Uncomplicated diverticular disease: innate and adaptive immunity in human gut mucosa before and after rifaximin

Rossella Cianci, Simona Frosali, Danilo Pagliari, Paola Cesaro, Lucio Petruzziello, Fabio Casciano, Raffaele Landolfi, Guido Costamagna, Franco Pandolfi, Rossella Cianci, Simona Frosali, Danilo Pagliari, Paola Cesaro, Lucio Petruzziello, Fabio Casciano, Raffaele Landolfi, Guido Costamagna, Franco Pandolfi

Abstract

Background/aim: Uncomplicated diverticular disease (UDD) is a frequent condition in adults. The pathogenesis of symptoms remains unknown. Bacteria are able to interact with Toll-like receptors (TLRs) and to induce inflammation through both innate immunity and T-cell recruitment. We investigated the pattern of TLRs 2 and 4 and the intestinal homing in patients with UDD before and after a course of Rifaximin.

Methods: Forty consecutive patients with UDD and 20 healthy asymptomatic subjects were enrolled. Among UDD patients, 20 were assigned to a 2-month course of treatment with Rifaximin 1.2 g/day for 15 days/month and 20 received placebo. Blood sample and colonic biopsies were obtained from patients and controls. The samples were collected and analyzed at baseline and at the end of treatment. Flow cytometry was performed using monoclonal antibodies (CD3, CD4, CD8, CD103, TCR-gamma/delta, CD14, TLR2, and TLR4).

Results: In UDD, TLR2 and TLR4 expression on immune cell subpopulations from blood and mucosa of the affected colon are altered as compared with controls. Rifaximin treatment induced significant modifications of altered conditions.

Conclusions: Our data show the role of TLRs in the development of inflammation in UDD. TLRs distribution is altered in UDD and these alterations are reversed after antibiotic treatment. This trial is registered with ClinicalTrials.gov: NCT02068482.

Figures

Figure 1
Figure 1
Expression of TLRs in lymphocytes. (a, b) TLR2 and TLR4 in peripheral blood lymphocytes, at baseline and after Rifaximin and placebo treatments. (c, d) TLR2 and TLR4 in sigma mucosa lymphocytes at baseline and after Rifaximin and placebo treatments. (e) TLR2 and TLR4 in transverse mucosa lymphocytes at baseline and after Rifaximin treatment. The values reported in box-and-whiskers plots show the minimum, the 25th percentile, the median, the 75th percentile, and the maximum. In particular, the whiskers go down to the smallest value (minimum) and up to the largest one (maximum), the top and bottom of the box are the 25th and 75th percentiles, and the line in the middle of the box is the median corresponding to 50th percentile (***P < 0.001, **P < 0.01, *P < 0.05 patients versus control (health or baseline)); §§P < 0.01, §P < 0.05 between treatments. Healthy control (CTR), UDD patients (PZ), before Rifaximin (BR), after Rifaximin (AR), before placebo (BP), and after placebo (AP).
Figure 2
Figure 2
Modulation of TLR2 and TLR4 lymphocytes in PBMC and gut. (a, b) The comparison of modulation of TLR2 and TLR4 expression in the different patient groups between PBMC and gut (before and after placebo or Rifaximin). (c-d) Modulation of TLR2 and TLR4 expression observed in a single representative patient. The values reported in box-and-whiskers plots show the minimum, the 25th percentile, the median, the 75th percentile, and the maximum. In particular, the whiskers go down to the smallest value (minimum) and up to the largest one (maximum), the top and bottom of the box are the 25th and 75th percentiles, and the line in the middle of the box is the median, corresponding to 50th percentile, evaluated before Rifaximin (BR), after Rifaximin (AR), before placebo (BP), and after placebo (AP).
Figure 3
Figure 3
Expression of TLRs in monocytes and granulocytes. (a, b) Percentage of TLR2 and TLR4 monocytes in peripheral blood, at baseline and after Rifaximin and placebo treatments. (c, d) Median of fluorescence (MFI) of TLR2 and TLR4 monocytes in peripheral blood at baseline and after Rifaximin and placebo treatments. (e, f) MFI of TLR2 and TLR4 granulocytes in peripheral blood at baseline and after Rifaximin and placebo treatments. The values reported in box-and-whiskers plots show the minimum, the 25th percentile, the median, the 75th percentile, and the maximum. In particular, the whiskers go down to the smallest value (minimum) and up to the largest one (maximum), the top and bottom of the box are the 25th and 75th percentiles, and the line in the middle of the box is the median corresponding to 50th percentile (***P < 0.001, **P < 0.01, and *P < 0.05 patients versus controls (health or baseline)). Healthy control (CTR), UDD patients (PZ), before Rifaximin (BR), after Rifaximin (AR), before placebo (BP), and after placebo (AP).
Figure 4
Figure 4
Expression of CD103 in lymphocytes of sigma mucosa. (a) Percentage of CD103 in CD4 lymphocytes subpopulation after Rifaximin and placebo treatments. (b) Percentage of CD103 in CD8 lymphocytes subpopulation after Rifaximin and placebo treatments. (c) Percentage of TCR-gamma/delta CD103 lymphocytes subpopulation after Rifaximin and placebo treatments. The values reported in box-and-whiskers plots show the minimum, the 25th percentile, the median, the 75th percentile, and the maximum. In particular, the whiskers go down to the smallest value (minimum) and up to the largest one (maximum), the top and bottom of the box are the 25th and 75th percentiles, and the line in the middle of the box is the median corresponding to 50th percentile (*P < 0.05 before versus after treatments in patients); before Rifaximin (BR), after Rifaximin (AR), before placebo (BP), and after placebo (AP).

