Gut Microbiome as Target for Innovative Strategies Against Food Allergy

Roberto Berni Canani, Lorella Paparo, Rita Nocerino, Carmen Di Scala, Giusy Della Gatta, Ylenia Maddalena, Aniello Buono, Cristina Bruno, Luana Voto, Danilo Ercolini, Roberto Berni Canani, Lorella Paparo, Rita Nocerino, Carmen Di Scala, Giusy Della Gatta, Ylenia Maddalena, Aniello Buono, Cristina Bruno, Luana Voto, Danilo Ercolini

Abstract

The dramatic increase in food allergy prevalence and severity globally requires effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota function and structure, and microbiome alterations (dysbiosis) have a pivotal role in the development of food allergy. Environmental factors, including a low-fiber/high-fat diet, cesarean delivery, antiseptic agents, lack of breastfeeding, and drugs can induce gut microbiome dysbiosis, and have been associated with food allergy. New experimental tools and technologies have provided information regarding the role of metabolites generated from dietary nutrients and selected probiotic strains that could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence has provided useful information on optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge of the crucial role played by nutrients and gut microbiota-derived metabolites is opening the way to a post-biotic approach in the stimulation of immune tolerance through epigenetic regulation. This review focused on the potential role of gut microbiome as the target for innovative strategies against food allergy.

Keywords: butyrate; dysbiosis; gut microbiota; gut microbiota metabolites; immune tolerance; mediterranean diet; probiotics; short chain fatty acids.

Figures

Figure 1
Figure 1
Gut microbiome as a target of intervention against food allergy. Several genetic, environmental, and dietary factors could modulate the gut microbiome-immune system axis influencing the occurrence of FA. For instance, increased family size, exposure to pets and/or rural environment, healthy diet (full of fibers, fermented foods, antioxidants, omega-3), breastfeeding and use of probiotics are associated with protection to FA. Conversely, C-section, prenatal, and early-life exposure to antibiotics/gastric acidity inhibitors/antiseptic agents, unhealthy diet (low fibers/high saturated fats and junk foods) may increase the risk for the development of FA. All these environmental factors act mainly on a modulation of gut microbiota structure and function which in turn could be responsible for the epigenetic regulation of genes involved in immune tolerance.
Figure 2
Figure 2
The Food Allergy pyramid. Children with FA present an increased risk to develop other conditions such as allergic disorders (atopic march), inflammatory bowel diseases (IBD), functional gastrointestinal disorders (FGIDs), and neuropsychiatric disorders. Several genetic factors are implicated in the pathogenesis of these conditions, but recent evidence suggest the pivotal role of gut microbiome dysbiosis (induced by environmental factors). Emerging evidence support the hypothesis of dysbiosis as the first hit in the development of alterations in intestinal barrier and immune system function (responsible for the occurrence of FA and atopic march) and dysregulation of the brain-gut endocrine-immune system axis (responsible for the occurrence of FGIDs, IBD, and neuropsychiatric disorders), at least in part through an activation of epigenetic mechanisms.
Figure 3
Figure 3
The structure of the gut microbiome-immune system axis. Within the gut microbiome-immune system axis the cross talk between microbes and the immune system may occur directly through microbial components or indirectly through the action of metabolites, such as SCFAs. A positive modulation of this axis can counteract the pathogenesis of FA by promoting epithelial integrity, gut permeability, mucus production, CD103+ tolerogenic DCs, Treg differentiation, cytokines production, and sIgA release from plasma cells.
Figure 4
Figure 4
Toward a gut microbiome-based precision medicine against food allergy. We are approaching an era where the metagenomic and metabolomic evaluation of gut microbiota in children at risk for FA will drive personalized intervention to preserve or restore an “eubiosis” state based on nutritional counseling and educational programs.

References

    1. Boyce JA, Assa'ad A, Burks AW, Assa'ad A, Burks AW, Jones SM, et al. Guidelines for the diagnosis and management of food allergy in the United States: report of the NIAID–sponsored expert panel. J Allergy Clin Immunology (2010) 126:1105–18. 10.1016/j.jaci.2010.10.007
    1. Loh W, Tang MLK. The epidemiology of food allergy in the global context. Int J Environ Res Public Health (2018) 15:2043. 10.3390/ijerph15092043
    1. McBride D, Keil T, Grabenhenrich L, Dubakiene R, Drasutiene G, Fiocchi A, et al. . The EuroPrevall birth cohort study on food allergy: baseline characteristics of 12,000 newborns and their families from nine European countries. Pediatr Allergy Immunol. (2012) 23:230–9. 10.1111/j.1399-3038.2011.01254.x
    1. Leung ASY, Wong GWK, Tang MLK. Food allergy in the developing world. J Allergy Clin Immunol. (2018) 141:76–8. 10.1016/j.jaci.2017.11.008
