2,724
Views
5
CrossRef citations to date
0
Altmetric
Research Paper

Gut microbiota modulates visceral sensitivity through calcitonin gene-related peptide (CGRP) production

, , , , , & ORCID Icon show all
Article: 2188874 | Received 01 Nov 2022, Accepted 27 Feb 2023, Published online: 20 Mar 2023

References

  • Guthrie E, Thompson D. Abdominal pain and functional gastrointestinal disorders. Bmj. 2002;325(7366):701–15. doi:10.1136/bmj.325.7366.701.
  • Enck P, Aziz Q, Barbara G, Farmer AD, Fukudo S, Mayer EA, Niesler B, Quigley EMM, Rajilić-Stojanović M, Schemann M, et al. Irritable bowel syndrome. Nat Rev Dis Primers. 2016;2(1):16014. doi:10.1038/nrdp.2016.14.
  • Lembo A. Irritable bowel syndrome medications side effects survey. J Clin Gastroenterol. 2004;38(9):776–781. doi:10.1097/01.mcg.0000139029.00451.c7.
  • Camilleri M, Boeckxstaens G. Dietary and pharmacological treatment of abdominal pain in IBS. Gut. 2017;66:966–974. doi:10.1136/gutjnl-2016-313425.
  • Fichna J, Storr MA. Brain-Gut Interactions in IBS. Front Pharmacol. 2012;3:127. doi:10.3389/fphar.2012.00127.
  • Grover M, Drossman DA. Functional abdominal pain. Curr Gastroenterol Rep. 2010;12(5):391–398. doi:10.1007/s11894-010-0125-0.
  • Ossipov MH, Morimura K, Porreca F. Descending pain modulation and chronification of pain. Curr Opin Support Palliat Care. 2014;8(2):143–151. doi:10.1097/spc.0000000000000055.
  • Yam MF, Loh Y, Tan C, Khadijah Adam S, Abdul Manan N, Basir R. General pathways of pain sensation and the major neurotransmitters involved in pain regulation. Int J Mol Sci. 2018;19(8):2164. doi:10.3390/ijms19082164.
  • Ossipov MH, Dussor GO, Porreca F. Central modulation of pain. J Clin Invest. 2010;120(11):3779–3787. doi:10.1172/jci43766.
  • Garland EL. Pain processing in the human nervous system: a selective review of nociceptive and biobehavioral pathways. Prim Care. 2012;39(3):561–571. doi:10.1016/j.pop.2012.06.013.
  • Schou WS, Ashina S, Amin FM, Goadsby PJ, Ashina M. Calcitonin gene-related peptide and pain: a systematic review. J Headache Pain. 2017;18(1):34. doi:10.1186/s10194-017-0741-2.
  • Zieglgänsberger W. Substance P and pain chronicity. Cell Tissue Res. 2019;375(1):227–241. doi:10.1007/s00441-018-2922-y.
  • Iyengar S, Ossipov MH, Johnson KW. The role of calcitonin gene–related peptide in peripheral and central pain mechanisms including migraine. Pain. 2017;158(4):543–559. doi:10.1097/j.pain.0000000000000831.
  • Vass Z, Dai CF, Steyger PS, Jancsó G, Trune DR, Nuttall AL. Co-localization of the vanilloid capsaicin receptor and substance P in sensory nerve fibers innervating cochlear and vertebro-basilar arteries. Neuroscience. 2004;124(4):919–927. doi:10.1016/j.neuroscience.2003.12.030.
  • Yang NJ, Chiu IM. Bacterial Signaling to the Nervous System through Toxins and Metabolites. J Mol Biol. 2017;429(5):587–605. doi:10.1016/j.jmb.2016.12.023.
  • Luczynski P, Tramullas M, Viola M, Shanahan F, Clarke G, O’Mahony S, Dinan TG, Cryan JF. Microbiota regulates visceral pain in the mouse. Elife. 2017;6. doi:10.7554/eLife.25887.
  • Verdú EF, et al. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut. 2006;55:182–190. doi:10.1136/gut.2005.066100.
  • O’mahony SM, V. D. Felice, K. Nally, H. M. Savignac, M. J. Claesson, P. Scully, J. Woznicki. Disturbance of the gut microbiota in early-life selectively affects visceral pain in adulthood without impacting cognitive or anxiety-related behaviors in male rats. Neuroscience. 2014;277:885–901. doi:10.1016/j.neuroscience.2014.07.054.
