Targeting zonulin and intestinal epithelial barrier function to prevent onset of arthritis

Narges Tajik, Michael Frech, Oscar Schulz, Fabian Schälter, Sébastien Lucas, Vugar Azizov, Kerstin Dürholz, Franziska Steffen, Yasunori Omata, Andreas Rings, Marko Bertog, Aroldo Rizzo, Aida Iljazovic, Marijana Basic, Arnd Kleyer, Stephan Culemann, Gerhard Krönke, Yubin Luo, Klaus Überla, Udo S Gaipl, Benjamin Frey, Till Strowig, Kerstin Sarter, Stephan C Bischoff, Stefan Wirtz, Juan D Cañete, Francesco Ciccia, Georg Schett, Mario M Zaiss, Narges Tajik, Michael Frech, Oscar Schulz, Fabian Schälter, Sébastien Lucas, Vugar Azizov, Kerstin Dürholz, Franziska Steffen, Yasunori Omata, Andreas Rings, Marko Bertog, Aroldo Rizzo, Aida Iljazovic, Marijana Basic, Arnd Kleyer, Stephan Culemann, Gerhard Krönke, Yubin Luo, Klaus Überla, Udo S Gaipl, Benjamin Frey, Till Strowig, Kerstin Sarter, Stephan C Bischoff, Stefan Wirtz, Juan D Cañete, Francesco Ciccia, Georg Schett, Mario M Zaiss

Abstract

Gut microbial dysbiosis is associated with the development of autoimmune disease, but the mechanisms by which microbial dysbiosis affects the transition from asymptomatic autoimmunity to inflammatory disease are incompletely characterized. Here, we identify intestinal barrier integrity as an important checkpoint in translating autoimmunity to inflammation. Zonulin family peptide (zonulin), a potent regulator for intestinal tight junctions, is highly expressed in autoimmune mice and humans and can be used to predict transition from autoimmunity to inflammatory arthritis. Increased serum zonulin levels are accompanied by a leaky intestinal barrier, dysbiosis and inflammation. Restoration of the intestinal barrier in the pre-phase of arthritis using butyrate or a cannabinoid type 1 receptor agonist inhibits the development of arthritis. Moreover, treatment with the zonulin antagonist larazotide acetate, which specifically increases intestinal barrier integrity, effectively reduces arthritis onset. These data identify a preventive approach for the onset of autoimmune disease by specifically targeting impaired intestinal barrier function.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. Zonulin family peptide levels and…
Fig. 1. Zonulin family peptide levels and intestinal barrier dysfunction in human rheumatoid arthritis.
a Serum zonulin family peptide (zonulin) levels in healthy controls (controls) (n = 41), cancer (n = 19), hepatitis C virus (HCV) (n = 21), and rheumatoid arthritis (RA) (n = 40) patients. b Serum zonulin levels in healthy controls (n = 41) and pre-RA patients (n = 32). c Serum zonulin levels in pre-RA patients with (n = 12) or without (n = 53) later development of RA (left panel); Kaplan–Meier graph showing loss of healthy state and progression to RA patients with low (<10) or elevated (≥10) zonulin levels over month relapsed (right panel). d Fold reduction of occludin expression determined in histological gut biopsy sections in healthy controls (n = 10), new-onset (n = 9) and established RA patients (n = 5) with representative microphotographs (occludin in brown) from three healthy control and three new-onset RA patients. Size bar: 20 µm. e Fold reduction of claudin-1 expression determined in histological gut biopsy sections in healthy controls (n = 10), new-onset (n = 10) and established RA patients (n = 5) with representative microphotographs (claudin-1 in brown) from three healthy control and three new-onset RA patients. Size bar: 20 µm. f Immunohistological quantification of gut infiltrating T cells (CD3), B cells (CD19) and macrophages (CD68) in gut biopsies from healthy controls or new-onset RA patients from stained gut biopsies (n = 10). g Representative H&E-stained gut biopsy microphotographs from a new-onset RA patient; arrows: signs of inflammation. Size bar: 20 µm. h Intestinal permeability assessed by lactulose/mannitol recovery ratio in urine of healthy controls (n = 10), and new-onset (n = 10) and established (n = 5) RA patients after 24 h. Data are expressed as the mean ± s.d. Statistical difference was determined by one-way ANOVA. *p < 0.05; ***p < 0.001. Source data are provided as a Source Data file.
