29
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lipoprotein(a) and Oxidized Phospholipids Promote Valve Calcification in Patients With Aortic Stenosis

      research-article
      , MD a , , MD b , , MD, PhD c , , PhD a , , MD, PhD c , , MD c , c , , MD a , , MD, PhD d , , PhD e , , MD, PhD e , , PhD f , , MD g , , MD, PhD c , , PhD h , , PhD i , , MD, PhD a , , MD, PhD j , , MD, PhD c ,
      Journal of the American College of Cardiology
      Elsevier Biomedical
      aortic valve stenosis, calcific aortic valve disease, lipoprotein(a), oxidized phospholipids, valvular interstitial cells, 18F-NaF, fluorine-18 sodium fluoride, apoB, apolipoprotein B-100, AS, aortic valve stenosis, AVR, aortic valve replacement, CT, computed tomography, Lp(a), lipoprotein(a), OxPL, oxidized phospholipids, PET, positron emission tomography, TBR, tissue-to-background ratio, VIC, valvular interstitial cell

      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

          Lipoprotein(a) [Lp(a)], a major carrier of oxidized phospholipids (OxPL), is associated with an increased incidence of aortic stenosis (AS). However, it remains unclear whether elevated Lp(a) and OxPL drive disease progression and are therefore targets for therapeutic intervention.

          Objectives

          This study investigated whether Lp(a) and OxPL on apolipoprotein B-100 (OxPL-apoB) levels are associated with disease activity, disease progression, and clinical events in AS patients, along with the mechanisms underlying any associations.

          Methods

          This study combined 2 prospective cohorts and measured Lp(a) and OxPL-apoB levels in patients with AS (V max >2.0 m/s), who underwent baseline 18F-sodium fluoride ( 18F-NaF) positron emission tomography (PET), repeat computed tomography calcium scoring, and repeat echocardiography. In vitro studies investigated the effects of Lp(a) and OxPL on valvular interstitial cells.

          Results

          Overall, 145 patients were studied (68% men; age 70.3 ± 9.9 years). On baseline positron emission tomography, patients in the top Lp(a) tertile had increased valve calcification activity compared with those in lower tertiles (n = 79; 18F-NaF tissue-to-background ratio of the most diseased segment: 2.16 vs. 1.97; p = 0.043). During follow-up, patients in the top Lp(a) tertile had increased progression of valvular computed tomography calcium score (n = 51; 309 AU/year [interquartile range: 142 to 483 AU/year] vs. 93 AU/year [interquartile range: 56 to 296 AU/year; p = 0.015), faster hemodynamic progression on echocardiography (n = 129; 0.23 ± 0.20 m/s/year vs. 0.14 ± 0.20 m/s/year] p = 0.019), and increased risk for aortic valve replacement and death (n = 145; hazard ratio: 1.87; 95% CI: 1.13 to 3.08; p = 0.014), compared with lower tertiles. Similar results were noted with OxPL-apoB. In vitro, Lp(a) induced osteogenic differentiation of valvular interstitial cells, mediated by OxPL and inhibited with the E06 monoclonal antibody against OxPL.

          Conclusions

          In patients with AS, Lp(a) and OxPL drive valve calcification and disease progression. These findings suggest lowering Lp(a) or inactivating OxPL may slow AS progression and provide a rationale for clinical trials to test this hypothesis.

          Central Illustration

          Related collections

          Most cited references16

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

          Genetic associations with valvular calcification and aortic stenosis.

          Limited information is available regarding genetic contributions to valvular calcification, which is an important precursor of clinical valve disease. We determined genomewide associations with the presence of aortic-valve calcification (among 6942 participants) and mitral annular calcification (among 3795 participants), as detected by computed tomographic (CT) scanning; the study population for this analysis included persons of white European ancestry from three cohorts participating in the Cohorts for Heart and Aging Research in Genomic Epidemiology consortium (discovery population). Findings were replicated in independent cohorts of persons with either CT-detected valvular calcification or clinical aortic stenosis. One SNP in the lipoprotein(a) (LPA) locus (rs10455872) reached genomewide significance for the presence of aortic-valve calcification (odds ratio per allele, 2.05; P=9.0×10(-10)), a finding that was replicated in additional white European, African-American, and Hispanic-American cohorts (P<0.05 for all comparisons). Genetically determined Lp(a) levels, as predicted by LPA genotype, were also associated with aortic-valve calcification, supporting a causal role for Lp(a). In prospective analyses, LPA genotype was associated with incident aortic stenosis (hazard ratio per allele, 1.68; 95% confidence interval [CI], 1.32 to 2.15) and aortic-valve replacement (hazard ratio, 1.54; 95% CI, 1.05 to 2.27) in a large Swedish cohort; the association with incident aortic stenosis was also replicated in an independent Danish cohort. Two SNPs (rs17659543 and rs13415097) near the proinflammatory gene IL1F9 achieved genomewide significance for mitral annular calcification (P=1.5×10(-8) and P=1.8×10(-8), respectively), but the findings were not replicated consistently. Genetic variation in the LPA locus, mediated by Lp(a) levels, is associated with aortic-valve calcification across multiple ethnic groups and with incident clinical aortic stenosis. (Funded by the National Heart, Lung, and Blood Institute and others.).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Antisense oligonucleotides targeting apolipoprotein(a) in people with raised lipoprotein(a): two randomised, double-blind, placebo-controlled, dose-ranging trials.

