Functional variants in the sucrase-isomaltase gene associate with increased risk of irritable bowel syndrome

Maria Henström, Lena Diekmann, Ferdinando Bonfiglio, Fatemeh Hadizadeh, Eva-Maria Kuech, Maren von Köckritz-Blickwede, Louise B Thingholm, Tenghao Zheng, Ghazaleh Assadi, Claudia Dierks, Martin Heine, Ute Philipp, Ottmar Distl, Mary E Money, Meriem Belheouane, Femke-Anouska Heinsen, Joseph Rafter, Gerardo Nardone, Rosario Cuomo, Paolo Usai-Satta, Francesca Galeazzi, Matteo Neri, Susanna Walter, Magnus Simrén, Pontus Karling, Bodil Ohlsson, Peter T Schmidt, Greger Lindberg, Aldona Dlugosz, Lars Agreus, Anna Andreasson, Emeran Mayer, John F Baines, Lars Engstrand, Piero Portincasa, Massimo Bellini, Vincenzo Stanghellini, Giovanni Barbara, Lin Chang, Michael Camilleri, Andre Franke, Hassan Y Naim, Mauro D'Amato, Maria Henström, Lena Diekmann, Ferdinando Bonfiglio, Fatemeh Hadizadeh, Eva-Maria Kuech, Maren von Köckritz-Blickwede, Louise B Thingholm, Tenghao Zheng, Ghazaleh Assadi, Claudia Dierks, Martin Heine, Ute Philipp, Ottmar Distl, Mary E Money, Meriem Belheouane, Femke-Anouska Heinsen, Joseph Rafter, Gerardo Nardone, Rosario Cuomo, Paolo Usai-Satta, Francesca Galeazzi, Matteo Neri, Susanna Walter, Magnus Simrén, Pontus Karling, Bodil Ohlsson, Peter T Schmidt, Greger Lindberg, Aldona Dlugosz, Lars Agreus, Anna Andreasson, Emeran Mayer, John F Baines, Lars Engstrand, Piero Portincasa, Massimo Bellini, Vincenzo Stanghellini, Giovanni Barbara, Lin Chang, Michael Camilleri, Andre Franke, Hassan Y Naim, Mauro D'Amato

Abstract

Objective: IBS is a common gut disorder of uncertain pathogenesis. Among other factors, genetics and certain foods are proposed to contribute. Congenital sucrase-isomaltase deficiency (CSID) is a rare genetic form of disaccharide malabsorption characterised by diarrhoea, abdominal pain and bloating, which are features common to IBS. We tested sucrase-isomaltase (SI) gene variants for their potential relevance in IBS.

Design: We sequenced SI exons in seven familial cases, and screened four CSID mutations (p.Val557Gly, p.Gly1073Asp, p.Arg1124Ter and p.Phe1745Cys) and a common SI coding polymorphism (p.Val15Phe) in a multicentre cohort of 1887 cases and controls. We studied the effect of the 15Val to 15Phe substitution on SI function in vitro. We analysed p.Val15Phe genotype in relation to IBS status, stool frequency and faecal microbiota composition in 250 individuals from the general population.

Results: CSID mutations were more common in patients than asymptomatic controls (p=0.074; OR=1.84) and Exome Aggregation Consortium reference sequenced individuals (p=0.020; OR=1.57). 15Phe was detected in 6/7 sequenced familial cases, and increased IBS risk in case-control and population-based cohorts, with best evidence for diarrhoea phenotypes (combined p=0.00012; OR=1.36). In the population-based sample, 15Phe allele dosage correlated with stool frequency (p=0.026) and Parabacteroides faecal microbiota abundance (p=0.0024). The SI protein with 15Phe exhibited 35% reduced enzymatic activity in vitro compared with 15Val (p<0.05).

Conclusions: SI gene variants coding for disaccharidases with defective or reduced enzymatic activity predispose to IBS. This may help the identification of individuals at risk, and contribute to personalising treatment options in a subset of patients.

Keywords: DIARRHOEA; GENETICS; IRRITABLE BOWEL SYNDROME; POLYMORPHIC VARIATION.