Fig. 2. Zonulin upregulation, decrease in tight…
Fig. 2. Zonulin upregulation, decrease in tight junction proteins and barrier dysfunction before onset of arthritis.
a Time course of serum zonulin levels in mice induced for collagen-induced arthritis (CIA) (n = 5). bd Time course of intestinal permeability assessed by b the serum FITC-Dextran, c lactulose/mannitol urine recovery ratio and d urine sucralose recovery ratio in mice induced for CIA (n = 5). e, f Quantification of ZO-1 (red) expression determined in histological sections of e the ileum (n = 6) and f the colon (n = 6) presented as floating bars (min to max) with line at median. g Representative microphotographs of ZO-1 (red) expression in small intestine. Size bar: 50 µm. h, i Quantification of occludin expression (red) in histological sections of h the ileum (n = 6) and i the colon (n = 6) presented as floating bars (min to max) with line at median with j representative microphotographs of occludin (red) expression in small intestine. Size bar: 50 µm. k, l Time course of the length measurement of k the small intestine and l the colon in mice induced for CIA. mo Time course of m duodenal, n jejunal and o ileal crypt elongation with p representative H&E-stained sections from the ileum. Size bar: 200 µm. q Time course of goblet cell numbers in the colon and r representative PAS-stained sections from the colon. Size bar: 200 µm. Data are derived from two (bj), three (a), or four independent (kr) experiments and expressed as the mean ± s.d. Statistical difference was determined by one-way ANOVA. *p < 0.05; **p < 0.01; ***p < 0.001. Dashed red line: time point of clinical disease onset. Source data are provided as a Source Data file.
Fig. 3. Dysbiotic microbiota from arthritis mice…
Fig. 3. Dysbiotic microbiota from arthritis mice transfers leaky barrier and mucosal inflammation to GF mice.
a Intestinal permeability assessed by the lactulose/mannitol urine recovery ratio in mice using different collagen-induced arthritis (CIA) immunization protocols namely collagen type II + CFA (CII + CFA), denaturated collagen type II + CFA (deCII + CFA) or collagen type II only (CII) (n = 5). b Arthritis scores in mice induced for CIA using different immunization protocols (n = 5). c Length measurement of the intestine at 20 days post immunization (dpi) in CIA mice (n = 4) presented as floating bars (min to max) with line at median. d Intestinal permeability assessed by the lactulose/mannitol urine recovery ratio in germ-free (GF) mice reconstituted with the donor microbiota from mice immunized with different CIA immunization protocols at 20 dpi (n = 4). e Time course of serum zonulin levels and western blot analysis of ZO-1 expression in Caco-2 cells after 24 h stimulation with intestinal fecal supernatants (FSN) (n = 3). f Transepithelial electrical resistance (TEER) changes in mice induced for CIA treated with the zonulin agonist (AT-1002) (n = 6). g Intestinal organoids from wild-type mice treated with fecal supernatant (FSN) from mice with collagen-induced arthritis at 35 dpi, FSN and zonulin antagonist, or PBS (control). Upon addition of Lucifer yellow (457 Da), confocal fluorescent images were captured every 5 min for 120 min. At 105 min, EGTA was added as a positive control for the ability of the organoids to take up Lucifer yellow. Fluorescence was determined in the organoid lumen and outside of the organoid. Relative intensity values were calculated (fluorescence inside/outside) and are shown for each time point. Each point represents mean values, measured in ten organoids derived from two independent experiments (n = 10). Representative images at time point 55 min are shown. Upper panel: brightfield; lower panel: Lucifer yellow (green), size bar: 50 μm. Data are derived from two (dg) or three (ac) independent experiments and expressed as the mean ± s.d. Statistical difference was determined by one-way (d, e) or two-way ANOVA (a, b, g). *p < 0.05; **p < 0.01; ***p < 0.001. Dashed red line: time point of clinical disease onset. Source data are provided as a Source Data file.