            Elevated lipoprotein(a) (Lp[a]) is a highly prevalent (around 20% of people) genetic risk factor for cardiovascular disease and calcific aortic valve stenosis, but no approved specific therapy exists to substantially lower Lp(a) concentrations. We aimed to assess the efficacy, safety, and tolerability of two unique antisense oligonucleotides designed to lower Lp(a) concentrations.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A randomized trial of intensive lipid-lowering therapy in calcific aortic stenosis.

              Calcific aortic stenosis has many characteristics in common with atherosclerosis, including hypercholesterolemia. We hypothesized that intensive lipid-lowering therapy would halt the progression of calcific aortic stenosis or induce its regression. In this double-blind, placebo-controlled trial, patients with calcific aortic stenosis were randomly assigned to receive either 80 mg of atorvastatin daily or a matched placebo. Aortic-valve stenosis and calcification were assessed with the use of Doppler echocardiography and helical computed tomography, respectively. The primary end points were change in aortic-jet velocity and aortic-valve calcium score. Seventy-seven patients were assigned to atorvastatin and 78 to placebo, with a median follow-up of 25 months (range, 7 to 36). Serum low-density lipoprotein cholesterol concentrations remained at 130+/-30 mg per deciliter in the placebo group and fell to 63+/-23 mg per deciliter in the atorvastatin group (P<0.001). Increases in aortic-jet velocity were 0.199+/-0.210 m per second per year in the atorvastatin group and 0.203+/-0.208 m per second per year in the placebo group (P=0.95; adjusted mean difference, 0.002; 95 percent confidence interval, -0.066 to 0.070 m per second per year). Progression in valvular calcification was 22.3+/-21.0 percent per year in the atorvastatin group, and 21.7+/-19.8 percent per year in the placebo group (P=0.93; ratio of post-treatment aortic-valve calcium score, 0.998; 95 percent confidence interval, 0.947 to 1.050). Intensive lipid-lowering therapy does not halt the progression of calcific aortic stenosis or induce its regression. This study cannot exclude a small reduction in the rate of disease progression or a significant reduction in major clinical end points. Long-term, large-scale, randomized, controlled trials are needed to establish the role of statin therapy in patients with calcific aortic stenosis. Copyright 2005 Massachusetts Medical Society.
                Bookmark

                Author and article information

                Contributors
                Journal
                J Am Coll Cardiol
                J. Am. Coll. Cardiol
                Journal of the American College of Cardiology
                Elsevier Biomedical
                0735-1097
                1558-3597
                07 May 2019
                07 May 2019
                : 73
                : 17
                : 2150-2162
                Affiliations
                [a ]Department of Vascular Medicine, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands
                [b ]Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California, San Diego, La Jolla, California
                [c ]British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
                [d ]Department of Cardiothoracic Surgery, UMC Utrecht, Utrecht, the Netherlands
                [e ]Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
                [f ]Centre de recherche de l’Institut Universitaire de cardiologie et de pneumologie de Québec–Université Laval, Québec City, Québec, Canada
                [g ]Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, California
                [h ]Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
                [i ]Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
                [j ]Department of Cardiology, Academic Medical Center, Amsterdam UMC, Amsterdam, the Netherlands
                Author notes
                [] Address for correspondence: Dr. Marc R. Dweck, British Heart Foundation Centre for Cardiovascular Sciences, University of Edinburgh, The Chancellor’s Building, Little France Crescent, Edinburgh. marc.dweck@ 123456ed.ac.uk @ 123456MarcDweck @ 123456Lpa_Doc
                Article
                S0735-1097(19)33881-1
                10.1016/j.jacc.2019.01.070
                6494952
                31047003
                5b743f68-1566-4887-af29-f8c5ef65abe2
                © 2019 The Authors

                This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

                History
                : 17 September 2018
                : 28 January 2019
                : 28 January 2019
                Categories
                Article

                Cardiovascular Medicine
                aortic valve stenosis,calcific aortic valve disease,lipoprotein(a),oxidized phospholipids,valvular interstitial cells,18f-naf, fluorine-18 sodium fluoride,apob, apolipoprotein b-100,as, aortic valve stenosis,avr, aortic valve replacement,ct, computed tomography,lp(a), lipoprotein(a),oxpl, oxidized phospholipids,pet, positron emission tomography,tbr, tissue-to-background ratio,vic, valvular interstitial cell

                Comments

                Comment on this article