39
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Congenital diaphragmatic hernias: from genes to mechanisms to therapies

      review-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

          Congenital diaphragmatic hernias (CDHs) and structural anomalies of the diaphragm are a common class of congenital birth defects that are associated with significant morbidity and mortality due to associated pulmonary hypoplasia, pulmonary hypertension and heart failure. In ∼30% of CDH patients, genomic analyses have identified a range of genetic defects, including chromosomal anomalies, copy number variants and sequence variants. The affected genes identified in CDH patients include transcription factors, such as GATA4, ZFPM2, NR2F2 and WT1, and signaling pathway components, including members of the retinoic acid pathway. Mutations in these genes affect diaphragm development and can have pleiotropic effects on pulmonary and cardiac development. New therapies, including fetal endoscopic tracheal occlusion and prenatal transplacental fetal treatments, aim to normalize lung development and pulmonary vascular tone to prevent and treat lung hypoplasia and pulmonary hypertension, respectively. Studies of the association between particular genetic mutations and clinical outcomes should allow us to better understand the origin of this birth defect and to improve our ability to predict and identify patients most likely to benefit from specialized treatment strategies.

          Abstract

          Summary: In this Review, Kardon et al. discuss the complex etiology of congenital diaphragmatic hernia, as revealed by comprehensive genomic analyses and modeling in mice, and highlight the need for new therapies to treat this developmental disorder.

          Related collections

          Most cited references206

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

          WT-1 is required for early kidney development.

          In humans, germline mutations of the WT-1 tumor suppressor gene are associated with both Wilms' tumors and urogenital malformations. To develop a model system for the molecular analysis of urogenital development, we introduced a mutation into the murine WT-1 tumor suppressor gene by gene targeting in embryonic stem cells. The mutation resulted in embryonic lethality in homozygotes, and examination of mutant embryos revealed a failure of kidney and gonad development. Specifically, at day 11 of gestation, the cells of the metanephric blastema underwent apoptosis, the ureteric bud failed to grow out from the Wolffian duct, and the inductive events that lead to formation of the metanephric kidney did not occur. In addition, the mutation caused abnormal development of the mesothelium, heart, and lungs. Our results establish a crucial role for WT-1 in early urogenital development.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5.

            Congenital heart defects (CHDs) are the most common developmental anomaly and are the leading non-infectious cause of mortality in newborns. Only one causative gene, NKX2-5, has been identified through genetic linkage analysis of pedigrees with non-syndromic CHDs. Here, we show that isolated cardiac septal defects in a large pedigree were linked to chromosome 8p22-23. A heterozygous G296S missense mutation of GATA4, a transcription factor essential for heart formation, was found in all available affected family members but not in any control individuals. This mutation resulted in diminished DNA-binding affinity and transcriptional activity of Gata4. Furthermore, the Gata4 mutation abrogated a physical interaction between Gata4 and TBX5, a T-box protein responsible for a subset of syndromic cardiac septal defects. Conversely, interaction of Gata4 and TBX5 was disrupted by specific human TBX5 missense mutations that cause similar cardiac septal defects. In a second family, we identified a frame-shift mutation of GATA4 (E359del) that was transcriptionally inactive and segregated with cardiac septal defects. These results implicate GATA4 as a genetic cause of human cardiac septal defects, perhaps through its interaction with TBX5.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Essential role for the c-met receptor in the migration of myogenic precursor cells into the limb bud.

              Limb muscles develop from cells that migrate from the somites. The signal that induces migration of myogenic precursor cells to the limb emanates from the mesenchyme of the limb bud. Here we report that the c-met-encoded receptor tyrosine kinase is essential for migration of myogenic precursor cells into the limb anlage and for migration into diaphragm and tip of tongue. In c-met homozygous mutant (-/-) mouse embryos, the limb bud and diaphragm are not colonized by myogenic precursor cells and, as a consequence, skeletal muscles of the limb and diaphragm do not form. In contrast, development of the axial skeletal muscles proceeds in the absence of c-met signalling. The specific ligand of the c-met protein, the motility and growth factor scatter factor/hepatocyte growth factor, is expressed in limb mesenchyme and can thus provide the signal for migration which is received by c-met. We have therefore identified a paracrine signalling system that regulates migration of myogenic precursor cells.
                Bookmark

                Author and article information

                Journal
                Dis Model Mech
                Dis Model Mech
                DMM
                dmm
                Disease Models & Mechanisms
                The Company of Biologists Ltd
                1754-8403
                1754-8411
                1 August 2017
                1 August 2017
                : 10
                : 8
                : 955-970
                Affiliations
                [1 ]Department of Human Genetics, University of Utah , Salt Lake City, UT 84112, USA
                [2 ]Departments of Pediatrics (Critical Care) and Biomedical Genetics, University of Rochester Medical Center , Rochester, NY 14642, USA
                [3 ]Department of Pediatrics, University of Wisconsin, Madison , WI 53792, USA
                [4 ]Department of Systems Biology, Columbia University Medical Center , New York, NY 10032, USA
                [5 ]Departments of Pediatrics, Columbia University Medical Center, New York, NY 10032, USA
                [6 ]Department of Pediatrics, University of California, San Diego , La Jolla, CA 92093, USA
                [7 ]Department of Medicine, Columbia University Medical Center , New York, NY 10032, USA
                Author notes
                Author information
                http://orcid.org/0000-0003-2144-4463
                http://orcid.org/0000-0002-1875-8931
                http://orcid.org/0000-0003-3438-5685
                Article
                DMM028365
                10.1242/dmm.028365
                5560060
                28768736
                babc1c86-f91b-47c2-86d2-4ad0a6a2a9ce
                © 2017. Published by The Company of Biologists Ltd

                This is an Open Access article distributed under the terms of the Creative Commons Attribution License ( http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

                History
                Funding
                Funded by: National Institutes of Health, http://dx.doi.org/10.13039/100000002;
                Award ID: R01 HL085459
                Funded by: March of Dimes Foundation, http://dx.doi.org/10.13039/100000912;
                Funded by: Wheeler Foundation;
                Award ID: R01HL122406
                Award ID: R01HL119946
                Award ID: R01HL113870
                Funded by: Wisconsin Partnership Program;
                Award ID: HD057036 and UL1 RR024156
                Funded by: CHERUBS;
                Funded by: National Greek Orthodox Ladies Philoptochos Society;
                Funded by: Vanech Family Foundation;
                Funded by: Larsen Family;
                Funded by: Fore Hanley Foundation;
                Funded by: Wilke Family;
                Categories
                Review

                Molecular medicine
                structural birth defects,congenital diaphragmatic hernia (cdh),diaphragm,pulmonary hypoplasia,pulmonary hypertension,congenital heart disease (chd),genetics

                Comments

                Comment on this article