Interrogating the Gut-Brain Axis in the Context of Inflammatory Bowel Disease: A Translational Approach

Stephen M Collins, Stephen M Collins

Abstract

This review examines preclinical and clinical studies relevant to our understanding of how the bidirectional gut-brain axis influences the natural history of inflammatory bowel disease. Preclinical studies provide proof of concept that preexisting behavioral illness, such as depression, results in increased susceptibility to inflammatory stimuli and that commonly used classes of antidepressants protect against this vulnerability. However, clinical studies suggesting behavioral illness as a risk factor for IBD and a protective role for antidepressants have relied primarily on symptom-reporting rather than objective measurements of inflammation. In terms of gut-to-brain signaling, there is emerging evidence from preclinical and clinical observation that intestinal inflammation alters brain functions, including the induction of mood disorders, alteration of circadian rhythm both centrally and peripherally, and changes in appetitive behaviors. Furthermore, preclinical studies suggest that effective treatment of intestinal inflammation improves associated behavioral impairment. Taken together, the findings of this review encourage a holistic approach to the management of patients with IBD, accommodating lifestyle issues that include the avoidance of sleep deprivation, optimized nutrition, and the monitoring and appropriate management of behavioral disorders. The review also acknowledges the need for better-designed clinical studies evaluating the impact of behavioral disorders and their treatments on the natural history of IBD, utilizing hard end points to assess changes in the inflammatory process as opposed to reliance on symptom-based assessments. The findings of the review also encourage a better understanding of changes in brain function and circadian rhythm induced by intestinal inflammation.

Keywords: antidepressants; circadian rhythm; cytokines; depression; inflammation; vagus nerve.

© 2020 Crohn’s & Colitis Foundation. Published by Oxford University Press on behalf of Crohn’s & Colitis Foundation.

Figures

FIGURE 1.
FIGURE 1.
The structure of this review is illustrated. It will address 6 questions pertaining to the role of the bidirectional gut-brain axis in the natural history of IBD. Each question is addressed by first examining evidence of biologic feasibility based on preclinical studies, followed by a review of the existing clinical literature pertinent to the question.
FIGURE 2.
FIGURE 2.
Bidirectional gut-brain interactions in IBD may lead to a vicious cycle that promotes intestinal inflammation and reduces well-being. Thus, a holistic approach to the management of IBD is encouraged, involving both gut-to-brain and brain-to-gut signaling.

