7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Nox2 in regulatory T cells promotes angiotensin II–induced cardiovascular remodeling

      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

          The superoxide-generating enzyme Nox2 contributes to hypertension and cardiovascular remodeling triggered by activation of the renin-angiotensin system. Multiple Nox2-expressing cells are implicated in angiotensin II–induced (Ang II–induced) pathophysiology, but the importance of Nox2 in leukocyte subsets is poorly understood. Here, we investigated the role of Nox2 in T cells, particularly Tregs. Mice globally deficient in Nox2 displayed increased numbers of Tregs in the heart at baseline, whereas Ang II–induced effector T cell (Teff) infiltration was inhibited. To investigate the role of Treg Nox2, we generated a mouse line with CD4-targeted Nox2 deficiency (Nox2 fl/flCD4Cre +). These animals showed inhibition of Ang II–induced hypertension and cardiac remodeling related to increased tissue-resident Tregs and reduction in infiltrating Teffs, including Th17 cells. The protection in Nox2 fl/flCD4Cre + mice was reversed by anti-CD25 antibody depletion of Tregs. Mechanistically, Nox2 –/y Tregs showed higher in vitro suppression of Teff proliferation than WT Tregs, increased nuclear levels of FoxP3 and NF-κB, and enhanced transcription of CD25, CD39, and CD73. Adoptive transfer of Tregs confirmed that Nox2-deficient cells had greater inhibitory effects on Ang II–induced heart remodeling than WT cells. These results identify a previously unrecognized role of Nox2 in modulating suppression of Tregs, which acts to enhance hypertension and cardiac remodeling.

          Related collections

          Most cited references30

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

          Biochemistry, physiology, and pathophysiology of NADPH oxidases in the cardiovascular system.

          The NADPH oxidase (Nox) enzymes are critical mediators of cardiovascular physiology and pathophysiology. These proteins are expressed in virtually all cardiovascular cells, and regulate such diverse functions as differentiation, proliferation, apoptosis, senescence, inflammatory responses and oxygen sensing. They target a number of important signaling molecules, including kinases, phosphatases, transcription factors, ion channels, and proteins that regulate the cytoskeleton. Nox enzymes have been implicated in many different cardiovascular pathologies: atherosclerosis, hypertension, cardiac hypertrophy and remodeling, angiogenesis and collateral formation, stroke, and heart failure. In this review, we discuss in detail the biochemistry of Nox enzymes expressed in the cardiovascular system (Nox1, 2, 4, and 5), their roles in cardiovascular cell biology, and their contributions to disease development.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Interleukin 17 promotes angiotensin II-induced hypertension and vascular dysfunction.

            We have shown previously that T cells are required for the full development of angiotensin II-induced hypertension. However, the specific subsets of T cells that are important in this process are unknown. T helper 17 cells represent a novel subset that produces the proinflammatory cytokine interleukin 17 (IL-17). We found that angiotensin II infusion increased IL-17 production from T cells and IL-17 protein in the aortic media. To determine the effect of IL-17 on blood pressure and vascular function, we studied IL-17(-/-) mice. The initial hypertensive response to angiotensin II infusion was similar in IL-17(-/-) and C57BL/6J mice. However, hypertension was not sustained in IL-17(-/-) mice, reaching levels 30-mm Hg lower than in wild-type mice by 4 weeks of angiotensin II infusion. Vessels from IL-17(-/-) mice displayed preserved vascular function, decreased superoxide production, and reduced T-cell infiltration in response to angiotensin II. Gene array analysis of cultured human aortic smooth muscle cells revealed that IL-17, in conjunction with tumor necrosis factor-alpha, modulated expression of >30 genes, including a number of inflammatory cytokines/chemokines. Examination of IL-17 in diabetic humans showed that serum levels of this cytokine were significantly increased in those with hypertension compared with normotensive subjects. We conclude that IL-17 is critical for the maintenance of angiotensin II-induced hypertension and vascular dysfunction and might be a therapeutic target for this widespread disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              T regulatory lymphocytes prevent angiotensin II-induced hypertension and vascular injury.

              Angiotensin (Ang) II induces hypertension by mechanisms mediated in part by adaptive immunity and T effector lymphocytes. T regulatory lymphocytes (Tregs) suppress T effector lymphocytes. We questioned whether Treg adoptive transfer would blunt Ang II-induced hypertension and vascular injury. Ten- to 12-week-old male C57BL/6 mice were injected IV with 3 ×10(5) Treg (CD4(+)CD25(+)) or T effector (CD4(+)CD25(-)) cells, 3 times at 2-week intervals, and then infused or not with Ang II (1 μg/kg per minute, SC) for 14 days. Ang II increased systolic blood pressure by 43 mm Hg (P<0.05), NADPH oxidase activity 1.5-fold in aorta and 1.8-fold in the heart (P<0.05), impaired acetylcholine vasodilatory responses by 70% compared with control (P<0.05), and increased vascular stiffness (P<0.001), mesenteric artery vascular cell adhesion molecule expression (2-fold; P<0.05), and aortic macrophage and T-cell infiltration (P<0.001). All of the above were prevented by Treg but not T effector adoptive transfer. Ang II caused a 43% decrease in Foxp3(+) cells in the renal cortex, whereas Treg adoptive transfer increased Foxp3(+) cells 2-fold compared with control. Thus, Tregs suppress Ang II-mediated vascular injury in part through anti-inflammatory actions. Immune mechanisms modulate Ang II-induced blood pressure elevation, vascular oxidative stress, inflammation, and endothelial dysfunction.
                Bookmark

                Author and article information

                Contributors
                Journal
                J Clin Invest
                J. Clin. Invest
                J Clin Invest
                The Journal of Clinical Investigation
                American Society for Clinical Investigation
                0021-9738
                1558-8238
                11 June 2018
                2 July 2018
                2 July 2018
                : 128
                : 7
                : 3088-3101
                Affiliations
                [1 ]King’s College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom.
                [2 ]King’s College London, Medical Research Council Centre for Transplantation, School of Immunology and Microbial Sciences, London, United Kingdom.
                Author notes
                Address correspondence to: Ajay M. Shah, Department of Cardiology, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, United Kingdom. Phone: 0044.207848.5189; Email: ajay.shah@ 123456kcl.ac.uk . Or to: Giovanna Lombardi, Immunoregulation Laboratory, MRC Centre for Transplantation, King’s College London, 5th Floor Tower Wing, Guy’s Hospital, London SE1 9RT, United Kingdom. Phone: 0044.207188.7674; Email: giovanna.lombardi@ 123456kcl.ac.uk .

                Authorship note: AE, SCT, and HMD contributed equally to this work. GL and AMS are co–senior authors.

                Author information
                http://orcid.org/0000-0002-2114-5810
                http://orcid.org/0000-0003-2722-2458
                http://orcid.org/0000-0002-8706-0019
                http://orcid.org/0000-0003-1980-1230
                http://orcid.org/0000-0002-7083-2430
                http://orcid.org/0000-0003-2801-1900
                http://orcid.org/0000-0002-6547-0631
                Article
                97490
                10.1172/JCI97490
                6025997
                29688896
                94fe660e-b896-48f0-afcd-ff8a194633c1
                Copyright © 2018 Emmerson et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 14 September 2017
                : 17 April 2018
                Funding
                Funded by: British Heart Foundation, https://doi.org/10.13039/501100000274;
                Award ID: RG/13/11/30384
                Award ID: RE/13/2/30182
                Categories
                Research Article

                cardiology,immunology,cardiovascular disease,hypertension,t cells

                Comments

                Comment on this article