DOI QR코드

DOI QR Code

Effects of red ginseng on gut, microbiota, and brain in a mouse model of post-infectious irritable bowel syndrome

  • Yu, Seonhye (Department of Microbiology, College of Medicine, Gachon University) ;
  • Chun, Eunho (Department of Health Sciences and Technology, GAIHST, Gachon University) ;
  • Ji, Yeounjung (Lee Gil Ya Cancer and Diabetes Institute, Gachon University) ;
  • Lee, Young Joo (Department of Bioscience and Biotechnology, Sejong University) ;
  • Jin, Mirim (Department of Microbiology, College of Medicine, Gachon University)
  • Received : 2020.07.09
  • Accepted : 2021.03.26
  • Published : 2021.11.15

Abstract

Background: Irritable bowel syndrome (IBS), the most common functional gastrointestinal disorder, is characterized by chronic abdominal pain and bowel habit changes. Although diverse complicated etiologies are involved in its pathogenesis, a dysregulated gut-brain axis may be an important factor. Red ginseng (RG), a traditional herbal medicine, is proven to have anti-inflammatory effects and improve brain function; however, these effects have not been investigated in IBS. Methods: Three-day intracolonic zymosan injections were used to induce post-infectious human IBS-like symptoms in mice. The animals were randomized to receive either phosphate-buffered saline (CG) or RG (30/100/300 mg/kg) for 10 days. Amitriptyline and sulfasalazine were used as positive controls. Macroscopic scoring was performed on day 4. Visceral pain and anxiety-like behaviors were assessed by colorectal distension and elevated plus maze and open field tests, respectively, on day 10. Next-generation sequencing of gut microbiota was performed, and biomarkers involved in gut-brain axis responses were analyzed. Results: Compared to CG, RG significantly decreased the macroscopic score, frequency of visceral pain, and anxiety-like behavior in the IBS mice. These effects were comparable to those after sulfasalazine and amitriptyline treatments. Moreover, RG significantly increased the proliferation of beneficial microbes, including Lactobacillus johnsonii, Lactobacillus reuteri, and Parabacteroides goldsteinii. RG significantly suppressed expression of IL-1β and c-fos in the gut and prefrontal cortex, respectively. Further, it restored the plasma levels of corticosterone to within the normal range, accompanied by an increase in adrenocorticotropic hormone. Conclusion: RG may be a potential therapeutic option for the management of human IBS.

Keywords

Acknowledgement

This work was supported by the (2019) grant from the Korean Society of Ginseng, Bio & Medical Technology Development Program of the National Research Foundation (NRF), funded by the Korean government (MSIT) (grant number: NRF-2019M3E5D5064771 and NRF-2018M3A9C4076473), and Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HI20C0015).

