The Overlapping Area of Non-Celiac Gluten Sensitivity (NCGS) and Wheat-Sensitive Irritable Bowel Syndrome (IBS): An Update

Carlo Catassi, Armin Alaedini, Christian Bojarski, Bruno Bonaz, Gerd Bouma, Antonio Carroccio, Gemma Castillejo, Laura De Magistris, Walburga Dieterich, Diana Di Liberto, Luca Elli, Alessio Fasano, Marios Hadjivassiliou, Matthew Kurien, Elena Lionetti, Chris J Mulder, Kamran Rostami, Anna Sapone, Katharina Scherf, Detlef Schuppan, Nick Trott, Umberto Volta, Victor Zevallos, Yurdagül Zopf, David S Sanders, Carlo Catassi, Armin Alaedini, Christian Bojarski, Bruno Bonaz, Gerd Bouma, Antonio Carroccio, Gemma Castillejo, Laura De Magistris, Walburga Dieterich, Diana Di Liberto, Luca Elli, Alessio Fasano, Marios Hadjivassiliou, Matthew Kurien, Elena Lionetti, Chris J Mulder, Kamran Rostami, Anna Sapone, Katharina Scherf, Detlef Schuppan, Nick Trott, Umberto Volta, Victor Zevallos, Yurdagül Zopf, David S Sanders

Abstract

Gluten-related disorders have recently been reclassified with an emerging scientific literature supporting the concept of non-celiac gluten sensitivity (NCGS). New research has specifically addressed prevalence, immune mechanisms, the recognition of non-immunoglobulin E (non-IgE) wheat allergy and overlap of NCGS with irritable bowel syndrome (IBS)-type symptoms. This review article will provide clinicians with an update that directly impacts on the management of a subgroup of their IBS patients whose symptoms are triggered by wheat ingestion.

Keywords: amylase-trypsin inhibitors (ATIs); celiac disease; gluten sensitivity; gluten-free diet; gluten-related disorders; wheat allergy.

Conflict of interest statement

This paper was made possible by support from Schär for traveling and lodging sponsorship for all co-authors to meet to discuss the object of this paper.

Figures

Figure 1
Figure 1
Potential triggers in wheat that may account for both intestinal and extra-intestinal symptoms.
Figure 2
Figure 2
Wheat components.
Figure 3
Figure 3
IBS treatment pathway.

References

    1. Ellis A., Linaker B.D. Non-coeliac gluten sensitivity? Lancet. 1978;1:1358–1359. doi: 10.1016/S0140-6736(78)92427-3.
    1. Catassi C., Elli L., Bonaz B., Bouma G., Carroccio A., Castillejo G., Cellier C., Cristofori F., de Magistris L., Dolinsek J., et al. Diagnosis of non-celiac gluten sensitivity (NCGS): The salerno experts’ criteria. Nutrients. 2015;7:4966–4977. doi: 10.3390/nu7064966.
    1. Sapone A., Bai J.C., Ciacci C., Dolinsek J., Green P.H., Hadjivassiliou M., Kaukinen K., Rostami K., Sanders D.S., Schumann M., et al. Spectrum of gluten-related disorders: Consensus on new nomenclature and classification. BMC Med. 2012;10:13. doi: 10.1186/1741-7015-10-13.
    1. Catassi C., Bai J.C., Bonaz B., Bouma G., Calabro A., Carroccio A., Castillejo G., Ciacci C., Cristofori F., Dolinsek J., et al. Non-celiac gluten sensitivity: The new frontier of gluten related disorders. Nutrients. 2013;5:3839–3853. doi: 10.3390/nu5103839.
    1. Tanpowpong P., Ingham T.R., Lampshire P.K., Kirchberg F.F., Epton M.J., Crane J., Camargo C.A., Jr., New Zealand Asthma and Allergy Cohort Study Group Coeliac disease and gluten avoidance in New Zealand children. Arch. Dis. Child. 2012;97:12–16. doi: 10.1136/archdischild-2011-300248.
    1. Rubio-Tapia A., Ludvigsson J.F., Brantner T.L., Murray J.A., Everhart J.E. The prevalence of celiac disease in the United States. Am. J. Gastroenterol. 2012;107:1538–1544. doi: 10.1038/ajg.2012.219.
