Elsevier

The Journal of Pediatrics

Volume 190, November 2017, Pages 27-37
The Journal of Pediatrics

Medical progress
Can We Understand the Pathobiology of Bronchopulmonary Dysplasia?

https://doi.org/10.1016/j.jpeds.2017.08.041Get rights and content

Section snippets

Understanding BPD Pathobiology – Where are We Now?

In the original form of BPD described by Northway et al, the characteristic pathologic features of severe inflammation, marked fibrosis, and airway dysplasia and muscularization, appeared to result from severe lung injury superimposed on immature lungs.1 In contrast, the “new” form of BPD, characterized by alveolar simplification, appears to result from a lesser magnitude of injury superimposed on highly immature lungs. The process of alveolarization is an intricate, highly orchestrated,

Injurious Stimuli That Disrupt Late Lung Development

Alveolarization is a complex developmental program requiring the temporal-specific activation of diverse signaling pathways, participation and coordination of multiple cell types interacting with the extracellular matrix, and influences of the lung microenvironment. Each component is necessary, but none are sufficient. As a result, the disruption of seemingly disparate molecular pathways by injurious stimuli (detailed below) have the ability to cause similar disruptions of alveolar and vascular

Impaired and Dysmorphic Pulmonary Vascular Growth

In addition to alveolar simplification, the pathobiology of the “new” BPD includes abnormalities of pulmonary microvascular development. Pathologic examination of lung tissue from infants dying from the “new” BPD,119 or long-term ventilated preterm infants found evidence of either decreased and abnormally distributed pulmonary microvessels, or increased but simplified and immature capillaries.120 Taken together, these studies suggest that vascular abnormalities in BPD may be variable with

Understanding BPD Pathobiology–Future Directions

Future studies to deepen our understanding of the pathobiology of BPD will continue to employ broad screens of human or animal model material, followed by targeted studies using interventional pharmacologic or genetic approaches to examine a causal role for a particular pathway or mediator. To this end, a large number of transcriptomic studies have been undertaken, addressing changes in messenger RNA and microRNA expression,159, 160, 161 and these data are only now being interrogated and

Summary

Since the original description of BPD, the past 50 years have seen tremendous efforts to characterize and understand the pathobiology of BPD, using both human BPD patient material and experimental animal models of arrested alveolarization that span the gamut from mouse to nonhuman primates. All of these approaches have generated a very solid foundation of knowledge for future studies, which must include a better appreciation of the nature of the pathologic processes at play, and the validation

First page preview

First page preview
Click to open first page preview

References (203)

  • W.H. Northway et al.

    Pulmonary disease following respirator therapy of hyaline-membrane disease. Bronchopulmonary dysplasia

    N Engl J Med

    (1967)
  • B.J. Stoll et al.

    Neonatal outcomes of extremely preterm infants from the NICHD Neonatal Research Network

    Pediatrics

    (2010)
  • A.H. Jobe

    The new bronchopulmonary dysplasia

    Curr Opin Pediatr

    (2011)
  • A.H. Jobe

    What is BPD in 2012 and what will BPD become?

    Early Hum Dev

    (2012)
  • B.J. Stoll et al.

    Trends in care practices, morbidity, and mortality of extremely preterm neonates, 1993-2012

    JAMA

    (2015)
  • J. Bourbon et al.

    Control mechanisms of lung alveolar development and their disorders in bronchopulmonary dysplasia

    Pediatr Res

    (2005)
  • W.H. Northway et al.

    Radiologic and histologic investigation of pulmonary oxygen toxicity in newborn guinea pigs

    Invest Radiol

    (1969)
  • L. Frank

    Developmental aspects of experimental pulmonary oxygen toxicity

    Free Radic Biol Med

    (1991)
  • L. Frank et al.

    Oxygen toxicity in neonatal and adult animals of various species

    J Appl Physiol Respir Environ Exerc Physiol

    (1978)
  • B.B. Warner et al.

    Functional and pathological effects of prolonged hyperoxia in neonatal mice

    Am J Physiol

    (1998)
  • S. Ahmad et al.

    Glutamine protects mitochondrial structure and function in oxygen toxicity

    Am J Physiol Lung Cell Mol Physiol

    (2001)
  • V. Ratner et al.

    Mitochondrial dysfunction contributes to alveolar developmental arrest in hyperoxia-exposed mice

    Am J Respir Cell Mol Biol

    (2009)
  • D. Irwin et al.

    Neonatal lung side population cells demonstrate endothelial potential and are altered in response to hyperoxia-induced lung simplification

    Am J Physiol Lung Cell Mol Physiol

    (2007)
  • V. Balasubramaniam et al.

