35
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Reversing Blood Flows Act through klf2a to Ensure Normal Valvulogenesis in the Developing Heart

      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 directionality of local blood flow in the zebrafish embryonic heart is essential for proper heart valve formation.

          Abstract

          Heart valve anomalies are some of the most common congenital heart defects, yet neither the genetic nor the epigenetic forces guiding heart valve development are well understood. When functioning normally, mature heart valves prevent intracardiac retrograde blood flow; before valves develop, there is considerable regurgitation, resulting in reversing (or oscillatory) flows between the atrium and ventricle. As reversing flows are particularly strong stimuli to endothelial cells in culture, an attractive hypothesis is that heart valves form as a developmental response to retrograde blood flows through the maturing heart. Here, we exploit the relationship between oscillatory flow and heart rate to manipulate the amount of retrograde flow in the atrioventricular (AV) canal before and during valvulogenesis, and find that this leads to arrested valve growth. Using this manipulation, we determined that klf2a is normally expressed in the valve precursors in response to reversing flows, and is dramatically reduced by treatments that decrease such flows. Experimentally knocking down the expression of this shear-responsive gene with morpholine antisense oligonucleotides (MOs) results in dysfunctional valves. Thus, klf2a expression appears to be necessary for normal valve formation. This, together with its dependence on intracardiac hemodynamic forces, makes klf2a expression an early and reliable indicator of proper valve development. Together, these results demonstrate a critical role for reversing flows during valvulogenesis and show how relatively subtle perturbations of normal hemodynamic patterns can lead to both major alterations in gene expression and severe valve dysgenesis.

          Author Summary

          The growth and development of vertebrates are critically dependent on efficient cardiac output to drive blood circulation. An essential step of heart development is the formation of heart valves, whose leaflets are made through a complex set of cellular rearrangements of endothelial cells. Endothelial cells experience high flow forces as blood circulates. Moreover, heart valves and associated structures can be malformed when flow forces are abnormal, suggesting that these flow forces are in fact required for proper valve formation. Whether it is the force of the blood flow, its directionality (forward or reverse), or both that are important is not clear. We studied the interplay during valve development between key genes known to be involved in the process and epigenetic influences such as flow forces. Using zebrafish, whose optical clarity allows analyzing blood flow patterns at high resolution, we identified the presence of reversing flows specifically at the level of valve precursors. By manipulating blood flow patterns, we show that reversing flows are essential for valve morphogenesis. Specifically, we show that the expression of the gene klf2a depends on the presence of reversing flows and is required for valve development. We predict that by influencing levels of klf2a, reversing flows constitute an important stimulus controlling the appropriate biological responses of endothelial cells during valve formation.

          Related collections

          Most cited references38

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

          Prolonged fluid shear stress induces a distinct set of endothelial cell genes, most specifically lung Krüppel-like factor (KLF2).

          The endothelium expresses a large repertoire of genes under apparent transcriptional control of biomechanical forces, many of which are neither cell-type nor flow specific. We set out to identify genes that are uniquely flow responsive in human vascular endothelial cells. Transcriptional profiling using commercial DNA microarrays identified 12 of 18 000 genes that were modulated at least 5-fold after 24 hours of steady laminar flow (25 dyne/cm(2)). After a 7-day exposure to unidirectional pulsatile flow (19 +/- 12 dyne/cm(2)), only 3 of 12 remained elevated at least 5-fold. A custom microarray of ~300 vascular cell-related gene fragments was constructed, and expression analysis revealed that many flow-induced genes are also induced by at least one of the following agents: tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), transforming growth factor-beta, vascular endothelial growth factor, or thrombin, indicating a more general role in adaptive or stress responses. Most flow-induced genes were also induced by TNF-alpha but not IL-1beta, suggesting the involvement of reactive oxygen species. A limited panel of genes that are unique for flow-exposed cultures was identified, including lung Krüppel-like factor (LKLF/KLF2) and cytochrome P450 1B1 (CYP1B1). In marked contrast, both these genes were substantially repressed by TNF-alpha. LKLF but not CYP1B1 mRNA was detected exclusively in the vascular endothelium of healthy human aorta by in situ hybridization and appeared to be flow regulated. To date LKLF is the first endothelial transcription factor that is uniquely induced by flow and might therefore be at the molecular basis of the physiological healthy, flow-exposed state of the endothelial cell.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Heart valve development: endothelial cell signaling and differentiation.