References

    1. Mulder DJ, Noble AJ, Justinich CJ, et al. . A tale of two diseases: the history of inflammatory bowel disease. J Crohns Colitis. 2014;8:341–348.
    1. Warren S, Sommers SC. Pathogenesis of ulcerative colitis. Am J Pathol. 1949;25:657–679.
    1. Truelove SC, Witts LJ. Cortisone in ulcerative colitis; final report on a therapeutic trial. Br Med J. 1955;2:1041–1048.
    1. Engel GL. The need for a new medical model: a challenge for biomedicine. Science. 1977;196:129–136.
    1. Bonaz BL, Bernstein CN. Brain-gut interactions in inflammatory bowel disease. Gastroenterology. 2013;144:36–49.
    1. Abautret-Daly Á, Dempsey E, Parra-Blanco A, et al. . Gut-brain actions underlying comorbid anxiety and depression associated with inflammatory bowel disease. Acta Neuropsychiatr. 2018;30:275–296.
    1. Mogilevski T, Burgell R, Aziz Q, et al. . Review article: the role of the autonomic nervous system in the pathogenesis and therapy of IBD. Aliment Pharmacol Ther. 2019;50:720–737.
    1. Bonaz B. Inflammatory bowel diseases: a dysfunction of brain-gut interactions? Minerva Gastroenterol Dietol. 2013;59:241–259.
    1. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol. 2012;10:735–742.
    1. Goehler LE, Gaykema RP, Opitz N, et al. . Activation in vagal afferents and central autonomic pathways: early responses to intestinal infection with Campylobacter jejuni. Brain Behav Immun. 2005;19:334–344.
    1. Tracey KJ. The inflammatory reflex. Nature. 2002;420:853–859.
    1. Ghia JE, Blennerhassett P, Collins SM. Impaired parasympathetic function increases susceptibility to inflammatory bowel disease in a mouse model of depression. J Clin Invest. 2008;118:2209–2218.
    1. Bonaz B, Sinniger V, Hoffmann D, et al. . Chronic vagus nerve stimulation in Crohn’s disease: a 6-month follow-up pilot study. Neurogastroenterol Motil. 2016;28:948–953.
    1. Meregnani J, Clarençon D, Vivier M, et al. . Anti-inflammatory effect of vagus nerve stimulation in a rat model of inflammatory bowel disease. Auton Neurosci. 2011;160:82–89.
    1. Sun P, Zhou K, Wang S, et al. . Involvement of MAPK/NF-κB signaling in the activation of the cholinergic anti-inflammatory pathway in experimental colitis by chronic vagus nerve stimulation. PLoS One. 2013;8:e69424.
    1. de Jonge WJ, Ulloa L. The alpha7 nicotinic acetylcholine receptor as a pharmacological target for inflammation. Br J Pharmacol. 2007;151:915–929.
    1. Walker JR, Ediger JP, Graff LA, et al. . The Manitoba IBD cohort study: a population-based study of the prevalence of lifetime and 12-month anxiety and mood disorders. Am J Gastroenterol. 2008;103:1989–1997.
    1. Mikocka-Walus A, Knowles SR, Keefer L, et al. . Controversies revisited: a systematic review of the comorbidity of depression and anxiety with inflammatory bowel diseases. Inflamm Bowel Dis. 2016;22:752–762.
    1. Byrne G, Rosenfeld G, Leung Y, et al. . Prevalence of anxiety and depression in patients with inflammatory bowel disease. Can J Gastroenterol Hepatol. 2017;2017:6496727.
    1. Geiss T, Schaefert RM, Berens S, et al. . Risk of depression in patients with inflammatory bowel disease. J Dig Dis. 2018;19:456–467.
    1. Neuendorf R, Harding A, Stello N, et al. . Depression and anxiety in patients with inflammatory bowel disease: a systematic review. J Psychosom Res. 2016;87:70–80.
    1. Jairath V, Zou G, Parker CE, et al. . Systematic review with meta-analysis: placebo rates in induction and maintenance trials of Crohn’s disease. Aliment Pharmacol Ther. 2017;45:1021–1042.
    1. Jairath V, Zou GY, Parker CE, et al. . Placebo response and remission rates in randomised trials of induction and maintenance therapy for ulcerative colitis. Cochrane Database Syst Rev. 2017;9:CD011572.
    1. Hodes GE, Kana V, Menard C, et al. . Neuroimmune mechanisms of depression. Nat Neurosci. 2015;18:1386–1393.