    Hyperoxia reduces bone marrow, circulating, and lung endothelial progenitor cells in the developing lung: implications for the pathogenesis of bronchopulmonary dysplasia

    Am J Physiol Lung Cell Mol Physiol

    (2007)
  • C. Delaney et al.

    Lack of EC-SOD worsens alveolar and vascular development in a neonatal mouse model of bleomycin-induced bronchopulmonary dysplasia and pulmonary hypertension

    Pediatr Res

    (2015)
  • M.N. Ahmed et al.

    Extracellular superoxide dismutase protects lung development in hyperoxia-exposed newborn mice

    Am J Respir Crit Care Med

    (2003)
  • R.L. Auten et al.

    Transgenic extracellular superoxide dismutase protects postnatal alveolar epithelial proliferation and development during hyperoxia

    Am J Physiol Lung Cell Mol Physiol

    (2006)
  • E. Nozik-Grayck et al.

    Lung EC-SOD overexpression attenuates hypoxic induction of Egr-1 and chronic hypoxic pulmonary vascular remodeling

    Am J Physiol Lung Cell Mol Physiol

    (2008)
  • V. Bhandari

    Hyperoxia-derived lung damage in preterm infants

    Semin Fetal Neonatal Med

    (2010)
  • B.W. Buczynski et al.

    The role of hyperoxia in the pathogenesis of experimental BPD

    Semin Perinatol

    (2013)
  • D. Denis et al.

    Prolonged moderate hyperoxia induces hyperresponsiveness and airway inflammation in newborn rats

    Pediatr Res

    (2001)
  • E.D. Schultz et al.

    Mast cells mediate hyperoxia-induced airway hyper-reactivity in newborn rats

    Pediatr Res

    (2010)
  • R.L. Auten et al.

    Anti-neutrophil chemokine preserves alveolar development in hyperoxia-exposed newborn rats

    Am J Physiol Lung Cell Mol Physiol

    (2001)
  • DengH. et al.

    Lung inflammation in hyperoxia can be prevented by antichemokine treatment in newborn rats

    Am J Respir Crit Care Med

    (2000)
  • M.A. Vozzelli et al.

    Antimacrophage chemokine treatment prevents neutrophil and macrophage influx in hyperoxia-exposed newborn rat lung

    Am J Physiol Lung Cell Mol Physiol

    (2004)
  • L.C. Eldredge et al.

    CD11b(+) Mononuclear Cells Mitigate Hyperoxia-Induced Lung Injury in Neonatal Mice

    Am J Respir Cell Mol Biol

    (2016)
  • J.A. Kitterman

    The effects of mechanical forces on fetal lung growth

    Clin Perinatol

    (1996)
  • LiuM. et al.

    Invited review: mechanochemical signal transduction in the fetal lung

    J Appl Physiol

    (2000)
  • D.M. Dane et al.

    Separating in vivo mechanical stimuli for postpneumonectomy compensation: physiological assessment

    J Appl Physiol

    (2013)
  • A.M. Hoffman et al.

    Matrix modulation of compensatory lung regrowth and progenitor cell proliferation in mice

    Am J Physiol Lung Cell Mol Physiol

    (2010)
  • D.V. Page et al.

    Anomalies associated with pulmonary hypoplasia

    Am Rev Respir Dis

    (1982)
  • A. Rotschild et al.

    Neonatal outcome after prolonged preterm rupture of the membranes

    Am J Obstet Gynecol

    (1990)
  • WangW. et al.

    Longitudinal, noninvasive monitoring of compensatory lung growth in mice after pneumonectomy via (3)He and (1)H magnetic resonance imaging

    Am J Respir Cell Mol Biol

    (2013)
  • A.B. Ysasi et al.

    Effect of unilateral diaphragmatic paralysis on postpneumonectomy lung growth

    Am J Physiol Lung Cell Mol Physiol

    (2013)
  • S.M. Young et al.

    Localization and stretch-dependence of lung elastase activity in development and compensatory growth

    J Appl Physiol

    (2015)
  • J. Sanchez-Esteban et al.

    Cyclic mechanical stretch inhibits cell proliferation and induces apoptosis in fetal rat lung fibroblasts

    Am J Physiol Lung Cell Mol Physiol

    (2002)
  • J. Sanchez-Esteban et al.

    Mechanical stretch promotes alveolar epithelial type II cell differentiation

    J Appl Physiol

    (2001)
  • T.P. Quinn et al.