    1. Berni Canani R, Nocerino R, Terrin G, Leone L, Troncone R. Hospital admissions for food–induced anaphylaxis in Italian children. Clin Exp Allergy (2012) 42:1813–4. 10.1111/cea.12036
    1. Turner PJ, Gowland MH, Sharma V, Ierodiakonou D, Harper N, Garcez T, et al. . Increase in anaphylaxis–related hospitalizations but no increase in fatalities: an analysis of United Kingdom national anaphylaxis data, 1992–2012. J Allergy Clin Immunol. (2015) 135:956–63.e1. 10.1016/j.jaci.2014.10.021
    1. Mullins RJ, Dear KB, Tang ML. Time trends in Australian hospital anaphylaxis admissions in 1998–1999 to 2011–2012. J Allergy Clin Immunol. (2015) 136:367–75. 10.1016/j.jaci.2015.05.009
    1. Nocerino R, Leone L, Cosenza L, Berni Canani R. Increasing rate of hospitalizations for food–induced anaphylaxis in Italian children: an analysis of the Italian Ministry of Health database. J Allergy Clin Immunol. (2015) 135:833–5. 10.1016/j.jaci.2014.12.1912
    1. Mullins RJ, Wainstein BK, Barnes EH, Liew WK, Campbell DE. Increases in anaphylaxis fatalities in Australia from 1997 to 2013. Clin Exp Allergy (2016) 46:1099–110. 10.1111/cea.12748
    1. Sicherer SH, Muñoz–Furlong A, Godbold JH, Sampson HA. US prevalence of self–reported peanut, tree nut, and sesame allergy: 11–year follow–up. J Allergy Clin Immunol. (2010) 125:1322–6. 10.1016/j.jaci.2010.03.029
    1. Ben–Shoshan M, Harrington DW, Soller L, Fragapane J, Joseph L, St Pierre Y, et al. . A population–based study on peanut, tree nut, fish, shellfish, and sesame allergy prevalence in Canada. J Allergy Clin Immunol. (2010) 125:1327–35. 10.1016/j.jaci.2010.03.015
    1. Chafen JJ, Newberry SJ, Riedl MA, Bravata DM, Maglione M, Suttorp MJ, et al. . Diagnosing and managing common food allergies: a systematic review. JAMA (2010) 303:1848–56. 10.1001/jama.2010.582
    1. Osborne NJ, Koplin JJ, Martin PE, Gurrin LC, Lowe AJ, Matheson MC, et al. . Prevalence of challenge–proven IgE–mediated food allergy using population–based sampling and predetermined challenge criteria in infants. J Allergy Clin Immunol. (2011) 127:668–76.e2. 10.1016/j.jaci.2011.01.039
    1. Gupta RS, Springston EE, Warrier MR, Smith B, Kumar R, Pongracic J, et al. . The prevalence, severity, and distribution of childhood food allergy in the United States. Pediatrics (2011) 128:e9–17. 10.1542/peds.2011-0204
    1. National Academies of Sciences Engineering and Medicine Finding a Path to Safety in Food Allergy: Assessment of Global Burden, Causes, Prevention, Management, and Public Policy. Washington, DC: National Academies Press; (2016).
    1. Sicherer SH, Sampson HA. Food allergy: a review and update on epidemiology, pathogenesis, diagnosis, prevention, and management. J Allergy Clin Immunol. (2018) 141:41–58. 10.1016/j.jaci.2017.11.003
    1. Noval Rivas M, Burton OT, Wise P, Zhang YQ, Hobson SA, Garcia Lloret M, et al. . A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol. (2013) 131:201–12. 10.1016/j.jaci.2012.10.026
    1. Berni Canani R, Gilbert JA, Nagler CR. The role of the commensal microbiota in the regulation of tolerance to dietary allergens. Curr Opin Allergy Clin Immunol. (2015) 15:243–9. 10.1097/ACI.0000000000000157
    1. Paparo L, Aitoro R, Nocerino R, di Scala C, Di Costanzo M, Cosenza L, et al. Epigenetic regulation of early nutrition on immune system. In: Preedy VR, Patel VB, editors. Handbook of Nutrition, Diet, and Epigenetics Cham: Springer International Publishing AG; (2018). 10.1007/978-3-319-31143-2_54-1
    1. Peters RL, Allen KJ, Dharmage SC, Lodge CJ, Koplin JJ, Ponsonby AL, et al. . Differential factors associated with challenge–proven food allergy phenotypes in a population cohort of infants: a latent class analysis. Clin Exp Allergy (2015) 45:953–63. 10.1111/cea.12478
    1. Sicherer SH, Mu noz–Furlong A, Sampson HA. Prevalence of sea food allergy in the United States determined by a random telephone survey. J Allergy Clin Immunol. (2004) 114:159–65. 10.1016/j.jaci.2004.04.018
    1. Panjari M, Koplin JJ, Dharmage SC, Peters RL, Gurrin LC, Sawyer SM, et al. . Nut allergy prevalence and differences between Asian born children and Australian born children of Asian descent: a state–wide survey of children at primary school entry in Victoria, Australia. Clin Exp Allergy (2016) 46:602–9. 10.1111/cea.12699
    1. Sicherer SH, Wood RA, Vickery BP, Jones SM, Liu AH, Fleischer DM, et al. . The natural history of egg allergy in an observational cohort. J Allergy Clin Immunol. (2014) 133:492–9. 10.1016/j.jaci.2013.12.1041
    1. Hourihane JO, Dean TP, Warner JO. Peanut allergy in relation to heredity, maternal diet, and other atopic diseases: results of a questionnaire survey, skin prick testing, and food challenges. BMJ (1996) 313:518–2. 10.1136/bmj.313.7056.518
    1. Lichtenstein P, Svartengren M. Genes, environments, and sex: factors of importance in atopic diseases in 7–9–year–old Swedish twins. Allergy (1997) 52:1079–86. 10.1111/j.1398-9995.1997.tb00179.x
