3,904
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

Pasteurized Akkermansia muciniphila improves irritable bowel syndrome-like symptoms and related behavioral disorders in mice

ORCID Icon, , , , , , ORCID Icon, , ORCID Icon, , ORCID Icon, , ORCID Icon, , ORCID Icon & ORCID Icon show all
Article: 2298026 | Received 20 Mar 2023, Accepted 18 Dec 2023, Published online: 03 Jan 2024

References

  • Drossman DA, Hasler WL. Rome IV—functional GI disorders: disorders of gut-brain interaction. Gastroenterology. 2016;150(6):1257–21. doi:10.1053/j.gastro.2016.03.035.
  • Hillestad EMR, van der Meeren A, Nagaraja BH, Bjørsvik BR, Haleem N, Benitez-Paez A, Sanz Y, Hausken T, Lied GA, Lundervold A, et al. Gut bless you: the microbiota-gut-brain axis in irritable bowel syndrome. World J Gastroentero. 2022;28(4):412–431. doi:10.3748/wjg.v28.i4.412.
  • Barbara G, Grover M, Bercik P, Corsetti M, Ghoshal UC, Ohman L, Rajilić-Stojanović M. Rome foundation working team report on post-infection irritable bowel syndrome. Gastroenterology. 2019;156(1):46–58.e7. doi:10.1053/j.gastro.2018.07.011.
  • Nicholl BI, Halder SL, Macfarlane GJ, Thompson DG, O’Brien S, Musleh M, McBeth J. Psychosocial risk markers for new onset irritable bowel syndrome–results of a large prospective population-based study. Pain. 2008;137(1):147–155. doi:10.1016/j.pain.2007.08.029.
  • Camilleri M, Boeckxstaens G. Irritable bowel syndrome: treatment based on pathophysiology and biomarkers. Gut. 2023;72(3):590–599. doi:10.1136/gutjnl-2022-328515.
  • Barreau F, Ferrier L, Fioramonti J, Bueno L. New insights in the etiology and pathophysiology of irritable bowel syndrome: contribution of neonatal stress models. Pediatr Res. 2007;62(3):240–245. doi:10.1203/PDR.0b013e3180db2949.
  • Mallaret G, Lashermes A, Meleine M, Boudieu L, Barbier J, Aissouni Y, Gelot A, Chassaing B, Gewirtz AT, Ardid D, et al. Involvement of toll-like receptor 5 in mouse model of colonic hypersensitivity induced by neonatal maternal separation. World J Gastroenterol. 2022;28(29):3903–3916. doi:10.3748/wjg.v28.i29.3903.
  • Hanning N, Edwinson AL, Ceuleers H, Peters SA, De Man JG, Hassett LC, De Winter BY, Grover M. Intestinal barrier dysfunction in irritable bowel syndrome: a systematic review. Therap Adv Gastroenterol. 2021;14:1756284821993586. doi:10.1177/1756284821993586.
  • Mondelaers SU, Theofanous SA, Florens MV, Perna E, Aguilera-Lizarraga J, Boeckxstaens G, Wouters M. Effect of genetic background and postinfectious stress on visceral sensitivity in citrobacter rodentium-infected mice. Neurogastroent Motil. 2016;28(5):647–658. doi:10.1111/nmo.12759.
  • Collins JW, Keeney KM, Crepin VF, Rathinam VAK, Fitzgerald KA, Finlay BB, Frankel G. Citrobacter rodentium: infection, inflammation and the microbiota. Nat Rev Microbiol. 2014;12(9):612–623. doi:10.1038/nrmicro3315.
  • Meynier M, Baudu E, Rolhion N, Defaye M, Daugey V, Modoux M, Wawrzyniak I, Villéger R, Méleine M, Borras Nogues E, et al. AhR/IL-22 pathway as new target for the treatment of post-infectious irritable bowel syndrome symptoms. Gut Microbes. 2022;14(1):14. doi:10.1080/19490976.2021.2022997.
