Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Targeting the endocannabinoid system for the treatment of abdominal pain in irritable bowel syndrome

Abstract

The management of visceral pain in patients with disorders of gut–brain interaction, notably irritable bowel syndrome, presents a considerable clinical challenge, with few available treatment options. Patients are increasingly using cannabis and cannabinoids to control abdominal pain. Cannabis acts on receptors of the endocannabinoid system, an endogenous system of lipid mediators that regulates gastrointestinal function and pain processing pathways in health and disease. The endocannabinoid system represents a logical molecular therapeutic target for the treatment of pain in irritable bowel syndrome. Here, we review the physiological and pathophysiological functions of the endocannabinoid system with a focus on the peripheral and central regulation of gastrointestinal function and visceral nociception. We address the use of cannabinoids in pain management, comparing them to other treatment modalities, including opioids and neuromodulators. Finally, we discuss emerging therapeutic candidates targeting the endocannabinoid system for the treatment of pain in irritable bowel syndrome.

Key points

  • The management of abdominal pain in disorders of gut–brain interaction, including irritable bowel syndrome (IBS), is challenging.

  • Patients are increasingly using cannabis and cannabinoids as an alternative therapy to treat pain and altered bowel habits in IBS.

  • Cannabis acts on the cannabinoid receptors (CB1 and CB2) of the endocannabinoid system (ECS), which consists of ligands for these receptors, anandamide and 2-arachidonoylglycerol as well as the biosynthetic and degradative enzymes for these ligands.

  • The ECS is a logical molecular target for the treatment of IBS as it regulates gastrointestinal motility, secretion, barrier function, inflammation, gut microbiota and visceral sensitivity.

  • Cannabinoid therapeutics have been developed but are of limited use in managing pain in IBS; the effects of cannabis have not been rigorously examined in much-needed large clinical trials.

  • Evidence supports the putative analgesic properties of a peripherally restricted CB2 receptor agonist in IBS. Future therapies could target additional ECS components for the treatment of IBS and other disorders of gut–brain interaction.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: CB1 and CB2 expression in cell types throughout the gut–brain–microbiota axis.
Fig. 2: CB1 and CB2 structure, second messenger signalling mechanisms, and downstream targets within cells.
Fig. 3: Endocannabinoid system targets of visceral pain.
Fig. 4: Actions of the endocannabinoid system as they relate to the gastrointestinal tract, motility, gut microbiota, immune function and visceral pain.
Fig. 5: Potential therapeutic intervention strategies to modulate the function of CB1 and CB2.

Similar content being viewed by others

References

  1. Drossman, D. A. Functional gastrointestinal disorders: history, pathophysiology, clinical features and Rome IV. Gastroenterology 150, 1262–1279 (2016).

    Article  Google Scholar 

  2. Sperber, A. D. et al. Worldwide prevalence and burden of functional gastrointestinal disorders, results of Rome Foundation Global Study. Gastroenterology 160, 99–114 (2021).

    Article  Google Scholar 

  3. Mearin, F. et al. Bowel disorders. Gastroenterology 150, 1393–1407 (2016).

    Article  Google Scholar 

  4. Vanuytsel, T., Tack, J. F. & Boeckxstaens, G. E. Treatment of abdominal pain in irritable bowel syndrome. J. Gastroenterol. 49, 1193–1205 (2014).

    Article  CAS  Google Scholar 

  5. Simren, M. et al. Visceral hypersensitivity is associated with GI symptom severity in functional GI disorders: consistent findings from five different patient cohorts. Gut 67, 255–262 (2018).

    Article  Google Scholar 

  6. Drossman, D. A. et al. Neuromodulators for functional gastrointestinal disorders (disorders of gut-brain interaction): a Rome Foundation Working Team Report. Gastroenterology 154, 1140–1171 (2018).

    Article  Google Scholar 

  7. Cohen, L. & Neuman, M. G. Cannabis and the gastrointestinal tract. J. Pharm. Pharm. Sci. 23, 301–313 (2020).

    Article  Google Scholar 

  8. Gotfried, J., Naftali, T. & Schey, R. Role of cannabis and its derivatives in gastrointestinal and hepatic disease. Gastroenterology 159, 62–80 (2020).

    Article  CAS  Google Scholar 

  9. Nasser, Y., Woo, M. & Andrews, C. N. Cannabis in gastroenterology: watch your head! A review of use in inflammatory bowel disease, functional gut disorders, and gut-related adverse effects. Curr. Treat. Options Gastroenterol. 18, 519–530 (2020).

    Article  Google Scholar 

  10. DiPatrizio, N. V. Endocannabinoids in the gut. Cannabis Cannabinoid Res. 1, 67–77 (2016).

    Article  CAS  Google Scholar 

  11. Maselli, D. B. & Camilleri, M. Pharmacology, clinical effects, and therapeutic potential of cannabinoids for gastrointestinal and liver diseases. Clin. Gastroenterol. Hepatol. 19, 1748–1758 (2021).

    Article  CAS  Google Scholar 

  12. Sharkey, K. A. & Wiley, J. W. The role of the endocannabinoid system in the brain-gut axis. Gastroenterology 151, 252–266 (2016).

    Article  CAS  Google Scholar 

  13. Pesce, M., Esposito, G. & Sarnelli, G. Endocannabinoids in the treatment of gasytrointestinal inflammation and symptoms. Curr. Opin. Pharmacol. 43, 81–86 (2018).

    Article  CAS  Google Scholar 

  14. Storr, M. A., Yuce, B., Andrews, C. N. & Sharkey, K. A. The role of the endocannabinoid system in the pathophysiology and treatment of irritable bowel syndrome. Neurogastroenterol. Motil. 20, 857–868 (2008).

    Article  CAS  Google Scholar 

  15. Mechoulam, R., Hanus, L. O., Pertwee, R. & Howlett, A. C. Early phytocannabinoid chemistry to endocannabinoids and beyond. Nat. Rev. Neurosci. 15, 757–764 (2014).

    Article  CAS  Google Scholar 

  16. Di Marzo, V. New approaches and challenges to targeting the endocannabinoid system. Nat. Rev. Drug Discov. 17, 623–639 (2018).

    Article  Google Scholar 

  17. Fowler, C. J., Doherty, P. & Alexander, S. P. H. Endocannabinoid turnover. Adv. Pharmacol. 80, 31–66 (2017).

    Article  CAS  Google Scholar 

  18. Blankman, J. L. & Cravatt, B. F. Chemical probes of endocannabinoid metabolism. Pharmacol. Rev. 65, 849–871 (2013).

    Article  Google Scholar 

  19. van Egmond, N., Straub, V. M. & van der Stelt, M. Targeting endocannabinoid signaling: FAAH and MAG lipase inhibitors. Annu. Rev. Pharmacol. Toxicol. 61, 441–463 (2021).

    Article  Google Scholar 

  20. Hill, M. N. & Patel, S. Translational evidence for the involvement of the endocannabinoid system in stress-related psychiatric illnesses. Biol. Mood Anxiety Disord. 3, 19 (2013).

    Article  Google Scholar 

  21. Lutz, B., Marsicano, G., Maldonado, R. & Hillard, C. J. The endocannabinoid system in guarding against fear, anxiety and stress. Nat. Rev. Neurosci. 16, 705–718 (2015).

    Article  CAS  Google Scholar 

  22. Kaczocha, M., Glaser, S. T. & Deutsch, D. G. Identification of intracellular carriers for the endocannabinoid anandamide. Proc. Natl Acad. Sci. USA 106, 6375–6380 (2009).

    Article  CAS  Google Scholar 

  23. Haj-Dahmane, S. et al. Fatty-acid-binding protein 5 controls retrograde endocannabinoid signaling at central glutamate synapses. Proc. Natl Acad. Sci. USA 115, 3482–3487 (2018).

    Article  Google Scholar 

  24. Cristino, L., Bisogno, T. & Di Marzo, V. Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat. Rev. Neurol. 16, 9–29 (2020).

    Article  Google Scholar 

  25. Abyadeh, M. et al. A proteomic view of cellular and molecular effects of cannabis. Biomolecules 11, 1411 (2021).

    Article  CAS  Google Scholar 

  26. Hua, T. et al. Activation and signaling mechanism revealed by cannabinoid receptor-Gi complex structures. Cell 180, 655–665 (2020).

    Article  CAS  Google Scholar 

  27. Krishna Kumar, K. et al. Structure of a signaling cannabinoid receptor 1-G protein complex. Cell 176, 448–458 (2019).

    Article  CAS  Google Scholar 

  28. Li, X. et al. Crystal structure of the human cannabinoid receptor CB2. Cell 176, 459–467 (2019).

    Article  CAS  Google Scholar 

  29. Moreira, F. A., Grieb, M. & Lutz, B. Central side-effects of therapies based on CB1 cannabinoid receptor agonists and antagonists: focus on anxiety and depression. Best Pract. Res. Clin. Endocrinol. Metab. 23, 133–144 (2009).

    Article  CAS  Google Scholar 

  30. Jimenez-Blasco, D. et al. Glucose metabolism links astroglial mitochondria to cannabinoid effects. Nature 583, 603–608 (2020).