    1. DiGiacomo D.V., Tennyson C.A., Green P.H., Demmer R.T. Prevalence of gluten-free diet adherence among individuals without celiac disease in the USA: Results from the continuous national health and nutrition examination survey 2009–2010. Scand. J. Gastroenterol. 2013;48:921–925. doi: 10.3109/00365521.2013.809598.
    1. Aziz I., Lewis N.R., Hadjivassiliou M., Winfield S.N., Rugg N., Kelsall A., Newrick L., Sanders D.S. A UK study assessing the population prevalence of self-reported gluten sensitivity and referral characteristics to secondary care. Eur. J. Gastroenterol. Hepatol. 2014;26:33–39. doi: 10.1097/01.meg.0000435546.87251.f7.
    1. Lis D.M., Stellingwerff T., Shing C.M., Ahuja K.D., Fell J.W. Exploring the popularity, experiences, and beliefs surrounding gluten-free diets in nonceliac athletes. Int. J. Sport Nutr. Exerc. Metab. 2015;25:37–45. doi: 10.1123/ijsnem.2013-0247.
    1. Volta U., Bardella M.T., Calabro A., Troncone R., Corazza G.R., The Study Group for Non-Celiac Gluten Sensitivity An italian prospective multicenter survey on patients suspected of having non-celiac gluten sensitivity. BMC Med. 2014;12:85. doi: 10.1186/1741-7015-12-85.
    1. Golley S., Corsini N., Topping D., Morell M., Mohr P. Motivations for avoiding wheat consumption in australia: Results from a population survey. Public Health Nutr. 2015;18:490–499. doi: 10.1017/S1368980014000652.
    1. Mardini H.E., Westgate P., Grigorian A.Y. Racial differences in the prevalence of celiac disease in the us population: National health and nutrition examination survey (NHANES) 2009–2012. Dig. Dis. Sci. 2015;60:1738–1742. doi: 10.1007/s10620-014-3514-7.
    1. Van Gils T., Nijeboer P., IJssennagger C.E., Sanders D.S., Mulder C.J., Bouma G. Prevalence and characterization of self-reported gluten sensitivity in The Netherlands. Nutrients. 2016;8:714. doi: 10.3390/nu8110714.
    1. Carroccio A., Giambalvo O., Blasca F., Iacobucci R., D’Alcamo A., Mansueto P. Self-reported non-celiac wheat sensitivity in high school students: Demographic and clinical characteristics. Nutrients. 2017;9:771. doi: 10.3390/nu9070771.
    1. Kaukinen K., Turjanmaa K., Maki M., Partanen J., Venalainen R., Reunala T., Collin P. Intolerance to cereals is not specific for coeliac disease. Scand. J. Gastroenterol. 2000;35:942–946. doi: 10.1080/003655200750022995.
    1. Kabbani T.A., Vanga R.R., Leffler D.A., Villafuerte-Galvez J., Pallav K., Hansen J., Mukherjee R., Dennis M., Kelly C.P. Celiac disease or non-celiac gluten sensitivity? An approach to clinical differential diagnosis. Am. J. Gastroenterol. 2014;109:741–746. doi: 10.1038/ajg.2014.41.
    1. Raju S.A., Mooney P.D., Aziz I., Kurien M., Sanders D.S. Letter: Gluten challenge in the era of noncoeliac gluten sensitivity—A change in clinical practice? Aliment. Pharmacol. Ther. 2016;43:656. doi: 10.1111/apt.13503.
    1. Coburn J.A., Vande Voort J.L., Lahr B.D., Van Dyke C.T., Kroning C.M., Wu T.T., Gandhi M.J., Murray J.A. Human leukocyte antigen genetics and clinical features of self-treated patients on a gluten-free diet. J. Clin. Gastroenterol. 2013;47:828–833. doi: 10.1097/MCG.0b013e31828f531c.
    1. Aziz I., Hadjivassiliou M., Sanders D.S. Self-reported gluten sensitivity: An international concept in need of consensus? Am. J. Gastroenterol. 2014;109:1498–1499. doi: 10.1038/ajg.2014.205.
    1. Turnbull J.L., Adams H.N., Gorard D.A. Review article: The diagnosis and management of food allergy and food intolerances. Aliment. Pharmacol. Ther. 2015;41:3–25. doi: 10.1111/apt.12984.