  • Ibeakanma C, Miranda-Morales M, Richards M, Bautista-Cruz F, Martin N, Hurlbut D, Vanner S. Citrobacter rodentium colitis evokes post-infectious hyperexcitability of mouse nociceptive colonic dorsal root ganglion neurons. J Physiol. 2009;587(14):3505–3521. doi:10.1113/jphysiol.2009.169110.
  • Dinan TG, Cryan JF. The microbiome-gut-brain axis in health and disease. Gastroenterol Clin North Am. 2017;46(1):77–89. doi:10.1016/j.gtc.2016.09.007.
  • Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia municiphila gen. Nov., sp. Nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol. 2004;54(5):1469–1476. doi:10.1099/ijs.0.02873-0.
  • Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:9066–9071.
  • Cani PD, Depommier C, Derrien M, Everard A, de Vos WM. Akkermansia muciniphila: paradigm for next-generation beneficial microorganisms. Nat Rev Gastroenterol Hepatol. 2022;19(10):625–637. doi:10.1038/s41575-022-00631-9.
  • Karlsson CLJ, Önnerfält J, Xu J, Molin G, Ahrné S, Thorngren-Jerneck K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity. 2012;20(11):2257–2261. doi:10.1038/oby.2012.110.
  • Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GM, Neyrinck AM, Possemiers S, Van Holle A, François P, De Vos WM, et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes. 2011;60(11):2775–2786. doi:10.2337/db11-0227.
  • Plovier H, Everard A, Druart C, Depommier C, Van Hul M, Geurts L, Chilloux J, Ottman N, Duparc T, Lichtenstein L, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med. 2017;23(1):107–113. doi:10.1038/nm.4236.
  • Depommier C, Van Hul M, Everard A, Delzenne NM, De Vos WM, Cani PD. Pasteurized Akkermansia muciniphila increases whole-body energy expenditure and fecal energy excretion in diet-induced obese mice. Gut Microbes. 2020;11(5):1231–1245. doi:10.1080/19490976.2020.1737307.
  • Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, Falony G, Raes J, Maiter D, Delzenne NM, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096–1103. doi:10.1038/s41591-019-0495-2.
  • Xu D, Chen VL, Steiner CA, Berinstein JA, Eswaran S, Waljee AK, Higgins PDR, Owyang C. Efficacy of fecal microbiota transplantation in irritable bowel syndrome: a systematic review and meta-analysis. Am J Gastroenterol. 2019;114(7):1043–1050. doi:10.14309/ajg.0000000000000198.
  • Ford AC, Harris LA, Lacy BE, Quigley EMM, Moayyedi P. Systematic review with meta-analysis: the efficacy of prebiotics, probiotics, synbiotics and antibiotics in irritable bowel syndrome. Aliment Pharmacol Ther. 2018;48(10):1044–1060. doi:10.1111/apt.15001.
  • Cruz-Aguliar RM, Wantia N, Clavel T, Vehreschild MJGT, Buch T, Bajbouj M, Haller D, Busch D, Schmid RM, Stein-Thoeringer CK. An open-labeled study on fecal microbiota transfer in irritable bowel syndrome patients reveals improvement in abdominal pain associated with the relative abundance of Akkermansia muciniphila. Digestion. 2019;100(2):127–138. doi:10.1159/000494252.
  • 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.
  • Xin Y, Wang J, Chu T, Zhou Y, Liu C, Xu A. Electroacupuncture alleviates neuroinflammation by inhibiting the HMGB1 signaling pathway in rats with sepsis-associated encephalopathy. Brain Sci. 2022;12(12):1732. doi:10.3390/brainsci12121732.
  • Iban-Arias R, Trageser KJ, Yang E-J, Griggs E, Radu A, Naughton S, Al Rahim M, Tatsunori O, Raval U, Palmieri J, et al. Exposure to world trade center dust exacerbates cognitive impairment and evokes a central and peripheral pro-inflammatory transcriptional profile in an animal model of alzheimer’s disease. J Alzheimers Dis. 2022;91(2):779–794. doi:10.3233/JAD-221046.
