30
views
0
recommends
+1 Recommend
1 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Candidate Gene Resequencing in a Large Bicuspid Aortic Valve-Associated Thoracic Aortic Aneurysm Cohort: SMAD6 as an Important Contributor

      research-article
      1 , 1 , 1 , 2 , 1 , 1 , 1 , 3 , 1 , 1 , 1 , 1 , 2 , 4 , 5 , 5 , 3 , 6 , 6 , 7 , 8 , 4 , 1 , 9 , 9 , 9 , 10 , 10 , 11 , 12 , 13 , 13 , 13 , 10 , 10 , 7 , 2 , 4 , 14 , 1 , 1 , 13 , Mibava Leducq Consortium
      Frontiers in Physiology
      Frontiers Media S.A.
      bicuspid aortic valve, thoracic aortic aneurysm, SMAD6, targeted gene panel, variant burden test

      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

          Bicuspid aortic valve (BAV) is the most common congenital heart defect. Although many BAV patients remain asymptomatic, at least 20% develop thoracic aortic aneurysm (TAA). Historically, BAV-related TAA was considered as a hemodynamic consequence of the valve defect. Multiple lines of evidence currently suggest that genetic determinants contribute to the pathogenesis of both BAV and TAA in affected individuals. Despite high heritability, only very few genes have been linked to BAV or BAV/TAA, such as NOTCH1, SMAD6, and MAT2A. Moreover, they only explain a minority of patients. Other candidate genes have been suggested based on the presence of BAV in knockout mouse models (e.g., GATA5, NOS3) or in syndromic (e.g., TGFBR1/2, TGFB2/3) or non-syndromic (e.g., ACTA2) TAA forms. We hypothesized that rare genetic variants in these genes may be enriched in patients presenting with both BAV and TAA. We performed targeted resequencing of 22 candidate genes using Haloplex target enrichment in a strictly defined BAV/TAA cohort ( n = 441; BAV in addition to an aortic root or ascendens diameter ≥ 4.0 cm in adults, or a Z-score ≥ 3 in children) and in a collection of healthy controls with normal echocardiographic evaluation ( n = 183). After additional burden analysis against the Exome Aggregation Consortium database, the strongest candidate susceptibility gene was SMAD6 ( p = 0.002), with 2.5% ( n = 11) of BAV/TAA patients harboring causal variants, including two nonsense, one in-frame deletion and two frameshift mutations. All six missense mutations were located in the functionally important MH1 and MH2 domains. In conclusion, we report a significant contribution of SMAD6 mutations to the etiology of the BAV/TAA phenotype.

          Related collections

          Most cited references47

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

          Mutations in NOTCH1 cause aortic valve disease.

          Calcification of the aortic valve is the third leading cause of heart disease in adults. The incidence increases with age, and it is often associated with a bicuspid aortic valve present in 1-2% of the population. Despite the frequency, neither the mechanisms of valve calcification nor the developmental origin of a two, rather than three, leaflet aortic valve is known. Here, we show that mutations in the signalling and transcriptional regulator NOTCH1 cause a spectrum of developmental aortic valve anomalies and severe valve calcification in non-syndromic autosomal-dominant human pedigrees. Consistent with the valve calcification phenotype, Notch1 transcripts were most abundant in the developing aortic valve of mice, and Notch1 repressed the activity of Runx2, a central transcriptional regulator of osteoblast cell fate. The hairy-related family of transcriptional repressors (Hrt), which are activated by Notch1 signalling, physically interacted with Runx2 and repressed Runx2 transcriptional activity independent of histone deacetylase activity. These results suggest that NOTCH1 mutations cause an early developmental defect in the aortic valve and a later de-repression of calcium deposition that causes progressive aortic valve disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Bicuspid aortic valve is heritable.

            Previous studies have established familial clustering of bicuspid aortic valve (BAV), presumably indicating genetic inheritance. Our objective was to statistically test whether the segregation pattern of BAV is consistent with genetic inheritance and to obtain an estimate of the size of the genetic effect (heritability). Bicuspid aortic valve occurs in 1% of the population, making it the most common cardiovascular malformation (CVM). Bicuspid aortic valve is frequently an antecedent to aortic valve stenosis or insufficiency and is often associated with other CVMs, including aortic root dilation. The genetic and developmental significance of these findings remains obscure. In 50 probands with BAV, we obtained a three-generation family history and echocardiograms on first-degree relatives. Heritability (h2) of BAV and BAV and/or other CVMs were estimated using maximum-likelihood-based variance decomposition extended to dichotomous traits implemented in the computer package Sequential Oligogenic Linkage Analysis Routines (SOLAR, San Antonio, Texas). A total of 309 probands and relatives participated. Bicuspid aortic valve was identified in 74 individuals (prevalence = 24%). A total of 97 individuals had BAV and/or other CVM (prevalence = 31%), including aortic coarctation, ventricular or atrial septal defect, abnormal mitral valve, aortic root dilation, or hypoplastic left heart syndrome. The heritability (h2) of BAV and BAV and/or other CVMs were 89% and 75%, respectively. The high heritability of BAV suggests that in this study population BAV determination is almost entirely genetic. The heritability of BAV plus other cardiovascular anomalies suggests that valve malformation can be primary to defective valvulogenesis or secondary to other elements of cardiogenesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A role for smad6 in development and homeostasis of the cardiovascular system.

