13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Attenuated PDGF signaling drives alveolar and microvascular defects in neonatal chronic lung disease

      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

          Neonatal chronic lung disease ( nCLD) affects a significant number of neonates receiving mechanical ventilation with oxygen‐rich gas ( MV‐O 2). Regardless, the primary molecular driver of the disease remains elusive. We discover significant enrichment for SNPs in the PDGF‐Rα gene in preterms with nCLD and directly test the effect of PDGF‐Rα haploinsufficiency on the development of nCLD using a preclinical mouse model of MV‐O 2. In the context of MV‐O 2, attenuated PDGF signaling independently contributes to defective septation and endothelial cell apoptosis stemming from a PDGF‐Rα‐dependent reduction in lung VEGF‐A. TGF‐β contributes to the PDGF‐Rα‐dependent decrease in myofibroblast function. Remarkably, endotracheal treatment with exogenous PDGF‐A rescues both the lung defects in haploinsufficient mice undergoing MV‐O 2. Overall, our results establish attenuated PDGF signaling as an important driver of nCLD pathology with provision of PDGF‐A as a protective strategy for newborns undergoing MV‐O 2.

          Related collections

          Most cited references33

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

          PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis.

          A mouse platelet-derived growth factor A chain (PDGF-A) null allele is shown to be homozygous lethal, with two distinct restriction points, one prenatally before E10 and one postnatally. Postnatally surviving PDGF-A-deficient mice develop lung emphysema secondary to the failure of alveolar septation. This is apparently caused by the loss of alveolar myofibroblasts and associated elastin fiber deposits. PDGF alpha receptor-positive cells in the lung having the location of putative alveolar myofibroblast progenitors were specifically absent in PDGF-A null mutants. We conclude that PDGF-A is crucial for alveolar myofibroblast ontogeny. We have previously shown that PDGF-B is required in the ontogeny of kidney mesangial cells. The PDGFs therefore appear to regulate the generation of specific populations of myofibroblasts during mammalian development. The two PDGF null phenotypes also reveal analogous morphogenetic functions for myofibroblast-type cells in lung and kidney organogenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The new bronchopulmonary dysplasia.

            Bronchopulmonary dysplasia (BPD) remains the most common severe complication of preterm birth. A number of recent animal models and clinical studies provide new information about pathophysiology and treatment. The epidemiology of BPD continues to demonstrate that birth weight and gestational age are most predictive of BPD. Correlations of BPD with chorioamnionitis are clouded by the complexity of the fetal exposures to inflammation. Excessive oxygen use in preterm infants can increase the risk of BPD but low saturation targets may increase death. Numerous recent trials demonstrate that many preterm infants can be initially stabilized after delivery with continuous positive airway response (CPAP) and then be selectively treated with surfactant for respiratory distress syndrome. The growth of the lungs of the infant with BPD through childhood remains poorly characterized. Recent experiences in neonatology suggest that combining less invasive care strategies that avoid excessive oxygen and ventilation, decrease postnatal infections, and optimize nutrition may decrease the incidence and severity of BPD.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Loss of HIF-2alpha and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice.

              Respiratory distress syndrome (RDS) due to insufficient production of surfactant is a common and severe complication of preterm delivery. Here, we report that loss of the hypoxia-inducible transcription factor-2alpha (HIF-2alpha) caused fatal RDS in neonatal mice due to insufficient surfactant production by alveolar type 2 cells. VEGF, a target of HIF-2alpha, regulates fetal lung maturation: because VEGF levels in alveolar cells were reduced in HIF-2alpha-deficient fetuses; mice with a deficiency of the VEGF(164) and VEGF(188) isoforms or of the HIF-binding site in the VEGF promotor died of RDS; intrauterine delivery of anti-VEGF-receptor-2 antibodies caused RDS and VEGF stimulated production of surfactant proteins by cultured type 2 pneumocytes. Intrauterine delivery or postnatal intratracheal instillation of VEGF stimulated conversion of glycogen to surfactant and protected preterm mice against RDS. The pneumotrophic effect of VEGF may have therapeutic potential for lung maturation in preterm infants.
                Bookmark

