DOI QR코드

DOI QR Code

From Gut to Brain: Alteration in Inflammation Markers in the Brain of Dextran Sodium Sulfate-induced Colitis Model Mice

  • Do, Jongho (Department of Psychiatry, Kyungpook National University Hospital) ;
  • Woo, Jungmin (Department of Psychiatry, Kyungpook National University Hospital)
  • Received : 2017.11.23
  • Accepted : 2018.01.12
  • Published : 2018.11.30

Abstract

Objective: Neuropsychiatric manifestations like depression and cognitive dysfunction commonly occur in inflammatory bowel disease (IBD). In the context of the brain-gut axis model, colitis can lead to alteration of brain function in a bottom-up manner. Here, the changes in the response of the hypothalamic-pituitary-adrenal axis and inflammation-related markers in the brain in colitis were studied. Methods: Dextran sodium sulfate (DSS) was used to generate a mouse model of colitis. Mice were treated with DSS for 3 or 7 days and sacrificed. We analyzed the gene expression of brain-derived neurotrophic factor (BDNF), cyclooxygenase 2 (COX-2), and glial fibrillary acidic protein (GFAP), and the expression of GFAP, in the hippocampus, hypothalamus, and amygdala. Additionally, the levels of C-reactive protein (CRP) and serum cortisol/corticosterone were measured. Results: Alteration of inflammatory-related markers varied depending on the brain region and exposure time. In the hippocampus, COX-2 mRNA, GFAP mRNA, and GFAP expression were upregulated during exposure to DSS. However, in the hypothalamus, COX-2 mRNA was upregulated only 3 days after treatment. In the amygdala, BDNF and COX-2 mRNAs were downregulated. CRP and corticosterone expression increased with DSS treatment at day 7. Conclusion: IBD could lead to neuroinflammation in a bottom-up manner, and this effect varied according to brain region. Stress-related hormones and serum inflammatory markers, such as CRP, were upregulated from the third day of DSS treatment. Therefore, early and active intervention is required to prevent psychological and behavioral changes caused by IBD, and region-specific studies can help understand the precise mechanisms by which IBD affects the brain.

