Role of brain imaging in disorders of brain-gut interaction: a Rome Working Team Report

Emeran A Mayer, Jennifer Labus, Qasim Aziz, Irene Tracey, Lisa Kilpatrick, Sigrid Elsenbruch, Petra Schweinhardt, Lukas Van Oudenhove, David Borsook, Emeran A Mayer, Jennifer Labus, Qasim Aziz, Irene Tracey, Lisa Kilpatrick, Sigrid Elsenbruch, Petra Schweinhardt, Lukas Van Oudenhove, David Borsook

Abstract

Imaging of the living human brain is a powerful tool to probe the interactions between brain, gut and microbiome in health and in disorders of brain-gut interactions, in particular IBS. While altered signals from the viscera contribute to clinical symptoms, the brain integrates these interoceptive signals with emotional, cognitive and memory related inputs in a non-linear fashion to produce symptoms. Tremendous progress has occurred in the development of new imaging techniques that look at structural, functional and metabolic properties of brain regions and networks. Standardisation in image acquisition and advances in computational approaches has made it possible to study large data sets of imaging studies, identify network properties and integrate them with non-imaging data. These approaches are beginning to generate brain signatures in IBS that share some features with those obtained in other often overlapping chronic pain disorders such as urological pelvic pain syndromes and vulvodynia, suggesting shared mechanisms. Despite this progress, the identification of preclinical vulnerability factors and outcome predictors has been slow. To overcome current obstacles, the creation of consortia and the generation of standardised multisite repositories for brain imaging and metadata from multisite studies are required.

Keywords: brain/gut interaction; functional bowel disorder; irritable bowel syndrome; magnetic resonance imaging.

Conflict of interest statement

Competing interests: EAM is on the scientific advisory boards of Axial Biotherapeutics, Bloom Science, Danone, Viome, Whole Biome and Mahana Therapeutics.

© Author(s) (or their employer(s)) 2019. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Proposed integrative model for disorders of gut–brain Interactions. Replacing the conventional focus on individual brain regions and cell types in the gut, this integrative model posits reciprocal interactions between brain networks (brain connectome) and networks made up of multiple cells in the gut, including the gut microbiota (gut connectome). Gut-to-brain communication is mediated by neural, endocrine and inflammatory pathways, while brain-to-gut communication relies mainly on autonomic nervous system output to the gut. Modified with permission from Enck et al.
Figure 2
Figure 2
Brain networks involved in centrai processing and modulation of visceral pain. Shown are the default mode network (DMN) and four task-related brain networks that have been described in the literature, for which structural and functional alterations and correlations with clinical and behavioural measures have been reported in IBS subjects. Correlations of the listed clinical and behavioural measures have been reported for the salience network, sensorimotor network, emotional arousal network, central executive network, central autonomic network and DMN. Arrows indicate: (A) shift of activity from the DMN to the task-related networks in response to input from the salience network; (B) switching between DMN and central executive network depending on input from the salience network; (C) engagement of emotional arousal network in response to central executive network activation; (D) engagement of central autonomic network in response to emotional arousal network activation; (E) central autonomic network activation with output in the form of descending pain modulation and autonomic nervous system activity to GI tract; (F) ascending viscerosensory signals from gut to sensorimotor network; and (G) assessment of information from sensorimotor network by salience network. The functions of these networks are described in detail in the text. Modified with permission from Mayer et al.
Figure 3
Figure 3
Effect of the HTR3A polymorphism c. −42C>T on amygdala reactivity to emotional and non-emotional stimuli. C/C genotype subjects displayed greater amygdala responses during an emotion matching and form matching task, suggesting a role of this gene polymorphism in influencing the emotional response to different laboratory tasks. With permission from Kilpatrick et al.
Figure 4
Figure 4
Reduced neurokinin-1 receptor binding in IBD. Whole-brain voxel-wise statistical parametric mapping analysis shows regions with lower levels of neurokinin-1 receptor binding in several brain regions in subjects with IBD (A) and patients with IBS (B), relative to healthy controls (voxel extent threshold p20). With permission from Jarcho et al.
Figure 5
Figure 5
Schematic of workflow from multimodal brain image acquisition to multiomics integration of brain and metadata. Acquisition of structural, anatomical (DTI), functional (resting state oscillations) and metabolic (MR spectroscopy, not shown) is followed by image processing and parcellation into multiple regions of interest (ROIs). These parcellated data undergo multiomics integration of different image modalities and clinical, behavioural and non-brain metadata using machine learning approaches. Such data-driven analysis approaches are expected to reveal distinct patters of brain-gut interactions. DTI, diffusion tensor imaging.