    Article  CAS  Google Scholar 

  31. Soria-Gomez, E. et al. Subcellular specificity of cannabinoid effects in striatonigral circuits. Neuron 109, 1513–1526 (2021).

    Article  CAS  Google Scholar 

  32. Duncan, M. et al. Cannabinoid CB2 receptors in the enteric nervous system modulate gastrointestinal contractility in lipopolysaccharide-treated rats. Am. J. Physiol. Gastrointest. Liver Physiol. 295, G78–G87 (2008).

    Article  CAS  Google Scholar 

  33. Merriam, F. V., Wang, Z. Y., Guerios, S. D. & Bjorling, D. E. Cannabinoid receptor 2 is increased in acutely and chronically inflamed bladder of rats. Neurosci. Lett. 445, 130–134 (2008).

    Article  CAS  Google Scholar 

  34. Wright, K. et al. Differential expression of cannabinoid receptors in the human colon: cannabinoids promote epithelial wound healing. Gastroenterology 129, 437–453 (2005).

    Article  Google Scholar 

  35. Han, S., Thatte, J., Buzard, D. J. & Jones, R. M. Therapeutic utility of cannabinoid receptor type 2 (CB(2)) selective agonists. J. Med. Chem. 56, 8224–8256 (2013).

    Article  CAS  Google Scholar 

  36. Castro, J. et al. Olorinab (APD371), a peripherally acting, highly selective, full agonist of the cannabinoid receptor 2, reduces colitis-induced acute and chronic visceral hypersensitivity in rodents. Pain 163, e72–e86 (2022).

    Article  CAS  Google Scholar 

  37. Jordan, C. J. & Xi, Z. X. Progress in brain cannabinoid CB2 receptor research: from genes to behavior. Neurosci. Biobehav. Rev. 98, 208–220 (2019).

    Article  CAS  Google Scholar 

  38. Howlett, A. C. & Abood, M. E. CB1 and CB2 receptor pharmacology. Adv. Pharmacol. 80, 169–206 (2017).

    Article  CAS  Google Scholar 

  39. Lutz, B. Neurobiology of cannabinoid receptor signaling. Dialogues Clin. Neurosci. 22, 207–222 (2020).

    Article  Google Scholar 

  40. Booth, W. T., Walker, N. B., Lowther, W. T. & Howlett, A. C. Cannabinoid receptor interacting protein 1a (CRIP1a): function and structure. Molecules 24, 3672 (2019).

    Article  CAS  Google Scholar 

  41. Camilleri, M. Cannabinoids and gastrointestinal motility: pharmacology, clinical effects, and potential therapeutics in humans. Neurogastroenterol. Motil. 30, e13370 (2018).

    Article  CAS  Google Scholar 

  42. Izzo, A. A. & Sharkey, K. A. Cannabinoids and the gut: new developments and emerging concepts. Pharmacol. Ther. 126, 21–38 (2010).

    Article  CAS  Google Scholar 

  43. Hons, I. M. et al. Plasticity of mouse enteric synapses mediated through endocannabinoid and purinergic signaling. Neurogastroenterol. Motil. 24, e113–e124 (2012).

    Article  CAS  Google Scholar 

  44. Boesmans, W., Ameloot, K., van den Abbeel, V., Tack, J. & Vanden Berghe, P. Cannabinoid receptor 1 signalling dampens activity and mitochondrial transport in networks of enteric neurones. Neurogastroenterol. Motil. 21, 958–e77 (2009).

    Article  CAS  Google Scholar 

  45. Fichna, J., Sibaev, A., Salaga, M., Sobczak, M. & Storr, M. The cannabinoid-1 receptor inverse agonist taranabant reduces abdominal pain and increases intestinal transit in mice. Neurogastroenterol. Motil. 25, e550–e559 (2013).

    Article  CAS  Google Scholar 

  46. Storr, M. A. et al. Differential effects of CB1 neutral antagonists and inverse agonists on gastrointestinal motility in mice. Neurogastroenterol. Motil. 22, 787–796 (2010).

    Article  CAS  Google Scholar 

  47. Drokhlyansky, E. et al. The human and mouse enteric nervous system at single-cell resolution. Cell 182, 1606–1622 (2020).

    Article  CAS  Google Scholar 

  48. Jiang, Y., Nie, Y., Li, Y. & Zhang, L. Association of cannabinoid type 1 receptor and fatty acid amide hydrolase genetic polymorphisms in Chinese patients with irritable bowel syndrome. J. Gastroenterol. Hepatol. 29, 1186–1191 (2014).

    Article  CAS  Google Scholar 

  49. Wong, B. S. et al. Pharmacogenetic trial of a cannabinoid agonist shows reduced fasting colonic motility in patients with nonconstipated irritable bowel syndrome. Gastroenterology 141, 1638–1647 (2011).

    Article  CAS  Google Scholar 

  50. Bashashati, M. et al. Inhibiting fatty acid amide hydrolase normalizes endotoxin-induced enhanced gastrointestinal motility in mice. Br. J. Pharmacol. 165, 1556–1571 (2012).

    Article  CAS  Google Scholar 

  51. Kimball, E. S., Wallace, N. H., Schneider, C. R., D’Andrea, M. R. & Hornby, P. J. Small intestinal cannabinoid receptor changes following a single colonic insult with oil of mustard in mice. Front. Pharmacol. 1, 132 (2010).

    Article  Google Scholar 

  52. Lin, M., Chen, L., Xiao, Y. & Yu, B. Activation of cannabinoid 2 receptor relieves colonic hypermotility in a rat model of irritable bowel syndrome. Neurogastroenterol. Motil. 31, e13555 (2019).

    Article  Google Scholar 

  53. Bashashati, M. et al. Inhibiting endocannabinoid biosynthesis: a novel approach to the treatment of constipation. Br. J. Pharmacol. 172, 3099–3111 (2015).

    Article  CAS  Google Scholar 

  54. Crowe, M. S. & Kinsey, S. G. MAGL inhibition modulates gastric secretion and motility following NSAID exposure in mice. Eur. J. Pharmacol. 807, 198–204 (2017).

    Article  CAS  Google Scholar 

  55. Fichna, J. et al. Selective inhibition of FAAH produces antidiarrheal and antinociceptive effect mediated by endocannabinoids and cannabinoid-like fatty acid amides. Neurogastroenterol. Motil. 26, 470–481 (2014).

    Article  CAS  Google Scholar 

  56. Taschler, U. et al. Monoglyceride lipase deficiency causes desensitization of intestinal cannabinoid receptor type 1 and increased colonic mu-opioid receptor sensitivity. Br. J. Pharmacol. 172, 4419–4429 (2015).

    Article  CAS  Google Scholar 

  57. Capasso, R. et al. Palmitoylethanolamide normalizes intestinal motility in a model of post-inflammatory accelerated transit: involvement of CB(1) receptors and TRPV1 channels. Br. J. Pharmacol. 171, 4026–4037 (2014).

    Article  CAS  Google Scholar 

  58. Zhang, S. C., Wang, W. L., Su, P. J., Jiang, K. L. & Yuan, Z. W. Decreased enteric fatty acid amide hydrolase activity is associated with colonic inertia in slow transit constipation. J. Gastroenterol. Hepatol. 29, 276–283 (2014).

    Article  Google Scholar 

  59. Camilleri, M. et al. Genetic variation in endocannabinoid metabolism, gastrointestinal motility, and sensation. Am. J. Physiol. Gastrointest. Liver Physiol. 294, G13–G19 (2008).

    Article  CAS  Google Scholar 

  60. Ameloot, K. et al. Endocannabinoid control of gastric sensorimotor function in man. Aliment. Pharmacol. Ther. 31, 1123–1131 (2010).

    CAS  Google Scholar 

  61. Scarpellini, E. et al. Effect of rimonabant on oesophageal motor function in man. Aliment. Pharmacol. Ther. 33, 730–737 (2011).

    Article  CAS  Google Scholar 

  62. Tack, J. et al. The gastrointestinal tract in hunger and satiety signalling. UEG J. 9, 727–734 (2021).

    Article  Google Scholar 

  63. Tyler, K., Hillard, C. J. & Greenwood-Van Meerveld, B. Inhibition of small intestinal secretion by cannabinoids is CB1 receptor-mediated in rats. Eur. J. Pharmacol. 409, 207–211 (2000).

    Article  CAS  Google Scholar 

  64. MacNaughton, W. K. et al. Distribution and function of the cannabinoid-1 receptor in the modulation of ion transport in the guinea pig ileum: relationship to capsaicin-sensitive nerves. Am. J. Physiol. Gastrointest. Liver Physiol. 286, G863–G871 (2004).

    Article  CAS  Google Scholar 

  65. Izzo, A. A. et al. An endogenous cannabinoid tone attenuates cholera toxin-induced fluid accumulation in mice. Gastroenterology 125, 765–774 (2003).

    Article  CAS  Google Scholar 

  66. Alhamoruni, A., Lee, A. C., Wright, K. L., Larvin, M. & O’Sullivan, S. E. Pharmacological effects of cannabinoids on the Caco-2 cell culture model of intestinal permeability. J. Pharmacol. Exp. Ther. 335, 92–102 (2010).