Keywords

References

  1. Bernstein CN, Fried M, Krabshuis JH, Cohen H, Eliakim R, Fedail S, et al. World Gastroenterology Organization Practice Guidelines for the diagnosis and management of IBD in 2010. Inflamm Bowel Dis 2010;16:112-124. https://doi.org/10.1002/ibd.21048
  2. Loftus EV Jr. Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology 2004;126:1504-1517. https://doi.org/10.1053/j.gastro.2004.01.063
  3. Irvine EJ, Feagan B, Rochon J, Archambault A, Fedorak RN, Groll A, et al. Quality of life: a valid and reliable measure of therapeutic efficacy in the treatment of inflammatory bowel disease. Canadian Crohn's Relapse Prevention Trial Study Group. Gastroenterology 1994;106:287-296. https://doi.org/10.1016/0016-5085(94)90585-1
  4. Husain A, Triadafilopoulos G. Communicating with patients with inflammatory bowel disease. Inflamm Bowel Dis 2004;10:444-450; discussion 451. https://doi.org/10.1097/00054725-200407000-00020
  5. Karwowski CA, Keljo D, Szigethy E. Strategies to improve quality of life in adolescents with inflammatory bowel disease. Inflamm Bowel Dis 2009;15:1755-1764. https://doi.org/10.1002/ibd.20919
  6. Bernstein CN, Blanchard JF, Rawsthorne P, Yu N. The prevalence of extraintestinal diseases in inflammatory bowel disease: a population-based study. Am J Gastroenterol 2001;96:1116-1122. https://doi.org/10.1111/j.1572-0241.2001.03756.x
  7. Zois CD, Katsanos KH, Kosmidou M, Tsianos EV. Neurologic manifestations in inflammatory bowel diseases: current knowledge and novel insights. J Crohns Colitis 2010;4:115-124. https://doi.org/10.1016/j.crohns.2009.10.005
  8. Levine JS, Burakoff R. Extraintestinal manifestations of inflammatory bowel disease. Gastroenterol Hepatol (N Y) 2011;7:235-241.
  9. Mikocka-Walus A, Knowles SR, Keefer L, Graff L. Controversies revisited: a systematic review of the comorbidity of depression and anxiety with inflammatory bowel diseases. Inflamm Bowel Dis 2016;22:752-762. https://doi.org/10.1097/MIB.0000000000000620
  10. Evrensel A, Ceylan ME. Fecal microbiota transplantation and its usage in neuropsychiatric disorders. Clin Psychopharmacol Neurosci 2016;14:231-237. https://doi.org/10.9758/cpn.2016.14.3.231
  11. Foster JA, McVey Neufeld KA. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci 2013;36:305-312. https://doi.org/10.1016/j.tins.2013.01.005
  12. Stilling RM, Dinan TG, Cryan JF. Microbial genes, brain & behaviour - epigenetic regulation of the gut-brain axis. Genes Brain Behav 2014;13:69-86. https://doi.org/10.1111/gbb.12109
  13. Logan AC, Jacka FN, Craig JM, Prescott SL. The microbiome and mental health: looking back, moving forward with lessons from allergic diseases. Clin Psychopharmacol Neurosci 2016;14:131-147. https://doi.org/10.9758/cpn.2016.14.2.131
  14. Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol Ther 2016;158:52-62. https://doi.org/10.1016/j.pharmthera.2015.11.012
  15. McEwen BS. Re-examination of the glucocorticoid hypothesis of stress and aging. Prog Brain Res 1992;93:365-381; discussion 382-383.
  16. Reagan LP, McEwen BS. Controversies surrounding glucocorticoid-mediated cell death in the hippocampus. J Chem Neuroanat 1997;13:149-167. https://doi.org/10.1016/S0891-0618(97)00031-8
  17. Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ. Learning enhances adult neurogenesis in the hippocampal formation. Nat Neurosci 1999;2:260-265. https://doi.org/10.1038/6365
  18. Sapolsky RM. Glucocorticoids, stress, and their adverse neurological effects: relevance to aging. Exp Gerontol 1999;34:721-732. https://doi.org/10.1016/S0531-5565(99)00047-9
  19. Conrad CD. Chronic stress-induced hippocampal vulnerability: the glucocorticoid vulnerability hypothesis. Rev Neurosci 2008;19:395-411.
  20. Reber SO. Stress and animal models of inflammatory bowel disease--an update on the role of the hypothalamo-pituitary-adrenal axis. Psychoneuroendocrinology 2012;37:1-19. https://doi.org/10.1016/j.psyneuen.2011.05.014
  21. Reichmann F, Hassan AM, Farzi A, Jain P, Schuligoi R, Holzer P. Dextran sulfate sodium-induced colitis alters stress-associated behaviour and neuropeptide gene expression in the amygdala-hippocampus network of mice. Sci Rep 2015;5:9970. https://doi.org/10.1038/srep09970
  22. Bettio LE, Freitas AE, Neis VB, Santos DB, Ribeiro CM, Rosa PB, et al. Guanosine prevents behavioral alterations in the forced swimming test and hippocampal oxidative damage induced by acute restraint stress. Pharmacol Biochem Behav 2014;127:7-14. https://doi.org/10.1016/j.pbb.2014.10.002
  23. Mifsud KR, Saunderson EA, Spiers H, Carter SD, Trollope AF, Mill J, et al. Rapid down-regulation of glucocorticoid receptor gene expression in the dentate gyrus after acute stress in vivo: role of DNA methylation and microRNA activity. Neuroendocrinology 2017;104:157-169. https://doi.org/10.1159/000445875
  24. Bonaz BL, Bernstein CN. Brain-gut interactions in inflammatory bowel disease. Gastroenterology 2013;144:36-49. https://doi.org/10.1053/j.gastro.2012.10.003
  25. Martin-Subero M, Anderson G, Kanchanatawan B, Berk M, Maes M. Comorbidity between depression and inflammatory bowel disease explained by immune-inflammatory, oxidative, and nitrosative stress; tryptophan catabolite; and gutbrain pathways. CNS Spectr 2016;21:184-198. https://doi.org/10.1017/S1092852915000449
  26. Ruifrok AC, Johnston DA. Quantification of histochemical staining by color deconvolution. Anal Quant Cytol Histol 2001;23:291-299.
  27. Zonis S, Pechnick RN, Ljubimov VA, Mahgerefteh M, Wawrowsky K, Michelsen KS, et al. Chronic intestinal inflammation alters hippocampal neurogenesis. J Neuroinflammation 2015;12:65. https://doi.org/10.1186/s12974-015-0281-0
  28. Font-Nieves M, Sans-Fons MG, Gorina R, Bonfill-Teixidor E, Salas-Perdomo A, Marquez-Kisinousky L, et al. Induction of COX-2 enzyme and down-regulation of COX-1 expression by lipopolysaccharide (LPS) control prostaglandin E2 production in astrocytes. J Biol Chem 2012;287:6454-6468. https://doi.org/10.1074/jbc.M111.327874