  • Eutamene H, Lamine F, Chabo C, Theodorou V, Rochat F, Bergonzelli GE, Corthésy-Theulaz I, Fioramonti J, Bueno L. Synergy between lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J Nutr. 2007;137(8):1901–1907. doi:10.1093/jn/137.8.1901.
  • Rousseaux C, Thuru X, Gelot A, Barnich N, Neut C, Dubuquoy L, Dubuquoy C, Merour E, Geboes K, Chamaillard M, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med. 2007;13(1):35–37. doi:10.1038/nm1521.
  • Kannampalli P, Pochiraju S, Chichlowski M, Berg BM, Rudolph C, Bruckert M, Miranda A, Sengupta JN. Probiotic Lactobacillus rhamnosus GG (LGG) and prebiotic prevent neonatal inflammation-induced visceral hypersensitivity in adult rats. Neurogastroenterol Motil. 2014;26(12):1694–1704. doi:10.1111/nmo.12450.
  • Miquel S, Martín R, Lashermes A, Gillet M, Meleine M, Gelot A, Eschalier A, Ardid D, Bermúdez-Humarán LG, Sokol H, et al. Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep. 2016;6(1):19399. doi:10.1038/srep19399.
  • Amaral FA, Sachs D, Costa VV, Fagundes CT, Cisalpino D, Cunha TM, Ferreira SH, Cunha FQ, Silva TA, Nicoli JR, et al. Commensal microbiota is fundamental for the development of inflammatory pain. Proc Natl Acad Sci U S A. 2008;105(6):2193–2197. doi:10.1073/pnas.0711891105.
  • Chiu IM, Heesters BA, Ghasemlou N, Von Hehn CA, Zhao F, Tran J, Wainger B, Strominger A, Muralidharan S, Horswill AR, et al. Bacteria activate sensory neurons that modulate pain and inflammation. Nature. 2013;501(7465):52–57. doi:10.1038/nature12479.
  • Pinho-Ribeiro FA, Baddal B, Haarsma R, O’seaghdha M, Yang NJ, Blake KJ, Portley M, Verri WA, Dale JB, Wessels MR, et al. Blocking neuronal signaling to immune cells treats streptococcal invasive infection. Cell. 2018;173(5):1083–1097.e1022. doi:10.1016/j.cell.2018.04.006.
  • Pérez-Berezo T, Pujo J, Martin P, Le Faouder P, Galano J-M, Guy A, Knauf C, Tabet JC, Tronnet S, Barreau F, et al. Identification of an analgesic lipopeptide produced by the probiotic Escherichia coli strain Nissle 1917. Nat Commun. 2017;8(1):1314. doi:10.1038/s41467-017-01403-9.
  • Yang NJ, Isensee J, Neel DV, Quadros AU, Zhang HXB, Lauzadis J, Liu SM, Shiers S, Belu A, Palan S, et al. Anthrax toxins regulate pain signaling and can deliver molecular cargoes into ANTXR2(+) DRG sensory neurons. Nat Neurosci. 2022;25(2):168–179. doi:10.1038/s41593-021-00973-8.
  • Tramullas M, Collins JM, Fitzgerald P, Dinan TG, O’ Mahony SM, Cryan JF. Estrous cycle and ovariectomy-induced changes in visceral pain are microbiota-dependent. iScience. 2021;24(8):102850. doi:10.1016/j.isci.2021.102850.
  • Sorge RE, LaCroix-Fralish ML, Tuttle AH, Sotocinal SG, Austin J-S, Ritchie J, Chanda ML, Graham AC, Topham L, Beggs S, et al. Spinal cord Toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J Neurosci. 2011;31(43):15450–15454. doi:10.1523/jneurosci.3859-11.2011.
  • Sorge RE, Mapplebeck JCS, Rosen S, Beggs S, Taves S, Alexander JK, Martin LJ, Austin J-S, Sotocinal SG, Chen D, et al. Different immune cells mediate mechanical pain hypersensitivity in male and female mice. Nat Neurosci. 2015;18(8):1081–1083. doi:10.1038/nn.4053.