    1. Savage JH, Lee–Sarwar KA, Sordillo J, Bunyavanich S, Zhou Y, O'Connor G, et al. . A prospective microbiome wide association study of food sensitization and food allergy in early childhood. Allergy (2018) 73:145–52. 10.1111/all.13232
    1. Prince BT, Mandel MJ, Nadeau K, Singh AM. Gut microbiome and the development of food allergy and allergic disease. Pediatr Clin North Am. (2015) 62:1479–92. 10.1016/j.pcl.2015.07.007
    1. Mitselou N, Hallberg J, Stephansson O, Almqvist C, Melén E, Ludvigsson JF. Cesarean delivery, preterm birth, and risk of food allergy: Nationwide Swedish cohort study of more than 1 million children. J Allergy Clin Immunol. (2018) 142:1510–14. 10.1016/j.jaci.2018.06.044
    1. Du Toit G, Roberts G, Sayre PH, Plaut M, Bahnson HT, Mitchell H, et al. . Identifying infants at high risk of peanut allergy: the Learning Early About Peanut Allergy (LEAP) screening study. J Allergy Clin Immunol. (2013) 131:135–43.e1–12. 10.1016/j.jaci.2012.09.015
    1. Biasucci G, Rubini M, Riboni S, Morelli L, Bessi E, Retetangos C. Mode of delivery affects the bacterial community in the newborn gut. Early Hum Dev. (2010) 86(Suppl 1):13–5. 10.1016/j.earlhumdev.2010.01.004
    1. Guibas GV, Moschonis G, Xepapadaki P, Roumpedaki E, Androutsos O, Manios Y, et al. . Conception via in vitro fertilization and delivery by Caesarean section are associated with paediatric asthma incidence. Clin Exp Allergy (2013) 43:1058–66. 10.1111/cea.12152
    1. Greenwood C, Morrow AL, Lagomarcino AJ, Altaye M, Taft DH, Yu Z, et al. . Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter. J Pediatr. (2014) 165:23–9. 10.1016/j.jpeds.2014.01.010
    1. Arboleya S, Sánchez B, Milani C, Duranti S, Solís G, Fernández N, et al. . Intestinal microbiota development in preterm neonates and effect of perinatal antibiotics. J Pediatric. (2015) 166:538–44. 10.1016/j.jpeds.2014.09.041
    1. Fouhy F, Guinane CM, Hussey S, Wall R, Ryan CA, Dempsey EM, et al. . High–throughput sequencing reveals the incomplete, short–term recovery of infant gut microbiota following parenteral antibiotic treatment with ampicillin and gentamicin. Antimicrob Agents Chemother. (2012) 56:5811–20. 10.1128/AAC.00789-12
    1. Tanaka S, Kobayashi T, Songjinda P, Tateyama A, Tsubouchi M, Kiyohara C, et al. . Influence of antibiotic exposure in the early postnatal period on the development of intestinal microbiota. FEMS Immunol Med Microbiol. (2009) 56:80–7. 10.1111/j.1574-695X.2009.00553.x
    1. Kummeling I, Stelma FF, Dagnelie PC, Snijders BE, Penders J, Huber M, et al. . Early life exposure to antibiotics and the subsequent development of eczema, wheeze, and allergic sensitization in the first 2 years of life: the KOALA Birth Cohort Study. Pediatrics (2007) 119:e225–31. 10.1542/peds.2006-0896
    1. Zivkovic AM, German JB, Lebrilla CB, Mills DA. Human milk glycobiome and its impact on the infant gastrointestinal microbiota. Proc Natl Acad Sci USA. (2011) 108(Suppl 1):4653–8. 10.1073/pnas.1000083107
    1. Silvers KM, Frampton CM, Wickens K, Pattemore PK, Ingham T, Fishwick D, et al. . Breastfeeding protects against current asthma up to 6 years of age. J Pediatr. (2012) 160:991–6. 10.1016/j.jpeds.2011.11.055
    1. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. . Linking long–term dietary patterns with gut microbial enterotypes. Science (2011) 334:105–8. 10.1126/science.1208344
    1. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom–Bru C, et al. . Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. (2014) 20:159–66. 10.1038/nm.3444
    1. Savage J, Sicherer S, Wood R. The natural history of food allergy. J Allergy Clin Immunol Pract. (2016) 4:196–203. 10.1016/j.jaip.2015.11.024
    1. Hill DA, Spergel JM. The atopic march: critical evidence and clinical relevance. Ann Allergy Asthma Immunol. (2018) 120:131–7. 10.1016/j.anai.2017.10.037
    1. Saps M, Lu P, Bonilla S. Cow's–milk allergy is a risk factor for the development of FGIDs in children. J Pediatr Gastroenterol Nutr. (2011) 52:166–9. 10.1097/MPG.0b013e3181e85b55
    1. Virta LJ, Kautiainen H, Kolho KL. Symptoms suggestive of cow's milk allergy in infancy and pediatric inflammatory bowel disease. Pediatr Allergy Immunol. (2016) 27:361–7. 10.1111/pai.12551
    1. Topal E, Catal F, Soylu N, Ozcan OO, Celiksoy MH, Babayigit A, et al. . Psychiatric disorders and symptoms severity in pre–school children with cow's milk allergy. Allergol Immunopathol. (2016) 44:445–9. 10.1016/j.aller.2016.03.001
    1. Gilbert JA, Blaser MJ, Caporaso JG, Jansson JK, Lynch SV, Knight R. Current understanding of the human microbiome. Nat Med. (2018) 24:392–400. 10.1038/nm.4517
    1. Fraher MH, O'Toole PW, Quigley EM. Techniques used to characterize the gut microbiota: a guide for the clinician. Nat Rev Gastroenterol Hepatol. (2012) 9:312–2. 10.1038/nrgastro.2012.44
    1. Nicholson JK, Lindon JC. Systems biology: metabolomics. Nature (2008) 455:1054–6. 10.1038/4551054a
    1. Moco S, Collino S, Rezzi S, Martin FP. Metabolomics perspectives in pediatric research. Pediatr Res. (2013) 73:570–6. 10.1038/pr.2013.1
    1. Vernocchi P, Del Chierico F, Putignani L. Gut microbiota profiling: metabolomics based approach to unravel compounds affecting human health. Front Microbiol. (2016) 7:1144. 10.3389/fmicb.2016.01144