    Article  CAS  Google Scholar 

  67. Alhamoruni, A., Wright, K. L., Larvin, M. & O’Sullivan, S. E. Cannabinoids mediate opposing effects on inflammation-induced intestinal permeability. Br. J. Pharmacol. 165, 2598–2610 (2012).

    Article  CAS  Google Scholar 

  68. Karwad, M. A. et al. The role of CB1 in intestinal permeability and inflammation. FASEB J. 31, 3267–3277 (2017).

    Article  CAS  Google Scholar 

  69. Muccioli, G. G. et al. The endocannabinoid system links gut microbiota to adipogenesis. Mol. Syst. Biol. 6, 392 (2010).

    Article  Google Scholar 

  70. Dothel, G. et al. micro-opioid receptor, beta-endorphin, and cannabinoid receptor-2 are increased in the colonic mucosa of irritable bowel syndrome patients. Neurogastroenterol. Motil. 31, e13688 (2019).

    Article  Google Scholar 

  71. Cabral, G. A., Ferreira, G. A. & Jamerson, M. J. Endocannabinoids and the immune system in health and disease. Handb. Exp. Pharmacol. 231, 185–211 (2015).

    Article  CAS  Google Scholar 

  72. Duffy, S. S., Hayes, J. P., Fiore, N. T. & Moalem-Taylor, G. The cannabinoid system and microglia in health and disease. Neuropharmacology 190, 108555 (2021).

    Article  CAS  Google Scholar 

  73. Turcotte, C., Blanchet, M. R., Laviolette, M. & Flamand, N. The CB2 receptor and its role as a regulator of inflammation. Cell Mol. Life Sci. 73, 4449–4470 (2016).

    Article  CAS  Google Scholar 

  74. Brierley, S. M. & Linden, D. R. Neuroplasticity and dysfunction after gastrointestinal inflammation. Nat. Rev. Gastroenterol. Hepatol. 11, 611–627 (2014).

    Article  Google Scholar 

  75. Storr, M. A. et al. Activation of the cannabinoid 2 receptor (CB2) protects against experimental colitis. Inflamm. Bowel Dis. 15, 1678–1685 (2009).

    Article  Google Scholar 

  76. Leinwand, K. L. et al. Cannabinoid receptor-2 ameliorates inflammation in murine model of Crohn’s disease. J. Crohns Colitis 11, 1369–1380 (2017).

    Article  Google Scholar 

  77. Cremon, C. et al. Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Aliment. Pharmacol. Ther. 45, 909–922 (2017).

    Article  CAS  Google Scholar 

  78. Alhouayek, M., Ameraoui, H. & Muccioli, G. G. Bioactive lipids in inflammatory bowel diseases - from pathophysiological alterations to therapeutic opportunities. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1866, 158854 (2021).

    Article  CAS  Google Scholar 

  79. Picardo, S., Kaplan, G. G., Sharkey, K. A. & Seow, C. H. Insights into the role of cannabis in the management of inflammatory bowel disease. Ther. Adv. Gastroenterol. 12, 1756284819870977 (2019).

    Article  CAS  Google Scholar 

  80. Fichna, J. et al. Cannabinoids alleviate experimentally induced intestinal inflammation by acting at central and peripheral receptors. PLoS ONE 9, e109115 (2014).

    Article  Google Scholar 

  81. Szabady, R. L. et al. Intestinal P-glycoprotein exports endocannabinoids to prevent inflammation and maintain homeostasis. J. Clin. Invest. 128, 4044–4056 (2018).

    Article  Google Scholar 

  82. Foley, S. E. et al. Gut microbiota regulation of P-glycoprotein in the intestinal epithelium in maintenance of homeostasis. Microbiome 9, 183 (2021).

    Article  CAS  Google Scholar 

  83. Brusberg, M. et al. CB1 receptors mediate the analgesic effects of cannabinoids on colorectal distension-induced visceral pain in rodents. J. Neurosci. 29, 1554–1564 (2009).

    Article  CAS  Google Scholar 

  84. Hong, S. et al. Reciprocal changes in vanilloid (TRPV1) and endocannabinoid (CB1) receptors contribute to visceral hyperalgesia in the water avoidance stressed rat. Gut 58, 202–210 (2009).

    Article  CAS  Google Scholar 

  85. Iwata, Y. et al. Identification of a highly potent and selective CB2 agonist, RQ-00202730, for the treatment of irritable bowel syndrome. Bioorg. Med. Chem. Lett. 25, 236–240 (2015).

    Article  CAS  Google Scholar 

  86. Kikuchi, A., Ohashi, K., Sugie, Y., Sugimoto, H. & Omura, H. Pharmacological evaluation of a novel cannabinoid 2 (CB2) ligand, PF-03550096, in vitro and in vivo by using a rat model of visceral hypersensitivity. J. Pharmacol. Sci. 106, 219–224 (2008).

    Article  CAS  Google Scholar 

  87. Sanson, M., Bueno, L. & Fioramonti, J. Involvement of cannabinoid receptors in inflammatory hypersensitivity to colonic distension in rats. Neurogastroenterol. Motil. 18, 949–956 (2006).

    Article  CAS  Google Scholar 

  88. Naguib, M. et al. MDA7: a novel selective agonist for CB2 receptors that prevents allodynia in rat neuropathic pain models. Br. J. Pharmacol. 155, 1104–1116 (2008).

    Article  CAS  Google Scholar 

  89. Enck, P. et al. Irritable bowel syndrome. Nat. Rev. Dis. Prim. 2, 16014 (2016).

    Article  Google Scholar 

  90. Shen, L., Yang, X. J., Qian, W. & Hou, X. H. The role of peripheral cannabinoid receptors type 1 in rats with visceral hypersensitivity induced by chronic restraint stress. J. Neurogastroenterol. Motil. 16, 281–290 (2010).

    Article  Google Scholar 

  91. Hillsley, K. et al. Activation of the cannabinoid 2 (CB2) receptor inhibits murine mesenteric afferent nerve activity. Neurogastroenterol. Motil. 19, 769–777 (2007).

    Article  CAS  Google Scholar 

  92. Sadeghi, M. et al. Contribution of membrane receptor signalling to chronic visceral pain. Int. J. Biochem. Cell Biol. 98, 10–23 (2018).

    Article  CAS  Google Scholar 

  93. Prato, V. et al. Functional and molecular characterization of mechanoinsensitive “Silent” nociceptors. Cell Rep. 21, 3102–3115 (2017).

    Article  CAS  Google Scholar 

  94. Schaefer, I., Prato, V., Arcourt, A., Taberner, F. J. & Lechner, S. G. Differential modulation of voltage-gated sodium channels by nerve growth factor in three major subsets of TrkA-expressing nociceptors. Mol. Pain 14, 1744806918814640 (2018).

    Article  CAS  Google Scholar 

  95. Dothel, G. et al. Nerve fiber outgrowth is increased in the intestinal mucosa of patients with irritable bowel syndrome. Gastroenterology 148, 1002–1011 (2015).

    Article  CAS  Google Scholar 

  96. Sarnelli, G. et al. Impaired duodenal palmitoylethanolamide release underlies acid-induced mast cell activation in functional dyspepsia. Cell Mol. Gastroenterol. Hepatol. 11, 841–855 (2021).

    Article  CAS  Google Scholar 

  97. Barbara, G. et al. Activated mast cells in proximity to colonic nerves correlate with abdominal pain in irritable bowel syndrome. Gastroenterology 126, 693–702 (2004).

    Article  Google Scholar 

  98. Barbara, G. et al. Mast cell-dependent excitation of visceral-nociceptive sensory neurons in irritable bowel syndrome. Gastroenterology 132, 26–37 (2007).

    Article  CAS  Google Scholar 

  99. Aguilera-Lizarraga, J. et al. Local immune response to food antigens drives meal-induced abdominal pain. Nature 590, 151–156 (2021).

    Article  CAS  Google Scholar 

  100. Farquhar-Smith, W. P., Jaggar, S. I. & Rice, A. S. Attenuation of nerve growth factor-induced visceral hyperalgesia via cannabinoid CB1 and CB2-like receptors. Pain 97, 11–21 (2002).

    Article  CAS  Google Scholar 

  101. Cantarella, G. et al. Endocannabinoids inhibit release of nerve growth factor by inflammation-activated mast cells. Biochem. Pharmacol. 82, 380–388 (2011).

    Article  CAS  Google Scholar 

  102. Costa, B., Comelli, F., Bettoni, I., Colleoni, M. & Giagnoni, G. The endogenous fatty acid amide, palmitoylethanolamide, has anti-allodynic and anti-hyperalgesic effects in a murine model of neuropathic pain: involvement of CB(1), TRPV1 and PPARgamma receptors and neurotrophic factors. Pain 139, 541–550 (2008).

    Article  CAS  Google Scholar 

  103. Petrosino, S. et al. Palmitoylethanolamide counteracts substance P-induced mast cell activation in vitro by stimulating diacylglycerol lipase activity. J. Neuroinflammation 16, 274 (2019).

    Article  CAS  Google Scholar 

  104. Domoto, R., Sekiguchi, F., Tsubota, M. & Kawabata, A. Macrophage as a peripheral pain regulator. Cells 10, 1881 (2021).