  29. Sil S, Ghosh T. Role of cox-2 mediated neuroinflammation on the neurodegeneration and cognitive impairments in colchicine induced rat model of Alzheimer's disease. J Neuroimmunol 2016;291:115-124. https://doi.org/10.1016/j.jneuroim.2015.12.003
  30. Bignami A, Dahl D. Astrocyte-specific protein and neuroglial differentiation. An immunofluorescence study with antibodies to the glial fibrillary acidic protein. J Comp Neurol 1974;153:27-38. https://doi.org/10.1002/cne.901530104
  31. O'Callaghan JP, Sriram K. Glial fibrillary acidic protein and related glial proteins as biomarkers of neurotoxicity. Expert Opin Drug Saf 2005;4:433-442. https://doi.org/10.1517/14740338.4.3.433
  32. Yang Z, Wang KK. Glial fibrillary acidic protein: from intermediate filament assembly and gliosis to neurobiomarker. Trends Neurosci 2015;38:364-374. https://doi.org/10.1016/j.tins.2015.04.003
  33. Eng LF, Yu AC, Lee YL. Astrocytic response to injury. Prog Brain Res 1992;94:353-365.
  34. Eng LF, Ghirnikar RS. GFAP and astrogliosis. Brain Pathol 1994;4:229-237. https://doi.org/10.1111/j.1750-3639.1994.tb00838.x
  35. Nagele RG, Wegiel J, Venkataraman V, Imaki H, Wang KC, Wegiel J. Contribution of glial cells to the development of amyloid plaques in Alzheimer's disease. Neurobiol Aging 2004;25:663-674. https://doi.org/10.1016/j.neurobiolaging.2004.01.007
  36. Jacobsen JP, Mork A. Chronic corticosterone decreases brainderived neurotrophic factor (BDNF) mRNA and protein in the hippocampus, but not in the frontal cortex, of the rat. Brain Res 2006;1110:221-225. https://doi.org/10.1016/j.brainres.2006.06.077
  37. Schaaf MJ, De Kloet ER, Vreugdenhil E. Corticosterone effects on BDNF expression in the hippocampus. Implications for memory formation. Stress 2000;3:201-208. https://doi.org/10.3109/10253890009001124
  38. O'Banion MK. Cyclooxygenase-2: molecular biology, pharmacology, and neurobiology. Crit Rev Neurobiol 1999;13:45-82. https://doi.org/10.1615/CritRevNeurobiol.v13.i1.30
  39. Schaaf MJ, de Jong J, de Kloet ER, Vreugdenhil E. Downregulation of BDNF mRNA and protein in the rat hippocampus by corticosterone. Brain Res 1998;813:112-120. https://doi.org/10.1016/S0006-8993(98)01010-5
  40. Yamagata K, Andreasson KI, Kaufmann WE, Barnes CA, Worley PF. Expression of a mitogen-inducible cyclooxygenase in brain neurons: regulation by synaptic activity and glucocorticoids. Neuron 1993;11:371-386. https://doi.org/10.1016/0896-6273(93)90192-T
  41. O'Callaghan JP, Brinton RE, McEwen BS. Glucocorticoids regulate the concentration of glial fibrillary acidic protein throughout the brain. Brain Res 1989;494:159-161. https://doi.org/10.1016/0006-8993(89)90156-X
  42. Lindsay RM, Thoenen H, Barde YA. Placode and neural crest-derived sensory neurons are responsive at early developmental stages to brain-derived neurotrophic factor. Dev Biol 1985;112:319-328. https://doi.org/10.1016/0012-1606(85)90402-6
  43. Hofer MM, Barde YA. Brain-derived neurotrophic factor prevents neuronal death in vivo. Nature 1988;331:261-262. https://doi.org/10.1038/331261a0
  44. Sannino G, Pasqualini L, Ricciardelli E, Montilla P, Soverchia L, Ruggeri B, et al. Acute stress enhances the expression of neuroprotection- and neurogenesis-associated genes in the hippocampus of a mouse restraint model. Oncotarget 2016;7:8455-8465.
  45. Gray JD, Milner TA, McEwen BS. Dynamic plasticity: the role of glucocorticoids, brain-derived neurotrophic factor and other trophic factors. Neuroscience 2013;239:214-227. https://doi.org/10.1016/j.neuroscience.2012.08.034
  46. Duman RS, Heninger GR, Nestler EJ. A molecular and cellular theory of depression. Arch Gen Psychiatry 1997;54:597-606. https://doi.org/10.1001/archpsyc.1997.01830190015002
  47. Suliman S, Hemmings SM, Seedat S. Brain-derived neurotrophic factor (BDNF) protein levels in anxiety disorders: systematic review and meta-regression analysis. Front Integr Neurosci 2013;7:55.
  48. Guilloux JP, Douillard-Guilloux G, Kota R, Wang X, Gardier AM, Martinowich K, et al. Molecular evidence for BDNF- and GABA-related dysfunctions in the amygdala of female subjects with major depression. Mol Psychiatry 2012;17:1130-1142. https://doi.org/10.1038/mp.2011.113
  49. Hamilton JP, Siemer M, Gotlib IH. Amygdala volume in major depressive disorder: a meta-analysis of magnetic resonance imaging studies. Mol Psychiatry 2008;13:993-1000. https://doi.org/10.1038/mp.2008.57
  50. Kimbrell TA, Ketter TA, George MS, Little JT, Benson BE, Willis MW, et al. Regional cerebral glucose utilization in patients with a range of severities of unipolar depression. Biol Psychiatry 2002;51:237-252. https://doi.org/10.1016/S0006-3223(01)01216-1
  51. Agostini A, Filippini N, Cevolani D, Agati R, Leoni C, Tambasco R, et al. Brain functional changes in patients with ulcerative colitis: a functional magnetic resonance imaging study on emotional processing. Inflamm Bowel Dis 2011;17:1769-1777. https://doi.org/10.1002/ibd.21549
  52. Breder CD, Dewitt D, Kraig RP. Characterization of inducible cyclooxygenase in rat brain. J Comp Neurol 1995;355:296-315. https://doi.org/10.1002/cne.903550208
  53. Chen C, Magee JC, Bazan NG. Cyclooxygenase-2 regulates prostaglandin E2 signaling in hippocampal long-term synaptic plasticity. J Neurophysiol 2002;87:2851-2857. https://doi.org/10.1152/jn.2002.87.6.2851
  54. Albrecht D. Angiotensin-(1-7)-induced plasticity changes in the lateral amygdala are mediated by COX-2 and NO. Learn Mem 2007;14:177-184. https://doi.org/10.1101/lm.425907
  55. Noh H, Jeon J, Seo H. Systemic injection of LPS induces region-specific neuroinflammation and mitochondrial dysfunction in normal mouse brain. Neurochem Int 2014;69:35-40. https://doi.org/10.1016/j.neuint.2014.02.008