  • 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.
  • Perez-Burgos A, Wang L, McVey Neufeld K-A, Mao Y-K, Ahmadzai M, Janssen LJ, Stanisz AM, Bienenstock J, Kunze WA. The TRPV1 channel in rodents is a major target for antinociceptive effect of the probiotic lactobacillus reuteri DSM 17938. J Physiol. 2015;593:3943–3957. doi:10.1113/JP270229.
  • Simrén M, Törnblom H, Palsson OS, van Tilburg MAL, Van Oudenhove L, Tack J, Whitehead WE. Visceral hypersensitivity is associated with GI symptom severity in functional GI disorders: consistent findings from five different patient cohorts. Gut. 2018;67:255–262. doi:10.1136/gutjnl-2016-312361.
  • Ford AC, Sperber AD, Corsetti M, Camilleri M. Irritable bowel syndrome. Lancet. 2020;396(10263):1675–1688. doi:10.1016/S0140-6736(20)31548-8.
  • Drossman DA, Tack J, Ford AC, Szigethy E, Törnblom H, Van Oudenhove L. Neuromodulators for functional gastrointestinal disorders (disorders of gut−brain interaction): a Rome foundation working team report. Gastroenterology. 2018;154(4):1140–1171.e1. doi:10.1053/j.gastro.2017.11.279.
  • Camilleri M, Boeckxstaens G. Dietary and pharmacological treatment of abdominal pain in IBS. Gut. 2017;66(5):966–974. doi:10.1136/gutjnl-2016-313425.
  • Wilmes L, Collins JM, O’Riordan KJ, O’Mahony SM, Cryan JF, Clarke G. Of bowels, brain and behavior: a role for the gut microbiota in psychiatric comorbidities in irritable bowel syndrome. Neurogastroenterol Motil. 2021;33(3):e14095. doi:10.1111/nmo.14095.
  • Coutinho SV, Plotsky PM, Sablad M, Miller JC, Zhou H, Bayati AI, McRoberts JA, Mayer EA. Neonatal maternal separation alters stress-induced responses to viscerosomatic nociceptive stimuli in rat. Am J Physiol Gastrointest Liver Physiol. 2002;282(2):G307–G316. doi:10.1152/ajpgi.00240.2001.
  • Meleine M, Boudieu L, Gelot A, Muller E, Lashermes A, Matricon J, Silberberg C, Theodorou V, Eschalier A, Ardid D, et al. Comparative effects of α2δ-1 ligands in mouse models of colonic hypersensitivity. World J Gastroenterol. 2016;22(31):7111–7123. doi:10.3748/wjg.v22.i31.7111.
  • Chen Y, Xiao S, Gong Z, Zhu X, Yang Q, Li Y, Gao S, Dong Y, Shi Z, Wang Y, et al. Wuji wan formula ameliorates diarrhea and disordered colonic motility in post-inflammation irritable bowel syndrome rats by modulating the gut microbiota. Front Microbiol. 2017;8:2307. doi:10.3389/fmicb.2017.02307.
  • Jacobs JP, Gupta A, Bhatt RR, Brawer J, Gao K, Tillisch K, Lagishetty V, Firth R, Gudleski GD, Ellingson BM, et al. Cognitive behavioral therapy for irritable bowel syndrome induces bidirectional alterations in the brain-gut-microbiome axis associated with gastrointestinal symptom improvement. Microbiome. 2021;9(1):236. doi:10.1186/s40168-021-01188-6.
  • Morais LH, Schreiber HL, Mazmanian SK. The gut microbiota–brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19(4):241–255. doi:10.1038/s41579-020-00460-0.
  • Ji N-N, Jiang H, Xia M. The influence of the enriched environment in different periods on neonatal maternal separation-induced visceral pain, anxiousness, and depressive behaviors. Transl Pediatr. 2022;11(9):1562–1569. doi:10.21037/tp-22-475.