Figure 6
Figure 6
Effect of a CRF-R1 antagonist on amygdala response and emotional arousal circuit. (A). Error plot showing standard mean errors for beta contrasts (threat – safe) following placebo (PLA) versus a 20 mg or a 200 mg dose of the CRF-R1 antagonist GW876008 for the left locus coeruleus complex in patients with IBS and healthy controls (HCs) during an experimental pain threat. Results show a dose-dependent reduction in the threat-induced amygdala response by the CRF-R1 antagonist. (B). Path coefficients for the effective connectivity analysis of an emotional-arousal circuit during a pain threat following placebo versus high dose of the CRF-R1 antagonist (200 mg GW876008) In healthy controls and IBS subjects. Significantly different parameter estimates are shown by green arrows, while those not significantly different are shown in black. With permission from Hubbard et al. alNS, anterior insula; aMCC, anterior midclngulate cortex; AMYG, amygdala; HPC, hippocampus; HT, hypothalamus; LCC, locus coeruleus complex; OFC, orbitomedial prefrontal cortex; sgACC, subgenual anterior cingulate cortex.

References

    1. Aziz Q, Thompson DG. Brain-gut axis in health and disease. Gastroenterology 1998;114:559–78.
    1. Mayer EA, Gupta A, Kilpatrick LA, et al. Imaging brain mechanisms in chronic visceral pain. Pain 2015; 156:S50–S63.
    1. Drossman DA, Hasler WL. Rome IV-Functional GI Disorders: Disorders of Gut-Brain Interaction. Gastroenterology 2016;150:1257–61.
    1. Tillisch K, Labus JS. Advances in imaging the brain-gut axis: functional gastrointestinal disorders. Gastroenterology 2011;140:407–11.
    1. Van Oudenhove L, Aziz Q. Recent insights on central processing and psychological processes in functional gastrointestinal disorders. Dig Liver Dis 2009;41:781–7.
    1. Drossman DA. Functional Gastrointestinal Disorders: History, Pathophysiology, Clinical Features, and Rome IV. Gastroenterology 2016;150:1262–79.
    1. Mayer EA, Bushnell MC. Functional Pain Disorders: Time for a Paradigm Shift? In: Mayer EA, Bushnell MC, eds. Functional Pain Syndromes: Presentation and Pathophysiology. Seattle: IASP Press, 2009:531–65.
    1. Jones MP, Tack J, Van Oudenhove L, et al. Mood and Anxiety Disorders Precede Development of Functional Gastrointestinal Disorders in Patients but Not in the Population. Clin Gastroenterol Hepatol 2017;15:1014–20.
    1. Mayer EA, Labus JS, Tillisch K, et al. Towards a systems view of IBS. Nat Rev Gastroenterol Hepatol 2015;12:592–605.
    1. Tracey I, Mantyh PW. The cerebral signature for pain perception and its modulation. Neuron 2007;55:377–91.
    1. Derbyshire SWG. A systematic review of neuroimaging data during visceral stimulation. Am J Gastroenterol 2003;98:12–20.
    1. Tillisch K, Mayer EA, Labus JS. Quantitative meta-analysis identifies brain regions activated during rectal distension in irritable bowel syndrome. Gastroenterology 2011;140:91–100.
    1. Al Omran Y, Aziz Q. Functional brain imaging in gastroenterology: to new beginnings. Nat Rev Gastroenterol Hepatol 2014; 11:565–76.
    1. Enck P, Aziz Q, Barbara G, et al. Irritable bowel syndrome. Nat Rev Dis Primers 2016;2:16014.
    1. Hobson AR, Aziz Q. Brain imaging and functional gastrointestinal disorders: has it helped our understanding? Gut 2004;53:1198–206.
    1. Aizawa E, Sato Y, Kochiyama T, et al. Altered cognitive function of prefrontal cortex during error feedback in patients with irritable bowel syndrome, based on FMRI and dynamic causal modeling. Gastroenterology 2012;143:1188–98.
    1. Bajaj S, Adhikari BM, Dhamala M. Higher frequency network activity flow predicts lower frequency node activity in intrinsic low-frequency BOLD fluctuations. PLoS One 2013;8:e64466.
    1. Bullmore E, Sporns O. The economy of brain network organization. Nat Rev Neurosci 2012;13:336–49.
    1. Greicius MD, Supekar K, Menon V, et al. Resting-state functional connectivity reflects structural connectivity in the default mode network. Cereb Cortex 2009;19:72–8.
    1. Guo CC, Kurth F, Zhou J, et al. One-year test-retest reliability of intrinsic connectivity network fMRI in older adults. Neuroimage 2012;61:1471–83.
    1. Hutchison RM, Womelsdorf T, Gati JS, et al. Resting-state connectivity identifies distinct functional networks in macaque cingulate cortex. Cereb Cortex 2012;22:1294–308.
    1. Rubinov M, Sporns O. Complex network measures of brain connectivity: uses and interpretations. Neuroimage 2010;52:1059–69.
    1. Seeley WW, Menon V, Schatzberg AF, et al. Dissociable intrinsic connectivity networks for salience processing and executive control. J Neurosci 2007;27:2349–56.
    1. Sporns O Networks of the Brain. Cambridge, Massachussets: The MIT Press, 2010.
    1. Bischoff SC, Barbara G, Buurman W, et al. Intestinal permeability – a new target for disease prevention and therapy. BMC Gastroenterol 2014;14:189.