Cited by

  1. Heart and Brain Interaction of Psychiatric Illness: A Review Focused on Heart Rate Variability, Cognitive Function, and Quantitative Electroencephalography vol.17, pp.4, 2018, https://doi.org/10.9758/cpn.2019.17.4.459
  2. Electroacupuncture and Moxibustion Regulate Hippocampus Glia and Mitochondria Activation in DSS-Induced Colitis Mice vol.2020, pp.None, 2020, https://doi.org/10.1155/2020/2530253
  3. Effect of N‐palmitoylethanolamine‐oxazoline on comorbid neuropsychiatric disturbance associated with inflammatory bowel disease vol.34, pp.3, 2018, https://doi.org/10.1096/fj.201901584rr
  4. Maladaptive Alterations of Defensive Response Following Developmental Complex Stress in Rats vol.18, pp.3, 2018, https://doi.org/10.9758/cpn.2020.18.3.412
  5. Variant-to-Gene-Mapping Analyses Reveal a Role for the Hypothalamus in Genetic Susceptibility to Inflammatory Bowel Disease vol.11, pp.3, 2018, https://doi.org/10.1016/j.jcmgh.2020.10.004
  6. Pequi Oil, a MUFA/Carotenoid‐Rich Oil, Exhibited Protective Effects against DSS‐Induced Ulcerative Colitis in Mice vol.123, pp.9, 2018, https://doi.org/10.1002/ejlt.202000332
  7. Identification of a choroid plexus vascular barrier closing during intestinal inflammation vol.374, pp.6566, 2018, https://doi.org/10.1126/science.abc6108
  8. Is cyclooxygenase‐1 involved in neuroinflammation? vol.99, pp.11, 2018, https://doi.org/10.1002/jnr.24934
  9. Apple Polyphenols Extract (APE) Alleviated Dextran Sulfate Sodium Induced Acute Ulcerative Colitis and Accompanying Neuroinflammation via Inhibition of Apoptosis and Pyroptosis vol.10, pp.11, 2018, https://doi.org/10.3390/foods10112711
  10. Lipopolysaccharide exacerbates chronic restraint stress-induced neurobehavioral deficits: Mechanisms by redox imbalance, ASK1-related apoptosis, autophagic dysregulation vol.144, pp.None, 2021, https://doi.org/10.1016/j.jpsychires.2021.10.021
  11. Is LRRK2 the missing link between inflammatory bowel disease and Parkinson’s disease? vol.7, pp.1, 2018, https://doi.org/10.1038/s41531-021-00170-1
  12. The impact of dextran sodium sulphate and probiotic pre-treatment in a murine model of Parkinson’s disease vol.18, pp.1, 2021, https://doi.org/10.1186/s12974-020-02062-2
  13. Animal models of brain-first and body-first Parkinson's disease vol.163, pp.None, 2018, https://doi.org/10.1016/j.nbd.2021.105599
  14. Neuroinflammatory remodeling of the anterior cingulate cortex as a key driver of mood disorders in gastrointestinal disease and disorders vol.133, pp.None, 2018, https://doi.org/10.1016/j.neubiorev.2021.12.020