References

  1. Halkjaer SI, Boolsen AW, Gunther S, Christensen AH, Petersen AM. Can fecal microbiota transplantation cure irritable bowel syndrome? World J Gastroenterol 2017;23:4112-20. https://doi.org/10.3748/wjg.v23.i22.4112.
  2. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol 2015;28:203-9.
  3. Moloney RD, Johnson AC, O'Mahony SN, et al. Stress and the microbiota-gutbrain axis in visceral pain: relevance to irritable bowel syndrome. CNS Neurosci Ther 2016;22:102-17. https://doi.org/10.1111/cns.12490.
  4. Enck P, Aziz Q, Barbara G, et al. Irritable bowel syndrome. Nat Rev Dis Primers 2016;2:16014. https://doi.org/10.1038/nrdp.2016.14.
  5. Banerjee A, Sarkhel S, Sarkar R, Dhali GK. Anxiety and depression in irritable bowel syndrome. Indian J Psychol Med 2017;39:741-5. https://doi.org/10.4103/IJPSYM.IJPSYM_46_17.
  6. Lee C, Doo E, Choi JM, et al. The increased level of depression and anxiety in irritable bowel syndrome patients compared with healthy controls: systematic review and meta-analysis. J Neurogastroenterol Motil 2017;23:349-62. https://doi.org/10.5056/jnm16220.
  7. Kabra N, Nadkarni A. Prevalence of depression and anxiety in irritable bowel syndrome: a clinic based study from India. Indian J Psychiatry 2013;55:77-80. https://doi.org/10.4103/0019-5545.105520.
  8. Chang L, Sundaresh S, Elliott J, et al. Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis in irritable bowel syndrome. Neurogastroenterol Motil 2009;21:149-59. https://doi.org/10.1111/j.1365-2982.2008.01171.x.
  9. Sugaya N, Izawa S, Saito K, et al. Effect of prolonged stress on the adrenal hormones of individuals with irritable bowel syndrome. Biopsychosoc Med 2015;9:4. https://doi.org/10.1186/s13030-015-0031-7.
  10. Li J, Zhong W, Wang W, et al. Ginsenoside metabolite compound K promotes recovery of dextran sulfate sodium-induced colitis and inhibits inflammatory responses by suppressing NF-kappaB activation. PLoS One 2014;9:e87810. https://doi.org/10.1371/journal.pone.0087810.
  11. Lee S, Rhee DK. Effects of ginseng on stress-related depression, anxiety, and the hypothalamic-pituitary-adrenal axis. J Ginseng Res 2017;41:589-94. https://doi.org/10.1016/j.jgr.2017.01.010.
  12. Kim HJ, Kim P, Shin CY. A comprehensive review of the therapeutic and pharmacological effects of ginseng and ginsenosides in central nervous system. J Ginseng Res 2013;37:8-29. https://doi.org/10.5142/jgr.2013.37.8.
  13. Chun E, Yoon S, Parveen A, Jin M. Alleviation of irritable bowel syndrome-like symptoms and control of gut and brain responses with oral administration of Dolichos lablab L. In a mouse model. Nutrients 2018;10. https://doi.org/10.3390/nu10101475.
  14. Vannucchi MG, Evangelista S. Experimental models of irritable bowel syndrome and the role of the enteric neurotransmission. J Clin Med 2018;7. https://doi.org/10.3390/jcm7010004.
  15. Wang Y, Bi Z, Wang E, Sun B, Zheng Y, Zhong LL, et al. Rodent model of irritable bowel syndrome. Int J Gastroenterol Disord Ther 2017.
  16. Fuentes IM, Christianson JA. Ion channels, ion channel receptors, and visceral hypersensitivity in irritable bowel syndrome. Neurogastroenterol Motil 2016;28:1613-8. https://doi.org/10.1111/nmo.12979.
  17. In G, Ahn NG, Bae BS, et al. In situ analysis of chemical components induced by steaming between fresh ginseng, steamed ginseng, and red ginseng. J Ginseng Res 2017;41:361-9. https://doi.org/10.1016/j.jgr.2016.07.004.
  18. Lee J, Park J, Lee YY, Lee Y. Comparative transcriptome analysis of the protective effects of Korean Red Ginseng against the influence of bisphenol a in the liver and uterus of ovariectomized mice. J Ginseng Res 2020;44:519-26. https://doi.org/10.1016/j.jgr.2020.01.008.
  19. Park BK, Chun E, Choi JJ, et al. Administration of wasabia koreana ameliorates irritable bowel syndrome-like symptoms in a zymosan-induced mouse model. J Med Food 2017;20:474-84. https://doi.org/10.1089/jmf.2016.3844.
