Intest Res.  2020 Jul;18(3):325-336. 10.5217/ir.2019.00093.

Melatonin in the colon modulates intestinal microbiota in response to stress and sleep deprivation

Affiliations
  • 1Division of Gastroenterology, Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Korea
  • 2Department of Health, Environment and Safety, Graduate School of Health Science, Eulji University, Seongnam, Korea
  • 3Departemnt of Biomedical Laboratory Science, Graduate School of Health Science, Eulji University, Seongnam, Korea
  • 4Department of Biomedical Laboratory Science, Eulji University, Seongnam, Korea
  • 5Department of Pathology, Eulji University School of Medicine, Seoul, Korea

Abstract

Background/Aims
Stress is closely related to the deterioration of digestive disease. Melatonin has potent anti-inflammatory properties. The objective of this study was to determine the effect of water stress (WS) and sleep deprivation (SD) on intestinal microbiota and roles of melatonin in stressful condition.
Methods
We used C57BL/6 mice and specially designed water bath for stress and SD for 10 days. We measured melatonin concentrations in serum, feces, and colon tissues by high-performance liquid chromatography. Genomic DNA was extracted from feces and amplified using primers targeting V3 to V4 regions of bacterial 16S ribosomal RNA genes.
Results
Compared to the control, melatonin concentration was lower in the WS and SD. Fecal concentration was 0.132 pg/mL in control, 0.062 pg/mL in WS, and 0.068 pg/mL in SD. In colon tissue, it was 0.45 pg/mL in control, 0.007 pg/mL in WS, and 0.03 pg/mL in SD. After melatonin treatment, melatonin concentrations in feces and colon tissue were recovered to the level of control. Metagenomic analysis of microbiota showed abundance in colitogenic microbiota in WS and SD. Melatonin injection attenuated this harmful effect. WS and SD showed decreased Lactobacillales and increased Erysipelotrichales and Enterobacteriales. Melatonin treatment increased Akkermansia muciniphila and Lactobacillus and decreased Bacteroides massiliensis and Erysipelotrichaceae.
Conclusions
This study showed that stress and SD could affect intestinal dysbiosis and increase colitogenic microbiota, which could contribute to the aggravating digestive disease. Melatonin concentrations in feces and colon tissue decreased under WS and SD. Melatonin treatment brought recovery of melatonin concentration in colon tissue and modulating dysbiosis of intestinal microbiota.

Keyword

Melatonin; Gastrointestinal microbiome; Psychological stress; Sleep deprivation

Figure

  • Fig. 1. Schematic diagram of the experiment design. Group I (n=6): control. Group II (n=6): water stress: these mice were housed in a specially designed water bath for stress for 10 days. Group III (n=6): water stress+melatonin, melatonin at a dose of 10 mg/kg was administered to these mice intraperitoneally for 10 days. Group IV (n=6): water stress+sleep deprivation. The experiment was divided into normal sleep group and sleep deprivation group by differentiating the radius of the plat form in which the mouse was sitting in the water bath. Group V (n=6): water stress+sleep deprivation+melatonin.

  • Fig. 2. Animal experimental results. (A) Changes in body weight in each group. Stress induced significant weight loss without intestinal histologic change. No significant reduction in weight loss during water stress was observed even though melatonin administration. (B) Colon length in each group. (C) Histological analyses of colon (H&E, ×100). There were no significant changes in colon length or microscopic inflammatory change. WS, water stress; SD, sleep deprivation; M, melatonin.

  • Fig. 3. Measurement of melatonin concentration in each group. (A) Serum, (B) feces, and (C) colon tissues. aCompare with control, WS, WS+SD; bCompare with control, WS+M, WS+SD+M. WS, water stress; SD, sleep deprivation; M, melatonin.

  • Fig. 4. Analysis of intestinal microbiota in each group. (A) In water stress and/or sleep deprivation, there were significantly increased abundance in order of colitogenic microbiota such as Enterobacteriales and Erysipelotrichales. (B) No significant change in the Richness, Shannon index, Simpson index, or the number of operational taxonomic units (OTUs) among groups. (C) Increased abundance of Proteobacteria and Bacteroidetes at phylum level in water stress and/or sleep deprivation group (inner circle: phylum; outer circle: species). WS, weight loss; SD, sleep deprivation; M, melatonin.

  • Fig. 5. Compared relative abundant species in intestinal microbiota among each group. LDA, linear discriminant analysis; WS, water stress; SD, sleep deprivation.