Fig. 4. Targeting intestinal barrier dysfunction before…
Fig. 4. Targeting intestinal barrier dysfunction before arthritis onset attenuates development of arthritis.
a, b Time course of a intestinal barrier permeability (n = 5) and b total arthritis scores in collagen-induced arthritis (CIA) or nontreated healthy (NT) mice with or without butyrate treatment (n = 5). c, d Effect of butyrate treatment of mice induced for CIA on their c serum zonulin levels (n = 5) and d intestinal length measurements (n = 5). e, f Time course of e intestinal barrier permeability (n = 5) and f total arthritis scores in CIA or nontreated healthy mice with or without CB1R treatment (n = 5). g, h Effect of CB1R agonist treatment of mice induced for CIA on g serum zonulin levels (n = 5) and h intestine length (n = 5). Data are derived from two (eh) or four (ad) independent experiments and expressed as the mean ± s.d. Statistical difference was determined by Students’ t test, two-tailed (c, d, g) or two-way ANOVA (a, b, e, f). *p < 0.05; **p < 0.01; ***p < 0.001. Dashed red line: time point of clinical disease onset. Source data are provided as a Source Data file.
Fig. 5. Short-term zonulin antagonist treatment improves…
Fig. 5. Short-term zonulin antagonist treatment improves bone homeostasis.
ac Time course of a intestinal barrier permeability (n = 5), b total arthritis scores (n = 5) and c serum zonulin levels in collagen-induced arthritis (CIA) mice treated with Larazotide (zonulin antagonist) between 17 and 27 days post immunization (dpi) (n = 5, box and whiskers showing 1–99 percentile). d Time course of arthritis scores in mice induced for CIA treated with the zonulin agonist (AT-1002) (n = 5). e, f Histological analysis of tarsal joints after zonulin antagonist treatment showing e inflamed area and respective H&E-stained sections (size bar: 500 µm) (n = 5, showing 1–99 percentile) as well as f osteoclast numbers with respective TRAP-stained sections (size bar: 500 µm) (n = 5, showing 1–99 percentile). g Bone densities in mice treated with zonulin antagonist and respective micro-CT images (n = 5). h Osteoclast numbers per bone parameter in the tibia of CIA mice treated with zonulin antagonist between 17 and 27 days post immunization (n = 5, showing 1–99 percentile). i Schematic overview of cell trafficking experiments using photoconvertible Kaede mice. j Quantification of photoconverted cells from Kaede mice in lymphoid organs and the synovial tissue following short-term zonulin antagonist treatment (n = 8). k Cell populations identified in MLN, spleen and synovial tissue from Kaede CIA mice at day 26 post immunization (n = 6). Data are derived from three independent experiments and expressed as the mean ± s.d. Statistical difference was determined by Students’ t test, two-tailed (g) or two-way ANOVA (af, h, i). *p < 0.05; **p < 0.01; ***p < 0.001. Dashed red line: time point of clinical disease onset. Source data are provided as a Source Data file.

References

    1. Endesfelder D, Engel M, Zu Castell W. Gut immunity and type 1 diabetes: a melange of microbes, diet, and host interactions? Curr. Diabetes Rep. 2016;16:60. doi: 10.1007/s11892-016-0753-3.
    1. Berer K, et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature. 2011;479:538–541. doi: 10.1038/nature10554.
    1. Zhang H, Liao X, Sparks JB, Luo XM. Dynamics of gut microbiota in autoimmune lupus. Appl. Environ. Microbiol. 2014;80:7551–7560. doi: 10.1128/AEM.02676-14.
