial flora: changing understandings in the molecular age. J Nutr. 2004; 134:459–464.
Article
14. Dueñas M, Muñoz-González I, Cueva C, Jiménez-Girón A, Sánchez-Patán F, Santos-Buelga C, Moreno-Arribas MV, Bartolomé B. A survey of modulation of gut microbiota by dietary polyphenols. Biomed Res Int. 2015; 2015:850902.
Article
15. Biesalski HK. Nutrition meets the microbiome: micronutrients and the microbiota. Ann N Y Acad Sci. 2016; 1372:53–64.
Article
16. Nishikimi M, Yagi K. Molecular basis for the deficiency in humans of gulonolactone oxidase, a key enzyme for ascorbic acid biosynthesis. Am J Clin Nutr. 1991; 54:1203S–1208S.
Article
17. Jacob RA, Sotoudeh G. Vitamin C function and status in chronic disease. Nutr Clin Care. 2002; 5:66–74.
Article
18. Block G. Vitamin C and cancer prevention: the epidemiologic evidence. Am J Clin Nutr. 1991; 53:270S–282S.
Article
19. Deicher R, Ziai F, Bieglmayer C, Schillinger M, Horl WH. Low total vitamin C plasma level is a risk factor for cardiovascular morbidity and mortality in hemodialysis patients. J Am Soc Nephrol. 2005; 16:1811–1818.
Article
20. Wang X, Willen R, Wadstrom T. Astaxanthin-rich algal meal and vitamin C inhibit Helicobacter pylori infection in BALB/cA mice. Antimicrob Agents Chemother. 2000; 44:2452–2457.
Article
21. Korantzopoulos P, Kolettis TM, Kountouris E, Dimitroula V, Karanikis P, Pappa E, Siogas K, Goudevenos JA. Oral vitamin C administration reduces early recurrence rates after electrical cardioversion of persistent atrial fibrillation and attenuates associated inflammation. Int J Cardiol. 2005; 102:321–326.
Article
22. Kasetsuwan N, Wu FM, Hsieh F, Sanchez D, McDonnell PJ. Effect of topical ascorbic acid on free radical tissue damage and inflammatory cell influx in the cornea after excimer laser corneal surgery. Arch Ophthalmol. 1999; 117:649–652.
Article
23. Yan H, Wang H, Zhang X, Li X, Yu J. Ascorbic acid ameliorates oxidative stress and inflammation in dextran sulfate sodium-induced ulcerative colitis in mice. Int J Clin Exp Med. 2015; 8:20245–20253.
24. Remely M, Ferk F, Sterneder S, Setayesh T, Roth S, Kepcija T, Noorizadeh R, Rebhan I, Greunz M, Beckmann J, Wagner KH, Knasmüller S, Haslberger AG. EGCG prevents high fat diet-induced changes in gut microbiota, decreases of DNA strand breaks, and changes in expression and DNA methylation of Dnmt1 and MLH1 in C57BL/6J male mice. Oxid Med Cell Longev. 2017; 2017:3079148.
25. Xu J, Xu C, Chen X, Cai X, Yang S, Sheng Y, Wang T. Regulation of an antioxidant blend on intestinal redox status and major microbiota in early weaned piglets. Nutrition. 2014; 30:584–589.
Article
26. Yu C, Wen XD, Zhang Z, Zhang CF, Wu XH, Martin A, Du W, He TC, Wang CZ, Yuan CS. American ginseng attenuates azoxymethane/dextran sodium sulfate-induced colon carcinogenesis in mice. J Ginseng Res. 2015; 39:14–21.
Article
27. Kim KM, Kim YS, Lim JY, Min SJ, Shin JH, Ko HC, Kim SJ, Lim Y, Kim Y. Sasa quelpaertensis leaf extract suppresses dextran sulfate sodium-induced colitis in mice by inhibiting the proinflammatory mediators and mitogen-activated protein kinase phosphorylation. Nutr Res. 2014; 34:894–905.
Article
28. Okayasu I, Hatakeyama S, Yamada M, Ohkusa T, Inagaki Y, Nakaya R. A novel method in the induction of reliable experimental acute and chronic ulcerative colitis in mice. Gastroenterology. 1990; 98:694–702.
Article
29. Dieleman LA, Palmen MJ, Akol H, Bloemena E, Peña AS, Meuwissen SG, Van Rees EP. Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines. Clin Exp Immunol. 1998; 114:385–391.
Article
30. Araki A, Kanai T, Ishikura T, Makita S, Uraushihara K, Iiyama R, Totsuka T, Takeda K, Akira S, Watanabe M. MyD88-deficient mice develop severe intestinal inflammation in dextran sodium sulfate colitis. J Gastroenterol. 2005; 40:16–23.