    1. Drossman DA. Psychosocial and psychophysiologic mechanisms in GI illness Kirsner JB, ed. The Growth of Gastroenterologic knowledge in the 20th Century. Philadelphia: PA: Lea & Febiger, 1994:419–32.
    1. Kennedy PJ, Clarke G, O’Neill A, O’Neill A, et al. Cognitive performance in irritable bowel syndrome: evidence of a stress-related impairment in visuospatial memory. Psychol Med 2014;44:1553–66.
    1. Mayer EA, Savidge T, Shulman RJ. Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology 2014;146:1500–12.
    1. Piché M, Arsenault M, Poitras P, et al. Widespread hypersensitivity is related to altered pain inhibition processes in irritable bowel syndrome. Pain 2010;148:49–58.
    1. Wilder-Smith CH. The balancing act: endogenous modulation of pain in functional gastrointestinal disorders. Gut 2011; 60:1589–99.
    1. Tijms BM, Seriès P, Willshaw DJ, et al. Similarity-based extraction of individual networks from gray matter MRI scans. Cereb Cortex 2012;22:1530–41.
    1. Barboza JL, Talley NJ, Moshiree B. Current and emerging pharmacotherapeutic options for irritable bowel syndrome. Drugs 2014;74:1849–70.
    1. Hagmann P, Kurant M, Gigandet X, et al. Mapping human whole-brain structural networks with diffusion MRI. PLoS One 2007;2:e597.
    1. Buckner RL, Andrews-Hanna JR, Schacter DL. The brain’s default network: anatomy, function, and relevance to disease. Ann NY Acad Sci 2008; 1124:1–38.
    1. Greicius MD, Krasnow B, Reiss AL, et al. Functional connectivity in the resting brain: a network analysis of the default mode hypothesis. Proc Natl Acad Sci U SA 2003;100:253–8.
    1. Mawe GM, Hoffman JM. Serotonin signalling in the gut—functions, dysfunctions and therapeutic targets. Nat Rev Gastroenterol Hepatol 2013; 10:473–86.
    1. Bohórguez DV, Liddle RA. The gut connectome: making sense of what you eat. J Clin Invest 2015; 125:888–90.
    1. Farmer AD, Aziz Q, Tack J, et al. The future of neuroscientific research in functional gastrointestinal disorders: integration towards multidimensional (visceral) pain endophenotypes? J Psychosom Res 2010;68:475–81.
    1. Fukudo S Stress and visceral pain: focusing on irritable bowel syndrome. Pain 2013; 154(Suppl 1):S63–S70.
    1. Hong JY, Kilpatrick LA, Labus J, et al. Patients with chronic visceral pain show sex-related alterations in intrinsic oscillations of the resting brain. J Neurosci 2013;33:11994–2002.
    1. Kilpatrick LA, Ornitz E, Ibrahimovic H, et al. Sex-related differences in prepulse inhibition of startle in irritable bowel syndrome (IBS). Biol Psychol 2010;84:272–8.
    1. Van Oudenhove L, Vandenberghe J, Vos R, et al. Factors associated with co-morbid irritable bowel syndrome and chronic fatigue-like symptoms in functional dyspepsia. Neurogastroenterol Motil 2011; 23:524–e202.
    1. Gupta A, Kilpatrick L, Labus J, et al. Early adverse life events and resting state neural networks in patients with chronic abdominal pain: evidence for sex differences. Psychosom Med 2014;76:404–12.
    1. Naliboff BD, Berman S, Suyenobu B, et al. Longitudinal change in perceptual and brain activation response to visceral stimuli in irritable bowel syndrome patients. Gastroenterology 2006;131:352–65.
    1. Gupta A, Labus J, Kilpatrick LA, et al. Interactions of early adversity with stress-related gene polymorphisms impact regional brain structure in females. Brain Struct Puna 2016;221:1667–79.
    1. Orand A, Gupta A, Shih W, et al. Catecholaminergic Gene Polymorphisms Are Associated with GI Symptoms and Morphological Brain Changes in Irritable Bowel Syndrome. PLoS One 2015;10:e0135910.
    1. Berman S, Suyenobu B, Naliboff BD, et al. Evidence for alterations in central noradrenergic signaling in irritable bowel syndrome. Neuroimage 2012;63:1854–63.
    1. Gupta A, Mayer EA, Bonyadi M, et al. NR3C1 and IL-1 β Polymorphisms Interact with Early Adverse Life Events in influencing Gray Matter Variations in Women with and without chronic abdominal pain. Society for Neuroscience. San Diego, California, USA 2013.
    1. Kilpatrick LA, Labus JS, Coveleskie K, et al. The HTR3A polymorphism c. −42C>T is associated with amygdala responsiveness in patients with irritable bowel syndrome. Gastroenterology 2011; 140:1943–51.
    1. Gupta A, Cole S, Labus JS, et al. Gene expression profiles in peripheral blood mononuclear cells correlate with salience network activity in chronic visceral pain: A pilot study. Neurogastroenterol Motil 2017;29:e13027.