References

    1. Kruis W, Meier E, Schumacher M, Mickisch O, Greinwald R, Mueller R. Randomised clinical trial: Mesalazine (Salofalk granules) for uncomplicated diverticular disease of the colon—a placebo-controlled study. Alimentary Pharmacology and Therapeutics. 2013;37(7):680–690.
    1. Isolauri E, Sutas Y, Kankaanpää P, Arvilommi H, Salminen S. Probiotics: effects on immunity. The American Journal of Clinical Nutrition. 2001;73(supplement 2):444S–450S.
    1. Barbara G, Stanghellini V, Brandi G, et al. Interactions between commensal bacteria and gut sensorimotor function in health and disease. The American Journal of Gastroenterology. 2005;100(11):2560–2568.
    1. Fiocchi C. Inflammatory bowel disease: etiology and pathogenesis. Gastroenterology. 1998;115(1):182–205.
    1. Fiorucci S, Distrutti E, Mencarelli A, Barbanti M, Palazzini E, Morelli A. Inhibition of intestinal bacterial translocation with rifaximin modulates lamina propria monocytic cells reactivity and protects against inflammation in a rodent model of colitis. Digestion. 2002;66(4):246–256.
    1. Sheth AA, Longo W, Floch MH. Diverticular disease and diverticulitis. The American Journal of Gastroenterology. 2008;103(6):1550–1556.
    1. Ferzoco LB, Raptopoulos V, Silen W. Acute diverticulitis. The New England Journal of Medicine. 1998;338(21):1521–1526.
    1. Cianci R, Iacopini F, Petruzziello L, Cammarota G, Pandolfi F, Costamagna G. Involvement of central immunity in uncomplicated diverticular disease. Scandinavian Journal of Gastroenterology. 2009;44(1):108–115.
    1. Blandizzi C, Viscomi GC, Marzo A, Scarpignato C. Is generic rifaximin still a poorly absorbed antibiotic? A comparison of branded and generic formulations in healthy volunteers. Pharmacological Research. 2014;85:39–44.
    1. Akira S. Toll-like receptor signaling. The Journal of Biological Chemistry. 2003;278(40):38105–38108.
    1. Werling D, Jann OC, Offord V, Glass EJ, Coffey TJ. Variation matters: TLR structure and species-specific pathogen recognition. Trends in Immunology. 2009;30(3):124–130.
    1. Pasare C, Medzhitov R. Toll-like receptors: linking innate and adaptive immunity. Advances in Experimental Medicine and Biology. 2005;560:11–18.
    1. Liu G, Zhao Y. Toll-like receptors and immune regulation: their direct and indirect modulation on regulatory CD4+ CD25+ T cells. Immunology. 2007;122(2):149–156.
    1. Caramalho I, Lopes-Carvalho T, Ostler D, Zelenay S, Haury M, Demengeot J. Regulatory T cells selectively express toll-like receptors and are activated by lipopolysaccharide. Journal of Experimental Medicine. 2003;197(4):403–411.
    1. Cottalorda A, Mercier BC, Mbitikon-Kobo FM, et al. TLR2 engagement on memory CD8+ T cells improves their cytokine-mediated proliferation and IFN-γ secretion in the absence of Ag. European Journal of Immunology. 2009;39(10):2673–2681.
    1. Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY. TLR2 is expressed activated T cells as a costimulatory receptor. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(9):3029–3034.