    1. De Filippis F, Vitaglione P, Cuomo R, Berni Canani R, Ercolini D, et al. . Dietary interventions to modulate the gut microbiome—how far away are we from precision medicine. Inflamm Bowel Dis. (2018) 24:2142–54. 10.1093/ibd/izy080
    1. Bashiardes S, Godneva A, Elinav E, Segal E, Bashiardes S, Godneva A, Elinav E, et al. . Towards utilization of the human genome and microbiome for personalized nutrition. Curr Opin Biotechnol. (2018) 51:57–63. 10.1016/j.copbio.2017.11.013
    1. Berni Canani R, Sangwan N, Stefka AT, Nocerino R, Paparo L, Aitoro R, et al. . Lactobacillus rhamnosus GG supplemented formula expands butyrate producing bacterial strains in food allergic infants. ISME J. (2016) 10:742–50. 10.1038/ismej.2015.151
    1. Fieten KB, Totté JEE, Levin E, Reyman M, Meijer Y, Knulst A, et al. . Fecal microbiome and food allergy in pediatric atopic dermatitis: a cross–sectional pilot study. Clin Allergol. (2018) 175:77–84. 10.1159/000484897
    1. Azad MB, Konya T, Guttman DS, Field CJ, Sears MR, HayGlass KT, et al. . Infant gut microbiota and food sensitization: associations in the first year of life. Clin Exp Allergy (2015) 45:632–43. 10.1111/cea.12487
    1. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. . Induction of colonic regulatory T cells by indigenous Clostridium species. Science (2011) 331:337–41. 10.1126/science.1198469
    1. Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, et al. . Early life antibiotic–driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. (2012) 13:440–7. 10.1038/embor.2012.32
    1. Bashir ME, Louie S, Shi HN, Nagler–Anderson C. Toll–like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. (2004) 172:6978–87. 10.4049/jimmunol.172.11.6978
    1. Geuking MB, Cahenzli J, Lawson MA, Ng DC, Slack E, Hapfelmeier S, et al. . Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity (2011) 34:794–806. 10.1016/j.immuni.2011.03.021
    1. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly–Y M, et al. . The microbial metabolites, short chain fatty acids, regulate colonic Treg cell homeostasis. Science (2013) 341:569–73. 10.1126/science.1241165
    1. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, et al. . Peripheral education of the immune system by colonic commensal microbiota. Nature (2011) 478:250–4. 10.1038/nature10434
    1. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature (2008) 453:620–5. 10.1038/nature07008
    1. Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, et al. . Commensal bacteria protect against food allergen sensitization. PNAS (2014) 111:13145–50. 10.1073/pnas.1412008111
    1. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. . Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature (2013) 500:232–6. 10.1038/nature12331
    1. Bjorksten B, Naaber P, Sepp E, Mikelsaar M. The intestinal microflora in allergic Estonian and Swedish 2–year–old children. Clin Exp Allergy (1999) 29:342–6.
    1. Thompson–Chagoyan OC, Vieites JM, Maldonado J, Edwards C, Gil A. Changes in faecal microbiota of infants with cow's milk protein allergy – a Spanish prospective case–control 6–month follow–up study. Pediatr Allergy Immunol. (2010) 21:e394–e400. 10.1111/j.1399-3038.2009.00961.x
    1. Thompson–Chagoyan OC, Fallani M, Maldonado J, Vieites JM, Khanna S, Edwards C, et al. . Faecal microbiota and short–chain fatty acid levels in faeces from infants with cow‘s milk protein allergy. Int Arch Allergy Immunol. (2011) 156:325–32. 10.1159/000323893
    1. Nakayama J, Kobayashi T, Tanaka S, Korenori Y, Tateyama A, Sakamoto N, et al. . Aberrant structures of fecal bacterial community in allergic Infants profiled by 16S rRNA gene pyrosequencing. FEMS Immunol Med Microbiol. (2011) 63:397–406. 10.1111/j.1574-695X.2011.00872.x
    1. Ling Z, Li Z, Liu X, Cheng Y, Luo Y, Tong X, et al. . Altered fecal microbiota composition associated with food allergy in infants. App Environ Microbiol. (2014) 80:2546–54. 10.1128/AEM.00003-14
    1. Chen CC, Chen KJ, Kong MS, Chang HJ, Huang JL. Alterations in the gut microbiota of children with food sensitization in early life. Pediatr Allergy Immunol. (2016) 27:254–62. 10.1111/pai.12522
    1. Bunyavanich S, Shen N, Grishin A, Wood R, Burks W, Dawson P, et al. . Early–life gut microbiome composition and milk allergy resolution. J Allergy Clin Immunol. (2016) 138:1122–30. 10.1016/j.jaci.2016.03.041
    1. Inoue R, Sawai T, Sawai C, Nakatani M, Romero–Pérez GA, Ozeki M, et al. . A preliminary study of gut dysbiosis in children with food allergy. Biosci Biotechnol Biochem. (2017) 81:2396–9. 10.1080/09168451.2017.1383849
    1. Kourosh A, Luna RA, Balderas M, Nance C, Anagnostou A, Devaraj S, et al. . Fecal microbiome signatures are different in food– allergic children compared to siblings and healthy children. Pediatr Allergy Immunol. (2018) 29:545–54. 10.1111/pai.12904
    1. Fazlollahi M, Chun Y, Grishin A, Wood RA, Burks AW, Dawson P, et al. . Early–life gut microbiome and egg allergy. Allergy (2018) 73:1515–24. 10.1111/all.13389