    1. Labus JS, Oezguen N, Hollister EB, et al. 752 Regional Brain Morphology Is Associated With Gut Microbial Metabolites in Irritable Bowel Syndrome (IBS). Gastroenterology 2015;148:S-142–S-142.
    1. Clayton JA. Studying both sexes: a guiding principle for biomedicine. Faseb J 2016;30:519–24.
    1. Kilpatrick L, Tillisch K. Irritable Bowel Syndrome, 2012.
    1. Gupta A, Mayer EA, Fling C, et al. Sex-based differences in brain alterations across chronic pain conditions. J Neurosci Res 2017;95:604–16.
    1. Vincent K, Tracey I. Sex hormones and pain: the evidence from functional imaging. CurrPain Headache Rep 2010;14:396–403.
    1. Vincent K, Warnaby C, Stagg CJ, et al. Brain imaging reveals that engagement of descending inhibitory pain pathways in healthy women in a low endogenous estradiol state varies with testosterone. Pain 2013; 154:515–24.
    1. Melchior M, Poisbeau P, Gaumond I, et al. Insights into the mechanisms and the emergence of sex-differences in pain. Neuroscience 2016;338:63–80.
    1. Choi JC, Park SK, Kim YH, et al. Different brain activation patterns to pain and pain-related unpleasantness during the menstrual cycle. Anesthesiology 2006; 105:120–7.
    1. Hg L, Dupont P, Geeraerts B, et al. Lack of endogenous opioid release during sustained visceral pain: a [11C]carfentanil PET study. Pain 2013;154:2072–7.
    1. Jarcho JM, Feier NA, Bert A, et al. Diminished neurokinin-1 receptor availability in patients with two forms of chronic visceral pain. Pain 2013;154:987–96.
    1. Hg L, Ceccarini J, Weltens N, et al. Increased cerebral cannabinoid-1 receptor availability is a stable feature of functional dyspepsia: a [FJMK-9470 PET study. Psychother Psychosom 2015;84:149–58.
    1. Tominaga K, Tsumoto C, Ataka S, et al. Regional brain disorders of serotonin neurotransmission are associated with functional dyspepsia. Life Sci 2015;137:150–7.
    1. Ende G Proton Magnetic Resonance Spectroscopy: Relevance of Glutamate and GABA to Neuropsychology. Neuropsychol Rev 2015;25:315–25.
    1. Niddam DM, Tsai SY, Lu CL, et al. Reduced hippocampal glutamate-glutamine levels in irritable bowel syndrome: preliminary findings using magnetic resonance spectroscopy. Am J Gastroenterol 2011; 106:1503–11.
    1. Labus JS, Dinov ID, Jiang Z, et al. Irritable bowel syndrome in female patients is associated with alterations in structural brain networks. Pain 2014;155:137–49.
    1. Labus JS, Hamadani K, Gupta A, et al. Su1571 Functional Network Properties of Brain Regions in Irritable Bowel Syndrome. Gastroenterology 2016;150:S529–529.
    1. Labus JS, Van Horn JD, Torgerson C, et al. 585 Architecture of Anatomical Brain Networks Differs in Irritable Bowel Syndrome Compared to Healthy Controls. Gastroenterology 2014; 146:S–109.
    1. Bullmore E, Sporns O. Complex brain networks: graph theoretical analysis of structural and functional systems. Nat Rev Neurosci 2009;10:186–98.
    1. Sporns O. Structure and function of complex brain networks. Dialogues Clin Neurosci 2013;15:247–62.
    1. Sporns O Network attributes for segregation and integration in the human brain. Curr Opin Neurobiol 2013;23:162–71.
    1. Labus JS, Naliboff B, Kilpatrick L, et al. Pain and Interoception Imaging Network (PAIN): A multimodal multisite, brain-imaging repository for chronic somatic and visceral pain disorders. Neuroimage 2016;124:1232–7.
    1. Dinov ID, Petrosyan P, Liu Z, et al. The perfect neuroimaging-genetics-computation storm: collision of petabytes of data, millions of hardware devices and thousands of software tools. Brain Imaging Behav 2014;8:311–22.
    1. Glasser MF, Sotiropoulos SN, Wilson JA, et al. The minimal preprocessing pipelines for the Human Connectome Project. Neuroimage 2013;80:105–24.
    1. Secretary TWHOotP. Fact Sheet: BRAIN Initiative, 2013.
    1. Markram H Human Brain Project: Henry Markram plans to spend €1 bn building a perfect model of the human brain. Neuroscience Observer. The Guardian 2013.
    1. Van Essen DC, Smith SM, Barch DM, et al. The WU-Minn Human Connectome Project: an overview. Neuroimage 2013;80:62–79.
    1. Allen N, Sudlow C, Downey P, et al. UK Biobank: Current status and what it means for epidemiology. Health Policy Technol 2012;1:123–6.
