201
|
Sriram S, Schreiber MD, Msall ME, Kuban KCK, Joseph RM, O' Shea TM, Allred EN, Leviton A. Cognitive Development and Quality of Life Associated With BPD in 10-Year-Olds Born Preterm. Pediatrics 2018; 141:e20172719. [PMID: 29773664 PMCID: PMC6317639 DOI: 10.1542/peds.2017-2719] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2018] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES To compare neurocognitive, language, executive function, academic achievement, neurologic and behavioral outcomes, and quality of life at age 10 years in children born extremely preterm who developed bronchopulmonary dysplasia (BPD) to children who did not develop BPD. METHODS The Extremely Low Gestational Age Newborns study population included 863 children born extremely preterm whose BPD status before discharge was known had an IQ (Differential Ability Scales II [DAS II]) assessment at 10 years. We evaluated the association of BPD with any cognitive (DAS II), executive function (NEuroPSYchological Assessment II), academic achievement (Wechsler Individual Achievement Test-III and Oral and Written Language Scales [OWLS]) as well as social dysfunctions (Social Responsiveness Scale). We used logistic regression models, adjusting for potential confounding factors, to assess the strength of association between the severity of BPD and each outcomes. RESULTS Three hundred and seventy-two (43%) children were oxygen-dependent at 36 weeks postconception age, whereas an additional 78 (9%) were also oxygen- and ventilator-dependent. IQ scores 2 or more SDs below the expected mean (ie, z scores ≤-2) occurred twice as commonly among children who had BPD as among those who did not. Children with severe BPD consistently had the lowest scores on DAS II, OWLS, Wechsler Individual Achievement Test-III, NEuroPSYchological Assessment II, and Social Responsiveness Scale assessments. CONCLUSIONS Among 10-year-old children born extremely preterm, those who had BPD were at increased risk of cognitive, language, and executive dysfunctions; academic achievement limitations; social skill deficits; and low scores on assessments of health-related quality of life.
Collapse
Affiliation(s)
- Sudhir Sriram
- Department of Pediatrics, Section of Neonatology, and
| | | | - Michael E Msall
- Section of Developmental and Behavioral Pediatrics, Kennedy Research Center on Intellectual and Developmental Disabilities, The University of Chicago, Chicago, Illinois
| | - Karl C K Kuban
- Division of Pediatric Neurology, Departments of Pediatrics and
| | - Robert M Joseph
- Anatomy and Neurobiology, School of Medicine, Boston University, Boston, Massachusetts
| | - T Michael O' Shea
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Elizabeth N Allred
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Alan Leviton
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts
| |
Collapse
|
202
|
TRPA1 channels: expression in non-neuronal murine lung tissues and dispensability for hyperoxia-induced alveolar epithelial hyperplasia. Pflugers Arch 2018; 470:1231-1241. [PMID: 29754249 DOI: 10.1007/s00424-018-2148-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/22/2018] [Accepted: 04/27/2018] [Indexed: 01/31/2023]
Abstract
Transient receptor potential A1 (TRPA1) channels were originally characterized in neuronal tissues but also identified in lung epithelium by staining with fluorescently coupled TRPA1 antibodies. Its exact function in non-neuronal tissues, however, is elusive. TRPA1 is activated in vitro by hypoxia and hyperoxia and is therefore a promising TRP candidate for sensing hyperoxia in pulmonary epithelial cells and for inducing alveolar epithelial hyperplasia. Here, we isolated tracheal, bronchial, and alveolar epithelial cells and show low but detectable TRPA1 mRNA levels in all these cells as well as TRPA1 protein by Western blotting in alveolar type II (AT II) cells. We quantified changes in intracellular Ca2+ ([Ca2+]i) levels induced by application of hyperoxic solutions in primary tracheal epithelial, bronchial epithelial, and AT II cells isolated from wild-type (WT) and TRPA1-deficient (TRPA1-/-) mouse lungs. In all cell types, we detected hyperoxia-induced rises in [Ca2+]i levels, which were not significantly different in TRPA1-deficient cells compared to WT cells. We also tested TRPA1 function in a mouse model for hyperoxia-induced alveolar epithelial hyperplasia. A characteristic significant increase in thickening of alveolar tissues was detected in mouse lungs after exposure to hyperoxia, but not in normoxic WT and TRPA1-/- controls. Quantification of changes in lung morphology in hyperoxic WT and TRPA1-/- mice, however, again revealed no significant changes. Therefore, TRPA1 expression does neither appear to be a key player for hyperoxia-induced changes in [Ca2+]i levels in primary lung epithelial cells, nor being essential for the development of hyperoxia-induced alveolar epithelial hyperplasia.
Collapse
|
203
|
Kaneko M, Sato M, Ogasawara K, Imamura T, Hashimoto K, Momoi N, Hosoya M. Serum cytokine concentrations, chorioamnionitis and the onset of bronchopulmonary dysplasia in premature infants. J Neonatal Perinatal Med 2018; 10:147-155. [PMID: 28409755 DOI: 10.3233/npm-171669] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To investigate the relationships between serum cytokine concentrations and chorioamnionitis (CAM) and CAM-related bronchopulmonary dysplasia (BPD) in premature infants. METHODS Serum was collected at 0 and 7 days after birth from 36 premature infants born at <32 weeks of gestation. We examined the relationships between 30 cytokine concentrations and CAM, BPD, and other perinatal factors. RESULTS On day 0, GM-CSF, IL-15, IL-17, IL-2, IL-2R, VEGF, and MIG concentrations were significantly higher in the CAM group (n = 17) than in the non-CAM group (n = 19). These concentrations had decreased by day 7 and were similar in both groups. The IL-12p70 concentration on day 0 was significantly lower in the BPD group (n = 16) than in the non-BPD group (n = 15). BPD incidence was similar between the CAM and non-CAM groups. CONCLUSIONS These data support the hypothesis that intrauterine inflammation is not a primary risk factor for BPD. The immunological environment at birth or soon after, rather than intrauterine fetal inflammation (e.g., CAM), is a primary risk factor for BPD onset in preterm infants. Decreased inflammatory responses are particularly relevant, as indicated by the relationship between BPD and low serum IL-12p70 concentrations on day 0.
Collapse
|
204
|
Placental transfusion in preterm neonates of 30-33 weeks' gestation: a randomized controlled trial. J Perinatol 2018; 38:496-504. [PMID: 29410545 DOI: 10.1038/s41372-018-0064-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/30/2017] [Accepted: 01/16/2018] [Indexed: 11/08/2022]
Abstract
OBJECTIVES To compare effect of placental transfusion by delayed cord clamping (DCC) or cord milking (CM) with early cord clamping (ECC) on a composite of mortality or abnormal neurological status at 40 weeks' post-menstrual age (PMA) and 24-30 months' chronological age in neonates of 30-33 weeks' gestation. STUDY DESIGN Randomized, controlled trial. OUTCOMES A composite of mortality or abnormal neurological status at 40 weeks PMA and survival free of neurodevelopmental abnormalities at 24-30 months' chronological age. RESULTS A total of 461 neonates were randomized to placental transfusion (n = 233) or to ECC (n = 228). Among those assigned to placental transfusion group, 173 underwent DCC while in the remaining 60, CM was done. Incidence of mortality or abnormal neurological status at 40 weeks PMA (43 (18%) vs 35 (15%), RR (95% CI) 1.2 (0.8, 1.8), p = 0.4) and survival free of neurodevelopmental impairment at 24-30 months of chronological age (99 (47%) vs. 100 (50%); RR (95% CI): 0.9 (0.8, 1.2); P = 0.9) was similar between the study groups. The placental transfusion group showed a trend towards lower incidence of necrotizing enterocolitis. CONCLUSION In 30-33 weeks' gestation preterm neonates, placental transfusion as compared to early cord clamping resulted in similar mortality or abnormal neurological status at 40 weeks PMA and at 24-30 months of chronological age.
Collapse
|
205
|
Pabelick CM, Thompson MA, Britt RD. Effects of Hyperoxia on the Developing Airway and Pulmonary Vasculature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 967:179-194. [PMID: 29047087 DOI: 10.1007/978-3-319-63245-2_11] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although it is necessary and part of standard practice, supplemental oxygen (40-90% O2) or hyperoxia is a significant contributing factor to development of bronchopulmonary dysplasia, persistent pulmonary hypertension, recurrent wheezing, and asthma in preterm infants. This chapter discusses hyperoxia and the role of redox signaling in the context of neonatal lung growth and disease. Here, we discuss how hyperoxia promotes dysfunction in the airway and the known redox-mediated mechanisms that are important for postnatal vascular and alveolar development. Whether in the airway or alveoli, redox pathways are important and greatly influence the neonatal lung.
Collapse
Affiliation(s)
- Christina M Pabelick
- Department of Anesthesiology, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA. .,Departments Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA.
| | - Michael A Thompson
- Department of Anesthesiology, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA
| | - Rodney D Britt
- Departments Physiology and Biomedical Engineering, College of Medicine, Mayo Clinic, 4-184 W Jos SMH, 200 First St SW, Rochester, MN, 55905, USA
| |
Collapse
|
206
|
Sucre JMS, Deutsch GH, Jetter CS, Ambalavanan N, Benjamin JT, Gleaves LA, Millis BA, Young LR, Blackwell TS, Kropski JA, Guttentag SH. A Shared Pattern of β-Catenin Activation in Bronchopulmonary Dysplasia and Idiopathic Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:853-862. [PMID: 29355514 PMCID: PMC5866104 DOI: 10.1016/j.ajpath.2017.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/14/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
Wnt/β-catenin signaling is necessary for normal lung development, and abnormal Wnt signaling contributes to the pathogenesis of both bronchopulmonary dysplasia (BPD) and idiopathic pulmonary fibrosis (IPF), fibrotic lung diseases that occur during infancy and aging, respectively. Using a library of human normal and diseased human lung samples, we identified a distinct signature of nuclear accumulation of β-catenin phosphorylated at tyrosine 489 and epithelial cell cytosolic localization of β-catenin phosphorylated at tyrosine 654 in early normal lung development and fibrotic lung diseases BPD and IPF. Furthermore, this signature was recapitulated in murine models of BPD and IPF. Image analysis of immunofluorescence colocalization demonstrated a consistent pattern of elevated nuclear phosphorylated β-catenin in the lung epithelium and surrounding mesenchyme in BPD and IPF, closely resembling the pattern observed in 18-week fetal lung. Nuclear β-catenin phosphorylated at tyrosine 489 associated with an increased expression of Wnt target gene AXIN2, suggesting that the observed β-catenin signature is of functional significance during normal development and injury repair. The association of specific modifications of β-catenin during normal lung development and again in response to lung injury supports the widely held concept that repair of lung injury involves the recapitulation of developmental programs. Furthermore, these observations suggest that β-catenin phosphorylation has potential as a therapeutic target for the treatment and prevention of both BPD and IPF.
Collapse
Affiliation(s)
- Jennifer M S Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee.