    1. Siddique I, Khan I. Mechanism of regulation of Na-H exchanger in inflammatory bowel disease: role of TLR-4 signaling mechanism. Digestive Diseases and Sciences. 2011;56(6):1656–1662.
    1. Zhao L, Kwon M, Huang S, et al. Differential modulation of nods signaling pathways by fatty acids in human colonic epithelial HCT116 cells. Journal of Biological Chemistry. 2007;282(16):11618–11628.
    1. Prabha C, Rajashree P, Sulochana DD. TLR2 and TLR4 expression on the immune cells of tuberculous pleural fluid. Immunology Letters. 2008;117(1):26–34.
    1. Babu S, Blauvelt CP, Kumaraswami V, Nutman TB. Cutting edge: diminished T cell TLR expression and function modulates the immune response in human filarial infection. Journal of Immunology. 2006;176(7):3885–3889.
    1. Cammarota G, Pandolfi F, Cianci R, Pagliari D, Landolfi R, Kunkel S. Cellular mediators of inflammation: tregs and T H 17 cells in gastrointestinal diseases. Mediators of Inflammation. 2009;2009132028
    1. Liu PT, Stenger S, Li H, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311(5768):1770–1773.
    1. Cianci R, Cammarota G, Frisullo G. Tissue-infiltrating lymphocytes analysis reveals large modifications of the duodenal immunological niche in coeliac disease after gluten-free diet. Clinical and Translational Gastroenterology. 2012;3:p. e28.
    1. Olsen T, Rismo R, Cui G, Goll R, Christiansen I, Florholmen J. TH1 and TH17 interactions in untreated inflamed mucosa of inflammatory bowel disease, and their potential to mediate the inflammation. Cytokine. 2011;56(3):633–640.
    1. Mencarelli A, Migliorati M, Barbanti M, et al. Pregnane-X-receptor mediates the anti-inflammatory activities of rifaximin on detoxification pathways in intestinal epithelial cells. Biochemical Pharmacology. 2010;80(11):1700–1707.
    1. Salomão R, Martins PS, Brunialti MKC, et al. TLR signaling pathway in patients with sepsis. Shock. 2008;30(supplement 1):73–76.
    1. Abu-Maziad A, Schaa K, Bell EF, et al. Role of polymorphic variants as genetic modulators of infection in neonatal sepsis. Pediatric Research. 2010;68(4):323–329.
    1. Agnese DM, Calvano JE, Hahm SJ, et al. Human toll-like receptor 4 mutations but not CD14 polymorphisms are associated with an increased risk of gram-negative infections. Journal of Infectious Diseases. 2002;186(10):1522–1525.
    1. Ismail AS, Severson KM, Vaishnava S, et al. Gammadelta intraepithelial lymphocytes are essential mediators of host-microbial homeostasis at the intestinal mucosal surface. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(21):8743–8748.
    1. Cianci R, Pagliari D, Landolfi R, et al. New insights on the role of T cells in the pathogenesis of celiac disease. Journal of Biological Regulators and Homeostatic Agents. 2012;26(2):171–179.
    1. Zuo A, Liang D, Shao H, Born WK, Kaplan HJ, Sun D. In vivo priming of IL-17+ uveitogenic T cells is enhanced by Toll ligand receptor (TLR)2 and TLR4 agonists via γδ T cell activation. Molecular Immunology. 2012;50(3):125–133.

Source: PubMed

3
Prenumerera