    Article  CAS  Google Scholar 

  105. Cattaruzza, F. et al. Cathepsin S is activated during colitis and causes visceral hyperalgesia by a PAR2-dependent mechanism in mice. Gastroenterology 141, 1864–1874 (2011).

    Article  CAS  Google Scholar 

  106. Jimenez-Vargas, N. N. et al. Protease-activated receptor-2 in endosomes signals persistent pain of irritable bowel syndrome. Proc. Natl Acad. Sci. USA 115, E7438–E7447 (2018).

    Article  CAS  Google Scholar 

  107. Grubišić, V. et al. Enteric glia modulate macrophage phenotype and visceral sensitivity following inflammation. Cell Rep. 32, 108100 (2020).

    Article  Google Scholar 

  108. Esposito, G. et al. Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-alpha activation. Gut 63, 1300–1312 (2014).

    Article  CAS  Google Scholar 

  109. Acharya, N. et al. Endocannabinoid system acts as a regulator of immune homeostasis in the gut. Proc. Natl Acad. Sci. USA 114, 5005–5010 (2017).

    Article  CAS  Google Scholar 

  110. Basso, L. et al. Granulocyte-colony-stimulating factor (G-CSF) signaling in spinal microglia drives visceral sensitization following colitis. Proc. Natl Acad. Sci. USA 114, 11235–11240 (2017).

    Article  CAS  Google Scholar 

  111. Yuan, T., Manohar, K., Latorre, R., Orock, A. & Greenwood-Van Meerveld, B. Inhibition of microglial activation in the amygdala reverses stress-induced abdominal pain in the male rat. Cell Mol. Gastroenterol. Hepatol. 10, 527–543 (2020).

    Article  Google Scholar 

  112. Yuan, T., Orock, A. & Greenwood-Van Meerveld, B. Amygdala microglia modify neuronal plasticity via complement C1q/C3-CR3 signaling and contribute to visceral pain in a rat model. Am. J. Physiol. Gastrointest. Liver Physiol. 320, G1081–G1092 (2021).

    Article  CAS  Google Scholar 

  113. Zhang, G. et al. Activation of corticotropin-releasing factor neurons and microglia in paraventricular nucleus precipitates visceral hypersensitivity induced by colorectal distension in rats. Brain Behav. Immun. 55, 93–104 (2016).

    Article  CAS  Google Scholar 

  114. Zhang, G. et al. Hippocampal microglial activation and glucocorticoid receptor down-regulation precipitate visceral hypersensitivity induced by colorectal distension in rats. Neuropharmacology 102, 295–303 (2016).

    Article  CAS  Google Scholar 

  115. Mayer, E. A., Labus, J. S., Tillisch, K., Cole, S. W. & Baldi, P. Towards a systems view of IBS. Nat. Rev. Gastroenterol. Hepatol. 12, 592–605 (2015).

    Article  CAS  Google Scholar 

  116. Martín-Pérez, C. et al. Endocannabinoid signaling of homeostatic status modulates functional connectivity in reward and salience networks. Psychopharmacology 239, 1311–1319 (2021).

    Article  Google Scholar 

  117. Sisk, L. M. et al. Genetic variation in endocannabinoid signaling is associated with differential network-level functional connectivity in youth. J. Neurosci. Res. 100, 731–743 (2022).

    Article  CAS  Google Scholar 

  118. Yasmin, F. et al. Stress-induced modulation of endocannabinoid signaling leads to delayed strengthening of synaptic connectivity in the amygdala. Proc. Natl Acad. Sci. USA 117, 650–655 (2020).

    Article  CAS  Google Scholar 

  119. Benedetti, F., Amanzio, M., Rosato, R. & Blanchard, C. Nonopioid placebo analgesia is mediated by CB1 cannabinoid receptors. Nat. Med. 17, 1228–1230 (2011).

    Article  CAS  Google Scholar 

  120. Peciña, M. et al. FAAH selectively influences placebo effects. Mol. Psychiatry 19, 385–391 (2014).

    Article  Google Scholar 

  121. Enck, P. & Klosterhalfen, S. Placebo responses and Placebo effects in functional gastrointestinal disorders. Front. Psychiatry 11, 797 (2020).

    Article  Google Scholar 

  122. Meerveld, B. G. & Johnson, A. C. Mechanisms of stress-induced visceral pain. J. Neurogastroenterol. Motil. 24, 7–18 (2018).

    Article  Google Scholar 

  123. Mahurkar-Joshi, S. & Chang, L. Epigenetic mechanisms in irritable bowel syndrome. Front. Psychiatry 11, 805 (2020).

    Article  Google Scholar 

  124. McEwen, B. S. et al. Mechanisms of stress in the brain. Nat. Neurosci. 18, 1353–1363 (2015).

    Article  CAS  Google Scholar 

  125. Rusconi, F., Rubino, T. & Battaglioli, E. Endocannabinoid-epigenetic cross-talk: a bridge toward stress coping. Int. J. Mol. Sci. 21, 6252 (2020).

    Article  CAS  Google Scholar 

  126. Tran, L., Chaloner, A., Sawalha, A. H. & Greenwood Van-Meerveld, B. Importance of epigenetic mechanisms in visceral pain induced by chronic water avoidance stress. Psychoneuroendocrinology 38, 898–906 (2013).

    Article  CAS  Google Scholar 

  127. Hong, S., Zheng, G. & Wiley, J. W. Epigenetic regulation of genes that modulate chronic stress-induced visceral pain in the peripheral nervous system. Gastroenterology 148, 148–157 (2015).

    Article  CAS  Google Scholar 

  128. Osadchiy, V., Martin, C. R. & Mayer, E. A. The gut-brain axis and the microbiome: mechanisms and clinical implications. Clin. Gastroenterol. Hepatol. 17, 322–332 (2019).

    Article  CAS  Google Scholar 

  129. Pittayanon, R. et al. Gut microbiota in patients with irritable bowel syndrome-a systematic review. Gastroenterology 157, 97–108 (2019).

    Article  Google Scholar 

  130. Simren, M. et al. Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut 62, 159–176 (2013).

    Article  Google Scholar 

  131. Esquerre, N. et al. Colitis-induced microbial perturbation promotes postinflammatory visceral hypersensitivity. Cell Mol. Gastroenterol. Hepatol. 10, 225–244 (2020).

    Article  Google Scholar 

  132. Reigstad, C. S. et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 29, 1395–1403 (2015).

    Article  CAS  Google Scholar 

  133. Sugisawa, E. et al. RNA sensing by gut Piezo1 is essential for systemic serotonin synthesis. Cell 182, 609–624 (2020).

    Article  CAS  Google Scholar 

  134. Anitha, M., Vijay-Kumar, M., Sitaraman, S. V., Gewirtz, A. T. & Srinivasan, S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling. Gastroenterology 143, 1006–1016 (2012).

    Article  CAS  Google Scholar 

  135. Brun, P. et al. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology 145, 1323–1333 (2013).

    Article  CAS  Google Scholar 

  136. Yarandi, S. S. et al. Intestinal bacteria maintain adult enteric nervous system and nitrergic neurons via Toll-like receptor 2-induced neurogenesis in mice. Gastroenterology 159, 200–213 (2020).

    Article  CAS  Google Scholar 

  137. Obata, Y. et al. Neuronal programming by microbiota regulates intestinal physiology. Nature 578, 284–289 (2020).

    Article  CAS  Google Scholar 

  138. Lyte, M. Microbial endocrinology: an ongoing personal journey. Adv. Exp. Med. Biol. 874, 1–24 (2016).

    Article  CAS  Google Scholar 

  139. Sharkey, K. A. & Savidge, T. C. Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton. Neurosci. 181, 94–106 (2014).

    Article  CAS  Google Scholar 

  140. DiPatrizio, N. V. & Piomelli, D. Intestinal lipid-derived signals that sense dietary fat. J. Clin. Invest. 125, 891–898 (2015).

    Article  Google Scholar 

  141. Everard, A. et al. Intestinal epithelial N-acylphosphatidylethanolamine phospholipase D links dietary fat to metabolic adaptations in obesity and steatosis. Nat. Commun. 10, 457 (2019).

    Article  CAS  Google Scholar 

  142. Castonguay-Paradis, S. et al. Dietary fatty acid intake and gut microbiota determine circulating endocannabinoidome signaling beyond the effect of body fat. Sci. Rep. 10, 15975 (2020).

    Article  CAS  Google Scholar 

  143. Christie, S., O’Rielly, R., Li, H., Wittert, G. A. & Page, A. J. Biphasic effects of methanandamide on murine gastric vagal afferent mechanosensitivity. J. Physiol. 598, 139–150 (2020).

    Article  CAS  Google Scholar 

  144. Lacroix, S. et al. Rapid and concomitant gut microbiota and endocannabinoidome response to diet-induced obesity in mice. mSystems 4, e00407-19 (2019).

    Article  Google Scholar 

  145. DiPatrizio, N. V. et al. Fasting stimulates 2-AG biosynthesis in the small intestine: role of cholinergic pathways. Am. J. Physiol. Regul. Integr. Comp. Physiol. 309, R805–R813 (2015).

    Article  CAS  Google Scholar 

  146. Hong, S. et al. Corticosterone mediates reciprocal changes in CB 1 and TRPV1 receptors in primary sensory neurons in the chronically stressed rat. Gastroenterology 140, 627–637 (2011).