    1. Dong P, Feng JJ, Yan DY, Lyu YJ, Xu X. Early–life gut microbiome and cow's milk allergy– a prospective case – control 6–month follow–up study. Saudi J Biol Sci. (2018) 25:875–80. 10.1016/j.sjbs.2017.11.051
    1. Berni Canani R, De Filippis F, Nocerino R, Paparo L, Di Scala C, Cosenza L, et al. . Gut microbiota composition and butyrate production in children affected by non–IgE–mediated cow's milk allergy. Sci Rep. (2018) 8:12500. 10.1038/s41598-018-30428-3
    1. Díaz M, Guadamuro L, Espinosa–Martos I, Mancabelli L, Jiménez S, Molinos–Norniella C, et al. . Microbiota and derived parameters in fecal samples of infants with non–IgE Cow's milk protein allergy under a restricted diet. Nutrients (2018) 10:1481. 10.3390/nu10101481
    1. Garcia–Larsen V, Ierodiakonou D, Jarrold K, Cunha S, Chivinge J, Robinson Z, et al. Diet during pregnancy and infancy risk of allergic or autoimmune disease: a systematic review and meta–analysis. PLoS Med. (2018) 15:e1002507 10.1371/journal.pmed.1002507
    1. Barker DJP. Developmental origins of chronic disease. Public Health (2012) 126:185–9. 10.1016/j.puhe.2011.11.014
    1. Netting MJ, Middleton PF, Makrides M. Does maternal diet during pregnancy and lactation affects outcomes in offspring? A systematic review of food–based approaches. Nutrition (2014) 30:1225–41. 10.1016/j.nut.2014.02.015
    1. Wopereis H, Oozeer R, Knipping K, Belzer C, Knol J. The first thousand days – intestinal microbiology of early life: establishing a symbiosis. Pediatr Allergy Immunol. (2014) 25:428–38. 10.1111/pai.12232
    1. Grimshaw KE, Maskell J, Oliver EM, Morris RC, Foote KD, Mills EN, et al. . Diet and food allergy development during infancy: birth cohort study findings using prospective food diary data. J Allergy Clin Immunol. (2014) 133:511–9. 10.1016/j.jaci.2013.05.035
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. (2016) 16:341–52. 10.1038/nri.2016.42
    1. Ma N, Guo P, Zhang J, He T, Kim SW, Zhang G, et al. Nutrients mediate intestinal bacteria–mucosal immune cross talk. Front Immunol. (2018) 9:5 10.3389/fimmu.2018.00005
    1. Castro–Rodriguez JA, Garcia–Marcos L. What are the effects of a Mediterranean diet on allergies and asthma in children? Front Pediatr. (2017) 5:72. 10.3389/fped.2017.00072
    1. Castro–Rodriguez JA, Ramirez–Hernandez M, Padilla O, Pacheco–Gonzalez RM, Pérez–Fernández V, Garcia–Marcos L. Effect of foods and Mediterranean diet during pregnancy and first years of life on wheezing, rhinitis and dermatitis in preschoolers. Allergol Immunopathol. (2016) 44:400–9. 10.1016/j.aller.2015.12.002
    1. Berni Canani R, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. (2011) 17:1519–28. 10.3748/wjg.v17.i12.1519
    1. Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, et al. . A dietary fiber–deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell (2016) 167:1339. 10.1016/j.cell.2016.10.043
    1. De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L, et al. . High–level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut (2016) 65:1812–21. 10.1136/gutjnl-2015-309957
    1. McKenzie C, Tan J, Macia L, Mackay CR. The nutrition–gut microbiome–physiology axis and allergic diseases. Immunol Rev. (2017) 278:277–95. 10.1111/imr.12556
    1. Louis P, Flint HJ. Diversity, metabolism and microbial ecology of butyrate–producing bacteria from the human large intestine. FEMS Microbiol Lett. (2009) 294:1–8. 10.1111/j.1574-6968.2009.01514.x
    1. Schauber J, Svanholm C, Termen S, Iffland K, Menzel T, Scheppach W, et al. Expression of the cathelicidin LL−37 is modulated by short chain fatty acids in colonocytes: relevance of signaling pathways. Gut (2003) 52:735–41.
    1. Peng L, Li ZR, Green RS, Holzman IR, Lin J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP–activated protein kinase in Caco−2 cell monolayers. J Nutr. (2009) 139:1619–25. 10.3945/jn.109.104638
    1. Kim M, Kim CH. Regulation of humoral immunity by gut microbial products. Gut Microbes (2017) 8:392–9. 10.1080/19490976.2017.1299311
    1. Kasubuchi M, Hasegawa S, Hiramatsu T, Ichimura A, Kimura I. Dietary gut microbial metabolites, short–chain fatty acids, and host metabolic regulation. Nutrients (2015) 7:2839–49. 10.3390/nu7042839
    1. Ohira H, Fujioka Y, Katagiri C, Mamoto R, Aoyama–Ishikawa M, Amako K, et al. . Butyrate attenuates inflammation and lipolysis generated by the interaction of adipocytes and macrophages. J Atheroscler Thromb. (2013) 20:425–42.
    1. Nastasi C, Candela M, Bonefeld CM, Geisler C, Hansen M, Krejsgaard T, et al. . The effect of short–chain fatty acids on human monocyte–derived dendritic cells. Sci Rep. (2015) 5:16148. 10.1038/srep16148
    1. Fontenelle B, Gilbert KM. n–Butyrate anergized effector CD4+T cell generation or activity. Scand J Immunol. (2012) 76:457–63. 10.1111/j.1365-3083.2012.02740.x
    1. Brown AJ, Goldsworthy SM, Barnes AA, Eilert MM, Tcheang L, Daniels D, et al. . The Orphan G protein–coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem. (2003) 278:312–9. 10.1074/jbc.M211609200
    1. Nilsson NE, Kotarsky K, Owman C, Olde B. Identification of a free fatty acid receptor, FFA2R, expressed on leukocytes and activated by short–chain fatty acids. Biochem Biophys Res Commun. (2003) 303:1047–52.