    1. Guerreiro R, Wojtas A, Bras J, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med 2013;368:117–27.
    1. Swarup V, Geschwind DH. Alzheimer’s disease: From big data to mechanism. Nature 2013;500:34–5.
    1. Nalls MA, McLean CY, Rick J, et al. Diagnosis of Parkinson’s disease on the basis of clinical and genetic classification: a population-based modelling study. Lancet Neurol 2015;14:1002–9.
    1. Baliki MN, Petre B, Torbey S, et al. Corticostriatal functional connectivity predicts transition to chronic back pain. Nat Neurosci 2012; 15:1117–9.
    1. Mansour AR, Baliki MN, Huang L, et al. Brain white matter structural properties predict transition to chronic pain. Pain 2013;154:2160–8.
    1. Vachon-Presseau E, Tétreault P, Petre B, et al. Corticolimbic anatomical characteristics predetermine risk for chronic pain. Brain 2016;139:1958–70.
    1. Denk F, McMahon SB, Tracey I. Pain vulnerability: a neurobiological perspective. Nat Neurosci 2014;17:192–200.
    1. Blankstein U, Chen J, Diamant NE, et al. Altered Brain Structure in Irritable Bowel Syndrome: Potential Contributions of Pre-Existing and Disease-Driven Factors. Gastroenterology 2010; 138:1783–9.
    1. Chen JY, Blankstein U, Diamant NE, et al. White matter abnormalities in imitable bowel syndrome and relation to individual factors. Brain Res 2011;1392:121–31.
    1. Erpelding N, Moayedi M, Davis KD. Cortical thickness correlates of pain and temperature sensitivity. Pain 2012;153:1602–9.
    1. Navratilova E, Xie JY, Meske D, et al. Endogenous opioid activity in the anterior cingulate cortex is required for relief of pain. J Neurosci 2015;35:7264–71.
    1. Wanigasekera V, Lee MC, Rogers R, et al. Baseline reward circuitry activity and trait reward responsiveness predict expression of opioid analgesia in healthy subjects. Proc Natl Acad Sci U S A 2012; 109:17705–10.
    1. Tracey I A vulnerability to chronic pain and its interrelationship with resistance to analgesia. Brain 2016;139:1869–72.
    1. Hanis RE, Napadow V, Huggins JP, et al. Pregabalin rectifies aberrant brain chemistry, connectivity, and functional response in chronic pain patients. Anesthesiology 2013;119:1453–64.
    1. Borsook D, Hargreaves R, Bountra C, et al. Lost but making progress-Where will new analgesic drugs come from? Sci Transi Med 2014;249:sr3.
    1. Camilleri M Review article: biomarkers and personalised therapy in functional lower gastrointestinal disorders. Aliment Pharmacol Ther 2015;42:818–28.
    1. Erpelding N, Simons L, Lebel A, et al. Rapid treatment-induced brain changes in pediatric CRPS. Brain Structure and Function 2016;221:1095–111.
    1. Becerra L, Sava S, Simons LE, et al. Intrinsic brain networks normalize with treatment in pediatric complex regional pain syndrome. Neuroimage 2014;6:347–69.
    1. Gupta A, Kilpatrick L, Labus J, et al. Early Adverse Life Events and Resting State Neural Networks in Patients With Chronic Abdominal Pain. Psychosom Med 2014;76:404–12.
    1. Hong J-Y, Kilpatrick LA, Labus JS, et al. Sex and Disease-Related Alterations of Anterior Insula Functional Connectivity in Chronic Abdominal Pain. Journal of Neuroscience 2014;34:14252–9.
    1. Hubbard CS, Becerra L, Heinz N, et al. Abdominal Pain, the Adolescent and Altered Brain Structure and Function. PLoS One 2016; 11 :e0156545.
    1. Piché M, Chen JI, Roy M, et al. Thicker posterior insula is associated with disease duration in women with imitable bowel syndrome (IBS) whereas thicker orbitofrontal cortex predicts reduced pain inhibition in both IBS patients and controls. J Pain 2013;14:1217–26.
    1. Labus JS, Van Horn JD, Gupta A, et al. Multivariate morphological brain signatures predict patients with chronic abdominal pain from healthy control subjects. Pain 2015;156:1545–54.
    1. Bourke JH, Wall MB. phMRI: methodological considerations for mitigating potential confounding factors. Front Neurosci 2015;9:167.
    1. Jenkins BG. Pharmacologic magnetic resonance imaging (phMRI): imaging drug action in the brain. Neuroimage 2012;62:1072–85.
    1. Jacobs BL, Azmitia EC. Structure and function of the brain serotonin system. Physiol Rev 1992;72:165–229.
    1. Hubbard CS, Labus JS, Bueller J, et al. Corticotropin-releasing factor receptor 1 antagonist alters regional activation and effective connectivity in an emotional-arousal circuit during expectation of abdominal pain. J Neurosci 2011;31:12491–500.
