Cognitive behavioral therapy for irritable bowel syndrome induces bidirectional alterations in the brain-gut-microbiome axis associated with gastrointestinal symptom improvement

Jonathan P Jacobs, Arpana Gupta, Ravi R Bhatt, Jacob Brawer, Kan Gao, Kirsten Tillisch, Venu Lagishetty, Rebecca Firth, Gregory D Gudleski, Benjamin M Ellingson, Jennifer S Labus, Bruce D Naliboff, Jeffrey M Lackner, Emeran A Mayer, Jonathan P Jacobs, Arpana Gupta, Ravi R Bhatt, Jacob Brawer, Kan Gao, Kirsten Tillisch, Venu Lagishetty, Rebecca Firth, Gregory D Gudleski, Benjamin M Ellingson, Jennifer S Labus, Bruce D Naliboff, Jeffrey M Lackner, Emeran A Mayer

Abstract

Background: There is growing recognition that bidirectional signaling between the digestive tract and the brain contributes to irritable bowel syndrome (IBS). We recently showed in a large randomized controlled trial that cognitive behavioral therapy (CBT) reduces IBS symptom severity. This study investigated whether baseline brain and gut microbiome parameters predict CBT response and whether response is associated with changes in the brain-gut-microbiome (BGM) axis.

Methods: Eighty-four Rome III-diagnosed IBS patients receiving CBT were drawn from the Irritable Bowel Syndrome Outcome Study (IBSOS; ClinicalTrials.gov NCT00738920) for multimodal brain imaging and psychological assessments at baseline and after study completion. Fecal samples were collected at baseline and post-treatment from 34 CBT recipients for 16S rRNA gene sequencing, untargeted metabolomics, and measurement of short-chain fatty acids. Clinical measures, brain functional connectivity and microstructure, and microbiome features associated with CBT response were identified by multivariate linear and negative binomial models.

Results: At baseline, CBT responders had increased fecal serotonin levels, and increased Clostridiales and decreased Bacteroides compared to non-responders. A random forests classifier containing 11 microbial genera predicted CBT response with high accuracy (AUROC 0.96). Following treatment, CBT responders demonstrated reduced functional connectivity in regions of the sensorimotor, brainstem, salience, and default mode networks and changes in white matter in the basal ganglia and other structures. Brain changes correlated with microbiome shifts including Bacteroides expansion in responders.

Conclusions: Pre-treatment intestinal microbiota and serotonin levels were associated with CBT response, suggesting that peripheral signals from the microbiota can modulate central processes affected by CBT that generate abdominal symptoms in IBS. CBT response is characterized by co-correlated shifts in brain networks and gut microbiome that may reflect top-down effects of the brain on the microbiome during CBT. Video abstract.

Keywords: Biomarkers; Brain-gut-microbiome axis; Cognitive behavioral therapy; Irritable bowel syndrome; Neuroimaging; Outcome prediction.