  • Pontifex MG, Connell E, Le Gall G, Pourtau L, Gaudout D, Angeloni C, Zallocco L, Ronci M, Giusti L, Müller M, et al. Saffron extract (Safr’inside™) improves anxiety related behaviour in a mouse model of low-grade inflammation through the modulation of the microbiota and gut derived metabolites. Food Funct. 2022;13(23):12219–12233. doi:10.1039/D2FO02739A.
  • Sun Y, Zhu H, Cheng R, Tang Z, Zhang M. Outer membrane protein Amuc_1100 of Akkermansia muciniphila alleviates antibiotic-induced anxiety and depression-like behavior in mice. Physiol Behavior. 2023;258:114023. doi:10.1016/j.physbeh.2022.114023.
  • Bae M, Cassilly CD, Liu X, Park S-M, Tusi BK, Chen X, Kwon J, Filipčík P, Bolze AS, Liu Z, et al. Akkermansia muciniphila phospholipid induces homeostatic immune responses. Nature. 2022;608(7921):168–173. doi:10.1038/s41586-022-04985-7.
  • Gu Z, Pei W, Shen Y, Wang L, Zhu J, Zhang Y, Fan S, Wu Q, Li L, Zhang Z. Akkermansia muciniphila and its outer protein Amuc_1100 regulates tryptophan metabolism in colitis. Food Funct. 2021;12(20):10184–10195. doi:10.1039/D1FO02172A.
  • Bachmann R, Van Hul M, Baldin P, Léonard D, Delzenne NM, Belzer C, Ouwerkerk JP, Repsilber D, Rangel I, Kartheuser A, et al. Akkermansia muciniphila reduces peritonitis and improves intestinal tissue wound healing after a colonic transmural defect by a MyD88-dependent mechanism. Cells. 2022;11(17):2666. doi:10.3390/cells11172666.
  • Lapointe TK, Basso L, Iftinca MC, Flynn R, Chapman K, Dietrich G, Vergnolle N, Altier C. TRPV1 sensitization mediates postinflammatory visceral pain following acute colitis. Am J Physiol-Gastr L. 2015;309(2):G87–99. doi:10.1152/ajpgi.00421.2014.
  • Akbar A, Yiangou Y, Facer P, Walters JRF, Anand P, Ghosh S. Increased capsaicin receptor TRPV1-expressing sensory fibres in irritable bowel syndrome and their correlation with abdominal pain. Gut. 2008;57(7):923–929. doi:10.1136/gut.2007.138982.
  • Najjar SA, Davis BM, Albers KM. Epithelial–neuronal communication in the colon: implications for visceral pain. Trends Neurosci. 2020;43(3):170–181. doi:10.1016/j.tins.2019.12.007.
  • Jalanka J, Lam C, Bennett A, Hartikainen A, Crispie F, Finnegan LA, Cotter PD, Spiller R. Colonic gene expression and fecal microbiota in diarrhea-predominant irritable bowel syndrome: increased toll-like receptor 4 but minimal inflammation and no response to Mesalazine. J Neurogastroenterol Motil. 2021;27(2):279–291. doi:10.5056/jnm20205.
  • Barbara G, Wang B, Stanghellini V, de Giorgio R, Cremon C, Di Nardo G, Trevisani M, Campi B, Geppetti P, Tonini M, et al. Mast cell-dependent excitation of visceral-nociceptive sensory neurons in irritable bowel syndrome. Gastroenterology. 2007;132(1):26–37. doi:10.1053/j.gastro.2006.11.039.
  • Sugiuar T, Bielefeldt K, Gebhart GF. TRPV1 function in mouse colon sensory neurons is enhanced by metabotropic 5-hydroxytryptamine receptor activation. J Neurosci. 2004;24(43):9521–9530. doi:10.1523/JNEUROSCI.2639-04.2004.