    1. Labus JS, Hubbard CS, Bueller J, et al. Impaired emotional learning and involvement of the corticotropin-releasing factor signaling system in patients with imitable bowel syndrome. Gastroenterology 2013; 145:1253–61.
    1. Berman SM, Chang L, Suyenobu B, et al. Condition-specific deactivation of brain regions by 5-HT3 receptor antagonist Alosetron. Gastroenterology 2002;123:969–77.
    1. Mayer EA, Berman S, Derbyshire SW, et al. The effect of the 5-HT3 receptor antagonist, alosetron, on brain responses to visceral stimulation in irritable bowel syndrome patients. Aliment Pharmacol Ther 2002; 16:1357–66.
    1. Tillisch K, Labus J, Nam B, et al. Neurokinin-1-receptor antagonism decreases anxiety and emotional arousal circuit response to noxious visceral distension in women with imitable bowel syndrome: a pilot study. Aliment Pharmacol Ther 2012;35:360–7.
    1. Arrubla J, Tse DH, Amkreutz C, et al. GABA concentration in posterior cingulate cortex predicts putamen response during resting state fMRI. PLoS One 2014;9:e106609.
    1. Cole DM, Beckmann CF, Oei NY, et al. Differential and distributed effects of dopamine neuromodulations on resting-state network connectivity. Neuroimage 2013;78:59–67.
    1. Cole DM, Beckmann CF, Searle GE, et al. Orbitofrontal connectivity with resting-state networks is associated with midbrain dopamine D3 receptor availability. Cereb Cortex 2012;22:2784–93.
    1. Cole DM, Oei NY, Soeter RP, et al. Dopamine-dependent architecture of cortico-subcortical network connectivity. Cereb Cortex 2013;23:1509–16.
    1. van den Heuvel MP, Mandl RC, Kahn RS, et al. Functionally linked resting-state networks reflect the underlying structural connectivity architecture of the human brain. Hum Brain Mapp 2009;30:3127–41.
    1. Borsook D, Erpelding N, Becerra L. Losses and gains: chronic pain and altered brain morphology. Expert Rev Neurother 2013; 13:1221–34.
    1. Castrén E, Hen R. Neuronal plasticity and antidepressant actions. Trends Neurosci 2013;36:259–67.
    1. Castrén E Neuronal network plasticity and recovery from depression. JAMA Psychiatry 2013;70:983–9.
    1. Lackner JM, Lou Coad M, Mertz HR, et al. Cognitive therapy for irritable bowel syndrome is associated with reduced limbic activity, GI symptoms, and anxiety. Behav Res Ther 2006;44:621–38.
    1. Zhu Y, Wu Z, Ma X, et al. Brain regions involved in moxibustion-induced analgesia in imitable bowel syndrome with diarrhea: a functional magnetic resonance imaging study. BMC Complement Altern Med 2014;14:500.
    1. Flor H Psychological pain interventions and neurophysiology: implications for a mechanism-based approach. Am Psychol 2014;69:188–96.
    1. Jensen KB, Berna C, Loggia ML, et al. The use of functional neuroimaging to evaluate psychological and other non-pharmacological treatments for clinical pain. Neurosci Lett 2012;520:156–64.
    1. Harris RE, Zubieta J-K, Scott DJ, et al. Traditional Chinese acupuncture and placebo (sham) acupuncture are differentiated by their effects on μ-opioid receptors (MORs). Neuroimage 2009;47:1077–85.
    1. Egorova N, Gollub RL, Kong J. Repeated verum but not placebo acupuncture normalizes connectivity in brain regions dysregulated in chronic pain. Neuroimage Clin 2015;9:430–5.
    1. Dhond RP, Kettner N, Napadow V. Neuroimaging Acupuncture Effects in the Human Brain. The Journal of Alternative and Complementary Medicine 2007;13:603–16.
    1. Vanhaudenhuyse A, Laureys S, Faymonville M-E. Neurophysiology of hypnosis. Neurophysiologie Clinique/Clinical Neurophysiology 2014;44:343–53.
    1. Del Casale A, Ferracuti S, Rapinesi al. Pain perception and hypnosis: findings from recent functional neuroimaging studies. Int J Clin Exp Hypn 2015;63:144–70.
    1. Gaylord SA, Palsson OS, Garland EL, et al. Mindfulness training reduces the severity of irritable bowel syndrome in women: results of a randomized controlled trial. Am J Gastroenterol 2011;106:1678–88.
    1. Ljótsson B, Falk L, Vesterlund AW, et al. Internet-delivered exposure and mindfulness based therapy for irritable bowel syndrome-a randomized controlled trial. Behav Res 7fter 2010;48:531–9.
    1. Larsson MBO, Tillisch K, Craig AD, et al. Brain Responses to Visceral Stimuli Reflect Visceral Sensitivity Thresholds in Patients With Irritable Bowel Syndrome. Gastroenterology 2012; 142:463–72.
    1. Van Oudenhove L, Vandenberghe J, Dupont P, et al. Regional brain activity in functional dyspepsia: a H(2)(15)0-PET study on the role of gastric sensitivity and abuse history. Gastroenterology 2010;139:36–47.