  • Francis-Malavé AM, Martínez-González S, Pichardo C, Wilson TD, Rivera-García LG, Brinster LR, Carrasquillo Y. Sex differences in pain-related behaviors and clinical progression of disease in mouse models of colonic pain. Pain. 2023;164(1):197-215. doi:10.1097/j.pain.0000000000002683.
  • Aloisi AM, Affaitati G, Ceccarelli I, Fiorenzani P, Lerza R, Rossi C, Pace MC, Chiefari M, Aurilio C, Giamberardino MA. Estradiol and testosterone differently affect visceral pain-related behavioural responses in male and female rats. Eur J Pain. 2010;14(6):602–607. doi:10.1016/j.ejpain.2009.10.016.
  • Ji Y, Hu B, Li J, Traub RJ. Opposing roles of estradiol and testosterone on stress-induced visceral hypersensitivity in rats. J Pain. 2018;19(7):764–776. doi:10.1016/j.jpain.2018.02.007.
  • Chen J, Li Q, Saliuk G, Bazhanov S, Winston JH. Estrogen and serotonin enhance stress-induced visceral hypersensitivity in female rats by up-regulating brain-derived neurotrophic factor in spinal cord. Neurogastroenterol Motil. 2021;33(10):e14117. doi:10.1111/nmo.14117.
  • Bartley EJ, Fillingim RB. Sex differences in pain: a brief review of clinical and experimental findings. Br J Anaesth. 2013;111(1):52–58. doi:10.1093/bja/aet127.
  • Chang L, Mayer EA, Labus JS, Schmulson M, Lee OY, Olivas TI, Stains J, Naliboff BD. Effect of sex on perception of rectosigmoid stimuli in irritable bowel syndrome. Am J Physiol Regul Integr Comp Physiol. 2006;291(2):R277–284. doi:10.1152/ajpregu.00729.2005.
  • Kano M, Farmer AD, Aziz Q, Giampietro VP, Brammer MJ, Williams SCR, Fukudo S, Coen SJ. Sex differences in brain response to anticipated and experienced visceral pain in healthy subjects. Am J Physiol Gastrointest Liver Physiol. 2013;304(8):G687–699. doi:10.1152/ajpgi.00385.2012.
  • Petitfils C, Maurel S, Payros G, Hueber A, Agaiz B, Gazzo G, Marrocco R, Auvray F, Langevin G, Motta J-P, et al. Identification of bacterial lipopeptides as key players in IBS. Gut. 2022; gutjnl-2022-328084. doi:10.1136/gutjnl-2022-328084.
  • De Palma G, Shimbori C, Reed DE, Yu Y, Rabbia V, Lu J, Jimenez-Vargas N, Sessenwein J, Lopez-Lopez C, Pigrau M, et al. Histamine production by the gut microbiota induces visceral hyperalgesia through histamine 4 receptor signaling in mice. Sci Transl Med. 2022;14(655):eabj1895. doi:10.1126/scitranslmed.abj1895.
  • Fattori V, Hohmann MS, Rossaneis AC, Pinho-Ribeiro FA, Verri WAC. Current understanding of its mechanisms and therapy of pain and other pre-clinical and clinical uses. Molecules. 2016;21(7):844. doi:10.3390/molecules21070844.
  • Gosens R, Zaagsma J, Meurs H, Halayko AJ. Muscarinic receptor signaling in the pathophysiology of asthma and COPD. Respir Res. 2006;7(1):73. doi:10.1186/1465-9921-7-73.
  • Farzaei MH, Bahramsoltani R, Abdollahi M, Rahimi R. The role of visceral hypersensitivity in irritable bowel syndrome: pharmacological targets and novel treatments. J Neurogastroenterol Motil. 2016;22(4):558–574. doi:10.5056/jnm16001.
  • Chiu IM, von Hehn CA, Woolf CJ. Neurogenic inflammation and the peripheral nervous system in host defense and immunopathology. Nat Neurosci. 2012;15(8):1063–1067. doi:10.1038/nn.3144.
  • Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol. 2015;28:203–209.
  • Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–141. doi:10.1016/j.cell.2014.03.011.
  • Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, Hibberd ML, Forssberg H, Pettersson S. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011;108(7):3047–3052. doi:10.1073/pnas.1010529108.
  • Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol Motil. 2011;23(3):255–264, e119. doi:10.1111/j.1365-2982.2010.01620.x.