    1. Vetulani J. Early maternal separation: a rodent model of depression and a prevailing human condition. Pharmacol Rep. 2013;65:1451–1461.
    1. Varghese AK, Verdú EF, Bercik P, et al. . Antidepressants attenuate increased susceptibility to colitis in a murine model of depression. Gastroenterology. 2006;130:1743–1753.
    1. De Palma G, Lynch MD, Lu J, et al. . Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice. Sci Transl Med. 2017;9:eaaf6397.
    1. Fattahian E, Hajhashemi V, Rabbani M, et al. . Anti-inflammatory effect of amitriptyline on ulcerative colitis in normal and reserpine-induced depressed rats. Iran J Pharm Res. 2016;15:125–137.
    1. Marrie RA, Walld R, Bolton JM, et al. ; CIHR Team in Defining the Burden and Managing the Effects of Psychiatric Comorbidity in Chronic Immunoinflammatory Disease Rising incidence of psychiatric disorders before diagnosis of immune-mediated inflammatory disease. Epidemiol Psychiatr Sci. 2019;28:333–342.
    1. Frolkis AD, Vallerand IA, Shaheen AA, et al. . Depression increases the risk of inflammatory bowel disease, which may be mitigated by the use of antidepressants in the treatment of depression. Gut. 2019;68:1606–1612.
    1. Qiu BS, Vallance BA, Blennerhassett PA, et al. . The role of CD4+ lymphocytes in the susceptibility of mice to stress-induced reactivation of experimental colitis. Nat Med. 1999;5:1178–1182.
    1. Gareau MG, Jury J, Perdue MH. Neonatal maternal separation of rat pups results in abnormal cholinergic regulation of epithelial permeability. Am J Physiol Gastrointest Liver Physiol. 2007;293:G198–G203.
    1. Ghia JE, Blennerhassett P, Deng Y, et al. . Reactivation of inflammatory bowel disease in a mouse model of depression. Gastroenterology. 2009;136:2280–2288.e1.
    1. Ghia JE, Park AJ, Blennerhassett P, et al. . Adoptive transfer of macrophage from mice with depression-like behavior enhances susceptibility to colitis. Inflamm Bowel Dis. 2011;17:1474–1489.
    1. Sgoifo A, Carnevali L, Alfonso Mde L, et al. . Autonomic dysfunction and heart rate variability in depression. Stress. 2015;18:343–352.
    1. Carreno FR, Frazer A. Vagal nerve stimulation for treatment-resistant depression. Neurotherapeutics. 2017;14:716–727.
    1. Poojary P, Saha A, Chauhan K, et al. . Predictors of hospital readmissions for ulcerative colitis in the United States: a national database study. Inflamm Bowel Dis. 2017;23:347–356.
    1. Gaines LS, Slaughter JC, Horst SN, et al. . Association between affective-cognitive symptoms of depression and exacerbation of Crohn’s disease. Am J Gastroenterol. 2016;111:864–870.
    1. Persoons P, Vermeire S, Demyttenaere K, et al. . The impact of major depressive disorder on the short- and long-term outcome of Crohn’s disease treatment with infliximab. Aliment Pharmacol Ther. 2005;22:101–110.
    1. Mittermaier C, Dejaco C, Waldhoer T, et al. . Impact of depressive mood on relapse in patients with inflammatory bowel disease: a prospective 18-month follow-up study. Psychosom Med. 2004;66:79–84.
    1. Mikocka-Walus A, Pittet V, Rossel JB, et al. ; Swiss IBD Cohort Study Group. Symptoms of depression and anxiety are independently associated with clinical recurrence of inflammatory bowel disease. Clin Gastroenterol Hepatol. 2016;14:829–835.e1.
    1. Kochar B, Barnes EL, Long MD, et al. . Depression is associated with more aggressive inflammatory bowel disease. Am J Gastroenterol. 2018;113:80–85.
    1. Calloway A, Dalal R, Beaulieu DB, et al. . Depressive symptoms predict anti-tumor necrosis factor therapy noncompliance in patients with inflammatory bowel disease. Dig Dis Sci. 2017;62:3563–3567.
    1. Narula N, Cooray M, Anglin R, et al. . Impact of high-dose vitamin d3 supplementation in patients with Crohn’s disease in remission: a pilot randomized double-blind controlled study. Dig Dis Sci. 2017;62:448–455.