Conflict of interest statement

EAM is a scientific advisory board member of Danone, Axial Biotherapeutics, Viome, Amare, Mahana Therapeutics, Pendulum, Bloom Biosciences, APC Microbiome Ireland. BME is an advisor for Hoffman La-Roche; Siemens; Nativis; Medicenna; MedQIA; Bristol Meyers Squibb; Imaging Endpoints; VBL; and Agios Pharmaceuticals. BME is a Paid Consultant for Nativis; MedQIA; Siemens; Hoffman La-Roche; Imaging Endpoints; Medicenna; and Agios. BME received grant funding from Siemens, Agios, and Janssen. JPJ, AG, RRB, JB, KG, KT, VL, RF, GG, JSL, BDN, and JML do not have any disclosures.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Baseline fecal microbiota and serotonin are associated with CBT response. A, B Principal coordinates (PC) analysis of 16S rRNA sequence data. Each dot represents the baseline microbiome composition of one IBS participant. Color denotes CBT responder status and dots are sized by the fraction of the microbiome comprised of the Bacteroides genus (A) or Clostridiales order (B). C Microbial genera with statistically significant association with CBT responder status (q<.05) are shown. The y axis shows the log2 of the fold change between responders vs. non-responders (NR). Dot size is proportional to mean relative abundance across all samples. D Baseline relative abundances (median scaled) in feces of the neurotransmitters serotonin, dopamine, and histamine. Lines indicate medians. * p<.05 by Mann–Whitney U test. e Non-metric multidimensional scaling analysis (NMDS) (stress=0.20) of global metabolomics profiles. Color denotes CBT responder status. P value calculated by Adonis, adjusting for sex and bowel habit subtype
Fig. 2
Fig. 2
Classifiers derived from baseline fecal microbiota profiles outperformed those based on clinical/demographic and neuroimaging data to predict CBT response. A Receiver operating characteristic curves of random forest classifiers for CBT response constructed from differentially abundant microbial genera, baseline clinical/demographic data (left panel), or brain data (right panel). The 95% confidence intervals are represented as colored regions surrounding these curves (blue=microbiome, red=clinical/demographics or brain). B Importance scores for the 11 microbial genera in the random forests classifier
Fig. 3
Fig. 3
Changes in functional connectivity in responders and non-responders to CBT. A Connectograms demonstrating pair-wise connectivity differences in responders and non-responders to CBT. Significant decreases in connectivity between brain regions are denoted by blue lines connecting the regions (color intensity indicates magnitude of effect). There were no significant increases in connectivity. SMN: sensorimotor network, BG: basal ganglia, DMN: default mode network, SAL: salience network, ERN: emotion regulation network, CAN: central autonomic network, CEN: central executive Network, OCC: occipital/visual network. B Regions that significantly differed between responders and non-responders to CBT. Responders to CBT: ACirIns (anterior insula/anterior segment of the circular sulcus of the insula), MPosCgG/S (anterior mid-cingulate cortex), InfCirIns (anterior insula/inferior segment of the circular sulcus of the insula), SupTGLp (lateral aspect of the superior temporal gyrus), HG (Heschl’s gyrus), TPI (planum temporale), BSt (brainstem). Non-responders to CBT: MFG (middle frontal gyrus), PosDCgG (dorsal posterior cingulate cortex), PosVCgG (ventral posterior cingulate cortex), InfOcG/S (inferior occipital gyrus and sulcus)
Fig. 4
Fig. 4
CBT responders had distinct changes in white matter integrity compared to non-responders. A Colored areas indicate regions within the left inferior longitudinal fasciculus that had a significant change in FA after CBT. Color corresponds to relative difference in FA change between CBT responders and non-responders. B, C Colored areas indicate regions of the bilateral basal ganglia and anterior thalamus (B) and isthmus of the corpus callosum (C) that had a significant change in ADC after CBT. Color corresponds to relative difference in ADC change between CBT responders and non-responders
Fig. 5
Fig. 5
CBT responders have altered intestinal microbiome composition after CBT characterized by Bacteroides expansion. A Fecal microbial alpha diversity is shown for CBT responders and non-responders (NR) at baseline (PRE) and after CBT (POST). Three metrics are used: Chao1 index (richness), Faith’s phylogenetic diversity (PD), and Shannon index (richness and evenness). * p<.05. B Principal coordinates analysis of 16S rRNA sequence data before and after CBT, stratified by CBT response status. Each dot represents a sample, colored by time point (red=baseline, blue=post-CBT) and sized by Bacteroides abundance. Arrows connect samples from the same participants, with post-treatment indicated by the arrowhead. P values calculated by Adonis, adjusting for participant. C Microbial genera with statistically significant association with CBT responder status (q<.05) are shown. The y axis shows the log2 of the fold change between responders vs. non-responders. Dot size is proportional to mean relative abundance across all samples