  • Mistry S, Paule CC, Varga A, Photiou A, Jenes A, Avelino A, Buluwela L, Nagy I. Prolonged exposure to bradykinin and prostaglandin E2 increases TRPV1 mRNA but does not alter TRPV1 and TRPV1b protein expression in cultured rat primary sensory neurons. Neurosci Lett. 2014;564:89–93. doi:10.1016/j.neulet.2014.02.006.
  • Wouters MM, Balemans D, Van Wanrooy S, Dooley J, Cibert-Goton V, Alpizar YA, Valdez-Morales EE, Nasser Y, Van Veldhoven PP, Vanbrabant W, et al. Histamine receptor H1–mediated sensitization of TRPV1 mediates visceral hypersensitivity and symptoms in patients with irritable bowel syndrome. Gastroenterology. 2016;150(4):875–887.e9. doi:10.1053/j.gastro.2015.12.034.
  • Shen W, Shen M, Zhao X, Zhu H, Yang Y, Lu S, Tan Y, Li G, Li M, Wang J, et al. Anti-obesity effect of capsaicin in mice fed with high-fat diet is associated with an increase in population of the gut bacterium Akkermansia muciniphila. Front Microbiol. 2017;8:272. doi:10.3389/fmicb.2017.00272.
  • Gong T, Wang H, Liu S, Zhang M, Xie Y, Liu X. Capsaicin regulates lipid metabolism through modulation of bile acid/gut microbiota metabolism in high-fat-fed SD rats. Food Nutr Res. 2022;66:66. doi:10.29219/fnr.v66.8289.
  • Fried DE, Gulbransen BD. In situ Ca2+ imaging of the enteric nervous system. J Vis Exp. 2015;95. doi:10.3791/52506-v.
  • Dinan TG, Cryan JF. The impact of gut microbiota on brain and behaviour: implications for psychiatry. Curr Opin Clin Nutr Metab Care. 2015;18(6):552–558. doi:10.1097/MCO.0000000000000221.
  • Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain. 1983;16(2):109–110. doi:10.1016/0304-3959(83)90201-4.
  • Larauche M, Gourcerol G, Million M, Adelson DW, Taché Y. Repeated psychological stress-induced alterations of visceral sensitivity and colonic motor functions in mice: influence of surgery and postoperative single housing on visceromotor responses. Stress. 2010;13(4):344–355. doi:10.3109/10253891003664166.
  • Picard E, Carvalho FA, Agosti F, Bourinet E, Ardid D, Eschalier A, Daulhac L, Mallet C. Inhibition of cav 3.2 calcium channels: a new target for colonic hypersensitivity associated with low-grade inflammation. Br J Pharmacol. 2019;176(7):950–963. doi:10.1111/bph.14608.
  • Rodríguez Echandía EL, Broitman ST, Fóscolo MR. Effect of the chronic ingestion of chlorimipramine and desipramine on the hole board response to acute stresses in male rats. Pharmacol Biochem Behav. 1987;26(2):207–210. doi:10.1016/0091-3057(87)90106-7.
  • Sierksma ASR, Van Den Hove DLA, Pfau F, Philippens M, Bruno O, Fedele E, Ricciarelli R, Steinbusch HWM, Vanmierlo T, Prickaerts J. Improvement of spatial memory function in APPswe/PS1dE9 mice after chronic inhibition of phosphodiesterase type 4D. Neuropharmacology. 2014;77:120–130. doi:10.1016/j.neuropharm.2013.09.015.
  • Barre A, Berthoux C, De Bundel D, Valjent E, Bockaert J, Marin P, Bécamel C Presynaptic serotonin 2A receptors modulate thalamocortical plasticity and associative learning. Proceedings of the National Academy of Sciences of the United States of America 2016;113:E1382–91 doi:10.1073/pnas.1525586113.
  • Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, Alexander H, Alm EJ, Arumugam M, Asnicar F, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37(8):852–857. doi:10.1038/s41587-019-0209-9.
  • Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581–583. doi:10.1038/nmeth.3869.
  • Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glöckner FO. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41(D1):D590–D596. doi:10.1093/nar/gks1219.