    1. Gracie DJ, Guthrie EA, Hamlin PJ, et al. . Bidirectionality of brain-gut interactions in patients with inflammatory bowel disease. Gastroenterology. 2018;154:1635–1646.e3.
    1. Gracie DJ, Williams CJ, Sood R, et al. . Poor correlation between clinical disease activity and mucosal inflammation, and the role of psychological comorbidity, in inflammatory bowel disease. Am J Gastroenterol. 2016;111:541–551.
    1. Bernstein CN, Singh S, Graff LA, et al. . A prospective population-based study of triggers of symptomatic flares in IBD. Am J Gastroenterol. 2010;105:1994–2002.
    1. Targownik LE, Sexton KA, Bernstein MT, et al. . The relationship among perceived stress, symptoms, and inflammation in persons with inflammatory bowel disease. Am J Gastroenterol. 2015;110:1001–1012; quiz 13.
    1. Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights into pathogenic and therapeutic implications. Gut. 2005;54:1481–1491.
    1. Xia BT, Beckmann N, Winer LK, et al. . Amitriptyline treatment mitigates sepsis-induced tumor necrosis factor expression and coagulopathy. Shock. 2019;51:356–363.
    1. Minaiyan M, Hajhashemi V, Rabbani M, et al. . Evaluation of anti-colitic effect of fluvoxamine against acetic acid-induced colitis in normal and reserpinized depressed rats. Eur J Pharmacol. 2015;746:293–300.
    1. Thorkelson G, Bielefeldt K, Szigethy E. empirically supported use of psychiatric medications in adolescents and adults with IBD. Inflamm Bowel Dis. 2016;22:1509–1522.
    1. Haapamäki J, Tanskanen A, Roine RP, et al. . Medication use among inflammatory bowel disease patients: excessive consumption of antidepressants and analgesics. Scand J Gastroenterol. 2013;48:42–50.
    1. Virta LJ, Kolho KL. Antidepressant use among paediatric patients with recent-onset inflammatory bowel disease: a nationwide case control study in Finland. J Paediatr Child Health. 2014;50:562–565.
    1. Mikocka-Walus AA, Turnbull DA, Moulding NT, et al. . “It doesn’t do any harm, but patients feel better”: a qualitative exploratory study on gastroenterologists’ perspectives on the role of antidepressants in inflammatory bowel disease. BMC Gastroenterol. 2007;7:38.
    1. Goodhand JR, Greig FI, Koodun Y, et al. . Do antidepressants influence the disease course in inflammatory bowel disease? A retrospective case-matched observational study. Inflamm Bowel Dis. 2012;18:1232–1239.
    1. Kristensen MS, Kjærulff TM, Ersbøll AK, et al. . The influence of antidepressants on the disease course among patients with Crohn’s disease and ulcerative colitis-a Danish nationwide register-based cohort study. Inflamm Bowel Dis. 2019;25:886–893.
    1. Fiest KM, Bernstein CN, Walker JR, et al. ; CIHR Team “Defining the burden and managing the effects of psychiatric comorbidity in chronic immunoinflammatory disease.” Systematic review of interventions for depression and anxiety in persons with inflammatory bowel disease. BMC Res Notes. 2016;9:404.
    1. Mikocka-Walus A, Prady SL, Pollok J, et al. . Adjuvant therapy with antidepressants for the management of inflammatory bowel disease. Cochrane Database Syst Rev. 2019;4:CD012680.
    1. Macer BJ, Prady SL, Mikocka-Walus A. Antidepressants in inflammatory bowel disease: a systematic review. Inflamm Bowel Dis. 2017;23:534–550.
    1. Bennebroek Evertsz’ F, Sprangers MAG, Sitnikova K, et al. . Effectiveness of cognitive-behavioral therapy on quality of life, anxiety, and depressive symptoms among patients with inflammatory bowel disease: a multicenter randomized controlled trial. J Consult Clin Psychol. 2017;85:918–925.
    1. Mikocka-Walus A, Bampton P, Hetzel D, et al. . Cognitive-behavioral therapy for inflammatory bowel disease: 24-month data from a randomised controlled trial. Int J Behav Med. 2017;24:127–135.