    Article  CAS  Google Scholar 

  147. Keenan, C. M. et al. AM841, a covalent cannabinoid ligand, powerfully slows gastrointestinal motility in normal and stressed mice in a peripherally restricted manner. Br. J. Pharmacol. 172, 2406–2418 (2015).

    Article  CAS  Google Scholar 

  148. Salaga, M. et al. The novel peripherally active cannabinoid type 1 and serotonin type 3 receptor agonist AM9405 inhibits gastrointestinal motility and reduces abdominal pain in mouse models mimicking irritable bowel syndrome. Eur. J. Pharmacol. 836, 34–43 (2018).

    Article  CAS  Google Scholar 

  149. Sticht, M. A. et al. Endocannabinoid regulation of homeostatic feeding and stress-induced alterations in food intake in male rats. Br. J. Pharmacol. 176, 1524–1540 (2019).

    Article  CAS  Google Scholar 

  150. Cryan, J. F. et al. The microbiota-gut-brain axis. Physiol. Rev. 99, 1877–2013 (2019).

    Article  CAS  Google Scholar 

  151. Manca, C. et al. Germ-free mice exhibit profound gut microbiota-dependent alterations of intestinal endocannabinoidome signaling. J. Lipid Res. 61, 70–85 (2020).

    Article  CAS  Google Scholar 

  152. Manca, C. et al. Alterations of brain endocannabinoidome signaling in germ-free mice. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 1865, 158786 (2020).

    Article  CAS  Google Scholar 

  153. Kennedy, E. A., King, K. Y. & Baldridge, M. T. Mouse microbiota models: comparing germ-free mice and antibiotics treatment as tools for modifying gut bacteria. Front. Physiol. 9, 1534 (2018).

    Article  Google Scholar 

  154. Aguilera, M., Cerda-Cuellar, M. & Martinez, V. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes 6, 10–23 (2015).

    Article  CAS  Google Scholar 

  155. Aguilera, M., Vergara, P. & Martinez, V. Stress and antibiotics alter luminal and wall-adhered microbiota and enhance the local expression of visceral sensory-related systems in mice. Neurogastroenterol. Motil. 25, e515–e529 (2013).

    Article  CAS  Google Scholar 

  156. Guida, F. et al. Antibiotic-induced microbiota perturbation causes gut endocannabinoidome changes, hippocampal neuroglial reorganization and depression in mice. Brain Behav. Immun. 67, 230–245 (2018).

    Article  CAS  Google Scholar 

  157. Rousseaux, C. et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat. Med. 13, 35–37 (2007).

    Article  CAS  Google Scholar 

  158. Markey, L. et al. Colonization with the commensal fungus Candida albicans perturbs the gut-brain axis through dysregulation of endocannabinoid signaling. Psychoneuroendocrinology 121, 104808 (2020).

    Article  CAS  Google Scholar 

  159. Chevalier, G. et al. Effect of gut microbiota on depressive-like behaviors in mice is mediated by the endocannabinoid system. Nat. Commun. 11, 6363 (2020).

    Article  CAS  Google Scholar 

  160. Minichino, A. et al. Endocannabinoid system mediates the association between gut-microbial diversity and anhedonia/amotivation in a general population cohort. Mol. Psychiatry 26, 6269–6276 (2021).

    Article  CAS  Google Scholar 

  161. Cluny, N. L., Keenan, C. M., Reimer, R. A., Le Foll, B. & Sharkey, K. A. Prevention of diet-induced obesity effects on body weight and gut microbiota in mice treated chronically with Delta9-tetrahydrocannabinol. PLoS ONE 10, e0144270 (2015).

    Article  Google Scholar 

  162. Mehrpouya-Bahrami, P. et al. Blockade of CB1 cannabinoid receptor alters gut microbiota and attenuates inflammation and diet-induced obesity. Sci. Rep. 7, 15645 (2017).

    Article  Google Scholar 

  163. Oza, M. et al. Acute and short-term administrations of Δ-9-tetrahydrocannabinol modulate major gut metabolomic regulatory pathways in C57BL/6 mice. Sci. Rep. 9, 10520 (2019).

    Article  Google Scholar 

  164. Al-Ghezi, Z. Z., Busbee, P. B., Alghetaa, H., Nagarkatti, P. S. & Nagarkatti, M. Combination of cannabinoids, delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD), mitigates experimental autoimmune encephalomyelitis (EAE) by altering the gut microbiome. Brain Behav. Immun. 82, 25–35 (2019).

    Article  CAS  Google Scholar 

  165. Ellermann, M. et al. Endocannabinoids inhibit the induction of virulence in enteric pathogens. Cell 183, 650–665 (2020).

    Article  CAS  Google Scholar 

  166. Dione, N. et al. Mgll knockout mouse resistance to diet-induced dysmetabolism is associated with altered gut microbiota. Cells 9, 2705 (2020).

    Article  CAS  Google Scholar 

  167. Cani, P. D. et al. Endocannabinoids — at the crossroads between the gut microbiota and host metabolism. Nat. Rev. Endocrinol. 12, 133–143 (2016).

    Article  CAS  Google Scholar 

  168. Spiller, R. Impact of diet on symptoms of the irritable bowel syndrome. Nutrients 13, 575 (2021).

    Article  CAS  Google Scholar 

  169. Uranga, J. A., Martínez, V. & Abalo, R. Mast cell regulation and irritable bowel syndrome: effects of food components with potential nutraceutical use. Molecules 25, 4314 (2020).

    Article  CAS  Google Scholar 

  170. Di Marzo, V. & Silvestri, C. Lifestyle and metabolic syndrome: contribution of the endocannabinoidome. Nutrients 11, 1956 (2019).

    Article  CAS  Google Scholar 

  171. Kleberg, K., Hassing, H. A. & Hansen, H. S. Classical endocannabinoid-like compounds and their regulation by nutrients. Biofactors 40, 363–372 (2014).

    Article  CAS  Google Scholar 

  172. Hansen, H. S. Role of anorectic N-acylethanolamines in intestinal physiology and satiety control with respect to dietary fat. Pharmacol. Res. 86, 18–25 (2014).

    Article  CAS  Google Scholar 

  173. Matias, I. et al. Dysregulation of peripheral endocannabinoid levels in hyperglycemia and obesity: effect of high fat diets. Mol. Cell Endocrinol. 286, S66–S78 (2008).

    Article  CAS  Google Scholar 

  174. Diep, T. A. et al. Dietary fat decreases intestinal levels of the anorectic lipids through a fat sensor. FASEB J. 25, 765–774 (2011).

    Article  CAS  Google Scholar 

  175. Komarnytsky, S. et al. Endocannabinoid system and its regulation by polyunsaturated fatty acids and full spectrum hemp oils. Int. J. Mol. Sci. 22, 5479 (2021).

    Article  CAS  Google Scholar 

  176. Larrieu, T. & Layé, S. Food for mood: relevance of nutritional omega-3 fatty acids for depression and anxiety. Front. Physiol. 9, 1047 (2018).

    Article  Google Scholar 

  177. Gigante, I. et al. Cannabinoid receptors overexpression in a rat model of irritable bowel syndrome (IBS) after treatment with a Ketogenic diet. Int. J. Mol. Sci. 22, 2880 (2021).

    Article  CAS  Google Scholar 

  178. Martínez, V. et al. Cannabidiol and other non-psychoactive cannabinoids for prevention and treatment of gastrointestinal disorders: useful nutraceuticals? Int. J. Mol. Sci. 21, 3067 (2020).

    Article  Google Scholar 

  179. van Orten-Luiten, A. B., de Roos, N. M., Majait, S., Witteman, B. J. M. & Witkamp, R. F. Effects of cannabidiol chewing gum on perceived pain and well-being of irritable bowel syndrome patients: a placebo-controlled crossover exploratory intervention study with symptom-driven dosing. Cannabis Cannabinoid Res. 7, 436–444 (2021).

    Article  Google Scholar 

  180. Van den Houte, K. et al. Prevalence and impact of self-reported irritable bowel symptoms in the general population. UEG J. 7, 307–315 (2019).

    Article  Google Scholar 

  181. Black, C. J. & Ford, A. C. Global burden of irritable bowel syndrome: trends, predictions and risk factors. Nat. Rev. Gastroenterol. Hepatol. 17, 473–486 (2020).

    Article  Google Scholar 

  182. Posserud, I. et al. Altered rectal perception in irritable bowel syndrome is associated with symptom severity. Gastroenterology 133, 1113–1123 (2007).

    Article  Google Scholar 

  183. Tornblom, H. et al. Colonic transit time and IBS symptoms: what’s the link? Am. J. Gastroenterol. 107, 754–760 (2012).

    Article  Google Scholar 

  184. Simren, M. et al. Cumulative effects of psychologic distress, visceral hypersensitivity, and abnormal transit on patient-reported outcomes in irritable bowel syndrome. Gastroenterology 157, 391–402 (2019).

    Article  Google Scholar 

  185. Azpiroz, F. Hypersensitivity in functional gastrointestinal disorders. Gut 51, i25–i28 (2002).

    Article  Google Scholar 

  186. Azpiroz, F. et al. Mechanisms of hypersensitivity in IBS and functional disorders. Neurogastroenterol. Motil. 19, 62–88 (2007).