  • Luo Y, Zeng B, Zeng L, Du X, Li B, Huo R, Liu L, Wang H, Dong M, Pan J, et al. Gut microbiota regulates mouse behaviors through glucocorticoid receptor pathway genes in the hippocampus. Transl Psychiatry. 2018;8(1):187. doi:10.1038/s41398-018-0240-5.
  • Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu X-N, Kubo C, Koga Y. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004;558(1):263–275. doi:10.1113/jphysiol.2004.063388.
  • Holzer P, Farzi A. Neuropeptides and the microbiota-gut-brain axis. Adv Exp Med Biol. 2014;817:195–219. doi:10.1007/978-1-4939-0897-4_9.
  • Castagliuolo I, Keates AC, Qiu B, Kelly CP, Nikulasson S, Leeman SE, Pothoulakis C. Increased substance P responses in dorsal root ganglia and intestinal macrophages during Clostridium difficile toxin a enteritis in rats. Proc Natl Acad Sci U S A. 1997;94(9):4788–4793. doi:10.1073/pnas.94.9.4788.
  • Lomasney KW, Houston A, Shanahan F, Dinan TG, Cryan JF, Hyland NP. Selective influence of host microbiota on cAMP-mediated ion transport in mouse colon. Neurogastroenterol Motil. 2014;26(6):887–890. doi:10.1111/nmo.12328.
  • Aguilera M, Cerdà-Cuéllar M, Martínez V. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6:10–23. doi:10.4161/19490976.2014.990790.
  • Aguilera M, Rossini V, Hickey A, Simnica D, Grady F, Felice VD, Moloney A, Pawley L, Fanning A, McCarthy L, et al. Inflammasome signaling regulates the microbial–neuroimmune axis and visceral pain in mice. Int J Mol Sci. 2021;22(15):8336. doi:10.3390/ijms22158336.
  • Hayes CL, Dong J, Galipeau HJ, Jury J, McCarville J, Huang X, Wang X-Y, Naidoo A, Anbazhagan AN, Libertucci J, et al. Commensal microbiota induces colonic barrier structure and functions that contribute to homeostasis. Sci Rep. 2018;8(1):14184. doi:10.1038/s41598-018-32366-6.
  • Hurley RW, Adams MC. Sex, gender, and pain: an overview of a complex field. Anesth Analg. 2008;107(1):309–317. doi:10.1213/01.ane.0b013e31816ba437.
  • Hartmann EM, Handwerker HO, Forster C. Gender differences in itch and pain-related sensations provoked by histamine, cowhage and capsaicin. Acta Derm Venereol. 2015;95(1):25–30. doi:10.2340/00015555-1894.
  • Bubb KJ, Wen H, Panayiotou CM, Finsterbusch M, Khan FJ, Chan MV, Priestley JV, Baker MD, Ahluwalia A. Activation of neuronal transient receptor potential vanilloid 1 channel underlies 20-hydroxyeicosatetraenoic acid–induced vasoactivity. Hypertension. 2013;62(2):426–433. doi:10.1161/hypertensionaha.111.00942.
  • Paige C, Plasencia-Fernandez I, Kume M, Papalampropoulou-Tsiridou M, Lorenzo L-E, David ET, He L, Mejia GL, Driskill C, Ferrini F, et al. A Female-Specific Role for Calcitonin Gene-Related Peptide (CGRP) in rodent pain models. J Neurosci. 2022;42(10):1930–1944. doi:10.1523/jneurosci.1137-21.2022.
  • Avona A, Burgos-Vega C, Burton MD, Akopian AN, Price TJ, Dussor G. Dural calcitonin gene-related peptide produces female-specific responses in rodent migraine models. J Neurosci. 2019;39(22):4323–4331. doi:10.1523/jneurosci.0364-19.2019.
  • Gottesman-Katz L, Latorre R, Vanner S, Schmidt BL, Bunnett NW. Targeting G protein-coupled receptors for the treatment of chronic pain in the digestive system. Gut. 2021;70:970–981. doi:10.1136/gutjnl-2020-321193.
  • Yano JM, Yu K, Donaldson G, Shastri G, Ann P, Ma L, Nagler C, Ismagilov R, Mazmanian S, Hsiao E. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161(2):264–276. doi:10.1016/j.cell.2015.02.047.