    1. Bischoff S., Crowe S.E. Gastrointestinal food allergy: New insights into pathophysiology and clinical perspectives. Gastroenterology. 2005;128:1089–1113. doi: 10.1053/j.gastro.2004.08.015.
    1. Carroccio A., Mansueto P., D’Alcamo A., Iacono G. Non-celiac wheat sensitivity as an allergic condition: Personal experience and narrative review. Am. J. Gastroenterol. 2013;108:1845–1852. doi: 10.1038/ajg.2013.353.
    1. Volta U., Tovoli F., Cicola R., Parisi C., Fabbri A., Piscaglia M., Fiorini E., Caio G. Serological tests in gluten sensitivity (nonceliac gluten intolerance) J. Clin. Gastroenterol. 2012;46:680–685. doi: 10.1097/MCG.0b013e3182372541.
    1. Carroccio A., Brusca I., Mansueto P., Pirrone G., Barrale M., Di Prima L., Ambrosiano G., Iacono G., Lospalluti M.L., La Chiusa S.M., et al. A cytologic assay for diagnosis of food hypersensitivity in patients with irritable bowel syndrome. Clin. Gastroenterol. Hepatol. 2010;8:254–260. doi: 10.1016/j.cgh.2009.11.010.
    1. Carroccio A., Mansueto P., Iacono G., Soresi M., D’Alcamo A., Cavataio F., Brusca I., Florena A.M., Ambrosiano G., Seidita A., et al. Non-celiac wheat sensitivity diagnosed by double-blind placebo-controlled challenge: Exploring a new clinical entity. Am. J. Gastroenterol. 2012;107:1898–1906. doi: 10.1038/ajg.2012.236.
    1. Fritscher-Ravens A., Schuppan D., Ellrichmann M., Schoch S., Rocken C., Brasch J., Bethge J., Bottner M., Klose J., Milla P.J. Confocal endomicroscopy shows food-associated changes in the intestinal mucosa of patients with irritable bowel syndrome. Gastroenterology. 2014;147:1012–1020. doi: 10.1053/j.gastro.2014.07.046.
    1. Carroccio A., Soresi M., D’Alcamo A., Sciume C., Iacono G., Geraci G., Brusca I., Seidita A., Adragna F., Carta M., et al. Risk of low bone mineral density and low body mass index in patients with non-celiac wheat-sensitivity: A prospective observation study. BMC Med. 2014;12:230. doi: 10.1186/s12916-014-0230-2.
    1. Sapone A., Lammers K.M., Casolaro V., Cammarota M., Giuliano M.T., De Rosa M., Stefanile R., Mazzarella G., Tolone C., Russo M.I., et al. Divergence of gut permeability and mucosal immune gene expression in two gluten-associated conditions: Celiac disease and gluten sensitivity. BMC Med. 2011;9:23. doi: 10.1186/1741-7015-9-23.
    1. Sapone A., Lammers K.M., Mazzarella G., Mikhailenko I., Carteni M., Casolaro V., Fasano A. Differential mucosal il-17 expression in two gliadin-induced disorders: Gluten sensitivity and the autoimmune enteropathy celiac disease. Int. Arch. Allergy Immunol. 2010;152:75–80. doi: 10.1159/000260087.
    1. Di Liberto D., Mansueto P., D’Alcamo A., Lo Pizzo M., Lo Presti E., Geraci G., Fayer F., Guggino G., Iacono G., Dieli F., et al. Predominance of type 1 innate lymphoid cells in the rectal mucosa of patients with non-celiac wheat sensitivity: Reversal after a wheat-free diet. Clin. Transl. Gastroenterol. 2016;7:e178. doi: 10.1038/ctg.2016.35.
    1. Hollon J., Puppa E.L., Greenwald B., Goldberg E., Guerrerio A., Fasano A. Effect of gliadin on permeability of intestinal biopsy explants from celiac disease patients and patients with non-celiac gluten sensitivity. Nutrients. 2015;7:1565–1576. doi: 10.3390/nu7031565.