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital and University of Washington, Seattle, Washington
| | - Christopher S Jetter
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | | | - John T Benjamin
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Linda A Gleaves
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee; Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, Tennessee
| | - Lisa R Young
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Division of Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Susan H Guttentag
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
207
|
Bentsen MH, Markestad T, Halvorsen T. Ventilator flow data predict bronchopulmonary dysplasia in extremely premature neonates. ERJ Open Res 2018; 4:00099-2017. [PMID: 29546045 PMCID: PMC5847811 DOI: 10.1183/23120541.00099-2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/24/2018] [Indexed: 01/08/2023] Open
Abstract
Early prediction of bronchopulmonary dysplasia (BPD) may facilitate tailored management for neonates at risk. We investigated whether easily accessible flow data from a mechanical ventilator can predict BPD in neonates born extremely premature (EP). In a prospective population-based study of EP-born neonates, flow data were obtained from the ventilator during the first 48 h of life. Data were logged for >10 min and then converted to flow-volume loops using custom-made software. Tidal breathing parameters were calculated and averaged from ≥200 breath cycles, and data were compared between those who later developed moderate/severe and no/mild BPD. Of 33 neonates, 18 developed moderate/severe and 15 no/mild BPD. The groups did not differ in gestational age, surfactant treatment or ventilator settings. The infants who developed moderate/severe BPD had evidence of less airflow obstruction, significantly so for tidal expiratory flow at 50% of tidal expiratory volume (TEF50) expressed as a ratio of peak tidal expiratory flow (PTEF) (p=0.007). A compound model estimated by multiple logistic regression incorporating TEF50/PTEF, birthweight z-score and sex predicted moderate/severe BPD with good accuracy (area under the curve 0.893, 95% CI 0.735-0.973). This study suggests that flow data obtained from ventilators during the first hours of life may predict later BPD in premature neonates. Future and larger studies are needed to validate these findings and to determine their clinical usefulness.
Collapse
Affiliation(s)
- Mariann H Bentsen
- Dept of Pediatrics, Haukeland University Hospital, Bergen, Norway.,Dept of Clinical Science, Faculty of Medicine and Odontology, University of Bergen, Bergen, Norway
| | - Trond Markestad
- Dept of Pediatrics, Haukeland University Hospital, Bergen, Norway.,Dept of Clinical Science, Faculty of Medicine and Odontology, University of Bergen, Bergen, Norway
| | - Thomas Halvorsen
- Dept of Pediatrics, Haukeland University Hospital, Bergen, Norway.,Dept of Clinical Science, Faculty of Medicine and Odontology, University of Bergen, Bergen, Norway
| |
Collapse
|
208
|
Stone CA, McEvoy CT, Aschner JL, Kirk A, Rosas-Salazar C, Cook-Mills JM, Moore PE, Walsh WF, Hartert TV. Update on Vitamin E and Its Potential Role in Preventing or Treating Bronchopulmonary Dysplasia. Neonatology 2018; 113:366-378. [PMID: 29514147 PMCID: PMC5980725 DOI: 10.1159/000487388] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022]
Abstract
Vitamin E is obtained only through the diet and has a number of important biological activities, including functioning as an antioxidant. Evidence that free radicals may contribute to pathological processes such as bronchopulmonary dysplasia (BPD), a disease of prematurity associated with increased lung injury, inflammation and oxidative stress, led to trials of the antioxidant vitamin E (α-tocopherol) to prevent BPD with variable results. These trials were all conducted at supraphysiologic doses and 2 of these trials utilized a formulation containing a potentially harmful excipient. Since 1991, when the last of these trials was conducted, both neonatal management strategies for minimizing oxygen and ventilator-related lung injury and our understanding of vitamin E isoforms in respiratory health have advanced substantially. It is now known that there are differences between the effects of vitamin E isoforms α-tocopherol and γ-tocopherol on the development of respiratory morbidity and inflammation. What is not known is whether improvements in physiologic concentrations of individual or combinations of vitamin E isoforms during pregnancy or following preterm birth might prevent or reduce BPD development. The answers to these questions require adequately powered studies targeting pregnant women at risk of preterm birth or their premature infants immediately following birth, especially in certain subgroups that are at increased risk of vitamin E deficiency (e.g., smokers). The objective of this review is to compile, update, and interpret what is known about vitamin E isoforms and BPD since these first studies were conducted, and suggest future research directions.
Collapse
Affiliation(s)
- Cosby A Stone
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cindy T McEvoy
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - Judy L Aschner
- Division of Neonatology, Department of Pediatrics, Albert Einstein College of Medicine and the Children's Hospital at Montefiore, Bronx, New York, USA
| | - Ashudee Kirk
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christian Rosas-Salazar
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joan M Cook-Mills
- Division of Allergy-Immunology, Department of Medicine, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Paul E Moore
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William F Walsh
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tina V Hartert
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
209
|
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in the newborns. Staging of BPD severity does not have a high predictive value for the outcomes. This study was aimed to assess the diagnostic value of chest computed tomography (CT) combined with x-ray for premature infants with BPD.Twenty-five premature infants with mild BPD and 20 premature infants with moderate to severe BPD treated at our hospital from January 2015 to December 2015 were randomly selected. The imaging features were compared between premature infants with different severity of BPD.In mild BPD group, the incidence of increased lung opacity (at 3-10 and 29 days) were significantly higher than those in infants with moderate to severe BPD (P = .034, P = .003, respectively). However, the incidences of stage III BPD (3-10 days) and stage IV BPD (11-27 days) were significantly lower in infants with mild BPD than those in infants with moderate to severe BPD (P = .013, P = .033, respectively). The chest x-ray score in the mild BPD group was significantly lower than that in moderate to severe BPD group [3.0 (1.0) vs 5.0 (1.0), P < .001]. Spearman rank correlation analysis indicated that chest x-ray score had significant correlation (r = 0.787, P < .001) with the clinical severity. In the mild BPD group, the chest CT scan score was 11.52 ± 3.49, which was considerably lower than that in the moderate to severe BPD group (24.70 ± 4.32) (P < .001). Moreover, the severity of BPD in the premature infants was significantly correlated to the chest CT scan score (r = 0.855, P < .001).Chest CT combined with x-ray is an effective method for predicting the severity of BPD in premature infants.
Collapse
|
210
|
Roberts KD, Brown R, Lampland AL, Leone TA, Rudser KD, Finer NN, Rich WD, Merritt TA, Czynski AJ, Kessel JM, Tipnis SM, Stepka EC, Mammel MC. Laryngeal Mask Airway for Surfactant Administration in Neonates: A Randomized, Controlled Trial. J Pediatr 2018; 193:40-46.e1. [PMID: 29174079 DOI: 10.1016/j.jpeds.2017.09.068] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/01/2017] [Accepted: 09/26/2017] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To determine if preterm infants with moderate respiratory distress syndrome on continuous positive airway pressure (CPAP) who received surfactant via a laryngeal mask airway (LMA) would have a decreased rate of intubation and mechanical ventilation compared with those on CPAP who did not receive surfactant. STUDY DESIGN In this prospective, multicenter, randomized controlled trial, 103 premature infants 280/7-356/7 weeks gestation, ≥1250 g and ≤36 hours old on CPAP requiring fraction of inspired oxygen 0.30-0.40 were assigned to receive surfactant administered through an LMA then placed back on CPAP (LMA group) or maintained on CPAP with no surfactant administered (control group). The primary outcome was treatment failure necessitating intubation and mechanical ventilation in the first 7 days of life. RESULTS Surfactant administration through an LMA (n = 50) significantly decreased the rate of intubation and mechanical ventilation compared with controls (n = 53): 38% vs 64%, respectively, OR 0.30 (95% CI 0.13, 0.70), P = .006, number needed to treat: 4). There were no serious adverse events associated with placement of the LMA or surfactant administration. CONCLUSIONS In premature neonates with moderate respiratory distress syndrome, surfactant administered through an LMA decreased the rate of intubation and mechanical ventilation. This intervention may have significant impact on clinical care in both high and low resource settings. TRIAL REGISTRATION ClinicalTrials.gov: NCT01116921.
Collapse
Affiliation(s)
- Kari D Roberts
- Department of Pediatrics, University of Minnesota, Minneapolis, MN.
| | - Roland Brown
- Department of Biostatistics, University of Minnesota, Minneapolis, MN
| | - Andrea L Lampland
- Department of Pediatrics, University of Minnesota, Minneapolis, MN; Department of Pediatrics, Children's Minnesota, St. Paul, MN
| | - Tina A Leone
- Department of Pediatrics, Columbia University, New York, NY
| | - Kyle D Rudser
- Department of Biostatistics, University of Minnesota, Minneapolis, MN
| | - Neil N Finer
- Department of Pediatrics, University of California-San Diego, San Diego, CA
| | - Wade D Rich
- Department of Pediatrics, Sharp Mary Birch, San Diego, CA
| | - T Allen Merritt
- Department of Pediatrics, Loma Linda University, Loma Linda, CA
| | - Adam J Czynski
- Department of Pediatrics, Loma Linda University, Loma Linda, CA
| | - Julie M Kessel
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI
| | - Sajani M Tipnis
- Department of Pediatrics, University of Mississippi, Jackson, MS
| | - Erin C Stepka
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Mark C Mammel
- Department of Pediatrics, University of Minnesota, Minneapolis, MN; Department of Pediatrics, Children's Minnesota, St. Paul, MN
| |
Collapse
|
211
|
Lingappan K, Maturu P, Liang YW, Jiang W, Wang L, Moorthy B, Couroucli XI. β-Naphthoflavone treatment attenuates neonatal hyperoxic lung injury in wild type and Cyp1a2-knockout mice. Toxicol Appl Pharmacol 2018; 339:133-142. [PMID: 29180065 PMCID: PMC5758404 DOI: 10.1016/j.taap.2017.11.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 11/19/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023]
Abstract
Exposure to supraphysiological concentrations of oxygen (hyperoxia) leads to bronchopulmonary dysplasia (BPD), one of the most common pulmonary morbidities in preterm neonates, which is more prevalent in males than females. Beta-naphthoflavone (BNF) is protective against hyperoxic lung injury in adult and neonatal wild type (WT) mice and in and mice lacking Cyp1a1gene. In this investigation, we tested the hypothesis that BNF treatment will attenuate neonatal hyperoxic lung injury in WT and Cyp1a2-/- mice, and elucidated the effect of sex-specific differences. Newborn WT or Cyp1a2-/- mice were treated with BNF (10mg/kg) or the vehicle corn oil (CO) i.p., from postnatal day (PND) 2 to 8 once every other day, while being maintained in room air or hyperoxia (85% O2) for 14days. Hyperoxia exposure lead to alveolar simplification and arrest in angiogenesis in WT as well as Cyp1a2-/- mice No significant differences were seen between WT and Cyp1a2-/- mice. Cyp1a2-/- female mice had better preservation of pulmonary angiogenesis at PND15 compared to similarly exposed males. BNF treatment attenuated lung injury and inflammation in both genotypes, and this was accompanied by a significant induction of hepatic and pulmonary CYP1A1 in WT but not in Cyp1a2-/- mice. BNF treatment increased NADPH quinone oxidoreductase (NQO1) mRNA levels in Cyp1a2-/- mouse livers compared to WT mice. These results suggest that BNF is protective in neonatal mice exposed to hyperoxia independent of CYP1A2 and this may entail the protective effect of phase II enzymes like NQO1.