Conflict of interest statement

Competing interests: The work was partially financed by an unrestricted grant from Medical Need Europe AB to MDA. MDA and HYN have received unrestricted research grants and lecturing honoraria from QOL Medical, and LC has served on a scientific advisory board for QOL Medical.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.

Figures

Figure 1
Figure 1
Properties of sucrase–isomaltase (SI) mutants and common coding polymorphisms. Left: schematic representation of SI protein structure and functional domains; the position of congenital sucrase–isomaltase deficiency (CSID) mutations and common coding variants is reported and colour-coded according to their functional effects (red=damaging, green=benign). Right: the variants, corresponding dbSNP IDs (http://www.ncbi.nlm.nih.gov/SNP), SI protein domain location (TM, transmembrane; I, isomaltase; S, sucrase), allele frequency, PHRED-like score (range 1–99, ranking a variant relative to all possible substitutions in the human genome) and predicted functional consequences are reported. #Exome Aggregation Consortium browser (http://exac.broadinstitute.org); ^Combined Annotation-Dependent Depletion database (http://cadd.gs.washington.edu/info). SNP, single nucleotide polymorphism.
Figure 2
Figure 2
Functional characterisation of the p.Val15Phe coding polymorphism. COS-1 cells were transiently transfected with either 15Val or 15Phe cDNAs and studied 48 hours after transfection. Individual values for 15Val and 15Phe cells from the same experiment are indicated with identical symbols. Net differences are reported with red bars as per cent average relative to 15Val arbitrarily set as 100% reference. (A) Cell surface localisation via immunofluorescence. Non-permeabilised cells were immunostained with a mixture of anti-sucrase–isomaltase (SI) antibodies and Alexa 488 secondary antibody, and analysed by confocal laser scanning microscopy on the xy (scale bar 25 µm) and xz planes (scale bar 10 µm). (B) Quantification of cell surface expression. SI surface proteins were labelled with biotin and immunoprecipitated using anti-Si antibodies after cell lysis. Immunoprecipitates were divided into two equal aliquots and analysed by immunoblotting with either anti-SI or anti-streptavidin antibodies. Relative quantification of surface-bound SI versus total cell SI was performed, and results are expressed in relation to values obtained for 15Val, which is set to 100%. (C) Quantification of association with sphingolipid/cholesterol-rich microdomains (lipid rafts) via detergent-resistant membrane (DRM) analysis. Following non-ionic detergent cell lysis, SI proteins were immunoprecipitated, fractionated by ultracentrifugation into insoluble (pellet, raft) and soluble (supernatant, non-raft) fractions, and DRM association (raft) quantified by immunoblotting with anti-SI antibodies. (D) Quantification of enzymatic activity. Sucrase activity was determined on immunoprecipitated SI proteins by measuring glucose release with the GOD–PAP method, upon normalisation for total protein amount by immunoblotting. *p

Figure 3

Correlation between p.Val15Phe genotype and…

Figure 3

Correlation between p.Val15Phe genotype and stool frequency. Mean (±SD) number of bowel movements…

Figure 3
Correlation between p.Val15Phe genotype and stool frequency. Mean (±SD) number of bowel movements per day (stool frequency, y-axis) is reported for Population-based Colonoscopy Study individuals (with available diary data) stratified according to the genotype at the p.Val15Phe single nucleotide polymorphism site (x-axis). The Spearman's p value for the correlation test is also reported.
Figure 3
Figure 3
Correlation between p.Val15Phe genotype and stool frequency. Mean (±SD) number of bowel movements per day (stool frequency, y-axis) is reported for Population-based Colonoscopy Study individuals (with available diary data) stratified according to the genotype at the p.Val15Phe single nucleotide polymorphism site (x-axis). The Spearman's p value for the correlation test is also reported.

References

    1. Sperber AD, Dumitrascu D, Fukudo S, et al. . The global prevalence of IBS in adults remains elusive due to the heterogeneity of studies: a Rome Foundation working team literature review. Gut Published Online First: 27 Jan 2016. doi: 10.1136/gutjnl-2015-311240 10.1136/gutjnl-2015-311240
    1. Chey WD, Kurlander J, Eswaran S. Irritable bowel syndrome: a clinical review. JAMA 2015;313:949–58. 10.1001/jama.2015.0954
    1. Drossman DA. The functional gastrointestinal disorders and the Rome III process. Gastroenterology 2006;130:1377–90. 10.1053/j.gastro.2006.03.008
    1. Camilleri M. Peripheral mechanisms in irritable bowel syndrome. N Engl J Med 2012;367:1626–35. 10.1056/NEJMra1207068
    1. Gazouli M, Wouters MM, Kapur-Pojskić L, et al. . Lessons learned—resolving the enigma of genetic factors in IBS. Nat Rev Gastroenterol Hepatol 2016;13:77–87. 10.1038/nrgastro.2015.206
    1. Ragnarsson G, Bodemar G. Pain is temporally related to eating but not to defaecation in the irritable bowel syndrome (IBS). Patients’ description of diarrhea, constipation and symptom variation during a prospective 6-week study. Eur J Gastroenterol Hepatol 1998;10:415–21.
    1. Böhn L, Störsrud S, Törnblom H, et al. . 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;108:634–41. 10.1038/ajg.2013.105
    1. Gibson PR, Varney J, Malakar S, et al. . Food components and irritable bowel syndrome. Gastroenterology 2015;148:1158–74. 10.1053/j.gastro.2015.02.005
    1. De Giorgio R, Volta U, Gibson PR. Sensitivity to wheat, gluten and FODMAPs in IBS: facts or fiction? Gut 2016;65:169–78. 10.1136/gutjnl-2015-309757
    1. Money ME, Walkowiak J, Virgilio C, et al. . Pilot study: a randomised, double blind, placebo controlled trial of pancrealipase for the treatment of postprandial irritable bowel syndrome-diarrhoea. Frontline Gastroenterol 2011;2:48–56. 10.1136/fg.2010.002253
    1. Karnsakul W, Luginbuehl U, Hahn D, et al. . Disaccharidase activities in dyspeptic children: biochemical and molecular investigations of maltase-glucoamylase activity. J Pediatr Gastroenterol Nutr 2002;35:551–6. 10.1097/00005176-200210000-00017
    1. Treem WR. Congenital sucrase-isomaltase deficiency. J Pediatr Gastroenterol Nutr 1995;21:1–14. 10.1097/00005176-199507000-00001
    1. Diaz-Sotomayor M, Quezada-Calvillo R, Avery SE, et al. . Maltase-glucoamylase modulates gluconeogenesis and sucrase-isomaltase dominates starch digestion glucogenesis. J Pediatr Gastroenterol Nutr 2013;57:704–12. 10.1097/MPG.0b013e3182a27438
    1. Alfalah M, Keiser M, Leeb T, et al. . Compound heterozygous mutations affect protein folding and function in patients with congenital sucrase-isomaltase deficiency. Gastroenterology 2009;136:883–92. 10.1053/j.gastro.2008.11.038
    1. Naim HY, Heine M, Zimmer K-P. Congenital sucrase-isomaltase deficiency: heterogeneity of inheritance, trafficking, and function of an intestinal enzyme complex. J Pediatr Gastroenterol Nutr 2012;55(Suppl 2):S13–20. 10.1097/01.mpg.0000421402.57633.4b
    1. Ouwendijk J, Moolenaar CE, Peters WJ, et al. . Congenital sucrase-isomaltase deficiency. Identification of a glutamine to proline substitution that leads to a transport block of sucrase-isomaltase in a pre-Golgi compartment. J Clin Invest 1996;97:633–41.
    1. Muldoon C, Maguire P, Gleeson F. Onset of sucrase-isomaltase deficiency in late adulthood. Am J Gastroenterol 1999;94:2298–9. 10.1111/j.1572-0241.1999.01320.x
    1. Ringrose RE, Preiser H, Welsh JD. Sucrase-isomaltase (palatinase) deficiency diagnosed during adulthood. Dig Dis Sci 1980;25:384–7. 10.1007/BF01308064
    1. Uhrich S, Wu Z, Huang J-Y, et al. . Four mutations in the SI gene are responsible for the majority of clinical symptoms of CSID. J Pediatr Gastroenterol Nutr 2012;55(Suppl 2):S34–5. 10.1097/01.mpg.0000421408.65257.b5
    1. Wouters MM, Lambrechts D, Knapp M, et al. . Genetic variants in CDC42 and NXPH1 as susceptibility factors for constipation and diarrhoea predominant irritable bowel syndrome. Gut 2014;63:1103–11. 10.1136/gutjnl-2013-304570
    1. Zucchelli M, Camilleri M, Andreasson AN, et al. . Association of TNFSF15 polymorphism with irritable bowel syndrome. Gut 2011;60:1671–7. 10.1136/gut.2011.241877
    1. Ek WE, Reznichenko A, Ripke S, et al. . Exploring the genetics of irritable bowel syndrome: a GWA study in the general population and replication in multinational case-control cohorts. Gut 2015;64:1774–82. 10.1136/gutjnl-2014-307997
    1. Beyder A, Mazzone A, Strege PR, et al. . Loss-of-function of the voltage-gated sodium channel NaV1.5 (channelopathies) in patients with irritable bowel syndrome. Gastroenterology 2014;146:1659–68. 10.1053/j.gastro.2014.02.054
    1. Grasberger H, Chang L, Shih W, et al. . Identification of a functional TPH1 polymorphism associated with irritable bowel syndrome bowel habit subtypes. Am J Gastroenterol 2013;108:1766–74. 10.1038/ajg.2013.304
    1. Orand A, Gupta A, Shih W, et al. . Catecholaminergic gene polymorphisms are associated with gi symptoms and morphological brain changes in irritable bowel syndrome. PLoS ONE 2015;10:e0135910 10.1371/journal.pone.0135910
    1. Walter SA, Kjellström L, Nyhlin H, et al. . Assessment of normal bowel habits in the general adult population: the Popcol study. Scand J Gastroenterol 2010;45:556–66. 10.3109/00365520903551332
    1. Kjellström L, Molinder H, Agréus L, et al. . A randomly selected population sample undergoing colonoscopy: prevalence of the irritable bowel syndrome and the impact of selection factors. Eur J Gastroenterol Hepatol 2014;26:268–75. 10.1097/MEG.0000000000000024
    1. Sander P, Alfalah M, Keiser M, et al. . Novel mutations in the human sucrase-isomaltase gene (SI) that cause congenital carbohydrate malabsorption. Hum Mutat 2006;27:119 10.1002/humu.9392
    1. Moolenaar CE, Naim HY. Cloning and expression of human intestinal sucrase-isomaltase. Biochem Soc Trans 1995;23:304S 10.1042/bst023304s
    1. Dahlqvist A. Assay of intestinal disaccharidases. Anal Biochem 1968;22:99–107. 10.1016/0003-2697(68)90263-7
    1. Kircher M, Witten DM, Jain P, et al. . A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet 2014;46:310–15. 10.1038/ng.2892
    1. Chassard C, Lacroix C. Carbohydrates and the human gut microbiota. Curr Opin Clin Nutr Metab Care 2013;16:453–60. 10.1097/MCO.0b013e3283619e63
    1. Halpert A, Dalton CB, Palsson O, et al. . What patients know about irritable bowel syndrome (IBS) and what they would like to know. National Survey on Patient Educational Needs in IBS and development and validation of the Patient Educational Needs Questionnaire (PEQ). Am J Gastroenterol 2007;102:1972–82.
    1. Bohn L, Storsrud S, Liljebo T, et al. . Diet low in FODMAPs reduces symptoms of irritable bowel syndrome as well as traditional dietary advice: a randomized controlled trial. Gastroenterology 2015;149:1399–407. 10.1053/j.gastro.2015.07.054
    1. Wu GD, Chen J, Hoffmann C, et al. . Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334:105–8. 10.1126/science.1208344
    1. Noor SO, Ridgway K, Scovell L, et al. . Ulcerative colitis and irritable bowel patients exhibit distinct abnormalities of the gut microbiota. BMC Gastroenterol 2010;10:134 10.1186/1471-230X-10-134
    1. Rangel I, Sundin J, Fuentes S, et al. . The relationship between faecal-associated and mucosal-associated microbiota in irritable bowel syndrome patients and healthy subjects. Aliment Pharmacol Ther 2015;42:1211–21. 10.1111/apt.13399

Source: PubMed

3
Abonneren