    1. Uhde M., Ajamian M., Caio G., De Giorgio R., Indart A., Green P.H., Verna E.C., Volta U., Alaedini A. Intestinal cell damage and systemic immune activation in individuals reporting sensitivity to wheat in the absence of coeliac disease. Gut. 2016;65:1930–1937. doi: 10.1136/gutjnl-2016-311964.
    1. Prospective, Double-Blind Diagnostic Study of Confocal Laser Endomicroscopy (CLE) for Wheat Sensitivity (WS) in Patients with Irritable Bowel Syndrome. German Clinical Trials Register. [(accessed on 18 August 2017)]; Available online: .
    1. Pinto-Sanchez M.I., Verdu E.F. Non-coeliac gluten sensitivity: Are we closer to separating the wheat from the chaff? Gut. 2016;65:1921–1922. doi: 10.1136/gutjnl-2016-312471.
    1. Junker Y., Zeissig S., Kim S.J., Barisani D., Wieser H., Leffler D.A., Zevallos V., Libermann T.A., Dillon S., Freitag T.L., et al. Wheat amylase trypsin inhibitors drive intestinal inflammation via activation of toll-like receptor 4. J. Exp. Med. 2012;209:2395–2408. doi: 10.1084/jem.20102660.
    1. Gibson P.R., Varney J., Malakar S., Muir J.G. Food components and irritable bowel syndrome. Gastroenterology. 2015;148:1158–1174. doi: 10.1053/j.gastro.2015.02.005.
    1. De Punder K., Pruimboom L. The dietary intake of wheat and other cereal grains and their role in inflammation. Nutrients. 2013;5:771–787. doi: 10.3390/nu5030771.
    1. Scherf K.A., Koehler P., Wieser H. Gluten and wheat sensitivities—An overview. J. Cereal Sci. 2016;67:2–11. doi: 10.1016/j.jcs.2015.07.008.
    1. Maiuri L., Ciacci C., Ricciardelli I., Vacca L., Raia V., Auricchio S., Picard J., Osman M., Quaratino S., Londei M. Association between innate response to gliadin and activation of pathogenic T cells in coeliac disease. Lancet. 2003;362:30–37. doi: 10.1016/S0140-6736(03)13803-2.
    1. Frossi B., Tripodo C., Guarnotta C., Carroccio A., De Carli M., De Carli S., Marino M., Calabro A., Pucillo C.E. Mast cells are associated with the onset and progression of celiac disease. J. Allergy Clin. Immunol. 2017;139:1266–1274. doi: 10.1016/j.jaci.2016.08.011.
    1. Hajas L., Scherf K.A., Torok K., Bugyi Z., Schall E., Poms R.E., Koehler P., Tomoskozi S. Variation in Protein Composition among Wheat (Triticum aestivum L.) Cultivars to Identify Cultivars Suitable as Reference Material for Wheat Gluten Analysis. Food Chem. 2017 doi: 10.1016/j.foodchem.2017.05.005.
    1. Tatham A.S., Shewry P.R. Allergens to wheat and related cereals. Clin. Exp. Allergy. 2008;38:1712–1726.
    1. Gomez L., Martin E., Hernandez D., Sanchez-Monge R., Barber D., del Pozo V., de Andres B., Armentia A., Lahoz C., Salcedo G., et al. Members of the alpha-amylase inhibitors family from wheat endosperm are major allergens associated with baker’s asthma. FEBS Lett. 1990;261:85–88. doi: 10.1016/0014-5793(90)80642-V.
    1. Zevallos V.F., Raker V., Tenzer S., Jimenez-Calvente C., Ashfaq-Khan M., Russel N., Pickert G., Schild H., Steinbrink K., Schuppan D. Nutritional wheat amylase-trypsin inhibitors promote intestinal inflammation via activation of myeloid cells. Gastroenterology. 2017;152:1100–1113. doi: 10.1053/j.gastro.2016.12.006.
    1. Dalla Pellegrina C., Perbellini O., Scupoli M.T., Tomelleri C., Zanetti C., Zoccatelli G., Fusi M., Peruffo A., Rizzi C., Chignola R. Effects of wheat germ agglutinin on human gastrointestinal epithelium: Insights from an experimental model of immune/epithelial cell interaction. Toxicol. Appl. Pharmacol. 2009;237:146–153. doi: 10.1016/j.taap.2009.03.012.