Collapse
Affiliation(s)
- Krithika Lingappan
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Paramahamsa Maturu
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Yanhong Wei Liang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Weiwu Jiang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Lihua Wang
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Bhagavatula Moorthy
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Xanthi I Couroucli
- Department of Pediatrics, Section of Neonatology, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
212
|
Rutkowska M, Hożejowski R, Helwich E, Borszewska-Kornacka MK, Gadzinowski J. Severe bronchopulmonary dysplasia - incidence and predictive factors in a prospective, multicenter study in very preterm infants with respiratory distress syndrome. J Matern Fetal Neonatal Med 2018; 32:1958-1964. [PMID: 29295665 DOI: 10.1080/14767058.2017.1422711] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Severe bronchopulmonary dysplasia (BPD) remains a major complication of prematurity and can have significant impact on long-term pulmonary sequelae and adverse neurodevelopmental outcomes. OBJECTIVE To assess the incidence and evaluate the predictive factors for severe BPD in very preterm infants with respiratory distress syndrome. METHODS Of the 846 premature infants born at ≤32-week gestation who developed respiratory distress syndrome (RDS), 707 infants with known oxygen dependency at 36 weeks gestational age were included in the analysis of BPD incidence. With univariate and multiple logistic regression models we evaluated the risk factors for the development of severe BPD and calculated odds ratios (ORs). RESULTS The overall incidence of BPD was 45.2%. Severe BPD accounted for 6%, with morbidity pertaining mainly to infants <29-week gestation (incidence 10%). The risk factors for severe BPD included male gender (OR 3.02 95%CI 1.30-7.46), intubation in the delivery room (OR 2.57, 95%CI 1.00-7.18), and invasive ventilation >7 days (OR 7.05, 95%CI 2.63-22.4). The protective factors were early continuous positive airway pressure (CPAP) in the univariate analysis and receiving surfactant <15 min after birth in the multivariate model. CONCLUSIONS Mechanical ventilation >7 days is the most prevalent risk factor for severe BPD. CPAP initiated in the delivery room and early surfactant are key preventive measures.
Collapse
Affiliation(s)
- Magdalena Rutkowska
- a Clinic of Neonatology and Intensive Care , Institute of Mother and Child , Warsaw , Poland
| | - Roman Hożejowski
- b Medical Department , Chiesi Poland Sp. z o.o. , Warsaw , Poland
| | - Ewa Helwich
- a Clinic of Neonatology and Intensive Care , Institute of Mother and Child , Warsaw , Poland
| | | | - Janusz Gadzinowski
- d Department of Neonatology , Poznań University of Medical Sciences , Poznań , Poland
| |
Collapse
|
213
|
Bronchopulmonary dysplasia as a risk factor for asthma in school children and adolescents: A systematic review. Allergol Immunopathol (Madr) 2018; 46:87-98. [PMID: 28668285 DOI: 10.1016/j.aller.2017.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 02/18/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a chronic lung disease that mainly affects extremely pre-term infants, and remains the most common complication of prematurity. Several studies have shown that prematurity predisposes to the development of asthma in school children and adolescents. Nevertheless, it is not clear to what extent a history of BPD involves an additional risk. METHODS A systematic review of studies assessing the association between BPD and asthma in school-children and adolescents was made. A literature search was carried out in the MEDLINE and EMBASE databases to retrieve articles published between 1 January 2000 and 31 August 2016. RESULTS A total of 17 studies comprising 7433 patients were included in the review. There was considerable heterogeneity in the definitions of BPD and asthma among studies. Overall, the prevalence of asthma was higher in children and adolescents with a history of prematurity and BPD compared with those who did not develop BPD. However, in only one of the studies did this difference reach statistical significance. The main limitation of this review was potential bias due to the lack of adjustment for confounding factors between exposure (BPD) and outcome (asthma) in most of the studies. CONCLUSION Based on the studies reviewed, it cannot be argued that BPD, as an independent factor of prematurity, increases the risk of asthma defined by clinical parameters in school-children and adolescents. Further studies of greater methodological quality and homogeneous diagnostic criteria of BPD and asthma are needed for improved assessment of this association.
Collapse
|
214
|
Stark A, Dammann C, Nielsen HC, Volpe MV. A Pathogenic Relationship of Bronchopulmonary Dysplasia and Retinopathy of Prematurity? A Review of Angiogenic Mediators in Both Diseases. Front Pediatr 2018; 6:125. [PMID: 29951473 PMCID: PMC6008318 DOI: 10.3389/fped.2018.00125] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/16/2018] [Indexed: 01/11/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are common and significant morbidities of prematurely born infants. These diseases have in common altered and pathologic vascular formation in the face of incomplete organ development. Therefore, it is reasonable to question whether factors affecting angiogenesis could have a joint pathogenic role for both diseases. Inhibition or induced expression of a single angiogenic factor is unlikely to be 100% causative or protective of either of BPD or ROP. It is more likely that interactions of multiple factors leading to disordered angiogenesis are present, increasing the likelihood of common pathways in both diseases. This review explores this possibility by assessing the evidence showing involvement of specific angiogenic factors in the vascular development and maldevelopment in each disease. Theoretical interactions of specific factors mutually contributing to BPD and ROP are proposed and, where possible, a timeline of the proposed relationships between BPD and ROP is developed. It is hoped that future research will be inspired by the theories put forth in this review to enhance the understanding of the pathogenesis in both diseases.
Collapse
Affiliation(s)
- Ashley Stark
- Tufts University School of Medicine, Boston, MA, United States
| | - Christiane Dammann
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States.,Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Heber C Nielsen
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States.,Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - MaryAnn V Volpe
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
215
|
Leroy S, Caumette E, Waddington C, Hébert A, Brant R, Lavoie PM. A Time-Based Analysis of Inflammation in Infants at Risk of Bronchopulmonary Dysplasia. J Pediatr 2018; 192:60-65.e1. [PMID: 29092751 DOI: 10.1016/j.jpeds.2017.09.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/28/2017] [Accepted: 09/07/2017] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To precisely delineate the timing and contribution of inflammation to bronchopulmonary dysplasia (BPD) in preterm infants during the neonatal period. STUDY DESIGN Longitudinal study of blood inflammatory biomarkers (interleukin [IL]-6, IL-8, and granulocyte colony-stimulating factor) measured between birth and 42 days of age, at high temporal (daily) resolution, in infants born at or below 30 weeks of gestation. Cytokine predictors of BPD at 36 weeks postmenstrual age were adjusted for infant-specific and time-dependent factors, using hierarchical mixed effects regressions models. RESULTS A total of 1518 data points were obtained in 62 infants (mean gestational age of 27 weeks). Infants who developed BPD later on presented increased inflammation after birth compared with infants without BPD. Inflammation was sustained, with gradual attenuation over 2 weeks (IL-8: OR: 6.5 [95% CI: 1.8-24]; granulocyte colony-stimulating factor: 3.3 [1.5-7.6]) and was higher in boys and in infants of lower birth weight. This inflammation preceded the clinical increased requirement in supplemental oxygen characteristic of BPD, and preceded the peak occurrence of neonatal sepsis or necrotizing enterocolitis. CONCLUSIONS Systemic inflammation occurs early in the neonatal period and precedes clinical symptoms in infants with BPD. These data provide a discrete vulnerability window period, supporting a role for targeted intensive care interventions during the early phase of BPD.
Collapse
Affiliation(s)
- Sandrine Leroy
- EA2415, Montpellier University, Montpellier, France; Mobile Pediatrics Intensive Care Unit, Avicenne hospital, AP-HP, Paris, France
| | | | - Chandra Waddington
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Audrey Hébert
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rollin Brant
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pascal M Lavoie
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
216
|
de Wijs-Meijler DPM, van Duin RWB, Duncker DJ, Scherrer U, Sartori C, Reiss IKM, Merkus D. Structural and functional changes of the pulmonary vasculature after hypoxia exposure in the neonatal period: a new swine model of pulmonary vascular disease. Am J Physiol Heart Circ Physiol 2017; 314:H603-H615. [PMID: 29127236 DOI: 10.1152/ajpheart.00362.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pulmonary vascular disease (PVD) represents an underestimated and increasing clinical burden not only in the neonatal period but also later in life, when exercise tolerance is decreased. Animal models performing long-term followup after a perinatal insult are lacking. This study aimed to develop and characterize a neonatal swine model with hypoxia-induced PVD during long-term followup after reexposure to normoxia and to investigate the exercise response in this model. Piglets were exposed to a normoxic ( n = 10) or hypoxic environment ( n = 9) for 4 wk. Neonatal hypoxia exposure resulted in pulmonary hypertension. Mean pulmonary artery pressure was elevated 1 day after reexposure to normoxia (30.2 ± 3.3 vs. 14.3 ± 0.9 mmHg) and remained significantly higher in the second week (32.8 ± 3.8 vs. 21.4 ± 1.2 mmHg), accompanied by decreased exercise tolerance. Exercise resulted in a trend toward an exaggerated increase of pulmonary artery pressure in hypoxia-exposed animals ( week 6, P = 0.086). Although pulmonary hypertension was transient, thickening of pulmonary arterioles was found at the end of followup. Furthermore, right ventricular dilation, lower right ventricular fractional area change ( week 8, 40.0 ± 2.7% vs. 29.5 ± 4.7%), and tricuspid annular plane systolic excursion ( week 8, 27.0 ± 2.5 vs. 22.9 ± 2.1 mm) persisted during followup. Male animals showed more severe PVD than female animals. In conclusion, we developed a neonatal swine model that allows examination of the long-term sequelae of damage to the developing neonatal lung, the course of the disease and the effect of therapy on long-term outcome. NEW & NOTEWORTHY The swine model of neonatal pulmonary vascular disease developed in the present study is the first that allows exercise testing and examination of long-term sequelae of a perinatal hypoxic insult, the course of the disease, and the effect of therapy on long-term outcome.
Collapse
Affiliation(s)
- Daphne P M de Wijs-Meijler
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam , Rotterdam , The Netherlands.,Division of Neonatology, Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam , Rotterdam , The Netherlands
| | - Richard W B van Duin
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam , Rotterdam , The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam , Rotterdam , The Netherlands
| | - Urs Scherrer
- Departments of Cardiology and Clinical Research, University Hospital Bern, Bern, Switzerland, and Facultad de Ciencias, Departamento de Biología, Universidad de Tarapacá , Arica , Chile
| | - Claudio Sartori
- Department of Internal Medicine, Centre Hospitalier Universitaire Vaudois , Lausanne , Switzerland
| | - Irwin K M Reiss
- Division of Neonatology, Department of Pediatrics, Sophia Children's Hospital, Erasmus MC, University Medical Center Rotterdam , Rotterdam , The Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam , Rotterdam , The Netherlands
| |
Collapse
|
217
|
Oak P, Hilgendorff A. The BPD trio? Interaction of dysregulated PDGF, VEGF, and TGF signaling in neonatal chronic lung disease. Mol Cell Pediatr 2017; 4:11. [PMID: 29116547 PMCID: PMC5676585 DOI: 10.1186/s40348-017-0076-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 10/17/2017] [Indexed: 12/26/2022] Open
Abstract
The development of neonatal chronic lung disease (nCLD), i.e., bronchopulmonary dysplasia (BPD) in preterm infants, significantly determines long-term outcome in this patient population. Risk factors include mechanical ventilation and oxygen toxicity impacting on the immature lung resulting in impaired alveolarization and vascularization. Disease development is characterized by inflammation, extracellular matrix remodeling, and apoptosis, closely intertwined with the dysregulation of growth factor signaling. This review focuses on the causes and consequences of altered signaling in central pathways like transforming growth factor (TGF), platelet-derived growth factor (PDGF), and vascular endothelial growth factor (VEGF) driving these above indicated processes, i.e., inflammation, matrix remodeling, and vascular development. We emphasize the shared and distinct role of these pathways as well as their interconnection in disease initiation and progression, generating important knowledge for the development of future treatment strategies.