    1. van den Brink G, Stapersma L, Bom AS, et al. . Effect of cognitive behavioral therapy on clinical disease course in adolescents and young adults with inflammatory bowel disease and subclinical anxiety and/or depression: results of a randomized trial. Inflamm Bowel Dis. 2019;25:1945–1956.
    1. Taft TH, Ballou S, Bedell A, et al. . Psychological considerations and interventions in inflammatory bowel disease patient care. Gastroenterol Clin North Am. 2017;46:847–858.
    1. Mawdsley JE, Jenkins DG, Macey MG, et al. . The effect of hypnosis on systemic and rectal mucosal measures of inflammation in ulcerative colitis. Am J Gastroenterol. 2008;103:1460–1469.
    1. Cheifetz AS, Gianotti R, Luber R, et al. . Complementary and alternative medicines used by patients with inflammatory bowel diseases. Gastroenterology. 2017;152:415–429.e15.
    1. Haj-Mirzaian A, Amiri S, Amini-Khoei H, et al. . Anxiety- and depressive-like behaviors are associated with altered hippocampal energy and inflammatory status in a mouse model of Crohn’s disease. Neuroscience. 2017;366:124–137.
    1. Nyuyki KD, Cluny NL, Swain MG, et al. . Altered brain excitability and increased anxiety in mice with experimental colitis: consideration of hyperalgesia and sex differences. Front Behav Neurosci. 2018;12:58.
    1. Bercik P, Verdu EF, Foster JA, et al. . Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology. 2010;139:2102–2112.e1.
    1. Friebe A, Brünahl C, Karimi K, et al. . Effects of complete vagotomy and blockage of cell adhesion molecules on interferon-α induced behavioral changes in mice. Behav Brain Res. 2013;240:1–10.
    1. Bercik P, Denou E, Collins J, et al. . The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology. 2011;141:599–609, 609.e1.
    1. Martin-Subero M, Anderson G, Kanchanatawan B, et al. . Comorbidity between depression and inflammatory bowel disease explained by immune-inflammatory, oxidative, and nitrosative stress; tryptophan catabolite; and gut-brain pathways. CNS Spectr. 2016;21:184–198.
    1. Agostini A, Ballotta D, Righi S, et al. . Stress and brain functional changes in patients with Crohn’s disease: a functional magnetic resonance imaging study. Neurogastroenterol Motil. 2017;29:1–10.
    1. Bao CH, Liu P, Liu HR, et al. . Alterations in brain grey matter structures in patients with crohn’s disease and their correlation with psychological distress. J Crohns Colitis. 2015;9:532–540.
    1. Huang JS, Terrones L, Simmons AN, et al. . Pilot study of functional magnetic resonance imaging responses to somatic pain stimuli in youth with functional and inflammatory gastrointestinal disease. J Pediatr Gastroenterol Nutr. 2016;63:500–507.
    1. Thomann AK, Griebe M, Thomann PA, et al. . Intrinsic neural network dysfunction in quiescent Crohn’s Disease. Sci Rep. 2017;7:11579.
    1. Dibner C, Schibler U, Albrecht U. The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol. 2010;72:517–549.
    1. Yu W, Hardin PE. Circadian oscillators of Drosophila and mammals. J Cell Sci. 2006;119:4793–4795.
    1. Scheiermann C, Kunisaki Y, Frenette PS. Circadian control of the immune system. Nat Rev Immunol. 2013;13:190–198.
    1. Pagel R, Bär F, Schröder T, et al. . Circadian rhythm disruption impairs tissue homeostasis and exacerbates chronic inflammation in the intestine. Faseb J. 2017;31:4707–4719.
    1. Amara J, Saliba Y, Hajal J, et al. . Circadian rhythm disruption aggravates DSS-induced colitis in mice with fecal calprotectin as a marker of colitis severity. Dig Dis Sci. 2019;64:3122–3133.
    1. Liu X, Yu R, Zhu L, et al. . Bidirectional regulation of circadian disturbance and inflammation in inflammatory bowel disease. Inflamm Bowel Dis. 2017;23:1741–1751.
    1. Albrecht U, Ripperger JA. Circadian clocks and sleep: impact of rhythmic metabolism and waste clearance on the brain. Trends Neurosci. 2018;41:677–688.
    1. Johnston JD. Physiological links between circadian rhythms, metabolism and nutrition. Exp Physiol. 2014;99:1133–1137.