References

    1. Hellstrom PM, Benno P. The Rome IV: irritable bowel syndrome - a functional disorder. Best Pract Res Clin Gastroenterol. 2019;40-41:101634. doi: 10.1016/j.bpg.2019.101634.
    1. Palsson OS, Whitehead WE, van Tilburg MA, Chang L, Chey W, Crowell MD, Keefer L, Lembo AJ, Parkman HP, Rao SS, et al. Rome IV diagnostic questionnaires and tables for investigators and clinicians. Gastroenterology. 2016;150(6):1481–1491. doi: 10.1053/j.gastro.2016.02.014.
    1. American College of Gastroenterology Task Force on Irritable Bowel S, Brandt LJ, Chey WD, Foxx-Orenstein AE, Schiller LR, Schoenfeld PS, Spiegel BM, Talley NJ, Quigley EM: An evidence-based position statement on the management of irritable bowel syndrome. Am J Gastroenterol 2009, 104 Suppl 1:S1-35.
    1. Francis CY, Morris J, Whorwell PJ. The irritable bowel severity scoring system: a simple method of monitoring irritable bowel syndrome and its progress. Aliment Pharmacol Ther. 1997;11(2):395–402. doi: 10.1046/j.1365-2036.1997.142318000.x.
    1. Spiegel B, Camilleri M, Bolus R, Andresen V, Chey WD, Fehnel S, Mangel A, Talley NJ, Whitehead WE: Psychometric evaluation of patient-reported outcomes in irritable bowel syndrome randomized controlled trials: a Rome Foundation report. Gastroenterology 2009, 137(6):1944-1953 e1941-1943.
    1. Mayer EA, Labus JS, Tillisch K, Cole SW, Baldi P. Towards a systems view of IBS. Nat Rev Gastroenterol Hepatol. 2015;12(10):592–605. doi: 10.1038/nrgastro.2015.121.
    1. Spohn SN, Mawe GM. Non-conventional features of peripheral serotonin signalling - the gut and beyond. Nat Rev Gastroenterol Hepatol. 2017;14(7):412–420. doi: 10.1038/nrgastro.2017.51.
    1. Mayer EA, Labus J, Aziz Q, Tracey I, Kilpatrick L, Elsenbruch S, Schweinhardt P, Van Oudenhove L, Borsook D. Role of brain imaging in disorders of brain-gut interaction: a Rome Working Team Report. Gut. 2019;68(9):1701–1715. doi: 10.1136/gutjnl-2019-318308.
    1. Simren M. Manipulating the gut microbiome as a treatment strategy for functional gastrointestinal disorders. Gastroenterology. 2018;155(4):960–962. doi: 10.1053/j.gastro.2018.09.008.
    1. Labus JS, Osadchiy V, Hsiao EY, Tap J, Derrien M, Gupta A, Tillisch K, Le Neve B, Grinsvall C, Ljungberg M, et al. Evidence for an association of gut microbial Clostridia with brain functional connectivity and gastrointestinal sensorimotor function in patients with irritable bowel syndrome, based on tripartite network analysis. Microbiome. 2019;7(1):45. doi: 10.1186/s40168-019-0656-z.
    1. Labus JS, Hollister EB, Jacobs J, Kirbach K, Oezguen N, Gupta A, Acosta J, Luna RA, Aagaard K, Versalovic J, Savidge T, Hsiao E, Tillisch K, Mayer EA. Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome. Microbiome. 2017;5(1):49. doi: 10.1186/s40168-017-0260-z.
    1. Van Oudenhove L, Crowell MD, Drossman DA, Halpert AD, Keefer L, Lackner JM, Murphy TB. Naliboff BD. Biopsychosocial aspects of functional gastrointestinal disorders. Gastroenterology: Levy RL; 2016.
    1. Ford AC, Lacy BE, Harris LA, Quigley EMM, Moayyedi P. Effect of antidepressants and psychological therapies in irritable bowel syndrome: an updated systematic review and meta-analysis. Am J Gastroenterol. 2019;114(1):21–39. doi: 10.1038/s41395-018-0222-5.
