27
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Inflammasome-Independent NALP3 Contributes to High-Salt Induced Endothelial Dysfunction

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Backgrounds and Aims: Na + is an important nutrient and its intake, mainly from salt (NaCl), is essential for normal physiological function. However, high salt intake may lead to vascular injury, independent of a rise in blood pressure (BP). Canonical NALP3 inflammasome activation is a caspase-1 medicated process, resulting in the secretion of IL-18 and IL-1β which lead to endothelial dysfunction. However, some researches uncovered a direct and inflammasome-independent role of NALP3 in renal injury. Thus, this study was designed to investigate the possible mechanisms of NALP3 in high salt induced endothelial dysfunction.

          Methods and Results: Changes in endothelial function were measured by investigating mice (C57BL/6J, NALP3 -/- and wild-type, WT) fed with normal salt diet (NSD) or high salt diet (HSD) for 12W, and thoracic aortic rings from C57BL/6J mice cultured in high-salt medium. Changes of tube formation ability, intracellular reactive oxygen species (ROS), and NALP3 inflammasome expression were detected using mouse aortic endothelial cells (MAECs) cultured in high-salt medium. Consumption of HSD for 12W did not affect BP or body weight in C57BL/6J mice. Endothelium-dependent relaxation (EDR) decreased significantly in C57BL/6J mice fed with HSD for 12W, and in isolated thoracic aortic rings cultured in high-salt medium for 24 h. Results from the aortic ring assay also revealed that the angiogenic function of thoracic aortas was impaired by either consumption of HSD or exposure to high-salt medium. NALP3 -/- mice fed with HSD showed a relatively mild decrease in EDR function when compared with WT mice. Tube length of thoracic aortic rings from NALP3 -/- mice was longer than those from WT mice after receiving high-salt treatment. Inhibiting NALP3 with a NALP3 antagonist, small interfering (si) RNA experiments using si-NALP3, and decomposing ROS significantly improved tube formation ability in MAECs under high salt medium. NALP3 expression was increased in MAECs cultured with high salt treatment and inhibiting NALP3 reversed the down-regulation of p-eNOS induced by high salt in MAECs.

          Conclusion: High salt intake impairs endothelial function, which is at least in part mediated by increasing NALP3 expression.

          Related collections

          Most cited references34

          • Record: found
          • Abstract: found
          • Article: not found

          The NLRP3 inflammasome: a sensor for metabolic danger?

          Interleukin-1beta (IL-1beta), reactive oxygen species (ROS), and thioredoxin-interacting protein (TXNIP) are all implicated in the pathogenesis of type 2 diabetes mellitus (T2DM). Here we review mechanisms directing IL-1beta production and its pathogenic role in islet dysfunction during chronic hyperglycemia. In doing so, we integrate previously disparate disease-driving mechanisms for IL-1beta, ROS, and TXNIP in T2DM into one unifying model in which the NLRP3 inflammasome plays a central role. The NLRP3 inflammasome also drives IL-1beta maturation and secretion in another disease of metabolic dysregulation, gout. Thus, we propose that the NLRP3 inflammasome contributes to the pathogenesis of T2DM and gout by functioning as a sensor for metabolic stress.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Use of the mouse aortic ring assay to study angiogenesis.

            Here we provide a protocol for quantitative three-dimensional ex vivo mouse aortic ring angiogenesis assays, in which developing microvessels undergo many key features of angiogenesis over a timescale similar to that observed in vivo. The aortic ring assay allows analysis of cellular proliferation, migration, tube formation, microvessel branching, perivascular recruitment and remodeling-all without the need for cellular dissociation-thus providing a more complete picture of angiogenic processes compared with traditional cell-based assays. Our protocol can be applied to aortic rings from embryonic stage E18 through to adulthood and can incorporate genetic manipulation, treatment with growth factors, drugs or siRNA. This robust assay allows assessment of the salient steps in angiogenesis and quantification of the developing microvessels, and it can be used to identify new modulators of angiogenesis. The assay takes 6-14 d to complete, depending on the age of the mice, treatments applied and whether immunostaining is performed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Linking endothelial dysfunction with endothelial cell activation.

              K K Liao (2013)
              The thin layer of cells that lines the interior of blood vessels, known as the endothelium, plays a complex role in vascular biology. The endothelium mediates blood vessel tone, hemostasis, neutrophil recruitment, hormone trafficking, and fluid filtration. Endothelial dysfunction, as defined by a lack of NO, has been linked to a variety of disease states, including atherosclerosis, diabetes mellitus, coronary artery disease, hypertension, and hypercholesterolemia. Indeed, restoration of endothelial function is one of the earliest recognizable benefits of statin therapy. In 1995, James Liao and colleagues published a study in the JCI demonstrating that NO is a vascular protective factor that limits endothelial activation and prevents leukocyte adhesion to the vessel wall.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                22 August 2018
                2018
                : 9
                : 968
                Affiliations
                [1] 1Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University , Shanghai, China
                [2] 2Department of Health Toxicology, Faculty of Naval University, Second Military Medical University , Shanghai, China
                Author notes

                Edited by: Martin C. Michel, Johannes Gutenberg-Universität Mainz, Germany

                Reviewed by: Brett M. Mitchell, Texas A&M Health Science Center, United States; Ming Xu, China Pharmaceutical University, China

                *Correspondence: Dong-Jie Li, djli@ 123456tongji.edu.cn Fu-Ming Shen, fumingshen@ 123456tongji.edu.cn

                This article was submitted to Cardiovascular and Smooth Muscle Pharmacology, a section of the journal Frontiers in Pharmacology

                Article
                10.3389/fphar.2018.00968
                6113916
                4b0009d7-923a-414c-a198-d1e2260ce777
                Copyright © 2018 Fu, Chen, Lu, Jiang, Wang, Li and Shen.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 02 November 2017
                : 06 August 2018
                Page count
                Figures: 7, Tables: 0, Equations: 0, References: 46, Pages: 11, Words: 0
                Funding
                Funded by: Natural Science Foundation of Shanghai 10.13039/100007219
                Award ID: No. 16ZR1444500
                Award ID: No. 14DZ1930806
                Categories
                Pharmacology
                Original Research

                Pharmacology & Pharmaceutical medicine
                high salt intake,endothelial-dependent relaxation,angiogenic function,nalp3,reactive oxygen species

                Comments

                Comment on this article