Collapse
Affiliation(s)
- Prajakta Oak
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, University Hospital of the University of Munich and Helmholtz Zentrum Muenchen, Munich, Germany.
- Department of Neonatology, Perinatal Center Grosshadern, Ludwig-Maximilians University, Munich, Germany.
- Center for Comprehensive Developmental Care, Dr. von Haunersches Children's Hospital University, Hospital Ludwig-Maximilians University, Munich, Germany.
| |
Collapse
|
218
|
Alvira CM, Morty RE. Can We Understand the Pathobiology of Bronchopulmonary Dysplasia? J Pediatr 2017; 190:27-37. [PMID: 29144252 PMCID: PMC5726414 DOI: 10.1016/j.jpeds.2017.08.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/28/2017] [Accepted: 08/16/2017] [Indexed: 01/17/2023]
Affiliation(s)
- Cristina M. Alvira
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California 94305
| | - Rory E. Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center campus of the German Center for Lung Research, Giessen, Germany,Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
219
|
Inocencio IM, Bischof RJ, Xiang SD, Zahra VA, Nguyen V, Lim T, LaRosa D, Barbuto J, Tolcos M, Plebanski M, Polglase GR, Moss TJ. Exacerbation of Ventilation-Induced Lung Injury and Inflammation in Preterm Lambs by High-Dose Nanoparticles. Sci Rep 2017; 7:14704. [PMID: 29089616 PMCID: PMC5665983 DOI: 10.1038/s41598-017-13113-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
Mechanical ventilation of preterm neonates causes lung inflammation and injury, with potential life-long consequences. Inert 50-nm polystyrene nanoparticles (PS50G) reduce allergic inflammation in the lungs of adult mice. We aimed to confirm the anti-inflammatory effects of PS50G in a sheep asthma model, and investigate the effects of prophylactic administration of PS50G on ventilation-induced lung injury (VILI) in preterm lambs. We assessed lung inflammatory cell infiltration, with and without PS50G, after airway allergen challenge in ewes sensitised to house dust mite. Preterm lambs (0.83 gestation) were delivered by caesarean section for immediate tissue collection (n = 5) or ventilation either with (n = 6) or without (n = 5) prophylactic intra-tracheal administration of PS50G nanoparticles (3% in 2 ml). Ventilation was continued for a total of 2 h before tissue collection for histological and biomolecular assessment of lung injury and inflammation. In ewes with experimental asthma, PS50G decreased eosinophilic infiltration of the lungs. Ventilated preterm lambs showed molecular and histological signs of lung injury and inflammation, which were exacerbated in lambs that received PSG50G. PS50G treatment decreased established inflammation in the lungs of asthmatic sheep. However, prophylactic administration of PSG50 exacerbated ventilation-induced lung injury and lung inflammation in preterm lambs.
Collapse
Affiliation(s)
- Ishmael M Inocencio
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.
| | - Robert J Bischof
- The Ritchie Centre, Hudson Institute of Medical Research, Biotechnology Research Laboratories, Department of Physiology, Monash University, Melbourne, Australia
| | - Sue D Xiang
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Alfred Hospital Campus, Monash University, Melbourne, Australia
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Vy Nguyen
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Tammy Lim
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Domenic LaRosa
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Jade Barbuto
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Mary Tolcos
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Alfred Hospital Campus, Monash University, Melbourne, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| | - Timothy J Moss
- The Ritchie Centre, Hudson Institute of Medical Research and Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
| |
Collapse
|
220
|
Ahmed E, Sansac C, Assou S, Gras D, Petit A, Vachier I, Chanez P, De Vos J, Bourdin A. Lung development, regeneration and plasticity: From disease physiopathology to drug design using induced pluripotent stem cells. Pharmacol Ther 2017; 183:58-77. [PMID: 28987320 DOI: 10.1016/j.pharmthera.2017.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lungs have a complex structure composed of different cell types that form approximately 17 million airway branches of gas-delivering bronchioles connected to 500 million gas-exchanging alveoli. Airways and alveoli are lined by epithelial cells that display a low rate of turnover at steady-state, but can regenerate the epithelium in response to injuries. Here, we review the key points of lung development, homeostasis and epithelial cell plasticity in response to injury and disease, because this knowledge is required to develop new lung disease treatments. Of note, canonical signaling pathways that are essential for proper lung development during embryogenesis are also involved in the pathophysiology of most chronic airway diseases. Moreover, the perfect control of these interconnected pathways is needed for the successful differentiation of induced pluripotent stem cells (iPSC) into lung cells. Indeed, differentiation of iPSC into airway epithelium and alveoli is based on the use of biomimetics of normal embryonic and fetal lung development. In vitro iPSC-based models of lung diseases can help us to better understand the impaired lung repair capacity and to identify new therapeutic targets and new approaches, such as lung cell therapy.
Collapse
Affiliation(s)
- Engi Ahmed
- Department of Respiratory Diseases, Hôpital Arnaud de Villeneuve, Montpellier F34000, France; CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France; INSERM, U1183, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France
| | - Caroline Sansac
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France
| | - Said Assou
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France; INSERM, U1183, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France
| | - Delphine Gras
- Dept of Respiratory Diseases APHM, INSERM CNRS U 1067, UMR7333, Aix-Marseille University, Marseille, France
| | - Aurélie Petit
- INSERM, U1046, PhyMedExp, Montpellier F34000, France
| | | | - Pascal Chanez
- Dept of Respiratory Diseases APHM, INSERM CNRS U 1067, UMR7333, Aix-Marseille University, Marseille, France
| | - John De Vos
- CHU Montpellier, Institute for Regenerative Medicine and Biotherapy, Hôpital Saint-Eloi, Montpellier F34000, France; INSERM, U1183, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France; CHU Montpellier, Unit for Cellular Therapy, Hospital Saint-Eloi, Montpellier F 34000, France.
| | - Arnaud Bourdin
- Department of Respiratory Diseases, Hôpital Arnaud de Villeneuve, Montpellier F34000, France; Université de MONTPELLIER, UFR de Médecine, Montpellier F34000, France; INSERM, U1046, PhyMedExp, Montpellier F34000, France.
| |
Collapse
|
221
|
Pan B, Xue X, Zhang D, Li M, Fu J. SOX4 arrests lung development in rats with hyperoxia‑induced bronchopulmonary dysplasia by controlling EZH2 expression. Int J Mol Med 2017; 40:1691-1698. [PMID: 29039454 PMCID: PMC5716405 DOI: 10.3892/ijmm.2017.3171] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 09/21/2017] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is currently the most common severe complication in premature infants and is characterized by the arrest of alveolar and vascular growth. Alveolar type II cells play an important role in the pathological foundation of BPD. An association of BPD with epithelial‑to‑mesenchymal transition (EMT) in type II cells exposed to hyperoxia was previously identified. SOX4, a transcription factor that is indispensable to embryogenesis, including lung development, participates in regulating EMT and cell survival, affecting tumorigenesis. The aim of the present study was to investigate the involvement of SOX4 in the occurrence of BPD, which, to the best of our knowledge, has not been previously determined. For this purpose, newborn rats were randomly divided into two treatment groups: The model group was exposed to hyperoxia (80-85% O2), while the control group was kept under normoxic conditions (21% O2). Lung tissues were collected on postnatal days 1, 3, 7, 14 and 21 and morphological changes in the lungs were examined by hematoxylin and eosin staining. The location of SOX4 in type II cells was detected by double immunofluorescence. The expression of SOX4 and enhancer of zeste homolog 2 (EZH2) in type II cells and lung tissues were detected by immunochemistry, western blotting and quantitative polymerase chain reaction analysis. The results demonstrated that, compared with the control group, the radial alveolar count decreased rapidly in the model group, accompanied by increased mean alveolar diameter and alveolar septal thickness. SOX4 and EZH2 were highly expressed in type II cells exposed to hyperoxia. However, in total lung tissues, SOX4 and EZH2 expression was profoundly decreased in the early stages and increased in the late stages following exposure to hyperoxia. The expression of the EZH2 protein was positively correlated with that of the SOX4 protein. In conclusion, at the alveolar stage, which is a critical period after birth for lung development, hyperoxia induced dysregulation of SOX4 and EZH2 in rat lungs, indicating that SOX4 may contribute to the disruption of lung development in BPD by regulating EZH2 expression.
Collapse
Affiliation(s)
- Bingting Pan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xindong Xue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Dan Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Mengyun Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
222
|
Rath P, Nardiello C, Morty RE. A new target for caffeine in the developing lung: endoplasmic reticulum stress? Am J Physiol Lung Cell Mol Physiol 2017; 313:L659-L663. [DOI: 10.1152/ajplung.00251.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/06/2017] [Accepted: 07/11/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Philipp Rath
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
| | - Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E. Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
223
|
|
224
|
Diagnostic Approach to Pulmonary Hypertension in Premature Neonates. CHILDREN-BASEL 2017; 4:children4090075. [PMID: 28837121 PMCID: PMC5615265 DOI: 10.3390/children4090075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/01/2017] [Accepted: 08/09/2017] [Indexed: 02/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a form of chronic lung disease in premature infants following respiratory distress at birth. With increasing survival of extremely low birth weight infants, alveolar simplification is the defining lung characteristic of infants with BPD, and along with pulmonary hypertension, increasingly contributes to both respiratory morbidity and mortality in these infants. Growth restricted infants, infants born to mothers with oligohydramnios or following prolonged preterm rupture of membranes are at particular risk for early onset pulmonary hypertension. Altered vascular and alveolar growth particularly in canalicular and early saccular stages of lung development following mechanical ventilation and oxygen therapy, results in developmental lung arrest leading to BPD with pulmonary hypertension (PH). Early recognition of PH in infants with risk factors is important for optimal management of these infants. Screening tools for early diagnosis of PH are evolving; however, echocardiography is the mainstay for non-invasive diagnosis of PH in infants. Cardiac computed tomography (CT) and magnetic resonance are being used as imaging modalities, however their role in improving outcomes in these patients is uncertain. Follow-up of infants at risk for PH will help not only in early diagnosis, but also in appropriate management of these infants. Aggressive management of lung disease, avoidance of hypoxemic episodes, and optimal nutrition determine the progression of PH, as epigenetic factors may have significant effects, particularly in growth-restricted infants. Infants with diagnosis of PH are managed with pulmonary vasodilators and those resistant to therapy need to be worked up for the presence of cardio-vascular anomalies. The management of infants and toddlers with PH, especially following premature birth is an emerging field. Nonetheless, combination therapies in a multi-disciplinary setting improves outcomes for these infants.