    1. Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, Browning DD, et al. . GPR109A is a G–protein–coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Res. (2009) 69:2826–32. 10.1158/0008-5472.CAN-08-4466
    1. Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, et al. . Olfactory receptor responding to gut microbiota– derived signals plays a role in renin secretion and blood pressure regulation. Proc Natl Acad Sci USA. (2013) 110:4410–5. 10.1073/pnas.1215927110
    1. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short–chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology (2013) 145:e1–10. 10.1053/j.gastro.2013.04.056
    1. Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, et al. . Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. (2003) 278:481–9. 10.1074/jbc.M301403200
    1. Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, et al. . Short–chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR–S6K pathway. Mucosal Immunol. (2015) 8:80–93. 10.1038/mi.2014.44
    1. Nakajima A, Nakatani A, Hasegawa S, Irie J, Ozawa K, Tsujimoto G, et al. . The short chain fatty acid receptor GPR43 regulates inflammatory signals in adipose tissue M2– type macrophages. PLoS ONE (2017) 12:e0179696. 10.1371/journal.pone.0179696
    1. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. . Metabolites produced by commensal bacteria promote peripheral regulatory T–cell generation. Nature 504:451–5. 10.1038/nature12726
    1. Goverse G, Molenaar R, Macia L, Tan J, Erkelens MN, Konijn T, et al. Diet–derived short chain fatty acids stimulate intestinal epithelial cells to induce mucosal tolerogenic dendritic cells. J Immunol. (2017) 198:2172–81. 10.4049/jimmunol.1600165
    1. Kim M, Qie Y, Park J, Kim CH. Gut microbial metabolites fuel host antibody responses. Cell Host Microbe (2016) 20:202–14. 10.1016/j.chom.2016.07.001
    1. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. . Commensal microbe–derived butyrate induces the differentiation of colonic regulatory T cells. Nature (2013) 504:446–50. 10.1038/nature12721
    1. Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, Porrett PM, et al. . Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med. (2007) 13:1299–307. 10.1038/nm1652
    1. Sandin A, Bråbäck L, Norin E, Björkstén B. Faecal short chain fatty acid pattern and allergy in early childhood. Acta Paediatr. (2009) 98:823–7. 10.1111/j.1651-2227.2008.01215.x
    1. Di Costanzo M, Paparo L, Aitoro R, Cosenza L, Nocerino R, Cozzolino T, et al. Potential Beneficial Effects of Butyrate against Food Allergy. In: Cong–Jun Li, editors. Butyrate: Food Sources, Functions and Health Benefits. New York, NY: Biochemistry Research Trends; (2014). p. 81–90.
    1. Aitoro R, Paparo L, Amoroso A, Di Costanzo M, Cosenza L, Granata V, et al. . Gut microbiota as a target for preventive and therapeutic intervention against food allergy. Nutrients (2017) 9:E672. 10.3390/nu9070672
    1. Nowak–Wegrzyn A, Chatchatee P. Mechanisms of tolerance induction. Ann Nutr Metab. (2017) 70:7–24. 10.1159/000457915
    1. Wang H, Ji Y, Wu G, Sun K, Sun Y, Li W, et al. . l–tryptophan activates mammalian target of rapamycin and enhances expression of tight junction proteins in intestinal porcine epithelial cells. J Nutr. (2015) 145:1156–62. 10.3945/jn.114.209817
    1. Hashimoto T, Perlot T, Rehman A, Trichereau J, Ishiguro H, Paolino M, et al. . ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature (2012) 487:477–81. 10.1038/nature11228
    1. Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, et al. Aryl hydrocarbon receptor control of a disease tolerance defense pathway. Nature (2014) 511:184–90. 10.1038/nature13323
    1. Pilotte L, Larrieu P, Stroobant V, Colau D, Dolusic E, Frédérick R, et al. . Reversal of tumoral immune resistance by inhibition of tryptophan 2,3– dioxygenase. Proc Natl Acad Sci USA (2012) 109:2497–502. 10.1073/pnas.1113873109
    1. Fallarino F, Grohmann U, Vacca C, Bianchi R, Orabona C, Spreca A, et al. . T cell apoptosis by tryptophan catabolism. Cell Death Differ. (2002) 9:1069–77. 10.1038/sj.cdd.4401073
    1. Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, et al. . Innate lymphoid cells – a proposal for uniform nomenclature. Nat Rev Immunol. (2013) 13:145–9. 10.1038/nri3365
    1. Lanis JM, Alexeev EE, Curtis VF, Kitzenberg DA, Kao DJ, Battista KD, et al. . Tryptophan metabolite activation of the aryl hydrocarbon receptor regulates IL−10 receptor expression on intestinal epithelia. Mucosal Immunol. (2017) 10:1133–44. 10.1038/mi.2016.133
    1. Venkatesh M, Mukherjee S, Wang H, Li H, Sun K, Benechet AP, et al. . Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and toll–like receptor 4. Immunity (2014) 41:296–310. 10.1016/j.immuni.2014.06.014
    1. Hammerschmidt–Kamper C, Biljes D, Merches K, Steiner I, Daldrup T, Bol–Schoenmakers M, et al. . Indole−3–carbinol, a plant nutrient and AhR–Ligand precursor, supports oral tolerance against OVA and improves peanut allergy symptoms in mice. PLoS ONE (2017) 12:e0180321. 10.1371/journal.pone.0180321
    1. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. . The International scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotics. Nat Rev Gastro Hepat. (2014) 11:506–14. 10.1038/nrgastro.2014.66
    1. Sudo N, Sawamura S, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immunol. (1997) 159:1739–45.
    1. Isolauri E, Arvola T, Sütas Y, Moilanen E, Salminen S. Probiotics in the management of atopic eczema. Clin Exp Allergy (2000) 30:1604–10. 10.1046/j.1365-2222.2000.00943.x
    1. Malin M, Verronen P, Korhonen H, Syväoja EL, Salminen S, Mykkänen H, et al. . Dietary therapy with Lactobacillus GG, bovine colostrum or bovine immune colostrum in patients with juvenile chronic arthritis: evaluation of effect on gut defense mechanism. Inflammopharmacology (1997) 5:219–36.