Article
31. Wen XD, Wang CZ, Yu C, Zhao L, Zhang Z, Matin A, Wang Y, Li P, Xiao SY, Du W, He TC, Yuan CS. Panax notoginseng attenuates experimental colitis in the azoxymethane/dextran sulfate sodium mouse model. Phytother Res. 2014; 28:892–898.
Article
32. Sánchez-Fidalgo S, Cárdeno A, Sánchez-Hidalgo M, Aparicio-Soto M, de la Lastra CA. Dietary extra virgin olive oil polyphenols supplementation modulates DSS-induced chronic colitis in mice. J Nutr Biochem. 2013; 24:1401–1413.
Article
33. Tanaka T, Kohno H, Suzuki R, Yamada Y, Sugie S, Mori H. A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Sci. 2003; 94:965–973.
Article
34. Wang D, Dubois RN. The role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene. 2010; 29:781–788.
Article
35. Sheibanie AF, Yen JH, Khayrullina T, Emig F, Zhang M, Tuma R, Ganea D. The proinflammatory effect of prostaglandin E2 in experimental inflammatory bowel disease is mediated through the IL-23--〉IL-17 axis. J Immunol. 2007; 178:8138–8147.
Article
36. Wang D, Dubois RN. Prostaglandins and cancer. Gut. 2006; 55:115–122.
Article
37. Gudis K, Tatsuguchi A, Wada K, Futagami S, Nagata K, Hiratsuka T, Shinji Y, Miyake K, Tsukui T, Fukuda Y, Sakamoto C. Microsomal prostaglandin E synthase (mPGES)-1, mPGES-2 and cytosolic PGES expression in human gastritis and gastric ulcer tissue. Lab Invest. 2005; 85:225–236.
Article
38. Murakami M, Nakashima K, Kamei D, Masuda S, Ishikawa Y, Ishii T, Ohmiya Y, Watanabe K, Kudo I. Cellular prostaglandin E2 production by membrane-bound prostaglandin E synthase-2 via both cyclooxygenases-1 and -2. J Biol Chem. 2003; 278:37937–37947.
Article
39. Meduri GU, Headley S, Kohler G, Stentz F, Tolley E, Umberger R, Leeper K. Persistent elevation of inflammatory cytokines predicts a poor outcome in ARDS. Plasma IL-1 beta and IL-6 levels are consistent and efficient predictors of outcome over time. Chest. 1995; 107:1062–1073.
Article
40. Härtel C, Strunk T, Bucsky P, Schultz C. Effects of vitamin C on intracytoplasmic cytokine production in human whole blood monocytes and lymphocytes. Cytokine. 2004; 27:101–106.
Article
41. Jung IK, Choi HJ, Kim SS, Hong SH. Prognostic significance of proliferating cell nuclear antigen (PCNA) expression in patients with colorectal carcinoma. J Korean Cancer Assoc. 1995; 27:550–558.
42. Suzuki R, Kohno H, Sugie S, Tanaka T. Dose-dependent promoting effect of dextran sodium sulfate on mouse colon carcinogenesis initiated with azoxymethane. Histol Histopathol. 2005; 20:483–492.
43. De Robertis M, Massi E, Poeta ML, Carotti S, Morini S, Cecchetelli L, Signori E, Fazio VM. The AOM/DSS murine model for the study of colon carcinogenesis: From pathways to diagnosis and therapy studies. J Carcinog. 2011; 10:9.
Article
44. Onodera H, Maetani S, Kawamoto K, Kan S, Kondo S, Imamura M. Pathologic significance of tumor progression in locally recurrent rectal cancer: different nature from primary cancer. Dis Colon Rectum. 2000; 43:775–781.
Article
45. Jackson PE, O'Connor PJ, Cooper DP, Margison GP, Povey AC. Associations between tissue-specific DNA alkylation, DNA repair and cell proliferation in the colon and colon tumour yield in mice treated with 1,2-dimethylhydrazine. Carcinogenesis. 2003; 24:527–533.
Article
46. Bussey HJ, DeCosse JJ, Deschner EE, Eyers AA, Lesser ML, Morson BC, Ritchie SM, Thomson JP, Wadsworth J. A randomized trial of ascorbic acid in polyposis coli. Cancer. 1982; 50:1434–1439.
Article
47. Jacobs EJ, Connell CJ, Patel AV, Chao A, Rodriguez C, Seymour J, McCullough ML, Calle EE, Thun MJ. Vitamin C and vitamin E supplement use and colorectal cancer mortality in a large American Cancer Society cohort. Cancer Epidemiol Biomarkers Prev. 2001; 10:17–23.
48. Cahill RJ, O'Sullivan KR, Mathias PM, Beattie S, Hamilton H, O'Morain C. Effects of vitamin antioxidant supplementation on cell kinetics of patients with adenomatous polyps. Gut. 1993; 34:963–967.