  20. Zhao Z, Kim YW, Zhang J, Lee JH, et al. Korean Red Ginseng attenuates anxiety-like behavior during ethanol withdrawal in rats. J Ginseng Res 2014;38:256-63. https://doi.org/10.1016/j.jgr.2014.05.010.
  21. Kim HJ, Won H, Im J, et al. Effects of Panax ginseng CA Meyer extract on the offspring of adult mice with maternal immune activation. Mol Med Rep 2018;18:3834-42. https://doi.org/10.3892/mmr.2018.9417.
  22. Jang SH, Park J, Kim SH, et al. Oral administration of red ginseng powder fermented with probiotic alleviates the severity of dextran-sulfate sodium-induced colitis in a mouse model. Chin J Nat Med 2017;15:192-201. https://doi.org/10.1016/S1875-5364(17)30035-3.
  23. Zhang MM, Liu SB, Chen T, et al. Effects of NB001 and gabapentin on irritable bowel syndrome-induced behavioral anxiety and spontaneous pain. Mol Brain 2014;7:47. https://doi.org/10.1186/1756-6606-7-47.
  24. Kimball ES, Palmer JM, D'Andrea MR, Hornby PJ, Wade PR. Acute colitis induction by oil of mustard results in later development of an IBS-like accelerated upper GI transit in mice. Am J Physiol Gastrointest Liver Physiol 2005;288:G1266-73. https://doi.org/10.1152/ajpgi.00444.2004.
  25. Kim YR, Volpert G, Shin KO, et al. Ablation of ceramide synthase 2 exacerbates dextran sodium sulphate-induced colitis in mice due to increased intestinal permeability. J Cell Mol Med 2017;21:3565-78. https://doi.org/10.1111/jcmm.13267.
  26. Qi DB, Li WM. Effects of electroacupuncture on expression of c-fos protein in the spinal dorsal horn of rats with chronic visceral hyperalgesia. Zhong Xi Yi Jie He Xue Bao 2012;10:1490-6. https://doi.org/10.3736/jcim20121224.
  27. Laird JM, Martinez-Caro L, Garcia-Nicas E, Cervero F. A new model of visceral pain and referred hyperalgesia in the mouse. Pain 2001;92:335-42. https://doi.org/10.1016/S0304-3959(01)00275-5
  28. Jung JH, Kim SJ. Anxiolytic action of taurine via intranasal administration in mice. Biomol Ther (Seoul) 2019;27:450-6. https://doi.org/10.4062/biomolther.2018.218.
  29. Yang T, Santisteban MM, et al. Gut dysbiosis is linked to hypertension. Hypertension 2015;65:1331-40. https://doi.org/10.1161/Hypertensionaha.115.05315.
  30. Macsharry J, O'Mahony L, Fanning A, et al. Mucosal cytokine imbalance in irritable bowel syndrome. Scand J Gastroenterol 2008;43:1467-76. https://doi.org/10.1080/00365520802276127.
  31. Zhang R, Zou N, Li J, et al. Elevated expression of c-fos in central nervous system correlates with visceral hypersensitivity in irritable bowel syndrome (IBS): a new target for IBS treatment. Int J Colorectal Dis 2011;26:1035-44. https://doi.org/10.1007/s00384-011-1153-4.
  32. Song L. Effects of Lactobacillus plantarum and Lactobacillus paracasei for the prevention and alleviation of zymosan-induced irritable bowel syndrome in mice. Seoul National University; 2019.
  33. Shouval DS, Biswas A, Kang YH, et al. Interleukin 1beta mediates intestinal inflammation in mice and patients with interleukin 10 receptor deficiency. Gastroenterology 2016;151:1100-4. https://doi.org/10.1053/j.gastro.2016.08.055.
  34. Coccia M, Harrison OJ, Schiering C, et al. IL-1beta mediates chronic intestinal inflammation by promoting the accumulation of IL-17A secreting innate lymphoid cells and CD4(+) Th17 cells. J Exp Med 2012;209:1595-609. https://doi.org/10.1084/jem.20111453.
  35. Ren K, Torres R. Role of interleukin-1beta during pain and inflammation. Brain Res Rev 2009;60:57-64. https://doi.org/10.1016/j.brainresrev.2008.12.020.
  36. Kang SW, Park JH, Seok H, Park HJ, et al. The effects of Korea red ginseng on inflammatory cytokines and apoptosis in rat model with chronic nonbacterial prostatitis. Biomed Res Int 2019;2019:2462561. https://doi.org/10.1155/2019/2462561.
  37. Pirnik Z, Mravec B, Kiss A. Fos protein expression in mouse hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei upon osmotic stimulus: colocalization with vasopressin, oxytocin, and tyrosine hydroxylase. Neurochem Int 2004;45:597-607. https://doi.org/10.1016/j.neuint.2004.04.003.