  • Fig. 6. (A-D) At species level, water stress (WS) with sleep deprivation (SD) group showed significant change such as decreased abundance of Lactobacillus murinus and increased abundance of Bacteroides massiliensis. Clostridium cocleatum group, Enterobacter asburiae group (P<0.05 by Kruskal-Wallis H test). aP<0.05.

  • Fig. 7. Changes in intestinal microbiota after melatonin treatment. (A, B) Administration of melatonin resulted in changes such as increased abundance of Akkermansia muciniphila and Lactobacillus but decreased abundance of Bacteroides massiliensis, and Erysipelotrichaceae (P<0.05 by Kruskal-Wallis H test). aP<0.05. WS, water stress; M, melatonin; SD, sleep deprivation.


Reference

1. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012; 491:119–124.
2. Sonnenberg A. Occupational distribution of inflammatory bowel disease among German employees. Gut. 1990; 31:1037–1040.
Article
3. Ranjbaran Z, Keefer L, Farhadi A, Stepanski E, Sedghi S, Keshavarzian A. Impact of sleep disturbances in inflammatory bowel disease. J Gastroenterol Hepatol. 2007; 22:1748–1753.
Article
4. Swanson GR, Burgess HJ, Keshavarzian A. Sleep disturbances and inflammatory bowel disease: a potential trigger for disease flare? Expert Rev Clin Immunol. 2011; 7:29–36.
Article
5. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018; 11:1–10.
Article
6. Chung SH, Park YS, Kim OS, et al. Melatonin attenuates dextran sodium sulfate induced colitis with sleep deprivation: possible mechanism by microarray analysis. Dig Dis Sci. 2014; 59:1134–1141.
Article
7. Kim TK, Park YS, Baik HW, et al. Melatonin modulates adiponectin expression on murine colitis with sleep deprivation. World J Gastroenterol. 2016; 22:7559–7568.
Article
8. Park YS, Chung SH, Lee SK, et al. Melatonin improves experimental colitis with sleep deprivation. Int J Mol Med. 2015; 35:979–986.
Article
9. Paulose JK, Wright JM, Patel AG, Cassone VM. Human gut bacteria are sensitive to melatonin and express endogenous circadian rhythmicity. PLoS One. 2016; 11:e0146643.
Article
10. Xu P, Wang J, Hong F, et al. Melatonin prevents obesity through modulation of gut microbiota in mice. J Pineal Res. 2017; 62:e12399.
Article
11. Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016; 61:253–278.
Article
12. Reiter RJ, Tan DX, Fuentes-Broto L. Melatonin: a multitasking molecule. Prog Brain Res. 2010; 181:127–151.
Article
13. Ren W, Liu G, Chen S, et al. Melatonin signaling in T cells: functions and applications. J Pineal Res. 2017; 62:e12394.
Article
14. Marquez E, Sánchez-Fidalgo S, Calvo JR, la de Lastra CA, Motilva V. Acutely administered melatonin is beneficial while chronic melatonin treatment aggravates the evolution of TNBS-induced colitis. J Pineal Res. 2006; 40:48–55.
Article
15. Bubenik GA, Brown GM. Pinealectomy reduces melatonin levels in the serum but not in the gastrointestinal tract of rats. Biol Signals. 1997; 6:40–44.
Article
16. Chen CQ, Fichna J, Bashashati M, Li YY, Storr M. Distribution, function and physiological role of melatonin in the lower gut. World J Gastroenterol. 2011; 17:3888–3898.
Article
17. Necefli A, Tulumoğlu B, Giriş M, et al. The effect of melatonin on TNBS-induced colitis. Dig Dis Sci. 2006; 51:1538–1545.
Article
18. Swanson GR, Gorenz A, Shaikh M, et al. Decreased melatonin secretion is associated with increased intestinal permeability and marker of endotoxemia in alcoholics. Am J Physiol Gastrointest Liver Physiol. 2015; 308:G1004–G1011.
Article
19. Fernández-Gil B, Moneim AE, Ortiz F, et al. Melatonin protects rats from radiotherapy-induced small intestine toxicity. PLoS One. 2017; 12:e0174474.
Article
20. Bernstein CN. The brain-gut axis and stress in inflammatory bowel disease. Gastroenterol Clin North Am. 2017; 46:839–846.
Article
21. Fernandez-Mendoza J, Baker JH, Vgontzas AN, Gaines J, Liao D, Bixler EO. Insomnia symptoms with objective short sleep duration are associated with systemic inflammation in adolescents. Brain Behav Immun. 2017; 61:110–116.
Article