    1. Lackner JM, Jaccard J, Keefer L, Brenner DM, Firth RS, Gudleski GD, Hamilton FA, Katz LA, Krasner SS, Ma CX, Radziwon CD, Sitrin MD. Improvement in gastrointestinal symptoms after cognitive behavior therapy for refractory irritable bowel syndrome. Gastroenterology. 2018;155(1):47–57. doi: 10.1053/j.gastro.2018.03.063.
    1. Martin CR, Osadchiy V, Kalani A, Mayer EA. The brain-gut-microbiome axis. Cell Mol Gastroenterol Hepatol. 2018;6(2):133–148. doi: 10.1016/j.jcmgh.2018.04.003.
    1. Lackner JM, Keefer L, Jaccard J, Firth R, Brenner D, Bratten J, Dunlap LJ, Ma C, Byroads M, Group IR The Irritable Bowel Syndrome Outcome Study (IBSOS): rationale and design of a randomized, placebo-controlled trial with 12 month follow up of self- versus clinician-administered CBT for moderate to severe irritable bowel syndrome. Contemp Clin Trials. 2012;33(6):1293–1310. doi: 10.1016/j.cct.2012.07.013.
    1. Drossman DA. The functional gastrointestinal disorders and the Rome III process. Gastroenterology. 2006;130(5):1377–1390. doi: 10.1053/j.gastro.2006.03.008.
    1. Lackner JM, Jaccard J, Group IBSOSR Factors associated with efficacy of cognitive behavior therapy vs education for patients with irritable bowel syndrome. Clin Gastroenterol Hepatol. 2019;17(8):1500–1508. doi: 10.1016/j.cgh.2018.10.033.
    1. Lackner JM, Jaccard J, Radziwon CD, Firth RS, Gudleski GD, Hamilton F, Katz LA, Keefer L, Krasner SS, Ma CX, Sitrin MD, Brenner DM. Durability and decay of treatment benefit of cognitive behavioral therapy for irritable bowel syndrome: 12-month follow-up. Am J Gastroenterol. 2019;114(2):330–338. doi: 10.1038/s41395-018-0396-x.
    1. Krzywinski M, Schein J, Birol I, Connors J, Gascoyne R, Horsman D, Jones SJ, Marra MA. Circos: an information aesthetic for comparative genomics. Genome Res. 2009;19(9):1639–1645. doi: 10.1101/gr.092759.109.
    1. Ellingson BM, Mayer E, Harris RJ, Ashe-McNally C, Naliboff BD, Labus JS, Tillisch K. Diffusion tensor imaging detects microstructural reorganization in the brain associated with chronic irritable bowel syndrome. Pain. 2013;154(9):1528–1541. doi: 10.1016/j.pain.2013.04.010.
    1. Woodworth D, Mayer E, Leu K, Ashe-McNalley C, Naliboff BD, Labus JS, Tillisch K, Kutch JJ, Farmer MA, Apkarian AV, Johnson KA, Mackey SC, Ness TJ, Landis JR, Deutsch G, Harris RE, Clauw DJ, Mullins C, Ellingson BM, MAPP Research Network Unique Microstructural Changes in the Brain Associated with Urological Chronic Pelvic Pain Syndrome (UCPPS) revealed by diffusion tensor mri, super-resolution track density imaging, and statistical parameter mapping: a MAPP Network Neuroimaging Study. PLoS One. 2015;10(10):e0140250. doi: 10.1371/journal.pone.0140250.
    1. Woodworth DC, Dagher A, Curatolo A, Sachdev M, Ashe-McNalley C, Naliboff BD, Labus JS, Landis JR, Kutch JJ, Mayer EA, Lee RS, Moses MA, Ellingson BM, the MAPP Research Network Changes in brain white matter structure are associated with urine proteins in urologic chronic pelvic pain syndrome (UCPPS): A MAPP Network study. PLoS One. 2018;13(12):e0206807. doi: 10.1371/journal.pone.0206807.