              Smad proteins are intracellular mediators of signalling initiated by Tgf-betasuperfamily ligands (Tgf-betas, activins and bone morphogenetic proteins (Bmps)). Smads 1, 2, 3, 5 and 8 are activated upon phosphorylation by specific type I receptors, and associate with the common partner Smad4 to trigger transcriptional responses. The inhibitory Smads (6 and 7) are transcriptionally induced in cultured cells treated with Tgf-beta superfamily ligands, and downregulate signalling in in vitro assays. Gene disruption in mice has begun to reveal specific developmental and physiological functions of the signal-transducing Smads. Here we explore the role of an inhibitory Smad in vivo by targeted mutation of Madh6 (which encodes the Smad6 protein). Targeted insertion of a LacZ reporter demonstrated that Smad6 expression is largely restricted to the heart and blood vessels, and that Madh6 mutants have multiple cardiovascular abnormalities. Hyperplasia of the cardiac valves and outflow tract septation defects indicate a function for Smad6 in the regulation of endocardial cushion transformation. The role of Smad6 in the homeostasis of the adult cardiovascular system is indicated by the development of aortic ossification and elevated blood pressure in viable mutants. These defects highlight the importance of Smad6 in the tissue-specific modulation of Tgf-beta superfamily signalling pathways in vivo.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Physiol
                Front Physiol
                Front. Physiol.
                Frontiers in Physiology
                Frontiers Media S.A.
                1664-042X
                13 June 2017
                2017
                : 8
                : 400
                Affiliations
                [1] 1Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital Antwerp, Belgium
                [2] 2Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Université de Montreal Montreal, QC, Canada
                [3] 3Department of Cardiac and Thoracic Vascular Surgery, University Hospital Schleswig-Holstein Lübeck, Germany
                [4] 4McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine Baltimore, MD, United States
                [5] 5Department of Clinical pathology, Lviv National Medical University after Danylo Halytsky Lviv, Ukraine
                [6] 6Cardiovascular Research, SickKids University Hospital Toronto, ON, Canada
                [7] 7Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institute Stockholm, Sweden
                [8] 8Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institute Stockholm, Sweden
                [9] 9Department of Clinical Genetics, Erasmus University Medical Center Rotterdam, Netherlands
                [10] 10Assistance Publique–Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Université Paris Descartes, Paris Sorbonne Cité; Institut National de la Santé et de la Recherche Médicale, UMRS Paris, France
                [11] 11Department of Biology and Medical Genetics, 2nd Faculty of Medicine-Charles University and Motol University Hospital Prague, Czechia
                [12] 12Institute of Clinical and Experimental Medicine Prague, Czechia
                [13] 13Department of Human Genetics, Radboud University Medical Centre Nijmegen, Netherlands
                [14] 14Howard Hughes Medical Institute Baltimore, MD, United States
                Author notes

                Edited by: Alessandro Della Corte, Second University of Naples, Monaldi Hospital, Italy

                Reviewed by: Simon Body, Brigham and Women's Hospital, United States; Bengt-Olof Nilsson, Lund University, Sweden

                *Correspondence: Bart L. Loeys bart.loeys@ 123456uantwerpen.be

                This article was submitted to Vascular Physiology, a section of the journal Frontiers in Physiology

                †These authors have contributed equally to this work.

                Article
                10.3389/fphys.2017.00400
                5469151
                28659821
                0a1f81f4-db6d-4c64-a50e-7fceecbe6998
                Copyright © 2017 Gillis, Kumar, Luyckx, Preuss, Cannaerts, van de Beek, Wieschendorf, Alaerts, Bolar, Vandeweyer, Meester, Wünnemann, Gould, Zhurayev, Zerbino, Mohamed, Mital, Mertens, Björck, Franco-Cereceda, McCallion, Van Laer, Verhagen, van de Laar, Wessels, Messas, Goudot, Nemcikova, Krebsova, Kempers, Salemink, Duijnhouwer, Jeunemaitre, Albuisson, Eriksson, Andelfinger, Dietz, Verstraeten, Loeys and Mibava Leducq Consortium.

                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) or licensor 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
                : 14 April 2017
                : 26 May 2017
                Page count
                Figures: 2, Tables: 2, Equations: 0, References: 68, Pages: 10, Words: 8314
                Funding
                Funded by: Universiteit Antwerpen 10.13039/501100007660
                Award ID: Lanceringproject
                Funded by: Fonds Wetenschappelijk Onderzoek 10.13039/501100003132
                Award ID: G.0221.12
                Award ID: 12P0617N
                Award ID: 11Y5817N
                Award ID: 18B1316N
                Funded by: Fondation Leducq 10.13039/501100001674
                Award ID: Leducq 12CVD03
                Funded by: European Research Council 10.13039/501100000781
                Award ID: ERC- StG-2012-30972-BRAVE
                Funded by: Agentschap voor Innovatie door Wetenschap en Technologie 10.13039/501100003132
                Award ID: IWT.141429
                Categories
                Physiology
                Original Research

                Anatomy & Physiology
                bicuspid aortic valve,thoracic aortic aneurysm,smad6,targeted gene panel,variant burden test

                Comments

                Comment on this article