22. Irwin MR, Opp MR. Sleep health: reciprocal regulation of sleep and innate immunity. Neuropsychopharmacology. 2017; 42:129–155.
Article
23. Legaki E, Gazouli M. Influence of environmental factors in the development of inflammatory bowel diseases. World J Gastrointest Pharmacol Ther. 2016; 7:112–125.
Article
24. Pirinen T, Kolho KL, Ashorn M, Aronen ET. Sleep and emotional and behavioral symptoms in adolescents with inflammatory bowel disease. Sleep Disord. 2014; 2014:379450.
Article
25. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009; 9:313–323.
Article
26. Kamada N, Núñez G. Regulation of the immune system by the resident intestinal bacteria. Gastroenterology. 2014; 146:1477–1488.
Article
27. Buffie CG, Pamer EG. Microbiota-mediated colonization resistance against intestinal pathogens. Nat Rev Immunol. 2013; 13:790–801.
Article
28. Shang Q, Sun W, Shan X, et al. Carrageenan-induced colitis is associated with decreased population of anti-inflammatory bacterium, Akkermansia muciniphila, in the gut microbiota of C57BL/6J mice. Toxicol Lett. 2017; 279:87–95.
Article
29. Derrien M, Belzer C, de Vos WM. Akkermansia muciniphila and its role in regulating host functions. Microb Pathog. 2017; 106:171–181.
Article
30. Boureau H, Decré D, Carlier JP, Guichet C, Bourlioux P. Identification of a Clostridium cocleatum strain involved in an antiClostridium difficile barrier effect and determination of its mucin-degrading enzymes. Res Microbiol. 1993; 144:405–410.
Article
31. Fenner L, Roux V, Mallet MN, Raoult D. Bacteroides massiliensis sp. nov., isolated from blood culture of a newborn. Int J Syst Evol Microbiol. 2005; 55(Pt 3):1335–1337.
Article
32. Ramanan D, Tang MS, Bowcutt R, Loke P, Cadwell K. Bacterial sensor Nod2 prevents inflammation of the small intestine by restricting the expansion of the commensal Bacteroides vulgatus. Immunity. 2014; 41:311–324.
Article
33. Mezzatesta ML, Gona F, Stefani S. Enterobacter cloacae complex: clinical impact and emerging antibiotic resistance. Future Microbiol. 2012; 7:887–902.
Article
34. Zhu B, Wang S, Li O, et al. High-quality genome sequence of human pathogen Enterobacter asburiae type strain 1497-78T. J Glob Antimicrob Resist. 2017; 8:104–105.
Article
35. Chuffa LG, Fioruci-Fontanelli BA, Mendes LO, et al. Melatonin attenuates the TLR4-mediated inflammatory response through MyD88- and TRIF-dependent signaling pathways in an in vivo model of ovarian cancer. BMC Cancer. 2015; 15:34.
Article
36. King SJ, McCole DF. Epithelial-microbial diplomacy: escalating border tensions drive inflammation in inflammatory bowel disease. Intest Res. 2019; 17:177–191.
Article
37. Zhang YG, Wu S, Xia Y, Sun J. Salmonella infection upregulates the leaky protein claudin-2 in intestinal epithelial cells. PLoS One. 2013; 8:e58606.
Article
38. Chelakkot C, Choi Y, Kim DK, et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp Mol Med. 2018; 50:e450.
Article
39. Ren W, Wang P, Yan J, et al. Melatonin alleviates weanling stress in mice: involvement of intestinal microbiota. J Pineal Res. 2018; 64:e12448.
Article
40. Chen L, Wilson JE, Koenigsknecht MJ, et al. NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth. Nat Immunol. 2017; 18:541–551.
Article
41. Nishino K, Nishida A, Inoue R, et al. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. J Gastroenterol. 2018; 53:95–106.
Article
42. Sartor RB, Wu GD. Roles for intestinal bacteria, viruses, and fungi in pathogenesis of inflammatory bowel diseases and therapeutic approaches. Gastroenterology. 2017; 152:327–339.
Article
43. Takahashi K, Nishida A, Fujimoto T, et al. Reduced abundance of butyrate-producing bacteria species in the fecal microbial community in Crohn’s disease. Digestion. 2016; 93:59–65.
Article
44. Segata N, Izard J, Waldron L, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011; 12:R60.
Article
45. Eun CS, Kwak MJ, Han DS, et al. Does the intestinal microbial community of Korean Crohn’s disease patients differ from that of western patients? BMC Gastroenterol. 2016; 16:28.
Article
Full Text Links
  • IR
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