    1. Tong M, Jacobs JP, McHardy IH, Braun J: Sampling of intestinal microbiota and targeted amplification of bacterial 16S rRNA genes for microbial ecologic analysis. Curr Protoc Immunol 2014, 107:7 41 41-47 41 11.
    1. Jacobs JP, Goudarzi M, Singh N, Tong M, McHardy IH, Ruegger P, Asadourian M, Moon BH, Ayson A, Borneman J, et al. A disease-associated microbial and metabolomics state in relatives of pediatric inflammatory bowel disease patients. Cell Mol Gastroenterol Hepatol. 2016;2(6):750–766. doi: 10.1016/j.jcmgh.2016.06.004.
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, Fierer N, Pena AG, Goodrich JK, Gordon JI, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7(5):335–336. doi: 10.1038/nmeth.f.303.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol. 2005;71(12):8228–8235. doi: 10.1128/AEM.71.12.8228-8235.2005.
    1. Anderson MJ. A new method for non-parametric multivariate analysis of variance. Austral Ecology. 2001;26(1):32–46. doi: 10.1111/j.1442-9993.2001.01070.pp.x.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi: 10.1186/s13059-014-0550-8.
    1. Smyth GK: Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 2004, 3:Article3.
    1. Breiman L. Machine Learning. 2001;45(1):5–32. doi: 10.1023/A:1010933404324.
    1. Labus JS, Gupta A, Coveleskie K, Tillisch K, Kilpatrick L, Jarcho J, Feier N, Bueller J, Stains J, Smith S, Suyenobu B, Naliboff B, Mayer EA. Sex differences in emotion-related cognitive processes in irritable bowel syndrome and healthy control subjects. Pain. 2013;154(10):2088–2099. doi: 10.1016/j.pain.2013.06.024.
    1. Berman S, Suyenobu B, Naliboff BD, Bueller J, Stains J, Wong H, Mandelkern M, Fitzgerald L, Ohning G, Gupta A, Labus JS, Tillisch K, Mayer EA. Evidence for alterations in central noradrenergic signaling in irritable bowel syndrome. Neuroimage. 2012;63(4):1854–1863. doi: 10.1016/j.neuroimage.2012.08.028.
    1. Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161(2):264–276. doi: 10.1016/j.cell.2015.02.047.
    1. Karlsson S, Banhidi ZG, Albertsson AC. Detection by high-performance liquid chromatography of polyamines formed by clostridial putrefaction of caseins. J Chromatogr. 1988;442:267–277. doi: 10.1016/S0021-9673(00)94474-1.
    1. Labus JS, Mayer EA, Jarcho J, Kilpatrick LA, Kilkens TO. Evers EA, Backes WH, Brummer RJ, van Nieuwenhoven MA. Acute tryptophan depletion alters the effective connectivity of emotional arousal circuitry during visceral stimuli in healthy women. Gut. 2011;60(9):1196–1203. doi: 10.1136/gut.2010.213447.
    1. Fung TC, Vuong HE, Luna CDG, Pronovost GN, Aleksandrova AA, Riley NG, Vavilina A, McGinn J, Rendon T, Forrest LR, Hsiao EY. Intestinal serotonin and fluoxetine exposure modulate bacterial colonization in the gut. Nat Microbiol. 2019;4(12):2064–2073. doi: 10.1038/s41564-019-0540-4.
    1. Shpaner M, Kelly C, Lieberman G, Perelman H, Davis M, Keefe FJ, Naylor MR. Unlearning chronic pain: a randomized controlled trial to investigate changes in intrinsic brain connectivity following cognitive behavioral therapy. Neuroimage Clin. 2014;5:365–376. doi: 10.1016/j.nicl.2014.07.008.

Source: PubMed

3
購読する