    1. Elsenbruch S, Rosenberger C, Enck P, et al. Affective disturbances modulate the neural processing of visceral pain stimuli in irritable bowel syndrome: an fMRI study. Gut 2010;59:489–95.
    1. Tillisch K, Labus J, Kilpatrick L, et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 2013;144:1394–401.
    1. Tillisch K, Mayer EA, Gupta A, et al. Brain Structure and Response to Emotional Stimuli as Related to Gut Microbial Profiles in Healthy Women. Psychosom Med 2017;79:905–13.
    1. Haroon E, Fleischer CC, Felger JC, et al. Conceptual convergence: increased inflammation is associated with increased basal ganglia glutamate in patients with major depression. Mol Psychiatry 2016.
    1. Čeko M, Shir Y, Ouellet JA, et al. Partial recovery of abnormal insula and dorsolateral prefrontal connectivity to cognitive networks in chronic low back pain after treatment. Hum Brain Mapp 2015;36:2075–92.
    1. Bräscher AK, Becker S, Hoeppli ME, et al. Different Brain Circuitries Mediating Controllable and Uncontrollable Pain. J Neurosci 2016;36:5013–25.
    1. Keay KA, Bandler R. Distinct central representations of inescapable and escapable pain: observations and speculation. Exp Physiol 2002;87:275–9.
    1. Brown GK, Nicassio PM. Development of a questionnaire for the assessment of active and passive coping strategies in chronic pain patients. Pain 1987;31:53–64.
    1. Snow-Turek AL, Norris MP, Tan G. Active and passive coping strategies in chronic pain patients. Pain 1996;64:455–62.
    1. Akassoglou K, Merlini M, Rafalski VA, et al. In Vivo Imaging of CNS Injury and Disease. J Neurosci 2017;37:10808–16.
    1. Greenwood-Van Meerveld B, Prusator DK, Johnson AC. Animal models of gastrointestinal and liver diseases. Animal models of visceral pain: pathophysiology, translational relevance, and challenges. Am J Physiol Gastrointest Liver Physiol 2015;308:G885–G903.
    1. Holschneider DP, Bradesi S, Mayer EA. The role of experimental models in developing new treatments for irritable bowel syndrome. Expert Rev Gastroenterol Hepatol 2011;5:43–57.
    1. Alger JR, Ellingson BM, Ashe-McNalley C, et al. Multisite, multimodal neuroimaging of chronic urological pelvic pain: Methodology of the MAPP Research Network. Neuroimage 2016;12:65–77.
    1. Landis JR, Williams DA, Lucia MS, et al. The MAPP research network: design, patient characterization and operations. BMC Urol 2014;14:58.
    1. Clemens JQ, Mullins C, Kusek JW, et al. The MAPP research network: a novel study of urologie chronic pelvic pain syndromes. BMC Urol 2014;14:57.
    1. Hong JY, Kilpatrick LA, Labus JS, et al. Sex and disease-related alterations of anterior insula functional connectivity in chronic abdominal pain. J Neurosci 2014;34:14252–9.
    1. Hong J-Y, Naliboff BD, Labus JS, et al. Sa2014 IBS Patients Show Altered Brain Responses During Uncertain, but Not Certain Expectation of Painful Stimulation of the Abdominal Wall. Gastroenterology 2015;148::S–384..
    1. Jiang Z, Dinov ID, Labus J, et al. Sex-related differences of cortical thickness in patients with chronic abdominal pain. PLoS One 2013;8:e73932.
    1. Qi R, Ke J, Schoepf UJ, et al. Topological Reorganization of the Default Mode Network in Irritable Bowel Syndrome. Mol Neurobiol 2016;53:6585–93.
    1. Icenhour A, Witt ST, Elsenbruch S, et al. Brain functional connectivity is associated with visceral sensitivity in women with Irritable Bowel Syndrome. Neuroimage 2017;15:449–57.
    1. Ellingson BM, Mayer E, Harris RJ, et al. Diffusion tensor imaging detects microstructural reorganization in the brain associated with chronic irritable bowel syndrome. Pain 2013;154:1528–41.
    1. Mayer EA, Aziz Q, Coen S, et al. Brain imaging approaches to the study of functional GI disorders: a Rome working team report. Neurogastroenterol Motil 2009;21:579–96.
    1. Hall GB, Kamath MV, Collins S, et al. Heightened central affective response to visceral sensations of pain and discomfort in IBS. Neurogastroenterol Motil 2010;22:276–e80.
    1. Elsenbruch S, Rosenberger C, Bingel U, et al. Patients with irritable bowel syndrome have altered emotional modulation of neural responses to visceral stimuli. Gastroenterology 2010;139:1310–9.
    1. Labus JS, Gupta A, Coveleskie K, et al. Sex differences in emotion-related cognitive processes in irritable bowel syndrome and healthy control subjects. Pain 2013;154:2088–99.