            During the past decade, single gene disruption in mice and large-scale mutagenesis screens in zebrafish have elucidated many fundamental genetic pathways that govern early heart patterning and differentiation. Specifically, a number of genes have been revealed serendipitously to play important and selective roles in cardiac valve development. These initially surprising results have now converged on a finite number of signaling pathways that regulate endothelial proliferation and differentiation in developing and postnatal heart valves. This review highlights the roles of the most well-established ligands and signaling pathways, including VEGF, NFATc1, Notch, Wnt/beta-catenin, BMP/TGF-beta, ErbB, and NF1/Ras. Based on the interactions among and relative timing of these pathways, a signaling network model for heart valve development is proposed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Cardiac chamber formation: development, genes, and evolution.

              Concepts of cardiac development have greatly influenced the description of the formation of the four-chambered vertebrate heart. Traditionally, the embryonic tubular heart is considered to be a composite of serially arranged segments representing adult cardiac compartments. Conversion of such a serial arrangement into the parallel arrangement of the mammalian heart is difficult to understand. Logical integration of the development of the cardiac conduction system into the serial concept has remained puzzling as well. Therefore, the current description needed reconsideration, and we decided to evaluate the essentialities of cardiac design, its evolutionary and embryonic development, and the molecular pathways recruited to make the four-chambered mammalian heart. The three principal notions taken into consideration are as follows. 1) Both the ancestor chordate heart and the embryonic tubular heart of higher vertebrates consist of poorly developed and poorly coupled "pacemaker-like" cardiac muscle cells with the highest pacemaker activity at the venous pole, causing unidirectional peristaltic contraction waves. 2) From this heart tube, ventricular chambers differentiate ventrally and atrial chambers dorsally. The developing chambers display high proliferative activity and consist of structurally well-developed and well-coupled muscle cells with low pacemaker activity, which permits fast conduction of the impulse and efficacious contraction. The forming chambers remain flanked by slowly proliferating pacemaker-like myocardium that is temporally prevented from differentiating into chamber myocardium. 3) The trabecular myocardium proliferates slowly, consists of structurally poorly developed, but well-coupled, cells and contributes to the ventricular conduction system. The atrial and ventricular chambers of the formed heart are activated and interconnected by derivatives of embryonic myocardium. The topographical arrangement of the distinct cardiac muscle cells in the forming heart explains the embryonic electrocardiogram (ECG), does not require the invention of nodes, and allows a logical transition from a peristaltic tubular heart to a synchronously contracting four-chambered heart. This view on the development of cardiac design unfolds fascinating possibilities for future research.
                Bookmark

                Author and article information

                Contributors
                Role: Academic Editor
                Journal
                PLoS Biol
                plos
                plosbiol
                PLoS Biology
                Public Library of Science (San Francisco, USA )
                1544-9173
                1545-7885
                November 2009
                November 2009
                17 November 2009
                : 7
                : 11
                : e1000246
                Affiliations
                [1 ]Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
                [2 ]Option in Bioengineering, California Institute of Technology, Pasadena, California, United States of America
                [3 ]Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara, California, United States of America
                Osaka University, Japan
                Author notes

                The author(s) have made the following declarations about their contributions: Conceived and designed the experiments: JV ASF ML MG SEF. Performed the experiments: JV ASF DW DP. Analyzed the data: JV ASF ML DW MG SEF. Contributed reagents/materials/analysis tools: DW. Wrote the paper: JV ASF ML MG SEF. Movie crafting: JV ASF ML.

                [¤]

                Current address: IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), Department of Cell Biology and Development, Inserm-U964, CNRS-UMR7104, Université de Strasbourg, Illkirch, F-67400 France

                Article
                09-PLBI-RA-1415R2
                10.1371/journal.pbio.1000246
                2773122
                19924233
                7622b860-fb73-4d0b-8f8a-2cf6b37edcd6
                Vermot et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
                History
                : 1 April 2009
                : 9 October 2009
                Page count
                Pages: 14
                Categories
                Research Article
                Cardiovascular Disorders/Hemodynamics
                Developmental Biology/Morphogenesis and Cell Biology

                Life sciences
                Life sciences

                Comments

                Comment on this article