    Cyclic mechanical stretch induces VEGF and FGF-2 expression in pulmonary vascular smooth muscle cells

    Am J Physiol Lung Cell Mol Physiol

    (2002)
  • C.S. Muratore et al.

    Stretch-induced upregulation of VEGF gene expression in murine pulmonary culture: a role for angiogenesis in lung development

    J Pediatr Surg

    (2000)
  • J.J. Coalson et al.

    A baboon model of bronchopulmonary dysplasia. II. Pathologic features

    Exp Mol Pathol

    (1982)
  • J.J. Coalson et al.

    Neonatal chronic lung disease in extremely immature baboons

    Am J Respir Crit Care Med

    (1999)
  • E. Mourgeon et al.

    Mechanical stretch stimulates macrophage inflammatory protein-2 secretion from fetal rat lung cells

    Am J Physiol Lung Cell Mol Physiol

    (2000)
  • I.B. Copland et al.

    High tidal volume ventilation causes different inflammatory responses in newborn versus adult lung

    Am J Respir Crit Care Med

    (2004)
  • M. May et al.

    Apoptosis and proliferation in lungs of ventilated and oxygen-treated preterm infants

    Eur Respir J

    (2004)
  • J.D. Ricard et al.

    Production of inflammatory cytokines in ventilator-induced lung injury: a reappraisal

    Am J Respir Crit Care Med

    (2001)
  • U. Thome et al.

    Comparison of pulmonary inflammatory mediators in preterm infants treated with intermittent positive pressure ventilation or high frequency oscillatory ventilation

    Pediatr Res

    (1998)
  • L. Tremblay et al.

    Injurious ventilatory strategies increase cytokines and c-fos m-RNA expression in an isolated rat lung model

    J Clin Invest

    (1997)
  • R.D. Bland et al.

    Mechanical ventilation uncouples synthesis and assembly of elastin and increases apoptosis in lungs of newborn mice. Prelude to defective alveolar septation during lung development?

    Am J Physiol Lung Cell Mol Physiol

    (2008)
  • A.A. Kroon et al.

    Prolonged mechanical ventilation induces cell cycle arrest in newborn rat lung

    PLoS ONE

    (2011)
  • R.D. Bland et al.

    Mechanical ventilation with 40% oxygen reduces pulmonary expression of genes that regulate lung development and impairs alveolar septation in newborn mice

    Am J Physiol Lung Cell Mol Physiol

    (2007)
  • Cited by (41)

    • Bronchopulmonary Dysplasia

      2023, Principles of Neonatology
    • Safety of sildenafil in premature infants at risk of bronchopulmonary dysplasia: Rationale and methods of a phase II randomized trial

      2022, Contemporary Clinical Trials Communications
      Citation Excerpt :

      Approximately 50,000 infants are born each year in the US < 29 weeks' gestational age, and roughly 35% of these infants develop BPD [3,4]. Clinically, BPD is a disease of chronic respiratory insufficiency related to premature birth and lung inflammation and injury from hyperoxia [5,6] and mechanical ventilation [7–9]. These factors lead to impaired alveolar-capillary development, alveolar simplification, interstitial fibrosis, and the development of pulmonary hypertension [10–15].

    • Vitamin E and preterm infants

      2022, Free Radical Biology and Medicine
      Citation Excerpt :

      At present, BPD is roughly accepted as chronic respiratory insufficiency unique to extremely preterm infants (<28 wks GA) persisting at least beyond 36wks PMA. In brief, BPD is the end of abnormal lung development due to aberrant repair (remodeling) after injury to the immature developing lung, induced by various insults, such as intrauterine inflammatory processes, postnatal exposure to oxygen, ventilator-mediated mechanical trauma, infection, and malnutrition (Fig. 5A [166–170]). However, disappointingly, a satisfactory consensus on the definition has not yet been established.

    View all citing articles on Scopus

    Supported by the Stanford Child Health Research Institute Tashia and John Morgridge Faculty Scholar Award (CMA); the NIH HL122918 (CMA), the Max Planck Society (REM); Rhön Klinikum AG (grant FI_66) (REM); University Hospital Giessen and Marburg (FO-KOOPV-62589134) (REM); the Federal Ministry of Higher Education, Research and the Arts of the State of Hessen (LOEWE Program UGMLC) (REM), the German Center for Lung Research (Deutsches Zentrum für Lungenforschung; DZL) (REM); and the German Research Foundation (Deutsche Forschungsgemeinschaft, DFG) through EXC147, SFB1213, KFO309, and Mo 1789/1 (REM). The authors declare no conflicts of interest.

    View full text