    1. Berman S, Munakata J, Naliboff BD, et al. Gender differences in regional brain response to visceral pressure in IBS patients. Eur J Pain 2000;4:157–72.
    1. Naliboff BD, Berman S, Chang L, et al. Sex-related differences in IBS patients: central processing of visceral stimuli. Gastroenterology 2003;124:1738–47.
    1. Labus JS, Mayer EA, Jarcho J, et al. Acute tryptophan depletion alters the effective connectivity of emotional arousal circuitry during visceral stimuli in healthy women. Gut 2011;60:1196–203.
    1. Straube T, Schmidt S, Weiss T, et al. Dynamic activation of the anterior cingulate cortex during anticipatory anxiety. Neuroimage 2009;44:975–81.
    1. Porro CA, Baraldi P, Pagnoni G, et al. Does anticipation of pain affect cortical nociceptive systems? 7 Neurosci 2002;22:3206–14.
    1. Koyama T, McHaffie JG, Laurienti PJ, et al. The subjective experience of pain: where expectations become reality. Proc Natl Acad Sci USA 2005; 102:12950–5.
    1. Bornhövd K, Quante M, Glauche V, et al. Painful stimuli evoke different stimulus-response functions in the amygdala, prefrontal insula and somatosensory cortex: a single-trial fMRI study. Brain 2002;125:1326–36.
    1. Verne GN, Himes NC, Robinson ME, et al. Central representation of visceral and cutaneous hypersensitivity in the irritable bowel syndrome. Pain 2003;103:99–110.
    1. Seifert F, Schuberth N, De Col R, et al. Brain activity during sympathetic response in anticipation and experience of pain. Hum Brain Mapp 2013;34:1768–82.
    1. Yágüez L, Coen S, Gregory U, et al. Brain response to visceral aversive conditioning: a functional magnetic resonance imaging study. Gastroenterology 2005;128:1819–29.
    1. Berman SM, Naliboff BD, Suyenobu B, et al. Reduced brainstem inhibition during anticipated pelvic visceral pain correlates with enhanced brain response to the visceral stimulus in women with irritable bowel syndrome. J Neurosci 2008;28:349–59.
    1. Stephan E, Pardo JV, Faris PL, et al. Functional neuroimaging of gastric distention. J Gastrointest Surg 2003;7:740–9.
    1. Lawal A, Kern M, Sanjeevi A, et al. Neurocognitive processing of esophageal central sensitization in the insula and cingulate gyrus. Am J Physiol Gastrointest Liver Physiol 2008;294:G787–G794.
    1. Labus JS, Naliboff BN, Fallon J, et al. Sex differences in brain activity during aversive visceral stimulation and its expectation in patients with chronic abdominal pain: a network analysis. Neuroimage 2008;41:1032–43.
    1. Rubio A, Van Oudenhove L, Pellissier S, et al. Uncertainty in anticipation of uncomfortable rectal distension is modulated by the autonomic nervous system - A fMRI study in healthy volunteers. Neuroimage 2014; 107010–22.
    1. Afzal M, Potokar JP, Probert CS, et al. Selective processing of gastrointestinal symptom-related stimuli in irritable bowel syndrome. Psychosom Med 2006;68:758–61.
    1. Gibbs-Gallagher N, Palsson OS, Levy RL, et al. Selective recall of gastrointestinalsensation words: evidence for a cognitive-behavioral contribution to irritable bowel syndrome. Am J Gastroenterol 2001; 96:1133–8.
    1. Phillips K, Wright BJ, Kent S. Irritable bowel syndrome and symptom severity: Evidence of negative attention bias, diminished vigour, and autonomic dysregulation. J Psychosom Res 2014;77:13–19.
    1. Tkalcic M, Domijan D, Pletikosic S, et al. Attentional biases in irritable bowel syndrome patients. Clin Res Hepatol Gastroenterol 2014;38:621–8.
    1. Labus JS, Naliboff BD, Berman SM, et al. Brain networks underlying perceptual habituation to repeated aversive visceral stimuli in patients with irritable bowel syndrome. Neuroimage 2009;47:952–60.
    1. Seminowicz DA, Shpaner M, Keaser ML, et al. Cognitive-behavioral therapy increases prefrontal cortex gray matter in patients with chronic pain. 7 Pain 2013;14:1573–84.
    1. Jeffery IB, O’Toole PW, Öhman L, et al. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut 2012;61:997–1006.
    1. Simrén M IBS with intestinal microbial dysbiosis: a new and clinically relevant subgroup? Gut 2014;63:1685–6.
    1. Baliki MN, Schnitzer TJ, Bauer WR, et al. Brain morphological signatures for chronic pain. PLoS One 2011;6:e26010.
    1. Klöppel S, Abdulkadir A, Jack CR, et al. Diagnostic neuroimaging across diseases. Neuroimage 2012;61:457–63.
    1. Rosa MJ, Seymour B. Decoding the matrix: benefits and limitations of applying machine learning algorithms to pain neuroimaging. Pain 2014;155:864–7.

Source: PubMed

3
購読する