10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Genetic and Pharmacologic Inhibition of the Neutrophil Elastase Inhibits Experimental Atherosclerosis

      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

          Background

          To investigate whether neutrophil elastase ( NE) plays a causal role in atherosclerosis, and the molecular mechanisms involved.

          Methods and Results

          NE genetic–deficient mice (Apolipoprotein E −/−/ NE −/− mice), bone marrow transplantation, and a specific NE inhibitor ( GW311616A) were employed in this study to establish the causal role of NE in atherosclerosis. Aortic expression of NE mRNA and plasma NE activity was significantly increased in high‐fat diet ( HFD)–fed wild‐type ( WT) (Apolipoprotein E −/−) mice but, as expected, not in NE‐deficient mice. Selective NE knockout markedly reduced HFD‐induced atherosclerosis and significantly increased indicators of atherosclerotic plaque stability. While plasma lipid profiles were not affected by NE deficiency, decreased levels of circulating proinflammatory cytokines and inflammatory monocytes (Ly6C hi/ CD11b +) were observed in NE‐deficient mice fed with an HFD for 12 weeks as compared with WT. Bone marrow reconstitution of WT mice with NE −/− bone marrow cells significantly reduced HFD‐induced atherosclerosis, while bone marrow reconstitution of NE −/− mice with WT bone marrow cells restored the pathological features of atherosclerotic plaques induced by HFD in NE‐deficient mice. In line with these findings, pharmacological inhibition of NE in WT mice through oral administration of NE inhibitor GW311616A also significantly reduced atherosclerosis. Mechanistically, we demonstrated that NE promotes foam cell formation by increasing ATP‐binding cassette transporter ABCA1 protein degradation and inhibiting macrophage cholesterol efflux.

          Conclusions

          We outlined a pathogenic role for NE in foam cell formation and atherosclerosis development. Consequently, inhibition of NE may represent a potential therapeutic approach to treating cardiovascular disease.

          Related collections

          Most cited references61

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

          Neutrophil serine proteases: specific regulators of inflammation.

          Neutrophils are essential for host defence against invading pathogens. They engulf and degrade microorganisms using an array of weapons that include reactive oxygen species, antimicrobial peptides, and proteases such as cathepsin G, neutrophil elastase and proteinase 3. As discussed in this Review, the generation of mice deficient in these proteases has established a role for these enzymes as intracellular microbicidal agents. However, I focus mainly on emerging data indicating that, after release, these proteases also contribute to the extracellular killing of microorganisms, and regulate non-infectious inflammatory processes by activating specific receptors and modulating the levels of cytokines.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation.

            Elevated leukocyte cell numbers (leukocytosis), and monocytes in particular, promote atherosclerosis; however, how they become increased is poorly understood. Mice deficient in the adenosine triphosphate-binding cassette (ABC) transporters ABCA1 and ABCG1, which promote cholesterol efflux from macrophages and suppress atherosclerosis in hypercholesterolemic mice, displayed leukocytosis, a transplantable myeloproliferative disorder, and a dramatic expansion of the stem and progenitor cell population containing Lin(-)Sca-1(+)Kit+ (LSK) in the bone marrow. Transplantation of Abca1(-/-) Abcg1(-/-) bone marrow into apolipoprotein A-1 transgenic mice with elevated levels of high-density lipoprotein (HDL) suppressed the LSK population, reduced leukocytosis, reversed the myeloproliferative disorder, and accelerated atherosclerosis. The findings indicate that ABCA1, ABCG1, and HDL inhibit the proliferation of hematopoietic stem and multipotential progenitor cells and connect expansion of these populations with leukocytosis and accelerated atherosclerosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Scavenger receptors class A-I/II and CD36 are the principal receptors responsible for the uptake of modified low density lipoprotein leading to lipid loading in macrophages.