Collapse
|
225
|
Lio A, Rosati P, Pastorino R, Cota F, Tana M, Tirone C, Aurilia C, Ricci C, Gambacorta A, Paladini A, Mappa I, Buongiorno S, Zannoni GF, Romagnoli C, Vento G. Fetal Doppler velocimetry and bronchopulmonary dysplasia risk among growth-restricted preterm infants: an observational study. BMJ Open 2017; 7:e015232. [PMID: 28729313 PMCID: PMC5541508 DOI: 10.1136/bmjopen-2016-015232] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To investigate whether fetal growth restriction (FGR) diagnosis, based on pathological prenatal fetal Doppler velocimetry, is associated with bronchopulmonary dysplasia (BPD) independently of being small for gestational age (SGA) per se at birth among very preterm infants. DESIGN Prospective, observational study. FGR was defined as failing fetal growth in utero and fetal Doppler velocimetry abnormalities. SETTING Policlinico Universitario Agostino Gemelli, Roma, Italy. PATIENTS Preterm newborns with gestational age ≤30 weeks and birth weight (BW) ≤1250 g. MAIN OUTCOME MEASURES Bronchopulmonary dysplasia. RESULTS In the study period, 178 newborns were eligible for the study. Thirty-nine infants (22%) were considered fetal growth-restricted infants. Among the 154 survived babies at 36 weeks postmenstrual age, 12 out of 36 (33%) of the FGR group developed BPD versus 8 out of 118 (7%) of the NO-FGR group (p<0.001). BPD rate was sixfold higher among the SGA-FGR infants compared with the SGA-NO-FGR infants. In a multivariable model, FGR was significantly associated with BPD risk (OR 5.1, CI 1.4 to 18.8, p=0.01), independently from BW z-score that still remains a strong risk factor (OR 0.5, CI 0.3 to 0.9, p=0.01). CONCLUSION Among SGA preterm infants, BPD risk dramatically increases when placenta dysfunction is the surrounding cause of low BW. Antenatal fetal Doppler surveillance could be a useful tool for studying placenta wellness and predicting BPD risk among preterm babies. Further research is needed to better understand how FGR affects lung development.
Collapse
Affiliation(s)
- Alessandra Lio
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paolo Rosati
- Department of Obstetrics and Gynecology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Pastorino
- Section of Hygiene, Institute of Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Cota
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Milena Tana
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Chiara Tirone
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudia Aurilia
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Cinzia Ricci
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Gambacorta
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Angela Paladini
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilenia Mappa
- Department of Obstetrics and Gynecology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Silvia Buongiorno
- Department of Obstetrics and Gynecology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gian Franco Zannoni
- Department of Pathology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Costantino Romagnoli
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Vento
- Division of Neonatology, Fondazione Policlinico Universitario A. Gemelli—Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
226
|
Carlton EF, Sontag MK, Younoszai A, DiMaria MV, Miller JI, Poindexter BB, Abman SH, Mourani PM. Reliability of Echocardiographic Indicators of Pulmonary Vascular Disease in Preterm Infants at Risk for Bronchopulmonary Dysplasia. J Pediatr 2017; 186:29-33. [PMID: 28411949 PMCID: PMC5493442 DOI: 10.1016/j.jpeds.2017.03.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 02/06/2017] [Accepted: 03/06/2017] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To determine the assessment and inter-rater reliability of echocardiographic evaluations of pulmonary vascular disease (PVD) in preterm infants at risk for bronchopulmonary dysplasia. STUDY DESIGN We prospectively studied echocardiograms from preterm infants (birthweights 500-1250 g) at 7 days of age and 36 weeks postmenstrual age (PMA). Echocardiograms were assessed by both a cardiologist on clinical service and a single research cardiologist. Interpretations were reviewed for inclusion of determinants of PVD and assessed for inter-rater reliability using the Prevalence Adjusted Bias Adjusted Kappa Score (PABAK). RESULTS One hundred eighty and 188 matching research and clinical echocardiogram reports were available for the 7-day and 36-week PMA studies. At least one of the specific qualitative measures of PVD was missing from 54% of the clinical reports. PVD was diagnosed at 7 days in 31% and 20% of research and clinical interpretations, respectively (PABAK score of 0.54). At 36 weeks, PH was diagnosed in 15.6% and 17.8% of research and clinical interpretations, respectively (PABAK score of 0.80). CONCLUSIONS Although all qualitative variables of PVD are not consistently provided in echocardiogram reports, the inter-rater reliability of cardiologists evaluating measures of PVD revealed strong agreement, especially at 36 weeks PMA. We speculate that establishment of a protocol for echocardiographic evaluation may improve the identification of PVD in preterm infants.
Collapse
Affiliation(s)
- Erin F Carlton
- Section of Critical Care, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO.
| | - Marci K Sontag
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO
| | - Adel Younoszai
- Section of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Michael V DiMaria
- Section of Cardiology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Joshua I Miller
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO
| | - Brenda B Poindexter
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Steven H Abman
- Section of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO; The Pediatric Heart-Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Peter M Mourani
- Section of Critical Care, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO; The Pediatric Heart-Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
227
|
Barrette AM, Roberts JK, Chapin C, Egan EA, Segal MR, Oses-Prieto JA, Chand S, Burlingame AL, Ballard PL. Antiinflammatory Effects of Budesonide in Human Fetal Lung. Am J Respir Cell Mol Biol 2017; 55:623-632. [PMID: 27281349 DOI: 10.1165/rcmb.2016-0068oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Lung inflammation in premature infants contributes to the development of bronchopulmonary dysplasia (BPD), a chronic lung disease with long-term sequelae. Pilot studies administering budesonide suspended in surfactant have found reduced BPD without the apparent adverse effects that occur with systemic dexamethasone therapy. Our objective was to determine budesonide potency, stability, and antiinflammatory effects in human fetal lung. We cultured explants of second-trimester fetal lung with budesonide or dexamethasone and used microscopy, immunoassays, RNA sequencing, liquid chromatography/tandem mass spectrometry, and pulsating bubble surfactometry. Budesonide suppressed secreted chemokines IL-8 and CCL2 (MCP-1) within 4 hours, reaching a 90% decrease at 12 hours, which was fully reversed 72 hours after removal of the steroid. Half-maximal effects occurred at 0.04-0.05 nM, representing a fivefold greater potency than for dexamethasone. Budesonide significantly induced 3.6% and repressed 2.8% of 14,500 sequenced mRNAs by 1.6- to 95-fold, including 119 genes that contribute to the glucocorticoid inflammatory transcriptome; some are known targets of nuclear factor-κB. By global proteomics, 22 secreted inflammatory proteins were hormonally regulated. Two glucocorticoid-regulated genes of interest because of their association with lung disease are CHI3L1 and IL1RL1. Budesonide retained activity in the presence of surfactant and did not alter its surface properties. There was some formation of palmitate-budesonide in lung tissue but no detectable metabolism to inactive 16α-hydroxy prednisolone. We concluded that budesonide is a potent and stable antiinflammatory glucocorticoid in human fetal lung in vitro, supporting a beneficial antiinflammatory response to lung-targeted budesonide:surfactant treatment of infants for the prevention of BPD.
Collapse
Affiliation(s)
| | - Jessica K Roberts
- 2 Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, Salt Lake City, Utah; and
| | | | - Edmund A Egan
- 3 Department of Pediatrics, University of Buffalo, Buffalo, New York
| | | | - Juan A Oses-Prieto
- 5 Chemistry & Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Shreya Chand
- 5 Chemistry & Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Alma L Burlingame
- 5 Chemistry & Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | | |
Collapse
|
228
|
Reiterer F, Schwaberger B, Freidl T, Schmölzer G, Pichler G, Urlesberger B. Lung-protective ventilatory strategies in intubated preterm neonates with RDS. Paediatr Respir Rev 2017; 23:89-96. [PMID: 27876355 DOI: 10.1016/j.prrv.2016.10.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022]
Abstract
This article provides a narrative review of lung-protective ventilatory strategies (LPVS) in intubated preterm infants with RDS. A description of strategies is followed by results on short-and long-term respiratory and neurodevelopmental outcomes. Strategies will include patient-triggered or synchronized ventilation, volume targeted ventilation, the technique of intubation, surfactant administration and rapid extubation to NCPAP (INSURE), the open lung concept, strategies of high-frequency ventilation, and permissive hypercapnia. Based on this review single recommendations on optimal LPVS cannot be made. Combinations of several strategies, individually applied, most probably minimize or avoid potential serious respiratory and cerebral complications like bronchopulmonary dysplasia and cerebral palsy.
Collapse
Affiliation(s)
- F Reiterer
- Division of Neonatology, Department of Pediatrics and Adolescence Medicine, Medical University Graz, Austria.
| | - B Schwaberger
- Division of Neonatology, Department of Pediatrics and Adolescence Medicine, Medical University Graz, Austria
| | - T Freidl
- Division of Neonatology, Department of Pediatrics and Adolescence Medicine, Medical University Graz, Austria
| | - G Schmölzer
- Centre for the Studies of Asphyxia and Resuscitation, Neonatal Research Unit, Royal Alexandra Hospital, Edmonton, Canada; Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - G Pichler
- Division of Neonatology, Department of Pediatrics and Adolescence Medicine, Medical University Graz, Austria
| | - B Urlesberger
- Division of Neonatology, Department of Pediatrics and Adolescence Medicine, Medical University Graz, Austria
| |
Collapse
|
229
|
Nuyt AM, Lavoie JC, Mohamed I, Paquette K, Luu TM. Adult Consequences of Extremely Preterm Birth: Cardiovascular and Metabolic Diseases Risk Factors, Mechanisms, and Prevention Avenues. Clin Perinatol 2017; 44:315-332. [PMID: 28477663 DOI: 10.1016/j.clp.2017.01.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Extremely preterm babies are exposed to various sources of injury during critical stages of development. The extremely preterm infant faces premature transition to ex utero physiology and undergoes adaptive mechanisms that may be deleterious in the long term because of permanent alterations in organ structure and function. Perinatal events can also directly cause structural injury. These disturbances induce morphologic and functional changes in their organ systems that might heighten their risks for later adult chronic diseases. This review examines the pathophysiology of programming of long-term health and diseases after preterm birth and associated perinatal risk factors.
Collapse
Affiliation(s)
- Anne Monique Nuyt
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada.
| | - Jean-Claude Lavoie
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada; Department of Nutrition, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| | - Ibrahim Mohamed
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| | - Katryn Paquette
- Division of Neonatology, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| | - Thuy Mai Luu
- Division of General Pediatrics, Department of Pediatrics, Faculty of Medicine, Research Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, 3175 chemin de la Côte-Sainte-Catherine, Montreal, Quebec H3T 1C5, Canada
| |
Collapse
|
230
|
Altit G, Dancea A, Renaud C, Perreault T, Lands LC, Sant'Anna G. Pathophysiology, screening and diagnosis of pulmonary hypertension in infants with bronchopulmonary dysplasia - A review of the literature. Paediatr Respir Rev 2017; 23:16-26. [PMID: 27986502 DOI: 10.1016/j.prrv.2016.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/02/2016] [Indexed: 10/20/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication of extreme prematurity, which has increased over the last 20 years. BPD is associated with increased morbidities and mortality. It has been increasingly recognized that BPD affects overall lung development including the pulmonary vasculature. More recent studies have demonstrated an increased awareness of pulmonary arterial hypertension (PH) in BPD patients and recent international guidelines have advocated for better screening. This review will describe the current understanding of the pathophysiology of PH in infants with BPD, the in-depth assessment of the available literature linking PH and BPD, and propose an approach of screening and diagnosis of PH in infants with BPD.