                Author and article information

                Contributors
                tdesai@stanford.edu
                a.hilgendorff@med.uni-muenchen.de
                Journal
                EMBO Mol Med
                EMBO Mol Med
                10.1002/(ISSN)1757-4684
                EMMM
                embomm
                EMBO Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1757-4676
                1757-4684
                18 September 2017
                November 2017
                : 9
                : 11 ( doiID: 10.1002/emmm.v9.11 )
                : 1504-1520
                Affiliations
                [ 1 ] Comprehensive Pneumology Center University Hospital of the University of Munich and Helmholtz Zentrum Muenchen Munich Germany
                [ 2 ] Department of Pediatric Surgery Erasmus Medical Center – Sophia Children's Hospital Rotterdam The Netherlands
                [ 3 ] Institute for Medical Informatics Justus‐Liebig‐University Giessen Germany
                [ 4 ] Department of Pediatrics Stanford University School of Medicine Stanford CA USA
                [ 5 ] Department of Neonatology Perinatal Center Grosshadern Ludwig‐Maximilians University Munich Germany
                [ 6 ] Department of General Pediatrics University Clinic of Schleswig‐Holstein Campus Lübeck Lübeck Germany
                [ 7 ] Department of General Pediatrics and Neonatology Justus‐Liebig‐University and Universities of Giessen and Marburg Lung Center (UGMLC) Giessen Germany
                [ 8 ] Institute for Medical Informatics, Statistics, and Epidemiology (IMISE) University of Leipzig Leipzig Germany
                [ 9 ] Department of Pediatrics and Neonatology Medical University Vienna Vienna Austria
                [ 10 ] Department of Internal Medicine Pulmonary and Critical Care Stanford University School of Medicine Stanford CA USA
                [ 11 ] Center for Comprehensive Developmental Care Dr. von Haunersches Children's Hospital University Hospital Ludwig‐Maximilians University Munich Germany
                Author notes
                [*] [* ] Corresponding author. Tel: +1 650 723 1696; Fax: +1 650 498 6288; E‐mail: tdesai@ 123456stanford.edu

                Corresponding author. Tel: +49 89 3187 4675; Fax: +49 89 3187 4661; E‐mail: a.hilgendorff@ 123456med.uni-muenchen.de

                [†]

                These authors contributed equally to this work

                Author information
                http://orcid.org/0000-0002-8794-5319
                http://orcid.org/0000-0002-3725-996X
                Article
                EMMM201607308
                10.15252/emmm.201607308
                5666314
                28923828
                0a0bd0a5-c0e4-4fd2-82bd-ad57701d9277
                © 2017 Helmholtz Zentrum München Published under the terms of the CC BY 4.0 license

                This is an open access article under the terms of the Creative Commons Attribution 4.0 License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 14 November 2016
                : 25 July 2017
                : 27 July 2017
                Page count
                Figures: 10, Tables: 0, Pages: 17, Words: 11265
                Funding
                Funded by: Deutsche Forschungsgemeinschaft (DFG)
                Award ID: HI 1315/5‐1
                Funded by: Bundesministerium für Bildung und Forschung (BMBF)
                Award ID: 01KI1010C
                Award ID: 01KI1010I
                Funded by: Helmholtz‐Gemeinschaft (HGF)
                Award ID: NWG VH‐NG‐829
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                emmm201607308
                November 2017
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.2.1 mode:remove_FC converted:02.11.2017

                Molecular medicine
                bronchopulmonary dysplasia,neonatal chronic lung disease,pdgf‐rα,transforming growth factor‐β,vegf‐a,cardiovascular system,respiratory system

                Comments

                Comment on this article