  38. Raskov H, Burcharth J, Pommergaard HC, Rosenberg J. Irritable bowel syndrome, the microbiota and the gut-brain axis. Gut Microbes 2016;7:365-83. https://doi.org/10.1080/19490976.2016.1218585.
  39. Zhu H, Zeng D, Wang Q, et al. Diarrhea-associated intestinal microbiota in captive sichuan golden snub-nosed monkeys (Rhinopithecus roxellana). Microbes Environ 2018;33:249-56. https://doi.org/10.1264/jsme2.ME17163.
  40. Braun T, Sengni AD, BenShoshan M, et al. Fecal microbial characterization of hospitalized patients with suspected infectious diarrhea shows significant dysbiosis. Sci Rep 2017;7:1088. https://doi.org/10.1038/s41598-017-01217-1.
  41. Wick EC, Sears CL. Bacteroides spp. and diarrhea. Curr Opin Infect Dis 2010;23:470-4. https://doi.org/10.1097/QCO.0b013e32833da1eb.
  42. Forbes JD, Van Domselaar G, Bernstein CN. The gut microbiota in immunemediated inflammatory diseases. Front Microbiol 2016;7:1081. https://doi.org/10.3389/fmicb.2016.01081.
  43. Ley RE. Gut microbiota in 2015: Prevotella in the gut: choose carefully. Nat Rev Gastroenterol Hepatol 2016;13:69-70. https://doi.org/10.1038/nrgastro.2016.4.
  44. Urbanska M, Gieruszczak-Bialek D, Szajewska H. Systematic review with meta-analysis: Lactobacillus reuteri DSM 17938 for diarrhoeal diseases in children. Aliment Pharmacol Ther 2016;43:1025-34. https://doi.org/10.1111/apt.13590.
  45. Jadresin O, Hojsak I, et al. Lactobacillus reuteri DSM 17938 in the treatment of functional abdominal pain in children: RCT study. J Pediatr Gastroenterol Nutr 2017;64:925-9. https://doi.org/10.1097/MPG.0000000000001478.
  46. Kamiya T, Wang L, Forsythe P, et al. Inhibitory effects of Lactobacillus reuteri on visceral pain induced by colorectal distension in sprague-dawley rats. Gut 2006;55:191-6. https://doi.org/10.1136/gut.2005.070987.
  47. Mu Q, Tavella VJ, Luo XM. Role of Lactobacillus reuteri in human health and diseases. Front Microbiol 2018;9:757. https://doi.org/10.3389/fmicb.2018.00757.
  48. Jang HM, Lee HJ, Jang SE, Han MJ, Kim DH. Evidence for interplay among antibacterial-induced gut microbiota disturbance, neuro-inflammation, and anxiety in mice. Mucosal Immunol 2018;11:1386-97. https://doi.org/10.1038/s41385-018-0042-3.
  49. Davoren MJ, Liu J, Castellanos J, Rodriguez-Malave NI, Schiestl RH. A novel probiotic, Lactobacillus johnsonii 456, resists acid and can persist in the human gut beyond the initial ingestion period. Gut Microbes 2019;10:458-80. https://doi.org/10.1080/19490976.2018.1547612.
  50. Eftekhari K, Vahedi Z, Kamali Aghdam M, Noemi Diaz D. A randomized double-blind placebo-controlled trial of Lactobacillus reuteri for chronic functional abdominal pain in children. Iran J Pediatr 2015;25:e2616. https://doi.org/10.5812/ijp.2616.
  51. Jang HM, Lee KE, Lee HJ, Kim DH. Immobilization stress- induced Escherichia coli causes anxiety by inducing NF-kappa B activation through gut microbiota disturbance. Sci Rep-Uk 2018;8. https://doi.org/10.1038/s41598-018-31764-0. ARTN 13897.
  52. Fujimura KE, Demmor T, Rauch M, et al. House dust exposure mediates gut microbiome Lactobacillus enrichment and airway immune defense against allergens and virus infection. Proc Natl Acad Sci U S A 2014;111:805-10. https://doi.org/10.1073/pnas.1310750111.
  53. Miranda PM, Bertolini Francesca, Kadarmideen Haja N. Investigation of gut microbiome association with inflammatory bowel disease and depression: a machine learning approach. 2019. F1000Research.
  54. Wu TR, Lin CS, Chang CJ, et al. Gut commensal Parabacteroides goldsteinii plays a predominant role in the anti-obesity effects of polysaccharides isolated from Hirsutella sinensis. Gut 2019;68:248-62. https://doi.org/10.1136/gutjnl-2017-315458.
  55. Chassaing B, Gewirtz AT. Mice harboring pathobiont-free microbiota do not develop intestinal inflammation that normally results from an innate immune deficiency. PLoS One 2018;13:e0195310. https://doi.org/10.1371/journal.pone.0195310.