    1. Haska L., Nyman M., Andersson R. Distribution and characterisation of fructan in wheat milling fractions. J. Cereal Sci. 2008;48:768–774. doi: 10.1016/j.jcs.2008.05.002.
    1. Brouns F., Delzenne N., Gibson G. The dietary fibers-FODMAPs controversy. Cereal Foods World. 2017;62:768–774. doi: 10.1094/CFW-62-3-0098.
    1. Biesiekierski J.R., Rosella O., Rose R., Liels K., Barrett J.S., Shepherd S.J., Gibson P.R., Muir J.G. Quantification of fructans, galacto-oligosacharides and other short-chain carbohydrates in processed grains and cereals. J. Hum. Nutr. Diet. 2011;24:154–176. doi: 10.1111/j.1365-277X.2010.01139.x.
    1. Whelan K., Abrahmsohn O., David G.J., Staudacher H., Irving P., Lomer M.C., Ellis P.R. Fructan content of commonly consumed wheat, rye and gluten-free breads. Int. J. Food Sci. Nutr. 2011;62:498–503. doi: 10.3109/09637486.2011.553588.
    1. Chey W.D. Food: The main course to wellness and illness in patients with irritable bowel syndrome. Am. J. Gastroenterol. 2016;111:366–371. doi: 10.1038/ajg.2016.12.
    1. Volta U., Pinto-Sanchez M.I., Boschetti E., Caio G., De Giorgio R., Verdu E.F. Dietary triggers in irritable bowel syndrome: Is there a role for gluten? J. Neurogastroenterol. Motil. 2016;22:547–557. doi: 10.5056/jnm16069.
    1. Hayes P.A., Fraher M.H., Quigley E.M. Irritable bowel syndrome: The role of food in pathogenesis and management. Gastroenterol. Hepatol. 2014;10:164–174.
    1. De Giorgio R., Volta U., Gibson P.R. Sensitivity to wheat, gluten and FODMAPs in IBS: Facts or fiction? Gut. 2016;65:169–178. doi: 10.1136/gutjnl-2015-309757.
    1. Natividad J.M., Huang X., Slack E., Jury J., Sanz Y., David C., Denou E., Yang P., Murray J., McCoy K.D., et al. Host responses to intestinal microbial antigens in gluten-sensitive mice. PLoS ONE. 2009;4:e6472. doi: 10.1371/journal.pone.0006472.
    1. Wu R.L., Vazquez-Roque M.I., Carlson P., Burton D., Grover M., Camilleri M., Turner J.R. Gluten-induced symptoms in diarrhea-predominant irritable bowel syndrome are associated with increased myosin light chain kinase activity and claudin-15 expression. Lab. Investig. 2017;97:14–23. doi: 10.1038/labinvest.2016.118.
    1. Volta U., Caio G., Karunaratne T.B., Alaedini A., De Giorgio R. Non-coeliac gluten/wheat sensitivity: Advances in knowledge and relevant questions. Expert Rev. Gastroenterol. Hepatol. 2017;11:9–18. doi: 10.1080/17474124.2017.1260003.
    1. Dalrymple J., Bullock I. Diagnosis and management of irritable bowel syndrome in adults in primary care: Summary of nice guidance. BMJ. 2015;350:h1216. doi: 10.1136/.
    1. Lovell R.M., Ford A.C. Global prevalence of and risk factors for irritable bowel syndrome: A meta-analysis. Clin. Gastroenterol. Hepatol. 2012;10:712–721. doi: 10.1016/j.cgh.2012.02.029.
    1. Canavan C., West J., Card T. The epidemiology of irritable bowel syndrome. Clin. Epidemiol. 2014;6:71–80.
    1. Whitehead W.E., Palsson O., Jones K.R. Systematic review of the comorbidity of irritable bowel syndrome with other disorders: What are the causes and implications? Gastroenterology. 2002;122:1140–1156. doi: 10.1053/gast.2002.32392.
    1. Akehurst R.L., Brazier J.E., Mathers N., O’Keefe C., Kaltenthaler E., Morgan A., Platts M., Walters S.J. Health-related quality of life and cost impact of irritable bowel syndrome in a UK primary care setting. Pharmacoeconomics. 2002;20:455–462. doi: 10.2165/00019053-200220070-00003.