Collapse
|
231
|
Kim HR, Kim JY, Yun BL, Lee B, Choi CW, Kim BI. Interstitial pneumonia pattern on day 7 chest radiograph predicts bronchopulmonary dysplasia in preterm infants. BMC Pediatr 2017; 17:125. [PMID: 28506211 PMCID: PMC5433188 DOI: 10.1186/s12887-017-0881-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 05/08/2017] [Indexed: 12/01/2022] Open
Abstract
Background Early identification of infants at higher risk of developing bronchopulmonary dysplasia (BPD) may enable a targeted approach to reduce BPD. We aimed to evaluate the hypothesis that the interstitial pneumonia pattern on the day 7 chest radiograph predicts BPD or death before 36 weeks postmenstrual age (PMA). Methods A retrospective cohort study was performed on 336 preterm infants (birth weight < 1500 g and gestational age < 32 postmenstrual weeks) who were admitted to a single tertiary academic center between January 2008 and December 2014. Day 7 chest radiographs were independently reviewed by two pediatric radiologists who were unaware of the clinical information regarding each individual infant. Results Data from 304 infants who survived more than 7 days after birth were collected. The interstitial pneumonia pattern on the day 7 chest radiograph was independently associated with BPD or death before 36 weeks PMA (odds ratio [OR] 4.0, 95% confidence interval [CI] 1.1–14.4). The interstitial pneumonia pattern on the day 7 chest radiograph predicted BPD or death with a specificity of 98%. Histologic chorioamnionitis was a preceding factor that was independently associated with the interstitial pneumonia pattern on the day 7 chest radiograph (OR 3.7, 95% CI 1.3–10.3). Conclusions The interstitial pneumonia pattern on the day 7 chest radiograph has a high specificity for predicting BPD or death and can be utilized to select high-risk preterm infants who will benefit from potentially preventive interventions against BPD. Electronic supplementary material The online version of this article (doi:10.1186/s12887-017-0881-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hye-Rim Kim
- Department of Pediatrics, Bundang CHA Medical Center, CHA University, Seongnam, Republic of Korea.,Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Pediatrics, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Ji Young Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Bo La Yun
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Byoungkook Lee
- Department of Pediatrics, Wonju Severance Christian Hospital, Wonju, Republic of Korea
| | - Chang Won Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Department of Pediatrics, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea.
| | - Beyong Il Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Pediatrics, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea
| |
Collapse
|
232
|
Nutritional approach to preterm infants on noninvasive ventilation: An update. Nutrition 2017; 37:14-17. [DOI: 10.1016/j.nut.2016.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/29/2016] [Accepted: 12/17/2016] [Indexed: 11/18/2022]
|
233
|
Bokiniec R, Własienko P, Borszewska-Kornacka M, Szymkiewicz-Dangel J. Evaluation of left ventricular function in preterm infants with bronchopulmonary dysplasia using various echocardiographic techniques. Echocardiography 2017; 34:567-576. [DOI: 10.1111/echo.13488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Renata Bokiniec
- Neonatal and Intensive Care Department; Medical University of Warsaw; Warsaw Poland
| | - Paweł Własienko
- Perinatal Cardiology Unit; Medical University of Warsaw; Warsaw Poland
| | | | | |
Collapse
|
234
|
Mills DR, Mao Q, Chu S, Falcon Girard K, Kraus M, Padbury JF, De Paepe ME. Effects of human umbilical cord blood mononuclear cells on respiratory system mechanics in a murine model of neonatal lung injury. Exp Lung Res 2017; 43:66-81. [PMID: 28353351 DOI: 10.1080/01902148.2017.1300713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mononuclear cells (MNCs) have well-documented beneficial effects in a wide range of adult pulmonary diseases. The effects of human umbilical cord blood-derived MNCs on neonatal lung injury, highly relevant for potential autologous application in preterm newborns at risk for bronchopulmonary dysplasia (BPD), remain incompletely established. The aim of this study was to determine the long-term morphologic and functional effects of systemically delivered MNCs in a murine model of neonatal lung injury. MATERIALS AND METHODS MNCs from cryopreserved cord blood (1 × 106 cells per pup) were given intravenously to newborn mice exposed to 90% O2 from birth; controls received cord blood total nucleated cells (TNCs) or granular cells, or equal volume vehicle buffer (sham controls). In order to avoid immune rejection, we used SCID mice as recipients. Lung mechanics (flexiVent™), engraftment, growth, and alveolarization were evaluated eight weeks postinfusion. RESULTS Systemic MNC administration to hyperoxia-exposed newborn mice resulted in significant attenuation of methacholine-induced airway hyperreactivity, leading to reduction of central airway resistance to normoxic levels. These bronchial effects were associated with mild improvement of alveolarization, lung compliance, and elastance. TNCs had no effects on alveolar remodeling and were associated with worsened methacholine-induced bronchial hyperreactivity. Granular cell administration resulted in a marked morphologic and functional emphysematous phenotype, associated with high mortality. Pulmonary donor cell engraftment was sporadic in all groups. CONCLUSIONS These results suggest that cord blood MNCs may have a cell type-specific role in therapy of pulmonary conditions characterized by increased airway resistance, such as BPD and asthma. Future studies need to determine the active MNC subtype(s), their mechanisms of action, and optimal purification methods to minimize granular cell contamination.
Collapse
Affiliation(s)
- David R Mills
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA
| | - Quanfu Mao
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA.,b Department of Pathology and Laboratory Medicine , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| | - Sharon Chu
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA.,b Department of Pathology and Laboratory Medicine , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| | | | - Morey Kraus
- c ViaCord LLC, a Perkin Elmer Company , Cambridge , Massachusetts , USA
| | - James F Padbury
- d Department of Pediatrics , Women and Infants Hospital , Providence , Rhode Island , USA.,e Department of Pediatrics , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| | - Monique E De Paepe
- a Department of Pathology , Women and Infants Hospital , Providence , Rhode Island , USA.,b Department of Pathology and Laboratory Medicine , Alpert Medical School of Brown University , Providence , Rhode Island , USA
| |
Collapse
|
235
|
Lau CSM, Chamberlain RS, Sun S. Less Invasive Surfactant Administration Reduces the Need for Mechanical Ventilation in Preterm Infants: A Meta-Analysis. Glob Pediatr Health 2017; 4:2333794X17696683. [PMID: 28540346 PMCID: PMC5433666 DOI: 10.1177/2333794x17696683] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 12/29/2016] [Indexed: 11/30/2022] Open
Abstract
Neonatal respiratory distress syndrome due to surfactant deficiency is associated with high morbidity and mortality in preterm infants, and the use of less invasive surfactant administration (LISA) has been increasingly studied. This meta-analysis found that LISA via thin catheter significantly reduced the need for mechanical ventilation within the first 72 hours (relative risk [RR] = 0.677; P = .021), duration of mechanical ventilation (difference in means [MD] = −39.302 hours; P < .001), duration of supplemental oxygen (MD = −68.874 hours; P < .001), and duration of nasal continuous positive airway pressure (nCPAP; MD = −28.423 hours; P = .010). A trend toward a reduction in the incidence of bronchopulmonary dysplasia was observed (RR = 0.656; P = .141). No significant difference in overall mortality, incidence of pneumothorax, or successful first attempts was observed. LISA via thin catheter significantly reduces the need for mechanical ventilation within the first 72 hours as well as the duration of mechanical ventilation, supplemental oxygen, and nCPAP. LISA via thin catheter appears promising in improving preterm infant outcomes.
Collapse
Affiliation(s)
- Christine S M Lau
- Saint Barnabas Medical Center, Livingston, NJ, USA.,Saint George's University, Grenada, West Indies
| | - Ronald S Chamberlain
- Saint Barnabas Medical Center, Livingston, NJ, USA.,Saint George's University, Grenada, West Indies.,New Jersey Medical School, Rutgers University, Newark, NJ, USA.,Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Shyan Sun
- Saint Barnabas Medical Center, Livingston, NJ, USA.,New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
236
|
Fehrholz M, Glaser K, Speer CP, Seidenspinner S, Ottensmeier B, Kunzmann S. Caffeine modulates glucocorticoid-induced expression of CTGF in lung epithelial cells and fibroblasts. Respir Res 2017; 18:51. [PMID: 28330503 PMCID: PMC5363056 DOI: 10.1186/s12931-017-0535-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/10/2017] [Indexed: 12/19/2022] Open
Abstract
Background Although caffeine and glucocorticoids are frequently used to treat chronic lung disease in preterm neonates, potential interactions are largely unknown. While anti-inflammatory effects of glucocorticoids are well defined, their impact on airway remodeling is less characterized. Caffeine has been ascribed to positive effects on airway inflammation as well as remodeling. Connective tissue growth factor (CTGF, CCN2) plays a key role in airway remodeling and has been implicated in the pathogenesis of chronic lung diseases such as bronchopulmonary dysplasia (BPD) in preterm infants. The current study addressed the impact of glucocorticoids on the regulation of CTGF in the presence of caffeine using human lung epithelial and fibroblast cells. Methods The human airway epithelial cell line H441 and the fetal lung fibroblast strain IMR-90 were exposed to different glucocorticoids (dexamethasone, budesonide, betamethasone, prednisolone, hydrocortisone) and caffeine. mRNA and protein expression of CTGF, TGF-β1-3, and TNF-α were determined by means of quantitative real-time PCR and immunoblotting. H441 cells were additionally treated with cAMP, the adenylyl cyclase activator forskolin, and the selective phosphodiesterase (PDE)-4 inhibitor cilomilast to mimic caffeine-mediated PDE inhibition. Results Treatment with different glucocorticoids (1 μM) significantly increased CTGF mRNA levels in H441 (p < 0.0001) and IMR-90 cells (p < 0.01). Upon simultaneous exposure to caffeine (10 mM), both glucocorticoid-induced mRNA and protein expression were significantly reduced in IMR-90 cells (p < 0.0001). Of note, 24 h exposure to caffeine alone significantly suppressed basal expression of CTGF mRNA and protein in IMR-90 cells. Caffeine-induced reduction of CTGF mRNA expression seemed to be independent of cAMP levels, adenylyl cyclase activation, or PDE-4 inhibition. While dexamethasone or caffeine treatment did not affect TGF-β1 mRNA in H441 cells, increased expression of TGF-β2 and TGF-β3 mRNA was detected upon exposure to dexamethasone or dexamethasone and caffeine, respectively. Moreover, caffeine increased TNF-α mRNA in H441 cells (6.5 ± 2.2-fold, p < 0.05) which has been described as potent inhibitor of CTGF expression. Conclusions In addition to well-known anti-inflammatory features, glucocorticoids may have adverse effects on long-term remodeling by TGF-β1-independent induction of CTGF in lung cells. Simultaneous treatment with caffeine may attenuate glucocorticoid-induced expression of CTGF, thereby promoting restoration of lung homeostasis.
Collapse
Affiliation(s)
- Markus Fehrholz
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany.