Article
49. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008; 22:659–661.
Article
50. Levine M, Conry-Cantilena C, Wang Y, Welch RW, Washko PW, Dhariwal KR, Park JB, Lazarev A, Graumlich JF, King J, Cantilena LR. Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proc Natl Acad Sci U S A. 1996; 93:3704–3709.
Article
51. Carr AC, Frei B. Toward a new recommended dietary allowance for vitamin C based on antioxidant and health effects in humans. Am J Clin Nutr. 1999; 69:1086–1107.
Article
52. Jenab M, Riboli E, Ferrari P, Sabate J, Slimani N, Norat T, Friesen M, Tjønneland A, Olsen A, Overvad K, Boutron-Ruault MC, Clavel-Chapelon F, Touvier M, Boeing H, Schulz M, Linseisen J, Nagel G, Trichopoulou A, Naska A, Oikonomou E, Krogh V, Panico S, Masala G, Sacerdote C, Tumino R, Peeters PH, Numans ME, Bueno-de-Mesquita HB, Büchner FL, Lund E, Pera G, Sanchez CN, Sánchez MJ, Arriola L, Barricarte A, Quirós JR, Hallmans G, Stenling R, Berglund G, Bingham S, Khaw KT, Key T, Allen N, Carneiro F, Mahlke U, Del Giudice G, Palli D, Kaaks R, Gonzalez CA. Plasma and dietary vitamin C levels and risk of gastric cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC-EURGAST). Carcinogenesis. 2006; 27:2250–2257.
Article
53. Strnadová E, Prokopic J. Changes in ascorbic acid content in various organs and serum of mice experimentally infected with Taenia crassiceps (Zeder, 1800) cysticerci. Folia Parasitol (Praha). 1985; 32:185–188.
54. Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem? Front Microbiol. 2016; 7:455.
Article
55. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014; 12:661–672.
Article
56. Ott SJ, Musfeldt M, Wenderoth DF, Hampe J, Brant O, Fölsch UR, Timmis KN, Schreiber S. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004; 53:685–693.
Article
57. Claesson MJ, Jeffery IB, Conde S, Power SE, O'Connor EM, Cusack S, Harris HM, Coakley M, Lakshminarayanan B, O'Sullivan O, Fitzgerald GF, Deane J, O'Connor M, Harnedy N, O'Connor K, O'Mahony D, van Sinderen D, Wallace M, Brennan L, Stanton C, Marchesi JR, Fitzgerald AP, Shanahan F, Hill C, Ross RP, O'Toole PW. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012; 488:178–184.
Article
58. Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, Goedert JJ, Hayes RB, Yang L. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 2013; 105:1907–1911.
Article
59. Zackular JP, Baxter NT, Iverson KD, Sadler WD, Petrosino JF, Chen GY, Schloss PD. The gut microbiome modulates colon tumorigenesis. MBio. 2013; 4:e00692–e00613.
Article
60. Berry D, Kuzyk O, Rauch I, Heider S, Schwab C, Hainzl E, Decker T, Müller M, Strobl B, Schleper C, Urich T, Wagner M, Kenner L, Loy A. Intestinal microbiota signatures associated with inflammation history in mice experiencing recurring colitis. Front Microbiol. 2015; 6:1408.
Article
61. Yeom Y, Kim BS, Kim SJ, Kim Y. Sasa quelpaertensis leaf extract regulates microbial dysbiosis by modulating the composition and diversity of the microbiota in dextran sulfate sodium-induced colitis mice. BMC Complement Altern Med. 2016; 16:481.
Article
62. Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, Yang L, Liu ZJ, Yuan YZ, Liu F, Wu JX, Zhong L, Fang DC, Zou W, Fang JY. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013; 97:1044–1052.
Article
63. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016; 7:189–200.
Article
64. Hemarajata P, Versalovic J. Effects of probiotics on gut microbiota: mechanisms of intestinal immunomodulation and neuromodulation. Therap Adv Gastroenterol. 2013; 6:39–51.
Article
65. de Moreno de LeBlanc A, LeBlanc JG, Perdigón G, Miyoshi A, Langella P, Azevedo V, Sesma F. Oral administration of a catalase-producing Lactococcus lactis can prevent a chemically induced colon cancer in mice. J Med Microbiol. 2008; 57:100–105.
Article
66. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014; 505:559–563.
Article
67. Willing BP, Dicksved J, Halfvarson J, Andersson AF, Lucio M, Zheng Z, Järnerot G, Tysk C, Jansson JK, Engstrand L. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010; 139:1844–1854.e1.
Article
68. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012; 489:220–230.
Article
Full Text Links
  • NRP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr