Gastroenterology

Gastroenterology

Volume 144, Issue 5, May 2013, Pages 903-911.e3
Gastroenterology

Original Research
Full Report: Clinical—Alimentary Tract
A Controlled Trial of Gluten-Free Diet in Patients With Irritable Bowel Syndrome-Diarrhea: Effects on Bowel Frequency and Intestinal Function

https://doi.org/10.1053/j.gastro.2013.01.049Get rights and content

Background & Aims

Patients with diarrhea-predominant irritable bowel syndrome (IBS-D) could benefit from a gluten-free diet (GFD).

Methods

We performed a randomized controlled 4-week trial of a gluten-containing diet (GCD) or GFD in 45 patients with IBS-D; genotype analysis was performed for HLA-DQ2 and HLA-DQ8. Twenty-two patients were placed on the GCD (11 HLA-DQ2/8 negative and 11 HLA-DQ2/8 positive) and 23 patients were placed on the GFD (12 HLA-DQ2/8 negative and 11 HLA-DQ2/8 positive). We measured bowel function daily, small-bowel (SB) and colonic transit, mucosal permeability (by lactulose and mannitol excretion), and cytokine production by peripheral blood mononuclear cells after exposure to gluten and rice. We collected rectosigmoid biopsy specimens from 28 patients, analyzed levels of messenger RNAs encoding tight junction proteins, and performed H&E staining and immunohistochemical analyses. Analysis of covariance models was used to compare data from the GCD and GFD groups.

Results

Subjects on the GCD had more bowel movements per day (P = .04); the GCD had a greater effect on bowel movements per day of HLA-DQ2/8–positive than HLA-DQ2/8–negative patients (P = .019). The GCD was associated with higher SB permeability (based on 0−2 h levels of mannitol and the lactulose:mannitol ratio); SB permeability was greater in HLA-DQ2/8–positive than HLA-DQ2/8–negative patients (P = .018). No significant differences in colonic permeability were observed. Patients on the GCD had a small decrease in expression of zonula occludens 1 in SB mucosa and significant decreases in expression of zonula occludens 1, claudin-1, and occludin in rectosigmoid mucosa; the effects of the GCD on expression were significantly greater in HLA-DQ2/8–positive patients. The GCD vs the GFD had no significant effects on transit or histology. Peripheral blood mononuclear cells produced higher levels of interleukin-10, granulocyte colony-stimulating factor, and transforming growth factor-α in response to gluten than rice (unrelated to HLA genotype).

Conclusions

Gluten alters bowel barrier functions in patients with IBS-D, particularly in HLA-DQ2/8–positive patients. These findings reveal a reversible mechanism for the disorder. Clinical trials.gov NCT01094041.

Section snippets

Study Design and Intervention

Between January 2010 and February 2012, we conducted a single-center, parallel-group, randomized, controlled, 4-week trial of GCD and GFD in 45 gluten-ingesting patients with IBS-D. They were recruited from a database of more than 800 patients with IBS who had been evaluated clinically by the investigators or clinical staff at Mayo Clinic. These patients reside within 150 miles of Mayo Clinic in Rochester, Minnesota, and the database is maintained in the laboratory of the principal

Demographics and Baseline Characteristics

Supplementary Figure 1 shows the trial flow and summarizes the baseline demographic data. There were no differences in age, sex distribution, Hospital Anxiety and Depression scores, body mass index, baseline 0–2 hour excretion of mannitol and lactulose, and baseline colonic transit in the groups assigned to each diet. As expected in this patient population, there was a female predominance.

The prestudy diet questionnaire showed that the patients were not ingesting a gluten-free diet before the

Discussion

In this 4-week, randomized, controlled, diet intervention study, the intervention was associated with several significant effects. Subjects on a GCD had increased stool frequency compared with a GFD; this effect was greater in HLA-DQ2– or HLA-DQ8–positive patients. Although the absolute difference in stool frequency was small, it is important to appreciate that this increased frequency is on a background of the typical increase in stool frequency (average, 2.6 bowel movements/day at baseline)

Acknowledgments

The authors thank Michael Ryks and Debbie Rhoten for technical support, and Cindy Stanislav for secretarial assistance.

Maria Vazquez-Roque was the fellow investigator, and was responsible for participant recruitment and wrote the article; Michael Camilleri was the principal investigator, wrote the protocol, and wrote the article; Thomas Smyrk was responsible for surgical pathology; Joseph Murray wrote the article; Jessica O'Neill was the study coordinator and was responsible for participant

References (29)

  • E.F. Verdu et al.

    Gliadin-dependent neuromuscular and epithelial secretory responses in gluten-sensitive HLA-DQ8 transgenic mice

    Am J Physiol

    (2008)
  • A.S. Rao et al.

    Urine sugars for in vivo gut permeability: validation and comparisons in irritable bowel syndrome-diarrhea and controls

    Am J Physiol

    (2011)
  • C. Martínez et al.

    The jejunum of diarrhea-predominant irritable bowel syndrome shows molecular alterations in the tight junction signaling pathway that are associated with mucosal pathobiology and clinical manifestations

    Am J Gastroenterol

    (2012)
  • S.P. Dunlop et al.

    Abnormal intestinal permeability in subgroups of diarrhea-predominant irritable bowel syndromes

    Am J Gastroenterol

    (2006)
  • Cited by (384)

    • Dietary Interventions and Brain–Gut Disorders

      2024, The Gut-Brain Axis, Second Edition
    • Intestinal permeability, autoimmunity and gluten

      2023, Medecine des Maladies Metaboliques
    View all citing articles on Scopus

    Conflicts of interest This author discloses the following: Joseph Murray has received grants from Alba Therapeutics for clinical trials with the drug larazotide. The remaining authors disclose no conflicts.

    Funding This study was supported in part by National Institutes of Health grants 1RC1-DK086182 and R01-DK092179 (M.C.) and by a National Institutes of Health Clinical Translational Science Awards Program grant (UL1 TR000135).

    View full text