    Article  CAS  Google Scholar 

  187. Barbara, G. et al. Mechanisms underlying visceral hypersensitivity in irritable bowel syndrome. Curr. Gastroenterol. Rep. 13, 308–315 (2011).

    Article  Google Scholar 

  188. Malcolm, A., Phillips, S. F., Kellow, J. E. & Cousins, M. J. Direct clinical evidence for spinal hyperalgesia in a patient with irritable bowel syndrome. Am. J. Gastroenterol. 96, 2427–2431 (2001).

    Article  CAS  Google Scholar 

  189. Mertz, H. Review article: visceral hypersensitivity. Aliment. Pharmacol. Ther. 17, 623–633 (2003).

    Article  CAS  Google Scholar 

  190. Bhattarai, Y., Muniz Pedrogo, D. A. & Kashyap, P. C. Irritable bowel syndrome: a gut microbiota-related disorder? Am. J. Physiol. Gastrointest. Liver Physiol. 312, G52–G62 (2017).

    Article  Google Scholar 

  191. Akbar, A. et al. Increased capsaicin receptor TRPV1-expressing sensory fibres in irritable bowel syndrome and their correlation with abdominal pain. Gut 57, 923–929 (2008).

    Article  CAS  Google Scholar 

  192. Shiha, M. G., Ashgar, Z., Fraser, E. M., Kurien, M. & Aziz, I. High prevalence of primary bile acid diarrhoea in patients with functional diarrhoea and irritable bowel syndrome-diarrhoea, based on Rome III and Rome IV criteria. EClinicalMedicine 25, 100465 (2020).

    Article  Google Scholar 

  193. Halvorson, H. A., Schlett, C. D. & Riddle, M. S. Postinfectious irritable bowel syndrome–a meta-analysis. Am. J. Gastroenterol. 101, 1894–1899 (2006).

    Article  Google Scholar 

  194. Jalanka-Tuovinen, J. et al. Faecal microbiota composition and host-microbe cross-talk following gastroenteritis and in postinfectious irritable bowel syndrome. Gut 63, 1737–1745 (2014).

    Article  Google Scholar 

  195. Paula, H. et al. Non-enteric infections, antibiotic use, and risk of development of functional gastrointestinal disorders. Neurogastroenterol. Motil. 27, 1580–1586 (2015).

    Article  CAS  Google Scholar 

  196. Heitkemper, M., Jarrett, M., Bond, E. F. & Chang, L. Impact of sex and gender on irritable bowel syndrome. Biol. Res. Nurs. 5, 56–65 (2003).

    Article  Google Scholar 

  197. Naliboff, B. D. et al. Sex-related differences in IBS patients: central processing of visceral stimuli. Gastroenterology 124, 1738–1747 (2003).

    Article  Google Scholar 

  198. Hubbard, C. S. et al. Estrogen-dependent visceral hypersensitivity following stress in rats: an fMRI study. Mol. Pain 12, 1744806916654145 (2016).

    Article  Google Scholar 

  199. Jiang, Y., Greenwood-Van Meerveld, B., Johnson, A. C. & Travagli, R. A. Role of estrogen and stress on the brain-gut axis. Am. J. Physiol. Gastrointest. Liver Physiol. 317, G203–G209 (2019).

    Article  CAS  Google Scholar 

  200. Mulak, A., Taché, Y. & Larauche, M. Sex hormones in the modulation of irritable bowel syndrome. World J. Gastroenterol. 20, 2433–2448 (2014).

    Article  CAS  Google Scholar 

  201. Bradford, K. et al. Association between early adverse life events and irritable bowel syndrome. Clin. Gastroenterol. Hepatol. 10, 385–390 (2012).

    Article  Google Scholar 

  202. Chaloner, A. & Greenwood-Van Meerveld, B. Sexually dimorphic effects of unpredictable early life adversity on visceral pain behavior in a rodent model. J. Pain 14, 270–280 (2013).

    Article  Google Scholar 

  203. Rastelli, D. et al. Diminished androgen levels are linked to irritable bowel syndrome and cause bowel dysfunction in mice. J. Clin. Invest. 132, e150789 (2022).

    Article  CAS  Google Scholar 

  204. Blanton, H. L. et al. Sex differences and the endocannabinoid system in pain. Pharmacol. Biochem. Behav. 202, 173107 (2021).

    Article  CAS  Google Scholar 

  205. Simren, M. & Tack, J. New treatments and therapeutic targets for IBS and other functional bowel disorders. Nat. Rev. Gastroenterol. Hepatol. 15, 589–605 (2018).

    Article  CAS  Google Scholar 

  206. Dapoigny, M., Abitbol, J. L. & Fraitag, B. Efficacy of peripheral kappa agonist fedotozine versus placebo in treatment of irritable bowel syndrome. A multicenter dose-response study. Dig. Dis. Sci. 40, 2244–2249 (1995).

    Article  CAS  Google Scholar 

  207. Delvaux, M. et al. The kappa agonist fedotozine relieves hypersensitivity to colonic distention in patients with irritable bowel syndrome. Gastroenterology 116, 38–45 (1999).

    Article  CAS  Google Scholar 

  208. Mangel, A. W. et al. Clinical trial: asimadoline in the treatment of patients with irritable bowel syndrome. Aliment. Pharmacol. Ther. 28, 239–249 (2008).

    Article  CAS  Google Scholar 

  209. Szarka, L. A. et al. Efficacy of on-demand asimadoline, a peripheral kappa-opioid agonist, in females with irritable bowel syndrome. Clin. Gastroenterol. Hepatol. 5, 1268–1275 (2007).

    Article  CAS  Google Scholar 

  210. Delvaux, M. et al. Effect of asimadoline, a kappa opioid agonist, on pain induced by colonic distension in patients with irritable bowel syndrome. Aliment. Pharmacol. Ther. 20, 237–246 (2004).

    Article  CAS  Google Scholar 

  211. Dukes, G. et al. Lack of effect of the NH3 receptor antagonist, talnetant SB223412, on symptoms of IBS: results of 2 randomized, double-blind, placebo-controlled dose-ranging trials. Gastroenterology 132, A60 (2007).

    Google Scholar 

  212. Houghton, L. A. et al. Effect of the NK(3) receptor antagonist, talnetant, on rectal sensory function and compliance in healthy humans. Neurogastroenterol. Motil. 19, 732–743 (2007).

    Article  CAS  Google Scholar 

  213. Delvaux, M. Role of visceral sensitivity in the pathophysiology of irritable bowel syndrome. Gut 51, i67–i71 (2002).

    Article  Google Scholar 

  214. Lecci, A., Altamura, M., Capriati, A. & Maggi, C. A. Tachykinin receptors and gastrointestinal motility: focus on humans. Eur. Rev. Med. Pharmacol. Sci. 12, 69–80 (2008).

    Google Scholar 

  215. Tack, J. et al. The neurokinin-2 receptor antagonist ibodutant improves overall symptoms, abdominal pain and stool pattern in female patients in a phase II study of diarrhoea-predominant IBS. Gut 66, 1403–1413 (2017).

    Article  CAS  Google Scholar 

  216. Lee, O., Munakata, J., Naliboff, B., Chang, L. & Mayer, E. A double-blind parallel group pilot study of the effect of CJ-11,974 and placebo on perceptual and emotional responses to rectosigmoid distension in IBSs patients. Gastroenterology. 118, https://doi.org/10.1016/S0016-5085(00)85527-4 (2000).

  217. Tillisch, K. et al. Neurokinin-1-receptor antagonism decreases anxiety and emotional arousal circuit response to noxious visceral distension in women with irritable bowel syndrome: a pilot study. Aliment. Pharmacol. Ther. 35, 360–367 (2012).

    Article  CAS  Google Scholar 

  218. Akyuz, F., Mimidis, K., Nikolai, H., Vos, R. & Tack, J. Influence of NK1 receptor antagonist aprepitant on rectal sensitivity and compliance in healthy volunteers. Gastroenterology 32, A684 (2007).

    Google Scholar 

  219. Zakko, S., Barton, G., Weber, E., Dunger-Baldauf, C. & Ruhl, A. Randomised clinical trial: the clinical effects of a novel neurokinin receptor antagonist, DNK333, in women with diarrhoea-predominant irritable bowel syndrome. Aliment. Pharmacol. Ther. 33, 1311–1321 (2011).

    Article  CAS  Google Scholar 

  220. Kelleher, D. et al. Randomized, double-blind, placebo (PLA)-controlled, crossover study to evaluate efficacy and safety of the beta 3-adrenergic receptor agonist solabegron (SOL) in patients with irritable bowel syndrome (IBS). Neurogastroenterol. Motil. 20, 131–132 (2008).

    Google Scholar 

  221. Drossman, D. A. et al. Randomized, double-blind, placebo-controlled trial of the 5-HT1A receptor antagonist AZD7371 tartrate monohydrate (robalzotan tartrate monohydrate) in patients with irritable bowel syndrome. Am. J. Gastroenterol. 103, 2562–2569 (2008).