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Silvia Seidenspinner
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Barbara Ottensmeier
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Steffen Kunzmann
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany.,Clinic of Neonatology, Buergerhospital Frankfurt am Main, Nibelungenallee 37-41, 60318, Frankfurt am Main, Germany
| |
Collapse
|
237
|
Pozarska A, Rodríguez-Castillo JA, Surate Solaligue DE, Ntokou A, Rath P, Mižíková I, Madurga A, Mayer K, Vadász I, Herold S, Ahlbrecht K, Seeger W, Morty RE. Stereological monitoring of mouse lung alveolarization from the early postnatal period to adulthood. Am J Physiol Lung Cell Mol Physiol 2017; 312:L882-L895. [PMID: 28314804 DOI: 10.1152/ajplung.00492.2016] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/06/2017] [Accepted: 03/08/2017] [Indexed: 11/22/2022] Open
Abstract
Postnatal lung maturation generates a large number of small alveoli, with concomitant thinning of alveolar septal walls, generating a large gas exchange surface area but minimizing the distance traversed by the gases. This demand for a large and thin gas exchange surface area is not met in disorders of lung development, such as bronchopulmonary dysplasia (BPD) histopathologically characterized by fewer, larger alveoli and thickened alveolar septal walls. Diseases such as BPD are often modeled in the laboratory mouse to better understand disease pathogenesis or to develop new interventional approaches. To date, there have been no stereology-based longitudinal studies on postnatal mouse lung development that report dynamic changes in alveoli number or alveolar septal wall thickness during lung maturation. To this end, changes in lung structure were quantified over the first 22 mo of postnatal life of C57BL/6J mice. Alveolar density peaked at postnatal day (P)39 and remained unchanged at 9 mo (P274) but was reduced by 22 mo (P669). Alveoli continued to be generated, initially at an accelerated rate between P5 and P14, and at a slower rate thereafter. Between P274 and P669, loss of alveoli was noted, without any reduction in lung volume. A progressive thinning of the alveolar septal wall was noted between P5 and P28. Pronounced sex differences were observed in alveoli number in adult (but not juvenile) mice, when comparing male and female mouse lungs. This sex difference was attributed exclusively to the larger volume of male mouse lungs.
Collapse
Affiliation(s)
- Agnieszka Pozarska
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Aglaia Ntokou
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Philipp Rath
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and
| | - Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Alicia Madurga
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Konstantin Mayer
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - István Vadász
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Susanne Herold
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Werner Seeger
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
238
|
Matsumura H, Ichiba H, Ohnishi S, Saito M, Shintaku H. Histologic Chorioamnionitis, Amniotic Fluid Interleukin 6, Krebs von den Lungen 6, and Transforming Growth Factor β 1 for the Development of Neonatal Bronchopulmonary Dysplasia. JAPANESE CLINICAL MEDICINE 2017; 8:1179066017696076. [PMID: 28469527 PMCID: PMC5398658 DOI: 10.1177/1179066017696076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 02/01/2017] [Indexed: 01/24/2023]
Abstract
Background: Chorioamnionitis (CAM) is an important risk factor for the development of bronchopulmonary dysplasia (BPD) in preterm infants. Objectives: To evaluate the effects of CAM on the development of BPD using interleukin 6 (IL-6), Krebs von den Lungen 6 (KL-6), and transforming growth factor β1 (TGF-β1) in the amniotic fluid as markers for inflammation, lung injury, and fibrosis/remodeling, respectively. Methods: Amniotic fluid concentrations of IL-6, KL-6, and TGF-β1 were measured with enzyme-linked immunosorbent assay or electrochemiluminescence immunoassay. Results: Of the 36 preterm infants, 18 were exposed to histologically confirmed CAM. Of these, 12 were later diagnosed as having BPD. The IL-6, KL-6, and TGF-β1 levels in the amniotic fluid significantly increased with increasing histologic severity of CAM. Moreover, these markers were higher in the BPD group with histologic CAM than those without. Conclusions: Our study suggests that CAM is likely to induce inflammatory, injury, and remodeling processes in the fetal lung.
Collapse
Affiliation(s)
- Hisako Matsumura
- Department of Neonatology, Osaka City General Hospital, Osaka, Japan
| | - Hiroyuki Ichiba
- Department of Neonatology, Osaka City General Hospital, Osaka, Japan
| | - Satoshi Ohnishi
- Department of Neonatology, Osaka City General Hospital, Osaka, Japan
| | - Mika Saito
- Department of Pediatrics, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
239
|
Abstract
Neonates with chronic respiratory failure have uncertain prognosis and can face significant treatment burden. As the trajectory of the illness becomes more concerning, consultation with a pediatric palliative service should be considered, especially as therapeutic options shift from standard to "innovative." Benefits include as follows: supporting emotionally conflicted providers and parents, maintaining transparency in determination of goals, and balancing medical progress with each individual patient's and family's best interests.
Collapse
Affiliation(s)
- Nicolas F M Porta
- Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, 225 E. Chicago Ave, Box #45, Chicago, IL 60611.
| |
Collapse
|
240
|
de Wijs-Meijler DP, Duncker DJ, Tibboel D, Schermuly RT, Weissmann N, Merkus D, Reiss IK. Oxidative injury of the pulmonary circulation in the perinatal period: Short- and long-term consequences for the human cardiopulmonary system. Pulm Circ 2017; 7:55-66. [PMID: 28680565 PMCID: PMC5448552 DOI: 10.1086/689748] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/22/2016] [Indexed: 01/09/2023] Open
Abstract
Development of the pulmonary circulation is a complex process with a spatial pattern that is tightly controlled. This process is vulnerable for disruption by various events in the prenatal and early postnatal periods. Disruption of normal pulmonary vascular development leads to abnormal structure and function of the lung vasculature, causing neonatal pulmonary vascular diseases. Premature babies are especially at risk of the development of these diseases, including persistent pulmonary hypertension and bronchopulmonary dysplasia. Reactive oxygen species play a key role in the pathogenesis of neonatal pulmonary vascular diseases and can be caused by hyperoxia, mechanical ventilation, hypoxia, and inflammation. Besides the well-established short-term consequences, exposure of the developing lung to injurious stimuli in the perinatal period, including oxidative stress, may also contribute to the development of pulmonary vascular diseases later in life, through so-called "fetal or perinatal programming." Because of these long-term consequences, it is important to develop a follow-up program tailored to adolescent survivors of neonatal pulmonary vascular diseases, aimed at early detection of adult pulmonary vascular diseases, and thereby opening the possibility of early intervention and interfering with disease progression. This review focuses on pathophysiologic events in the perinatal period that have been shown to disrupt human normal pulmonary vascular development, leading to neonatal pulmonary vascular diseases that can extend even into adulthood. This knowledge may be particularly important for ex-premature adults who are at risk of the long-term consequences of pulmonary vascular diseases, thereby contributing disproportionately to the burden of adult cardiovascular disease in the future.
Collapse
Affiliation(s)
- Daphne P. de Wijs-Meijler
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Division of Neonatology, Department of Pediatrics, Sophia Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J. Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dick Tibboel
- Intensive Care Unit, Department of Pediatric Surgery, Sophia Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ralph T. Schermuly
- University of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Department of Internal Medicine, Members of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Norbert Weissmann
- University of Giessen and Marburg Lung Center (UGMLC), Excellence Cluster Cardio-Pulmonary Systems (ECCPS), Department of Internal Medicine, Members of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Irwin K.M. Reiss
- Division of Neonatology, Department of Pediatrics, Sophia Children’s Hospital, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
241
|
Klotz D, Schaefer C, Stavropoulou D, Fuchs H, Schumann S. Leakage in nasal high-frequency oscillatory ventilation improves carbon dioxide clearance-A bench study. Pediatr Pulmonol 2017; 52:367-372. [PMID: 27526104 DOI: 10.1002/ppul.23534] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/13/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Nasal high frequency oscillatory ventilation (nHFOV) is a promising mode of non-invasive neonatal respiratory support. To combine the effects of nasal continuous positive airway pressure (nCPAP) and high frequency oscillatory ventilation, an oscillatory pressure waveform is superposed to a nCPAP via a nasal or nasopharyngeal interface. nHFOV has been described to facilitate carbon dioxide (CO2 ) elimination compared to nCPAP. The influence of unintended leakage on CO2 elimination has not been investigated in nHFOV before. We explored the effects of oral leakage on CO2 elimination during nHFOV in a physical model of the neonatal respiratory system. METHODS A neonatal ventilator was connected to an airway- and lung model using binasal prongs as interface. The model comprised a continuous CO2 influx. Alveolar CO2 partial pressure was continuously measured. Gas flow rates and pressures were measured simultaneously at the prongs, pharynx, lung, and at the leakage. Effects of combined nasopharyngeal leakage (0, 5, or 10 L/min) on CO2 elimination, gas flow rate and pressure were determined at various ventilation frequencies (6, 8, 10, and 12 Hz) and amplitudes (10%, 20%, and 30% of maximum ventilator performance) at a mean airway pressure of 10 cmH2 O. RESULTS nHFOV with moderate leakage was more effective in CO2 elimination than without leakage (P < 0.001) for all tested amplitudes and frequencies. Maximum leakage resulted in highly variable, partly ineffective CO2 elimination. CONCLUSIONS A moderate oral leakage rather improves than impairs gas exchange during non-invasive ventilatory support with nHFOV. Pediatr Pulmonol. 2017;52:367-372. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniel Klotz
- Department of Neonatology, Center for Pediatrics, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Schaefer
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Anesthesiology and Critical Care, Medical Center-University of Freiburg, Freiburg, Germany
| | - Dimitra Stavropoulou
- Department of Neonatology, Center for Pediatrics, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans Fuchs
- Department of Neonatology, Center for Pediatrics, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Schumann
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Anesthesiology and Critical Care, Medical Center-University of Freiburg, Freiburg, Germany
| |
Collapse
|
242
|
Durlak W, Klimek M, Kwinta P. Regional lung ventilation pattern in preschool children with bronchopulmonary dysplasia is modified by bronchodilator response. Pediatr Pulmonol 2017; 52:353-359. [PMID: 28221733 DOI: 10.1002/ppul.23540] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 07/11/2016] [Accepted: 07/14/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) remains a significant long-term complication of prematurity. A standardized method of pulmonary function testing is still not available in preschool children with BPD. We investigated the feasibility of Electrical Impedance Segmentography (EIS) monitoring in this group and the impact of bronchodilator response (BDR) to salbutamol on the pattern of lung ventilation. METHODS We conducted a follow-up study of 4-year-old premature children who had been treated in the tertiary NICU. The cohort was divided into two groups based on the presence of BPD. EIS monitoring was performed before and 15 min after the administration of 400 µg of salbutamol (pMDI with spacer) in all subjects during spontaneous tidal breathing in upright position. Data were expressed as median segmental impedance amplitude differences and segmental ventilation inhomogeneity index (II) changes. RESULTS We included 51 children in our analysis: 33 with BPD (median birth weight-840 g; median gestational age-27 weeks) and 18 without BPD (1,290 g; 30 weeks, respectively). There was a significant increase in median segmental impedance amplitude after salbutamol in gravity non-dependent segments in children with BPD: upper left (UL): 462 versus 534 AU; (P = 0.003); upper right (UR): 481 versus 595 AU (P < 0.001) and II in these segments: UL: 0.046 versus 0.078 (P = 0.003) UR: 0.049 versus 0.064 (P = 0.006). There were no changes in the lower segments. There were no changes in ventilation pattern in children without BPD. CONCLUSION BDR to salbutamol increases breath amplitude in gravity non-dependent segments of the lungs during spontaneous tidal breathing in preschool children with BPD. Pediatr Pulmonol. 2017;52:353-359. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wojciech Durlak
- Department of Pediatrics, Jagiellonian University, Wielicka 265, Cracow, 30-663, Poland
| | - Małgorzata Klimek
- Department of Pediatrics, Jagiellonian University, Wielicka 265, Cracow, 30-663, Poland
| | - Przemko Kwinta
- Department of Pediatrics, Jagiellonian University, Wielicka 265, Cracow, 30-663, Poland
| |
Collapse
|
243
|
Barrington KJ, Fortin‐Pellerin E, Pennaforte T, Cochrane Neonatal Group. Fluid restriction for treatment of preterm infants with chronic lung disease. Cochrane Database Syst Rev 2017; 2:CD005389. [PMID: 28176308 PMCID: PMC6464249 DOI: 10.1002/14651858.cd005389.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Fluid restriction is often recommended as part of the management of infants with early or established bronchopulmonary dysplasia (BPD). OBJECTIVES To determine whether fluid restriction as part of the therapeutic intervention for early or established BPD improves clinical outcomes. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2016, Issue 1) in the Cochrane Library (searched 16 February 2016), MEDLINE via PubMed (1966 to 16 February 2016), Embase (1980 to 16 February 2016), and CINAHL (1982 to 16 February 2016). We also searched clinical trials' databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Prospective randomised clinical trials comparing two distinct fluid administration volumes in preterm infants with early or established BPD. DATA COLLECTION AND ANALYSIS We used the standard methods of Cochrane Neonatal. For the included trial, we extracted data and assessed the risk of bias, and used GRADE methods to assess the quality of the evidence. The outcomes considered in this review are effects on mortality or requirement for oxygen at 36 weeks' postmenstrual age (primary outcome measure), the duration of supplemental oxygen therapy, proportion of infants discharged from hospital on oxygen, duration of assisted ventilation, duration of hospitalisation, weight gain, feeding tolerance, apnoea, necrotizing enterocolitis, renal dysfunction or nephrocalcinosis, lung mechanics, and use of diuretic therapy (secondary outcome measures). MAIN RESULTS One trial was found, including 60 preterm infants at 28 days of age with persistent oxygen requirements. Infants were randomised to either 180 mL/kg/day of standard formula or 145 mL/kg/day of concentrated formula. This single study did not provide data regarding our primary outcome. No effects of the intervention were found on any of our secondary outcomes. The quality of the evidence from this study was graded low. AUTHORS' CONCLUSIONS There is no evidence to support the practice of fluid restriction in infants with early or established BPD.