              Modification of low density lipoprotein (LDL) can result in the avid uptake of these lipoproteins via a family of macrophage transmembrane proteins referred to as scavenger receptors (SRs). The genetic inactivation of either of two SR family members, SR-A or CD36, has been shown previously to reduce oxidized LDL uptake in vitro and atherosclerotic lesions in mice. Several other SRs are reported to bind modified LDL, but their contribution to macrophage lipid accumulation is uncertain. We generated mice lacking both SR-A and CD36 to determine their combined impact on macrophage lipid uptake and to assess the contribution of other SRs to this process. We show that SR-A and CD36 account for 75-90% of degradation of LDL modified by acetylation or oxidation. Cholesteryl ester derived from modified lipoproteins fails to accumulate in macrophages taken from the double null mice, as assessed by histochemistry and gas chromatography-mass spectrometry. These results demonstrate that SR-A and CD36 are responsible for the preponderance of modified LDL uptake in macrophages and that other scavenger receptors do not compensate for their absence.
                Bookmark

                Author and article information

                Contributors
                q.xiao@qmul.ac.uk
                Journal
                J Am Heart Assoc
                J Am Heart Assoc
                10.1002/(ISSN)2047-9980
                JAH3
                ahaoa
                Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease
                John Wiley and Sons Inc. (Hoboken )
                2047-9980
                08 February 2018
                February 2018
                : 7
                : 4 ( doiID: 10.1002/jah3.2018.7.issue-4 )
                : e008187
                Affiliations
                [ 1 ] Centre for Clinical Pharmacology William Harvey Research Institute Barts and The London School of Medicine and Dentistry Queen Mary University of London London United Kingdom
                [ 2 ] Centre for Microvascular Research William Harvey Research Institute Barts and The London School of Medicine and Dentistry Queen Mary University of London London United Kingdom
                [ 3 ] Key Laboratory of Cardiovascular Diseases The Second Affiliated Hospital School of Basic Medical Sciences Guangzhou Medical University Guangzhou Guangdong China
                [ 4 ] Key Laboratory of Protein Modification and Degradation School of Basic Medical Sciences Guangzhou Medical University Guangzhou Guangdong China
                [ 5 ] Department of Cardiothoracic Surgery Yongchuan Hospital of Chongqing Medical University Chongqing China
                [ 6 ] Department of Cardiology the First Affiliated Hospital School of Medicine Zhejiang University Hangzhou Zhejiang China
                [ 7 ] Department of Cardiovascular Sciences University of Leicester Leicester United Kingdom
                Author notes
                [*] [* ] Correspondence to: Qingzhong Xiao, MD, PhD, Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Heart Centre, Charterhouse Square, London EC1M 6BQ, United Kingdom. E‐mail: q.xiao@ 123456qmul.ac.uk
                [†]

                Dr Wen and Dr An contributed equally to this work and are joint first authors.

                Article
                JAH32920
                10.1161/JAHA.117.008187
                5850208
                29437605
                8b2c2728-8601-47cc-a722-51e28007f099
                © 2018 The Authors. Published on behalf of the American Heart Association, Inc., by Wiley.

                This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 01 December 2017
                : 21 December 2017
                Page count
                Figures: 14, Tables: 1, Pages: 29, Words: 13634
                Funding
                Funded by: British Heart Foundation
                Award ID: FS/09/044/28007
                Award ID: PG/11/40/28891
                Award ID: PG/13/45/30326
                Award ID: PG/15/11/31279
                Award ID: PG/15/86/31723
                Award ID: PG/16/1/31892
                Funded by: Wellcome Trust
                Award ID: 098291/Z/12/Z
                Categories
                Original Research
                Original Research
                Vascular Medicine
                Custom metadata
                2.0
                jah32920
                February 2018
                Converter:WILEY_ML3GV2_TO_NLMPMC version:version=5.3.2.2 mode:remove_FC converted:20.02.2018

                Cardiovascular Medicine
                atherosclerosis,atp‐binding cassette transporter abca1,bone marrow transplant,foam cells,gw311616a,inflammation,macrophage,neutrophil elastase,neutrophil elastase inhibitor,protease,peripheral vascular disease,vascular disease

                Comments

                Comment on this article