    Article  CAS  Google Scholar 

  222. Smith, S. C. & Wagner, M. S. Clinical endocannabinoid deficiency (CECD) revisited: can this concept explain the therapeutic benefits of cannabis in migraine, fibromyalgia, irritable bowel syndrome and other treatment-resistant conditions? Neuro Endocrinol. Lett. 35, 198–201 (2014).

    Google Scholar 

  223. Chang, L. et al. Efficacy and safety of olorinab, a peripherally acting, highly selective, full agonist of the cannabinoid receptor 2, for the treatment of abdominal pain in patients with irritable bowel syndrome: results from a phase 2b randomized study. UEG J. 9 (S8), 81 (2021).

    Google Scholar 

  224. National Academies of Sciences, Engineering, and Medicine; Health and Medicine Division; Board on Population Health and Public Health Practice. Committee on the Health Effects of Marijuana: an Evidence Review and Research Agenda. The Health Effects of Cannabis and Cannabinoids: The Current State of Evidence and Recommendations for Research (National Academies Press, 2017).

  225. Adejumo, A. C., Ajayi, T. O., Adegbala, O. M. & Bukong, T. N. Higher odds of irritable bowel syndrome among hospitalized patients using cannabis: a propensity-matched analysis. Eur. J. Gastroenterol. Hepatol. 31, 756–765 (2019).

    Article  Google Scholar 

  226. Russo, E. B. Clinical endocannabinoid deficiency (CECD): can this concept explain therapeutic benefits of cannabis in migraine, fibromyalgia, irritable bowel syndrome and other treatment-resistant conditions? Neuro Endocrinol. Lett. 25, 31–39 (2004).

    CAS  Google Scholar 

  227. Patel, R. S., Goyal, H., Satodiya, R. & Tankersley, W. E. Relationship of Cannabis use disorder and irritable bowel syndrome (IBS): an analysis of 6.8 million hospitalizations in the United States. Subst. Use Misuse 55, 281–290 (2020).

    Article  Google Scholar 

  228. Pandey, S. et al. Endocannabinoid system in irritable bowel syndrome and cannabis as a therapy. Complement. Ther. Med. 48, 102242 (2020).

    Article  Google Scholar 

  229. Choi, C. et al. Cannabis use is associated with reduced 30-day all-cause readmission among hospitalized patients with irritable bowel syndrome: a nationwide analysis. J. Clin. Gastroenterol. 56, 257–265 (2022).

    Article  Google Scholar 

  230. Fichna, J. et al. Endocannabinoid and cannabinoid-like fatty acid amide levels correlate with pain-related symptoms in patients with IBS-D and IBS-C: a pilot study. PLoS ONE 8, e85073 (2013).

    Article  Google Scholar 

  231. Esfandyari, T. et al. Effects of a cannabinoid receptor agonist on colonic motor and sensory functions in humans: a randomized, placebo-controlled study. Am. J. Physiol. Gastrointest. Liver Physiol. 293, G137–G145 (2007).

    Article  CAS  Google Scholar 

  232. Klooker, T. K., Leliefeld, K. E., Van Den Wijngaard, R. M. & Boeckxstaens, G. E. The cannabinoid receptor agonist delta-9-tetrahydrocannabinol does not affect visceral sensitivity to rectal distension in healthy volunteers and IBS patients. Neurogastroenterol. Motil. 23, 30–35 (2011).

    Article  CAS  Google Scholar 

  233. Osborn, L. A. et al. Self-medication of somatic and psychiatric conditions using botanical marijuana. J. Psychoact. Drugs 47, 345–350 (2015).

    Article  Google Scholar 

  234. Gonzalez, S., Cebeira, M. & Fernandez-Ruiz, J. Cannabinoid tolerance and dependence: a review of studies in laboratory animals. Pharmacol. Biochem. Behav. 81, 300–318 (2005).

    Article  CAS  Google Scholar 

  235. Perisetti, A. et al. Cannabis hyperemesis syndrome: an update on the pathophysiology and management. Ann. Gastroenterol. 33, 571–578 (2020).

    Google Scholar 

  236. Higgins, P. et al. P418 Safety and efficacy of olorinab, a peripherally restricted, highly-selective, cannabinoid receptor 2 agonist in a phase 2A study in chronic abdominal pain associated with Crohn’s disease. J. Crohns Colitis 13, S318–S318 (2019).

    Article  Google Scholar 

  237. Ly, H. G. et al. Increased cerebral cannabinoid-1 receptor availability is a stable feature of functional dyspepsia: a [F]MK-9470 PET study. Psychother. Psychosom. 84, 149–158 (2015).

    Article  Google Scholar 

  238. US National Library of Medicine. ClinicalTrials.gov https://ClinicalTrials.gov/show/NCT02875678 (2017).

  239. Weltens, N., Depoortere, I., Tack, J. & Van Oudenhove, L. Effect of acute Δ9-tetrahydrocannabinol administration on subjective and metabolic hormone responses to food stimuli and food intake in healthy humans: a randomized, placebo-controlled study. Am. J. Clin. Nutr. 109, 1051–1063 (2019).

    Article  Google Scholar 

  240. Rock, E. M., Limebeer, C. L., Pertwee, R. G., Mechoulam, R. & Parker, L. A. Therapeutic potential of cannabidiol, cannabidiolic acid, and cannabidiolic acid methyl ester as treatments for nausea and vomiting. Cannabis Cannabinoid Res. 6, 266–274 (2021).

    Article  CAS  Google Scholar 

  241. Sharkey, K. A., Darmani, N. A. & Parker, L. A. Regulation of nausea and vomiting by cannabinoids and the endocannabinoid system. Eur. J. Pharmacol. 722, 134–146 (2014).

    Article  CAS  Google Scholar 

  242. Grimison, P. et al. Oral THC:CBD cannabis extract for refractory chemotherapy-induced nausea and vomiting: a randomised, placebo-controlled, phase II crossover trial. Ann. Oncol. 31, 1553–1560 (2020).

    Article  CAS  Google Scholar 

  243. Jehangir, A. & Parkman, H. P. Cannabinoid use in patients with gastroparesis and related disorders: prevalence and benefit. Am. J. Gastroenterol. 114, 945–953 (2019).

    Article  Google Scholar 

  244. McCarty, T. R., Chouairi, F., Hathorn, K. E., Chan, W. W. & Thompson, C. C. Trends and socioeconomic health outcomes of cannabis use among patients with gastroparesis: a United States nationwide inpatient sample analysis. J. Clin. Gastroenterol. 56, 324–330 (2022).

    Article  Google Scholar 

  245. Tu, Q., Heitkemper, M. M., Jarrett, M. E. & Buchanan, D. T. Sleep disturbances in irritable bowel syndrome: a systematic review. Neurogastroenterol. Motil. 29, https://doi.org/10.1111/nmo.12946 (2017).

  246. Simpson, N. S., Scott-Sutherland, J., Gautam, S., Sethna, N. & Haack, M. Chronic exposure to insufficient sleep alters processes of pain habituation and sensitization. Pain 159, 33–40 (2018).

    Article  Google Scholar 

  247. Smith, M. T.Jr. et al. Sex differences in measures of central sensitization and pain sensitivity to experimental sleep disruption: implications for sex differences in chronic pain. Sleep 42, zsy209 (2019).

    Article  Google Scholar 

  248. Babson, K. A., Sottile, J. & Morabito, D. Cannabis, cannabinoids, and sleep: a review of the literature. Curr. Psychiatry Rep. 19, 23 (2017).

    Article  Google Scholar 

  249. Méndez-Díaz, M., Ruiz-Contreras, A. E., Cortés-Morelos, J. & Prospéro-García, O. Cannabinoids and sleep/wake control. Adv. Exp. Med. Biol. 1297, 83–95 (2021).

    Article  Google Scholar 

  250. Kaul, M., Zee, P. C. & Sahni, A. S. Effects of cannabinoids on sleep and their therapeutic potential for sleep disorders. Neurotherapeutics 18, 217–227 (2021).

    Article  Google Scholar 

  251. Mondino, A. et al. Effects of cannabis consumption on sleep. Adv. Exp. Med. Biol. 1297, 147–162 (2021).

    Article  CAS  Google Scholar 

  252. Eijsbouts, C. et al. Genome-wide analysis of 53,400 people with irritable bowel syndrome highlights shared genetic pathways with mood and anxiety disorders. Nat. Genet. 53, 1543–1552 (2021).

    Article  CAS  Google Scholar 

  253. Grigorenko, E. et al. Assessment of cannabinoid induced gene changes: tolerance and neuroprotection. Chem. Phys. Lipids 121, 257–266 (2002).

    Article  CAS  Google Scholar 

  254. Hryhorowicz, S., Walczak, M., Zakerska-Banaszak, O., Słomski, R. & Skrzypczak-Zielińska, M. Pharmacogenetics of cannabinoids. Eur. J. Drug Metab. Pharmacokinet. 43, 1–12 (2018).

    Article  CAS  Google Scholar 

  255. Wong, B. S. et al. Randomized pharmacodynamic and pharmacogenetic trial of dronabinol effects on colon transit in irritable bowel syndrome-diarrhea. Neurogastroenterol. Motil. 24, 358–e169 (2012).