    1. Kaila M, Isolauri E, Soppi E, Virtanen E, Laine S, Arvilommi H. Enhancement of the circulating antibody secreting cellresponse in human diarrhea by a human Lactobacillus strain. Pediatr Res. (1992) 32:141–4.
    1. Hardy H, Harris J, Lyon E, Beal J, Foey AD. Probiotics, Prebiotics and Immunomodulation of gutmucosal defences: homeostatis and Immunopathology. Nutrients (2013) 5:1869–12. 10.3390/nu5061869
    1. Kim JY, Choi YO, Ji GE. Effect of oral probiotics (Bifidobacteriumlactis AD011 andLactobacillus acidophilus AD031) administration on ovalbumin–induced food allergy mouse model. J Microbiol Biotechnol. (2008) 18:1393–400.
    1. Torii A, Torii S, Fujiwara S, Tanaka H, Inagaki N, Nagai H, et al. Lactobacillus acidophilus strain L−92 regulates theproduction of Th1 cytokine as well as Th2 cytokines. Allergol Int. (2007) 56:293–301. 10.2332/allergolint.O-06-459
    1. Niers LE, Timmerman HM, Rijkers GT, van Bleek GM, van Uden NO, Knol EF, et al. . Identification of strong interleukin−10 inducinglactic acid bacteria which down–regulate T helper type 2 cytokines. Clin Exp Allergy (2005) 35:1481–89. 10.1111/j.1365-2222.2005.02375.x
    1. Takahashi N, Kitazawa H, Iwabuchi N, Xiao JZ, Miyaji K, Iwatsuki K, et al. . Oral administration of an immunostimulatoryDNA sequence from Bifidobacterium longum improves Th1/Th2 balance in a murine model. Biosci. Biotechnol Biochem. (2006) 70:2013–7. 10.1271/bbb.60260
    1. Ghadimi D, Helwig U, Schrezenmeir J, Heller KJ, de Vrese M. Epigenetic imprinting by commensal probiotics inhibitis the IL−23/IL−17 axis in a vitro model of the intestinal mucosal immune system. J Leukoc Biol. (2012) 92:895–911. 10.1189/jlb.0611286
    1. Maassen CB, van Holten–Neelen C, Balk F, den Bak–Glashouwer MJ, Leer RJ, Laman JD, et al. . Strain–dependent induction of cytokine profiles in the gut by orally administered Lactobacillus strains. Vaccine (2000) 18:2613–23. 10.1016/S0264-410X(99)00378-3
    1. Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, van Capel TM, et al. . Selective probiotic bacteria induce IL−10– producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell– specific intercellular adhesion molecule 3–grabbing nonintegrin. J Allergy Clin Immunol. (2005) 115:1260–7. 10.1016/j.jaci.2005.03.036
    1. Cross ML, Gill HS. Can immunoregulatory lactic acid bacteria be used as dietary supplements to limit allergies? Int Arch Allergy Immunol. (2001) 125:112–9. 10.1159/000053804
    1. Aitoro R, Simeoli R, Amoroso A, Paparo L, Nocerino R, Pirozzi C, et al. Extensively hydrolyzed casein formula alone or with L. rhamnosus GG reduces β-lactoglobulin sensitization in mice. Pediatr Allergy Immunol. (2017) 28:230–7. 10.1111/pai.12687
    1. Hol J, van Leer EH, ElinkSchuurman BE, de Ruiter LF, Samsom JN, Hop W, et al. The acquisition of tolerance towards cow's milk through probiotic supplementation: a randomized controlled trial. J Allergy Clin Immunol. (2008) 121:1448–54. 10.1016/j.jaci.2008.03.018
    1. Flinterman AE, Knol EF, Van Ieperen AG, Timmerman HM, Knulst AC, Bruijnzeel-Koomen CAFM, et al. . Probiotics have a different immunomodulatory potential in vitro versus ex vivo upon oral administration in children with food allergy. Int Arch Allergy Immunol. (2007) 143:237–44. 10.1159/000099467
    1. Braat H, van den Brande J, van Tol E, Hommes D, Peppelenbosch M, van Deventer S. Lactobacillus rhamnosus induces peripheral hyporesponsiveness in stimulated CD4+ T cells via modulation of dendritic cell function. Am J Clan Nutr. (2004) 80:1618–25. 10.1093/ajcn/80.6.1618
    1. Borthakur A, Gill RK, Tyagi S, Koutsouris A, Alrefai WA, Hecht GA, et al. . The probiotic Lactobacillus acidophilus stimulates chloride/hydroxyl exchange activity in human intestinal epithelial cells. J Nutr. (2008) 138:1355–9. 10.1093/jn/138.7.1355
    1. Borchers AT, Keen CL, Gershwin ME. The influence of yogurt/Lactobacillus on the innate and acquired immune response. Clin Rev Allergy Immunol. (2009) 22:207–30. 10.1007/s12016-002-0009-7
    1. Fu L, Peng J, Zhao S, Zhang Y, Su X, Wang Y. Lactic acid bacteria-specific induction of CD4+Foxp3+T cells ameliorates shrimp tropomyosin induced allergic response in mice via suppression of mTOR signaling. Sci Rep. (2017) 7:1987. 10.1038/s41598-017-02260-8
    1. Berni Canani R, Paparo L, Nocerino R, Cosenza L, Pezzella V, Di Costanzo M, et al. Differences in DNA methylation profile of Th1 and Th2 cytokine genes are associated with tolerance acquisition in children with IgE–mediated cow's milk allergy. Clin Epigenetics (2015) 7:38 10.1186/2Fs13148-015-0070-8
    1. Paparo L, Nocerino R, Cosenza L, Aitoro R, D'Argenio V, Del Monaco V, et al. . Epigenetic features of FoxP3 in children with cow's milk allergy. Clin Epigenetics (2016) 8:86. 10.1186/s13148-016-0252-z
    1. Karlsson H, Larsson P, Wold AE, Rudin A. Pattern of cytokine responses to Gram–positive and Gram–Negative commensal bacteria is profoundly changed when monocytes differentiate into dendritic cells. Infect Immun. (2004) 72:2671–8. 10.1128/IAI.72.5.2671-2678.2004
    1. Mohamadzadeh M, Olson S, Kalina WV, Ruthel G, Demmin GL, Warfield KL, et al. . Lactobacilli activate human dendritic cells that skew T cells toward T helper 1 polarization. Proc Natl Acad Sci USA. (2005) 102:2880–5. 10.1073/pnas.0500098102
    1. Zhang J, Su H, Li Q, Wu H, Liu M, Huang J, et al. . Oral administration of Clostridium butyricum CGMCC0313.1 inhibits β-lactoglobulin–induced intestinal anaphylaxis in a mouse model of food allergy. Gut Pathog. (2017) 9:11. 10.1186/s13099-017-0160-6
    1. Yang B, Xiao L, Liu S, Liu X, Luo Y, Ji Q, et al. . Exploration of the effect of probiotics supplementation on intestinal microbiota of food allergic mice. Am J Translat Res. (2017) 9:376–85.