    1. Weintraub Y, Cohen S, Chapnik N, et al. . Clock gene disruption is an initial manifestation of inflammatory bowel diseases. Clin Gastroenterol Hepatol. 2020;18:115–122.e1.
    1. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. . Sleep duration affects risk for ulcerative colitis: a prospective cohort study. Clin Gastroenterol Hepatol. 2014;12:1879–1886.
    1. Mazzoccoli G, Palmieri O, Corritore G, et al. . Association study of a polymorphism in clock gene PERIOD3 and risk of inflammatory bowel disease. Chronobiol Int. 2012;29:994–1003.
    1. Tang Y, Preuss F, Turek FW, et al. . Sleep deprivation worsens inflammation and delays recovery in a mouse model of colitis. Sleep Med. 2009;10:597–603.
    1. 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.
    1. 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.
    1. Hood MM, Wilson R, Gorenz A, et al. . Sleep quality in ulcerative colitis: associations with inflammation, psychological distress, and quality of life. Int J Behav Med. 2018;25:517–525.
    1. Graff LA, Vincent N, Walker JR, et al. . A population-based study of fatigue and sleep difficulties in inflammatory bowel disease. Inflamm Bowel Dis. 2011;17:1882–1889.
    1. Tracey KJ, Wei H, Manogue KR, et al. . Cachectin/tumor necrosis factor induces cachexia, anemia, and inflammation. J Exp Med. 1988;167:1211–1227.
    1. McHugh K, Castonguay TW, Collins SM, et al. . Characterization of suppression of food intake following acute colon inflammation in the rat. Am J Physiol. 1993;265:R1001–R1005.
    1. McHugh KJ, Collins SM, Weingarten HP. Central interleukin-1 receptors contribute to suppression of feeding after acute colitis in the rat. Am J Physiol. 1994;266:R1659–R1663.
    1. El-Haj T, Poole S, Farthing MJ, et al. . Anorexia in a rat model of colitis: interaction of interleukin-1 and hypothalamic serotonin. Brain Res. 2002;927:1–7.
    1. Barbier M, Cherbut C, Aubé AC, et al. . Elevated plasma leptin concentrations in early stages of experimental intestinal inflammation in rats. Gut. 1998;43:783–790.
    1. Singh S, Blanchard A, Walker JR, et al. . Common symptoms and stressors among individuals with inflammatory bowel diseases. Clin Gastroenterol Hepatol. 2011;9:769–775.
    1. Hartman C, Marderfeld L, Davidson K, et al. . Food intake adequacy in children and adolescents with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2016;63:437–444.
    1. Ilzarbe L, Fàbrega M, Quintero R, et al. . Inflammatory bowel disease and eating disorders: a systematized review of comorbidity. J Psychosom Res. 2017;102:47–53.
    1. Ballinger A, Kelly P, Hallyburton E, et al. . Plasma leptin in chronic inflammatory bowel disease and HIV: implications for the pathogenesis of anorexia and weight loss. Clin Sci (Lond). 1998;94:479–483.
    1. Hoppin AG, Kaplan LM, Zurakowski D, et al. . Serum leptin in children and young adults with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 1998;26:500–505.
    1. Krügel U, Fischer J, Radicke S, et al. . Antidepressant effects of TNF-α blockade in an animal model of depression. J Psychiatr Res. 2013;47:611–616.
    1. Horst S, Chao A, Rosen M, et al. . Treatment with immunosuppressive therapy may improve depressive symptoms in patients with inflammatory bowel disease. Dig Dis Sci. 2015;60:465–470.
    1. Stevens BW, Borren NZ, Velonias G, et al. . Vedolizumab therapy is associated with an improvement in sleep quality and mood in inflammatory bowel diseases. Dig Dis Sci. 2017;62:197–206.
    1. Gray MA, Chao CY, Staudacher HM, et al. . Anti-TNFα therapy in IBD alters brain activity reflecting visceral sensory function and cognitive-affective biases. PLoS One. 2018;13:e0193542.
    1. Mannino G, Caradonna F, Cruciata I, et al. . Melatonin reduces inflammatory response in human intestinal epithelial cells stimulated by interleukin-1β. J Pineal Res. 2019;67:e12598.

Source: PubMed

3
订阅