    Article  CAS  Google Scholar 

  256. Bedse, G., Hill, M. N. & Patel, S. 2-Arachidonoylglycerol modulation of anxiety and stress adaptation: from grass roots to novel therapeutics. Biol. Psychiatry 88, 520–530 (2020).

    Article  CAS  Google Scholar 

  257. O’Sullivan, S. E., Yates, A. S. & Porter, R. K. The peripheral cannabinoid receptor type 1 (CB1) as a molecular target for modulating body weight in man. Molecules 26, 6178 (2021).

    Article  Google Scholar 

  258. Hossain, M. Z., Ando, H., Unno, S. & Kitagawa, J. Targeting peripherally restricted cannabinoid receptor 1, cannabinoid receptor 2, and endocannabinoid-degrading enzymes for the treatment of neuropathic pain including neuropathic orofacial pain. Int. J. Mol. Sci. 21, 1423 (2020).

    Article  CAS  Google Scholar 

  259. Alger, B. E. Endocannabinoids at the synapse a decade after the dies mirabilis (29 March 2001): what we still do not know. J. Physiol. 590, 2203–2212 (2012).

    Article  CAS  Google Scholar 

  260. Castillo, P. E., Younts, T. J., Chavez, A. E. & Hashimotodani, Y. Endocannabinoid signaling and synaptic function. Neuron 76, 70–81 (2012).

    Article  CAS  Google Scholar 

  261. Di Marzo, V., Blumberg, P. M. & Szallasi, A. Endovanilloid signaling in pain. Curr. Opin. Neurobiol. 12, 372–379 (2002).

    Article  Google Scholar 

  262. Gabrielsson, L., Mattsson, S. & Fowler, C. J. Palmitoylethanolamide for the treatment of pain: pharmacokinetics, safety and efficacy. Br. J. Clin. Pharmacol. 82, 932–942 (2016).

    Article  CAS  Google Scholar 

  263. Petrosino, S. & Di Marzo, V. The pharmacology of palmitoylethanolamide and first data on the therapeutic efficacy of some of its new formulations. Br. J. Pharmacol. 174, 1349–1365 (2017).

    Article  CAS  Google Scholar 

  264. Russo, R. et al. Gut-brain axis: role of lipids in the regulation of inflammation, pain and CNS diseases. Curr. Med. Chem. 25, 3930–3952 (2018).

    Article  CAS  Google Scholar 

  265. Iannotti, F. A. & Di Marzo, V. The gut microbiome, endocannabinoids and metabolic disorders. J. Endocrinol. 248, R83–R97 (2021).

    Article  CAS  Google Scholar 

  266. Laprairie, R. B., Bagher, A. M., Kelly, M. E. & Denovan-Wright, E. M. Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br. J. Pharmacol. 172, 4790–4805 (2015).

    Article  CAS  Google Scholar 

  267. Pacher, P., Kogan, N. M. & Mechoulam, R. Beyond THC and endocannabinoids. Annu. Rev. Pharmacol. Toxicol. 60, 637–659 (2020).

    Article  CAS  Google Scholar 

  268. Spanagel, R. & Bilbao, A. Approved cannabinoids for medical purposes — comparative systematic review and meta-analysis for sleep and appetite. Neuropharmacology 196, 108680 (2021).

    Article  CAS  Google Scholar 

  269. Abuhasira, R., Shbiro, L. & Landschaft, Y. Medical use of cannabis and cannabinoids containing products — regulations in Europe and North America. Eur. J. Intern. Med. 49, 2–6 (2018).

    Article  Google Scholar 

  270. D’Hooghe, M. et al. Sativex(R) (nabiximols) cannabinoid oromucosal spray in patients with resistant multiple sclerosis spasticity: the Belgian experience. BMC Neurol. 21, 227 (2021).

    Article  Google Scholar 

  271. Leite, C. E., Mocelin, C. A., Petersen, G. O., Leal, M. B. & Thiesen, F. V. Rimonabant: an antagonist drug of the endocannabinoid system for the treatment of obesity. Pharmacol. Rep. 61, 217–224 (2009).

    Article  CAS  Google Scholar 

  272. Brodie, M. J. et al. A phase 2 randomized controlled trial of the efficacy and safety of cannabidivarin as add-on therapy in participants with inadequately controlled focal seizures. Cannabis Cannabinoid Res. 6, 528–536 (2021).

    Article  CAS  Google Scholar 

  273. van den Elsen, G. A. H. et al. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am. J. Geriatr. Psychiatry 23, 1214–1224 (2015).

    Article  Google Scholar 

  274. Yacyshyn, B. et al. Su1930–safety and efficacy of olorinab, a peripherally restricted, highly selective, cannabinoid receptor 2 agonist in a phase 2A study in chronic abdominal pain associated with Crohn’s disease. Gastroenterology 156, S-665 (2019).

    Article  Google Scholar 

  275. Bloch, M. H., Landeros-Weisenberger, A., Johnson, J. A. & Leckman, J. F. A phase-2 pilot study of a therapeutic combination of Δ9-tetrahydracannabinol and palmitoylethanolamide for adults with Tourette’s syndrome. J. Neuropsychiatry Clin. Neurosci. 33, 328–336 (2021).

    Article  Google Scholar 

  276. Light, K. & Karboune, S. Emulsion, hydrogel and emulgel systems and novel applications in cannabinoid delivery: a review. Crit. Rev. Food Sci. Nutr. https://doi.org/10.1080/10408398.2021.1926903 (2021).

    Article  Google Scholar 

  277. Liktor-Busa, E., Keresztes, A., LaVigne, J., Streicher, J. M. & Largent-Milnes, T. M. Analgesic potential of terpenes derived from Cannabis sativa. Pharmacol. Rev. 73, 98–126 (2021).

    Article  CAS  Google Scholar 

  278. Park, C., Zuo, J., Somayaji, V., Lee, B. J. & Lobenberg, R. Development of a novel cannabinoid-loaded microemulsion towards an improved stability and transdermal delivery. Int. J. Pharm. 604, 120766 (2021).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors are grateful to the National Health and Medical Research Council of Australia (Investigator Leadership Grant, APP2008727, and Development Grant APP2014250 to S.M.B.), the Australian Research Council (Discovery Project, DP180101395 to S.M.B.), Department of Veterans Affairs Senior Research Career Scientist Award (BX003610 to B.V.G.M.), Department of Veterans Affairs (Merit Grant BX002188 to B.G.V.M.), National Institutes of Health (1U01 NS113869 and 1U01NS113871 to S.M.B.; R01 NIDDK119125-01A1 to B.V.G.M.), Oklahoma Center for the Advancement of Science (HR-18-040 to B.V.G.M.), Canadian Institutes of Health Research (FDN148380 to K.A.S.), the University of Leuven (Methusalem Grant to J.T.) and the Fund for Scientific Research Flanders (Research Grants to J.T.) for support of the original research performed in the authors’ laboratories.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Keith A. Sharkey.

Ethics declarations

Competing interests

S.M.B. has provided scientific advice and received research grant support from AusHealth, Arena Pharmaceuticals, Ironwood Pharmaceuticals, Takeda Pharmaceuticals, and Sosie Heptares. B.V.G.M. has provided scientific advice and assistance to Nocion Therapeutics Inc., Teva Branded Pharmaceuticals Products R & D, Bayer Healthcare, Ironwood Pharmaceuticals, EA Pharma Co. Ltd and Blue Therapeutics. G.S. has provided scientific advice and assistance to Nextbiomics and has served on a Speaker bureau for AlfaSigma, Takeda, Farmagens and Interalia Pharmaceuticals. K.A.S. has provided scientific advice and assistance to Arena Pharmaceuticals and GW Pharmaceuticals and has served on a speaker bureau for Abbvie and received research support from Abalone Inc. M.S. has provided scientific advice and assistance to CaraCare, Bayer AG, Dr. Schwabe, Echo-Pharma, and Medice and has served on a Speaker bureau for Falkfoundation, Microbiotica, Bayer AG, Dr. Schwabe, Medice, Kyowa Kirin, Hexal, and HlH Biopharma. J.T. has provided scientific advice and assistance to Adare, AlfaSigma, Arena, Bayer, Christian Hansen, Clasado, Danone, Devintec, Falk, Grünenthal, Ironwood, Janssen, Kiowa Kirin, Menarini, Mylan, Neurogastrx, Neutec, Novartis, Nutricia, Ricordati Shionogi, Takeda, Truvion, Tsumura, Zealand, and Zeria pharmaceuticals, has served on a Speaker bureau for Abbott, Allergan, AstraZeneca, Janssen, Kiowa Kirin, Menarini, Mylan, Novartis, Shire, Takeda, Wellspect and Zeria, and has received research support from Biohit, Kiowa Kirin, Shire, Sofar and Takeda Pharmaceuticals.

Peer review

Peer review information

Nature Reviews Gastroenterology & Hepatology thanks Emeran Mayer, Ravi Bhatt, Raquel Abalo and the other anonymous reviewer for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brierley, S.M., Greenwood-Van Meerveld, B., Sarnelli, G. et al. Targeting the endocannabinoid system for the treatment of abdominal pain in irritable bowel syndrome. Nat Rev Gastroenterol Hepatol 20, 5–25 (2023). https://doi.org/10.1038/s41575-022-00682-y

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-022-00682-y

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing