Clin Res Cardiol (2023). https://doi.org/10.1007/s00392-023-02180-w

Endothelial Dysfunction in Experimental Celiac Disease is Mediated by Gut-Derived Vascular Inflammation and Oxidative Stress
K. Keppeler1, S. Lange1, J. Helmstädter1, L. Strohm1, I. Kuntic1, M. Kuntic1, H. Ubbens1, M. T. Bayo Jimenez1, D. Mihalikova1, S. Finger2, M. Oelze1, S. Karbach3, D. Schuppan4, P. Wenzel3, P. S. Wild3, D. Leistner5, T. Münzel3, A. Daiber1, S. Steven5
1Labor für Molekulare Kardiologie, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz; 2Centrum für Thrombose und Hämostase, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz; 3Zentrum für Kardiologie, Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz; 4Institut für Translationale Immunologie (TIM), Universitätsmedizin der Johannes Gutenberg-Universität Mainz, Mainz; 5Med. Klinik III - Kardiologie, Angiologie, Universitätsklinikum Frankfurt, Frankfurt am Main;
Objective: Celiac disease (CeD) is an autoimmune enteropathy triggered by dietary gluten, with a prevalence of around 1%. Since many patients show heterogeneous symptoms, it is vastly underdiagnosed. Clinical studies comparing CeD patients to healthy controls have been conducted and hypothesized CeD as a cardiovascular risk factor. Herewith we aimed to explore the cardiovascular effects of CeD in a murine model, with special emphasis on endothelial dysfunction, vascular inflammation, and oxidative stress, known triggers for cardiovascular disease and its mechanistic causes. 
Methodology and Results: NOD.DQ8 mice, with a genetic predisposition for CeD, were raised on a zein-based, gluten-free diet. During the experiment, mice received gavage with a peptic-tryptic digest of gliadin and were switched to a gluten-containing diet (Gliadin group). Gliadin is a protein fraction of gluten and is recognized as an antigen in CeD. A control group (Zein group) continued a gluten-free diet and received accordingly digested zein via gavage.


Figure 1: Treatment scheme, illustrated with the Gliadin Group. 
(CTX-cholera toxin; GFD-gluten-free diet; PT-pepsin-trypsin; TG2-tissue transglutaminase)

Infiltration by intraepithelial T-lymphocytes into the epithelium of the small intestine in the Gliadin group was shown by qIHC. Tail-cuff measurements revealed elevated blood pressure in the Gliadin group. Endothelium-dependent relaxation (ACh) was impaired in the Gliadin group compared to Zein, whereas no change in endothelium-independent relaxation (GTN) was observed, tested by isometric tension recordings. Pro-inflammatory genes (Il6, Infy, Il1b, Tnfa, Nox2), were upregulated in intestinal and aortic tissue of Gliadin mice measured by quantitative rtPCR. Flow cytometry revealed elevated CD11b+ cells in the aortic wall. In cardiac tissue, Gliadin treatment increased the oxidative stress parameters 3-nitrotyrosine (3-NT) and 4-hydroxy-2-nonenal (4-HNE) shown via dot blots. To investigate the inflammatory gut-vascular link plasma proteomics (Olink) were performed and revealed elevated levels of IL-17A, which were confirmed by plasma ELISA. The cardiovascular complications were completely reversible by a gliadin-free diet. 
Conclusion: In a CeD mouse model, we demonstrate that a gliadin-containing diet elevates blood pressure and impairs vascular function. CeD elevates the cardiovascular risk by intestinal inflammation that conveys to the cardiovascular system. It leads to vascular inflammation/oxidative stress, and thereby impairs endothelial function. A potential mediator might be IL-17A, whose role in other autoimmunity-based cardiovascular risk factors (e.g., psoriasis) has already been demonstrated.
 

https://dgk.org/kongress_programme/jt2023/aP2100.html