    1. Jubair WK, et al. Modulation of inflammatory arthritis in mice by gut microbiota through mucosal inflammation and autoantibody generation. Arthritis Rheumatol. 2018;70:1220–1233. doi: 10.1002/art.40490.
    1. Liu X, et al. Role of the gut microbiome in modulating arthritis progression in mice. Sci. Rep. 2016;6:30594. doi: 10.1038/srep30594.
    1. Bernard NJ. Rheumatoid arthritis: Prevotella copri associated with new-onset untreated RA. Nat. Rev. Rheumatol. 2014;10:2. doi: 10.1038/nrrheum.2013.187.
    1. Scher JU, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife. 2013;2:e01202. doi: 10.7554/eLife.01202.
    1. Ogrendik M. Efficacy of roxithromycin in adult patients with rheumatoid arthritis who had not received disease-modifying antirheumatic drugs: a 3-month, randomized, double-blind, placebo-controlled trial. Clin. Therapeut. 2009;31:1754–1764. doi: 10.1016/j.clinthera.2009.08.014.
    1. Ogrendik M, Karagoz N. Treatment of rheumatoid arthritis with roxithromycin: a randomized trial. Postgrad. Med. 2011;123:220–227. doi: 10.3810/pgm.2011.09.2478.
    1. Stone M, Fortin PR, Pacheco-Tena C, Inman RD. Should tetracycline treatment be used more extensively for rheumatoid arthritis? Metaanalysis demonstrates clinical benefit with reduction in disease activity. J. Rheumatol. 2003;30:2112–2122.
    1. Zhang X, et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat. Med. 2015;21:895–905. doi: 10.1038/nm.3914.
    1. Skoldstam L, Hagfors L, Johansson G. An experimental study of a Mediterranean diet intervention for patients with rheumatoid arthritis. Ann. Rheum. Dis. 2003;62:208–214. doi: 10.1136/ard.62.3.208.
    1. Hager J, et al. The role of dietary fiber in rheumatoid arthritis patients: a feasibility study. Nutrients. 2019;11:2392. doi: 10.3390/nu11102392.
    1. Campisi L, et al. Apoptosis in response to microbial infection induces autoreactive TH17 cells. Nat. Immunol. 2016;17:1084–1092. doi: 10.1038/ni.3512.
    1. Deane KD, El-Gabalawy H. Pathogenesis and prevention of rheumatic disease: focus on preclinical RA and SLE. Nat. Rev. Rheumatol. 2014;10:212–228. doi: 10.1038/nrrheum.2014.6.
    1. El Asmar R, et al. Host-dependent zonulin secretion causes the impairment of the small intestine barrier function after bacterial exposure. Gastroenterology. 2002;123:1607–1615. doi: 10.1053/gast.2002.36578.
    1. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 2011;365:2205–2219. doi: 10.1056/NEJMra1004965.
    1. Neurath MF, Finotto S, Glimcher LH. The role of Th1/Th2 polarization in mucosal immunity. Nat. Med. 2002;8:567–573. doi: 10.1038/nm0602-567.
    1. Chen Z, et al. Th2 and eosinophil responses suppress inflammatory arthritis. Nat. Commun. 2016;7:11596. doi: 10.1038/ncomms11596.
    1. Zaiss MM, et al. Regulatory T cells protect from local and systemic bone destruction in arthritis. J. Immunol. 2010;184:7238–7246. doi: 10.4049/jimmunol.0903841.
    1. Zaiss MM, et al. Treg cells suppress osteoclast formation: a new link between the immune system and bone. Arthritis Rheum. 2007;56:4104–4112. doi: 10.1002/art.23138.
    1. Lucas S, et al. Short-chain fatty acids regulate systemic bone mass and protect from pathological bone loss. Nat. Commun. 2018;9:55. doi: 10.1038/s41467-017-02490-4.
    1. Zaiss MM, et al. The intestinal microbiota contributes to the ability of helminths to modulate allergic inflammation. Immunity. 2015;43:998–1010. doi: 10.1016/j.immuni.2015.09.012.
    1. Lewis K, et al. Enhanced translocation of bacteria across metabolically stressed epithelia is reduced by butyrate. Inflamm. Bowel Dis. 2010;16:1138–1148. doi: 10.1002/ibd.21177.
    1. Ploger S, et al. Microbial butyrate and its role for barrier function in the gastrointestinal tract. Ann. N. Y. Acad. Sci. 2012;1258:52–59. doi: 10.1111/j.1749-6632.2012.06553.x.
    1. Gopalakrishnan S, et al. Mechanism of action of ZOT-derived peptide AT-1002, a tight junction regulator and absorption enhancer. Int. J. Pharm. 2009;365:121–130. doi: 10.1016/j.ijpharm.2008.08.047.
    1. Zoppi S, et al. Endogenous cannabinoid system regulates intestinal barrier function in vivo through cannabinoid type 1 receptor activation. Am. J. Physiol. Gastrointest. Liver Physiol. 2012;302:G565–571. doi: 10.1152/ajpgi.00158.2011.
    1. Rutkowska M, Fereniec-Goltbiewska L. ACEA (arachidonyl-2-chloroethylamide), the selective cannabinoid CB1 receptor agonist, protects against aspirin-induced gastric ulceration. Die Pharmazie. 2006;61:341–342.
    1. Sturgeon C, Fasano A. Zonulin, a regulator of epithelial and endothelial barrier functions, and its involvement in chronic inflammatory diseases. Tissue Barriers. 2016;4:e1251384. doi: 10.1080/21688370.2016.1251384.
    1. Gopalakrishnan S, et al. Larazotide acetate regulates epithelial tight junctions in vitro and in vivo. Peptides. 2012;35:86–94. doi: 10.1016/j.peptides.2012.02.015.
    1. Fasano A. Zonulin and its regulation of intestinal barrier function: the biological door to inflammation, autoimmunity, and cancer. Physiol. Rev. 2011;91:151–175. doi: 10.1152/physrev.00003.2008.
    1. Tomura M, et al. Activated regulatory T cells are the major T cell type emigrating from the skin during a cutaneous immune response in mice. J. Clin. Investig. 2010;120:883–893. doi: 10.1172/JCI40926.
    1. Ciccia F, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann. Rheum. Dis. 2017;76:1123–1132. doi: 10.1136/annrheumdis-2016-210000.
    1. Malickova K, et al. Fecal zonulin is elevated in Crohn’s disease and in cigarette smokers. Practical Lab. Med. 2017;9:39–44. doi: 10.1016/j.plabm.2017.09.001.
    1. Watts T, et al. Role of the intestinal tight junction modulator zonulin in the pathogenesis of type I diabetes in BB diabetic-prone rats. Proc. Natl. Acad. Sci. USA. 2005;102:2916–2921. doi: 10.1073/pnas.0500178102.
    1. Saitou M, et al. Occludin-deficient embryonic stem cells can differentiate into polarized epithelial cells bearing tight junctions. J. Cell Biol. 1998;141:397–408. doi: 10.1083/jcb.141.2.397.
    1. Shen L, Weber CR, Raleigh DR, Yu D, Turner JR. Tight junction pore and leak pathways: a dynamic duo. Annu. Rev. Physiol. 2011;73:283–309. doi: 10.1146/annurev-physiol-012110-142150.
    1. Van Itallie CM, Fanning AS, Bridges A, Anderson JM. ZO-1 stabilizes the tight junction solute barrier through coupling to the perijunctional cytoskeleton. Mol. Biol. Cell. 2009;20:3930–3940. doi: 10.1091/mbc.e09-04-0320.
    1. Jubair WK, et al. Modulation of inflammatory arthritis in mice by gut microbiota through mucosal inflammation and autoantibody generation. Arthritis Rheum. 2018;70:1220–1233. doi: 10.1002/art.40490.
    1. Tripathi A, et al. Identification of human zonulin, a physiological modulator of tight junctions, as prehaptoglobin-2. Proc. Natl. Acad. Sci. USA. 2009;106:16799–16804. doi: 10.1073/pnas.0906773106.
    1. Wang W, Uzzau S, Goldblum SE, Fasano A. Human zonulin, a potential modulator of intestinal tight junctions. J. Cell Sci. 2000;113(Pt 24):4435–4440.
    1. Di Pierro M, et al. Zonula occludens toxin structure-function analysis. Identification of the fragment biologically active on tight junctions and of the zonulin receptor binding domain. J. Biol. Chem. 2001;276:19160–19165. doi: 10.1074/jbc.M009674200.
    1. McInnes IB, Schett G. Pathogenetic insights from the treatment of rheumatoid arthritis. Lancet. 2017;389:2328–2337. doi: 10.1016/S0140-6736(17)31472-1.
    1. Figueiredo CP, et al. Antimodified protein antibody response pattern influences the risk for disease relapse in patients with rheumatoid arthritis tapering disease modifying antirheumatic drugs. Ann. Rheum. Dis. 2017;76:399–407. doi: 10.1136/annrheumdis-2016-209297.
    1. Gerlag DM, et al. EULAR recommendations for terminology and research in individuals at risk of rheumatoid arthritis: report from the Study Group for Risk Factors for Rheumatoid Arthritis. Ann. Rheum. Dis. 2012;71:638–641. doi: 10.1136/annrheumdis-2011-200990.
    1. Ciccia F, et al. Potential involvement of IL-9 and Th9 cells in the pathogenesis of rheumatoid arthritis. Rheumatology. 2015;54:2264–2272. doi: 10.1093/rheumatology/kev252.
    1. Mansley MK, Korbmacher C, Bertog M. Inhibitors of the proteasome stimulate the epithelial sodium channel (ENaC) through SGK1 and mimic the effect of aldosterone. Pflug. Arch. 2018;470:295–304. doi: 10.1007/s00424-017-2060-5.
    1. Mansley MK, Neuhuber W, Korbmacher C, Bertog M. Norepinephrine stimulates the epithelial Na+ channel in cortical collecting duct cells via alpha2-adrenoceptors. Am. J. Physiol. Ren. Physiol. 2015;308:F450–458. doi: 10.1152/ajprenal.00548.2014.
    1. Bertog M, et al. Basolateral proteinase-activated receptor (PAR-2) induces chloride secretion in M-1 mouse renal cortical collecting duct cells. J. Physiol. 1999;521(Pt 1):3–17. doi: 10.1111/j.1469-7793.1999.00003.x.
    1. Sato T, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265. doi: 10.1038/nature07935.
    1. Bardenbacher M, et al. Permeability analyses and three dimensional imaging of interferon gamma-induced barrier disintegration in intestinal organoids. Stem Cell Res. 2019;35:101383. doi: 10.1016/j.scr.2019.101383.
    1. Dhariwal A, et al. MicrobiomeAnalyst: a web-based tool for comprehensive statistical, visual and meta-analysis of microbiome data. Nucleic Acids Res. 2017;45:W180–W188. doi: 10.1093/nar/gkx295.
    1. Caporaso JG, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc. Natl. Acad. Sci. USA. 2011;108(Suppl 1):4516–4522. doi: 10.1073/pnas.1000080107.
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat. Methods. 2013;10:996–998. doi: 10.1038/nmeth.2604.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–596. doi: 10.1093/nar/gks1219.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 2007;73:5261–5267. doi: 10.1128/AEM.00062-07.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.

Source: PubMed

3
Abonnieren