    1. Maiga MA, Morin S, Bernard H, Rabot S, Adel–Patient K, Hazebrouck S. Neonatal mono–colonization of germ–free mice with Lactobacillus casei enhances casein immunogenicity after oral sensitization to cow's milk. Mol Nutr Food Res. (2017) 61. 10.1002/mnfr.201600862
    1. Liu MY, Yang ZY, Dai WK, Huang JQ, Li YH, Zhang J, et al. Protective effect of Bifidobacterium infantis CGMCC313–2 on ovalbumin–induced airway asthma and b–lactoglobulin induced intestinal food allergy mouse models. World J Gastroenterol. (2017) 23:2149–58. 10.3748/wjg.v23.i12.2149
    1. Schiavi E, Barletta B, Butteroni C, Corinti S, Boirivant M, Di Felice G. Oral therapeutic administration of a probiotic mixture suppresses established Th2 responses and systemic anaphylaxis in a murine model of food allergy. Allergy (2011) 66:499–508. 10.1111/j.1398-9995.2010.02501.x
    1. Thang CL, Baurhoo B, Boye JI, Simpson BK, Zhao X. Effects of Lactobacillus rhamnosus GG supplementation on cow's milk allergy in a mouse model. Allergy Asthma Clin Immunol. (2011) 7:20. 10.1186/1710-1492-7-20
    1. Berni Canani R, Nocerino R, Terrin G, Coruzzo A, Cosenza L, Leone L, et al. . Effect of Lactobacillus GG on tolerance acquisition in infants with cow's milk allergy a randomized trial. J Allergy ClinImmunol. (2012) 129:580–2. 10.1016/j.jaci.2011.10.004
    1. Berni Canani R, Nocerino R, Terrin G, Frediani T, Lucarelli S, Cosenza L, et al. Formula selection for management of children with cow milk allergy influences the rate of acquisition of tolerance: a prospective multicenter study. J Pediatric. (2013) 163:771–7. 10.1016/j.jpeds.2013.03.008
    1. Berni Canani R, Di Costanzo M, Bedogni G, Amoroso A, Cosenza L, Di Scala C, et al. Extensively hydrolyzed casein formula containing Lactobacillus rhamnosus GG reduces the occurrence of other allergic manifestation sin children with cow's milk allergy: 3–year randomized controlled trial. J Allergy Clin Immunol. (2017) 139:1906–13. 10.1016/j.jaci.2016.10.050
    1. Mammas IN, Greenough A, Theodoridou M, Kramvis A, Rusan M, Melidou A, et al. Probiotics and allergy in infants –an update review. Pediatr Allergy Immunol. (2010) 21:e659–66. 10.1111/j.1399-3038.2010.01061.x
    1. Ghadimi D, Fölster–Holst R, de Vrese M, Winkler P, Heller KJ, Schrezenmeir J. Effects of probiotic bacteria and their genomic DNA on TH1/TH2–cytokine production by peripheral blood mononuclear cells (PBMCs) of healthy and allergic subjects. Immunobiology (2008) 213:677–92. 10.1016/j.imbio.2008.02.001
    1. Donato KA, Gareau MG, Wang YJ. Lactobacillus rhamnosus GG attenuates interferon–γ and tumor–necrosis factor–α- induced barrier dysfunction and pro–inflammatory signaling. Microbiology (2010) 156:3288–97. 10.1099/mic.0.040139-0
    1. Mileti E, Matteoli G, Iliev ID, Rescigno M. Comparison of the immunomodulatory properties of three probiotics strains of Lactobacilli using complex culture systems: prediction for in vivo efficacy. PLoS ONE (2009) 4:e7056 10.1371/journal.pone.0007056
    1. Baldassarre ME, Laforgia N, Fanelli M, Laneve A, Grosso R, Lifschitz C. Lactobacillus GG improves recovery in infants with blood in the stools and presumptive allergic colitis compared with extensively hydrolyzed formula alone. J Pediatr. (2010) 156:397–401. 10.1016/j.jpeds.2009.09.012
    1. Rachid R, Keet CA. Current status and unanswered questions for food allergy treatments. J Allergy Clin Immunol Pract. (2018) 6:377–382. 10.1016/j.jaip.2017.10.023
    1. Tang ML, Ponsonby AL, Orsini F, Tey D, Robinson M, Su EL, et al. . Administration of a probiotic with peanut oral immunotherapy: a randomized trial. J Allergy Clin Immunol. (2015) 135:737–44. 10.1016/j.jaci.2014.11.034

Source: PubMed

3
Se inscrever