    1. Drossman D.A. Functional gastrointestinal disorders: History, pathophysiology, clinical features and Rome IV. Gastroenterology. 2016;150:1262–1279. doi: 10.1053/j.gastro.2016.02.032.
    1. Morcos A., Dinan T., Quigley E.M. Irritable bowel syndrome: Role of food in pathogenesis and management. J. Dig. Dis. 2009;10:237–246. doi: 10.1111/j.1751-2980.2009.00392.x.
    1. Bohn L., Storsrud S., Tornblom H., Bengtsson U., Simren M. Self-reported food-related gastrointestinal symptoms in IBS are common and associated with more severe symptoms and reduced quality of life. Am. J. Gastroenterol. 2013;108:634–641. doi: 10.1038/ajg.2013.105.
    1. McKenzie Y.A., Thompson J., Gulia P., Lomer M.C. British dietetic association systematic review of systematic reviews and evidence-based practice guidelines for the use of probiotics in the management of irritable bowel syndrome in adults (2016 update) J. Hum. Nutr. Diet. 2016;29:576–592. doi: 10.1111/jhn.12386.
    1. Jones V.A., McLaughlan P., Shorthouse M., Workman E., Hunter J.O. Food intolerance: A major factor in the pathogenesis of irritable bowel syndrome. Lancet. 1982;2:1115–1117. doi: 10.1016/S0140-6736(82)92782-9.
    1. King T.S., Elia M., Hunter J.O. Abnormal colonic fermentation in irritable bowel syndrome. Lancet. 1998;352:1187–1189. doi: 10.1016/S0140-6736(98)02146-1.
    1. Shepherd S.J., Gibson P.R. Fructose malabsorption and symptoms of irritable bowel syndrome: Guidelines for effective dietary management. J. Am. Diet. Assoc. 2006;106:1631–1639. doi: 10.1016/j.jada.2006.07.010.
    1. Marsh A., Eslick E.M., Eslick G.D. Does a diet low in FODMAPs reduce symptoms associated with functional gastrointestinal disorders? A comprehensive systematic review and meta-analysis. Eur. J. Nutr. 2016;55:897–906. doi: 10.1007/s00394-015-0922-1.
    1. Francis C.Y., Morris J., Whorwell P.J. The irritable bowel severity scoring system: A simple method of monitoring irritable bowel syndrome and its progress. Aliment. Pharmacol. Ther. 1997;11:395–402. doi: 10.1046/j.1365-2036.1997.142318000.x.
    1. Staudacher H.M., Lomer M.C.E., Whelan K., Irving P. Pth-064 implementation of the low-fermentable oligo-, di-, mono-saccharides and polyols diet in the UK: How easy is it and does it work? Abstract pth-064. Gut. 2010;59 doi: 10.1136/gut.2009.209049y.
    1. Staudacher H., Ross F.S., Briscoe Z.M., Irving P.M., Whelan K., Lomer M.C. Ptu-183 advice from a dietitian regarding the low FODMAP diet broadly maintains nutrient intake and does not alter fibre intake. Gut. 2015;64 doi: 10.1136/gutjnl-2015-309861.298.
    1. Rao S.S., Yu S., Fedewa A. Systematic review: Dietary fibre and FODMAP-restricted diet in the management of constipation and irritable bowel syndrome. Aliment. Pharmacol. Ther. 2015;41:1256–1270. doi: 10.1111/apt.13167.
    1. Staudacher H.M., Lomer M.C., Anderson J.L., Barrett J.S., Muir J.G., Irving P.M., Whelan K. Fermentable carbohydrate restriction reduces luminal bifidobacteria and gastrointestinal symptoms in patients with irritable bowel syndrome. J. Nutr. 2012;142:1510–1518. doi: 10.3945/jn.112.159285.
    1. Halmos E.P., Christophersen C.T., Bird A.R., Shepherd S.J., Gibson P.R., Muir J.G. Diets that differ in their FODMAP content alter the colonic luminal microenvironment. Gut. 2015;64:93–100. doi: 10.1136/gutjnl-2014-307264.
    1. Bohn L., Storsrud S., Liljebo T., Collin L., Lindfors P., Tornblom H., Simren M. Diet low in FODMAPs reduces symptoms of irritable bowel syndrome as well as traditional dietary advice: A randomized controlled trial. Gastroenterology. 2015;149:1399–1407. doi: 10.1053/j.gastro.2015.07.054.
    1. Whigham L., Joyce T., Harper G., Irving P.M., Staudacher H.M., Whelan K., Lomer M.C. Clinical effectiveness and economic costs of group versus one-to-one education for short-chain fermentable carbohydrate restriction (low FODMAP diet) in the management of irritable bowel syndrome. J. Hum. Nutr. Diet. 2015;28:687–696. doi: 10.1111/jhn.12318.
    1. Chey W.D., Whelan K. Dietary guidelines for irritable bowel syndrome are important for gastroenterologists, dietitians and people with irritable bowel syndrome. J. Hum. Nutr. Diet. 2016;29:547–548. doi: 10.1111/jhn.12413.
    1. Eswaran S.L., Chey W.D., Han-Markey T., Ball S., Jackson K. A randomized controlled trial comparing the low FODMAP diet vs. Modified nice guidelines in US adults with IBS-D. Am. J. Gastroenterol. 2016;111:1824–1832. doi: 10.1038/ajg.2016.434.
    1. Biesiekierski J.R., Peters S.L., Newnham E.D., Rosella O., Muir J.G., Gibson P.R. No effects of gluten in patients with self-reported non-celiac gluten sensitivity after dietary reduction of fermentable, poorly absorbed, short-chain carbohydrates. Gastroenterology. 2013;145:320–328. doi: 10.1053/j.gastro.2013.04.051.
    1. Aziz I., Hadjivassiliou M., Sanders D.S. The spectrum of noncoeliac gluten sensitivity. Nat. Rev. Gastroenterol. Hepatol. 2015;12:516–526. doi: 10.1038/nrgastro.2015.107.
    1. Peters S.L., Biesiekierski J.R., Yelland G.W., Muir J.G., Gibson P.R. Randomised clinical trial: Gluten may cause depression in subjects with non-coeliac gluten sensitivity—An exploratory clinical study. Aliment. Pharmacol. Ther. 2014;39:1104–1112. doi: 10.1111/apt.12730.
    1. Wahnschaffe U., Ullrich R., Riecken E.O., Schulzke J.D. Celiac disease-like abnormalities in a subgroup of patients with irritable bowel syndrome. Gastroenterology. 2001;121:1329–1338. doi: 10.1053/gast.2001.29572.
    1. Wahnschaffe U., Schulzke J.D., Zeitz M., Ullrich R. Predictors of clinical response to gluten-free diet in patients diagnosed with diarrhea-predominant irritable bowel syndrome. Clin. Gastroenterol. Hepatol. 2007;5:844–850. doi: 10.1016/j.cgh.2007.03.021.
    1. Biesiekierski J.R., Newnham E.D., Irving P.M., Barrett J.S., Haines M., Doecke J.D., Shepherd S.J., Muir J.G., Gibson P.R. Gluten causes gastrointestinal symptoms in subjects without celiac disease: A double-blind randomized placebo-controlled trial. Am. J. Gastroenterol. 2011;106:508–514. doi: 10.1038/ajg.2010.487.
    1. Vazquez-Roque M.I., Camilleri M., Smyrk T., Murray J.A., O’Neill J., Carlson P., Lamsam J., Eckert D., Janzow D., Burton D., et al. Association of hla-dq gene with bowel transit, barrier function, and inflammation in irritable bowel syndrome with diarrhea. Am. J. Physiol. Gastrointest. Liver Physiol. 2012;303:G1262–G1269. doi: 10.1152/ajpgi.00294.2012.
    1. Vazquez-Roque M.I., Camilleri M., Smyrk T., Murray J.A., Marietta E., O’Neill J., Carlson P., Lamsam J., Janzow D., Eckert D., et al. A controlled trial of gluten-free diet in patients with irritable bowel syndrome-diarrhea: Effects on bowel frequency and intestinal function. Gastroenterology. 2013;144:903–911. doi: 10.1053/j.gastro.2013.01.049.
    1. Aziz I., Trott N., Briggs R., North J.R., Hadjivassiliou M., Sanders D.S. Efficacy of a gluten-free diet in subjects with irritable bowel syndrome-diarrhea unaware of their HLA-DQ2/8 genotype. Clin. Gastroenterol. Hepatol. 2016;14:696–703. doi: 10.1016/j.cgh.2015.12.031.
    1. Di Sabatino A., Volta U., Salvatore C., Biancheri P., Caio G., De Giorgio R., Di Stefano M., Corazza G.R. Small amounts of gluten in subjects with suspected nonceliac gluten sensitivity: A randomized, double-blind, placebo-controlled, cross-over trial. Clin. Gastroenterol. Hepatol. 2015;13:1604–1612. doi: 10.1016/j.cgh.2015.01.029.
    1. Shahbazkhani B., Sadeghi A., Malekzadeh R., Khatavi F., Etemadi M., Kalantri E., Rostami-Nejad M., Rostami K. Non-celiac gluten sensitivity has narrowed the spectrum of irritable bowel syndrome: A double-blind randomized placebo-controlled trial. Nutrients. 2015;7:4542–4554. doi: 10.3390/nu7064542.
    1. Zanini B., Basche R., Ferraresi A., Ricci C., Lanzarotto F., Marullo M., Villanacci V., Hidalgo A., Lanzini A. Randomised clinical study: Gluten challenge induces symptom recurrence in only a minority of patients who meet clinical criteria for non-coeliac gluten sensitivity. Aliment. Pharmacol. Ther. 2015;42:968–976. doi: 10.1111/apt.13372.
    1. Zanwar V.G., Pawar S.V., Gambhire P.A., Jain S.S., Surude R.G., Shah V.B., Contractor Q.Q., Rathi P.M. Symptomatic improvement with gluten restriction in irritable bowel syndrome: A prospective, randomized, double blinded placebo controlled trial. Intest. Res. 2016;14:343–350. doi: 10.5217/ir.2016.14.4.343.
    1. Elli L., Tomba C., Branchi F., Roncoroni L., Lombardo V., Bardella M.T., Ferretti F., Conte D., Valiante F., Fini L., et al. Evidence for the presence of non-celiac gluten sensitivity in patients with functional gastrointestinal symptoms: Results from a multicenter randomized double-blind placebo-controlled gluten challenge. Nutrients. 2016;8:84. doi: 10.3390/nu8020084.
    1. Barmeyer C., Schumann M., Meyer T., Zielinski C., Zuberbier T., Siegmund B., Schulzke J.D., Daum S., Ullrich R. Long-term response to gluten-free diet as evidence for non-celiac wheat sensitivity in one third of patients with diarrhea-dominant and mixed-type irritable bowel syndrome. Int. J. Colorectal Dis. 2017;32:29–39. doi: 10.1007/s00384-016-2663-x.
    1. De Palma G., Nadal I., Collado M.C., Sanz Y. Effects of a gluten-free diet on gut microbiota and immune function in healthy adult human subjects. Br. J. Nutr. 2009;102:1154–1160. doi: 10.1017/S0007114509371767.
    1. Nistal E., Caminero A., Vivas S., Ruiz de Morales J.M., Saenz de Miera L.E., Rodriguez-Aparicio L.B., Casqueiro J. Differences in faecal bacteria populations and faecal bacteria metabolism in healthy adults and celiac disease patients. Biochimie. 2012;94:1724–1729. doi: 10.1016/j.biochi.2012.03.025.
    1. Wild D., Robins G.G., Burley V.J., Howdle P.D. Evidence of high sugar intake, and low fibre and mineral intake, in the gluten-free diet. Aliment. Pharmacol. Ther. 2010;32:573–581. doi: 10.1111/j.1365-2036.2010.04386.x.
    1. Kinsey L., Burden S.T., Bannerman E. A dietary survey to determine if patients with coeliac disease are meeting current healthy eating guidelines and how their diet compares to that of the british general population. Eur. J. Clin. Nutr. 2008;62:1333–1342. doi: 10.1038/sj.ejcn.1602856.
    1. Martin L., Van Vuuren C., Seamark L., Williams M., Staudacher H., Irving P.M., Whelan K., Lomer M.C. Long-term effectiveness of short chain fermentable carbohydrate (FODMAP) restriction in patients with IBS; Proceedings of the 2nd Digestive Disorders Federation Conference; London, UK. 22–25 June 2015.

Source: PubMed

3
Suscribir