Collapse
Affiliation(s)
- Keith J Barrington
- CHU Ste‐JustineDepartment of Pediatrics3175 Cote Ste CatherineMontrealQCCanadaH3T 1C5
| | - Etienne Fortin‐Pellerin
- Sainte Justine University Health CenterDepartment of Neonatology3175 Cote Sainte CatherineMontrealQCCanadaH3T 1C5
| | | | | |
Collapse
|
244
|
Inhaled hydrofluoalkane-beclomethasone dipropionate in bronchopulmonary dysplasia. A double-blind, randomized, controlled pilot study. J Perinatol 2017; 37:197-202. [PMID: 27735931 DOI: 10.1038/jp.2016.177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The efficacy of inhaled steroids in spontaneously breathing infants with established bronchopulmonary dysplasia (BPD) is debatable. The inhaled steroid hydrofluoalkane-beclomethasone dipropionate (QVAR) is unique in its small particle size that results in higher lung deposition. Our objective was to determine if inhaled QVAR could decrease respiratory rehospitalizations of infants with established BPD. STUDY DESIGN Double-blind, randomized placebo-controlled, multicenter pilot study. Preterm infants with moderate-to-severe BPD were randomized to inhaled QVAR 100 μg per dose or placebo twice daily via Aerochamber with face mask. Treatment was administered daily from recruitment at 36 weeks post menstrual age until 3 months post discharge. Analysis was carried out by intention to treat. RESULTS The QVAR (n=18) and placebo (n=20) groups were comparable in birth and recruitment characteristics. Length of stay (108.5±26.3 vs 108.7±36.0 days) and infants requiring oxygen at discharge (5/17 vs 6/19) or at study end (0/17 vs 2/19) were comparable. Respiratory rehospitalizations/infant (0.1±0.5 vs 0.4±0.6), rehospitalization days (0.5±1.5 vs 4.1±10.3), and post-discharge additive inhaled (0.3±0.9 vs 6.4±21.5 days), systemic (0.7±2.8 vs 1.0±1.4 days) and combined (inhaled/systemic) steroids (1.0±2.9 vs 7.8±25.8 days) tended to be lower in the QVAR compared with the placebo group. Blood pressure, height and weight gain, and urine cortisol/creatinine ratio at study end were comparable between groups. CONCLUSIONS Our study was unable to detect a significant effect of inhaled QVAR on the respiratory course of established BPD. The study was underpowered. Possible benefits of QVAR could be masked by a tendency toward higher use of additional steroids in the placebo group.
Collapse
|
245
|
Zhang Y, Cheng HP, Bao TP, Wang XG, Tian ZF. [Expression of long non-coding RNA NANCI in lung tissues of neonatal mice with hyperoxia-induced lung injury and its regulatory effect on NKX2.1]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:215-221. [PMID: 28202123 PMCID: PMC7389476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/12/2016] [Indexed: 08/01/2024]
Abstract
OBJECTIVE To investigate the expression of long non-coding RNA NANCI in lung tissues of neonatal mice with hyperoxia-induced lung injury and its regulatory effect on NKX2.1. METHODS A total of 48 neonatal C57BL/6J mice were randomly divided into an air group and a hyperoxia group, with 24 mice in each group. Each group was further divided into 7-day, 14-day, and 21-day subgroups, with 8 mice in each subgroup. The mice in the air group were fed in the indoor environment (FiO2=21%) and those in the hyperoxia group were fed in a high-oxygen box (oxygen concentration: >95%). The mice were sacrificed at each time point and lung tissue samples were collected. Hematoxylin and eosin staining was used to observe pathological changes in lung tissues. RT-qPCR and Western blot were used to measure the mRNA and protein expression of NANCI and NKX2.1. RESULTS The air group had the highest mRNA expression of NANCI and NKX2.1 at 7 days and the same level of mRNA expression at 14 and 21 days. Compared with the air group, the hyperoxia group had significant reductions in the degree of alveolarization and radial alveolar count (RAC) in lung tissues (P<0.05), and in the hyperoxia group, RAC gradually decreased over the time of hyperoxia exposure (P<0.05). The hyperoxia group had significantly lower mRNA and protein expression of NANCI and NKX2.1 than the air group at all time points (P<0.05). In both groups, the relative mRNA and protein expression of NANCI and NKX2.1 gradually decreased over the time of hyperoxia exposure (P<0.05). The expression of NKX2 was positively correlated with that of NANCI (r=0.585, P=0.003), and the expression of NKX2 and NANCI was positively correlated with RAC in the hyperoxia group (r=0.655 and 0.541 respectively, P<0.05). CONCLUSIONS NANCI may be involved in the development of immature lung tissues. Lung injury is gradually aggravated over the time of hyperoxia exposure. The levels of NANCI and NKX2.1 are associated with the severity of lung injury, suggesting that the NANCI/NKX2.1 target gene signaling pathway might be involved in the development of hyperoxia-induced lung injury in neonatal mice.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neonatology, Huai'an First Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China.
| | | | | | | | | |
Collapse
|
246
|
Zhang Y, Cheng HP, Bao TP, Wang XG, Tian ZF. [Expression of long non-coding RNA NANCI in lung tissues of neonatal mice with hyperoxia-induced lung injury and its regulatory effect on NKX2.1]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:215-221. [PMID: 28202123 PMCID: PMC7389476 DOI: 10.7499/j.issn.1008-8830.2017.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/12/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To investigate the expression of long non-coding RNA NANCI in lung tissues of neonatal mice with hyperoxia-induced lung injury and its regulatory effect on NKX2.1. METHODS A total of 48 neonatal C57BL/6J mice were randomly divided into an air group and a hyperoxia group, with 24 mice in each group. Each group was further divided into 7-day, 14-day, and 21-day subgroups, with 8 mice in each subgroup. The mice in the air group were fed in the indoor environment (FiO2=21%) and those in the hyperoxia group were fed in a high-oxygen box (oxygen concentration: >95%). The mice were sacrificed at each time point and lung tissue samples were collected. Hematoxylin and eosin staining was used to observe pathological changes in lung tissues. RT-qPCR and Western blot were used to measure the mRNA and protein expression of NANCI and NKX2.1. RESULTS The air group had the highest mRNA expression of NANCI and NKX2.1 at 7 days and the same level of mRNA expression at 14 and 21 days. Compared with the air group, the hyperoxia group had significant reductions in the degree of alveolarization and radial alveolar count (RAC) in lung tissues (P<0.05), and in the hyperoxia group, RAC gradually decreased over the time of hyperoxia exposure (P<0.05). The hyperoxia group had significantly lower mRNA and protein expression of NANCI and NKX2.1 than the air group at all time points (P<0.05). In both groups, the relative mRNA and protein expression of NANCI and NKX2.1 gradually decreased over the time of hyperoxia exposure (P<0.05). The expression of NKX2 was positively correlated with that of NANCI (r=0.585, P=0.003), and the expression of NKX2 and NANCI was positively correlated with RAC in the hyperoxia group (r=0.655 and 0.541 respectively, P<0.05). CONCLUSIONS NANCI may be involved in the development of immature lung tissues. Lung injury is gradually aggravated over the time of hyperoxia exposure. The levels of NANCI and NKX2.1 are associated with the severity of lung injury, suggesting that the NANCI/NKX2.1 target gene signaling pathway might be involved in the development of hyperoxia-induced lung injury in neonatal mice.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neonatology, Huai'an First Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China.
| | | | | | | | | |
Collapse
|
247
|
Abstract
In human neonates rapid adaptation from an aqueous intrauterine environment to permanent air breathing is the rate-limiting step for extrauterine life, failure of which justifies the existence of neonatal intensive care units. The lung develops at about 4-6 weeks' gestation in humans as a ventral outpouching of the primitive foregut into the surrounding ventral mesenchyme, termed the laryngotracheal groove. At its posterior end lie progenitor cells that form a pair of bronchial tubes, from which arise all the distal epithelial structures of the lung. In humans, formation of the distal gas exchange surfaces begins in utero at about 20 weeks' gestation and is substantially established by term. Stereotypic branching of the proximal airway ends relatively early at 16-18 weeks at the bronchoalveolar duct junctions. Distally, about 5 finger-like alveolar ducts arise from each bronchoalveolar duct junction and ramify outwards towards the pleura. The majority of alveolar air sacs arise from the sides of the alveolar ducts and each of these alveoli can have up to 5 daughter alveoli arising from the outer surface as subsequent buds. At the end of each alveolar duct lie the mouths of 5 interconnected alveoli. Each family of alveoli arising from each bronchoalveolar duct junction has a different shape depending upon the limitations imposed by the pleural surface as well as the interstitial fascial planes.
Collapse
|
248
|
Omer M, Molloy EJ. QUESTION 2: Is permissive hypercapnia beneficial to preterm infants? Arch Dis Child 2017; 102:113-115. [PMID: 27881376 DOI: 10.1136/archdischild-2016-312050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/07/2016] [Indexed: 11/04/2022]
Affiliation(s)
- Murwan Omer
- Department of Paediatrics, Coombe Women and Infants University Hospital, Dublin, Ireland
| | - Eleanor J Molloy
- Department of Neonatology, Our lady's Children's Hospital, Trinity College, the University of Dublin, Dublin, Ireland
| |
Collapse
|
249
|
Gien J, Kinsella J, Thrasher J, Grenolds A, Abman SH, Baker CD. Retrospective Analysis of an Interdisciplinary Ventilator Care Program Intervention on Survival of Infants with Ventilator-Dependent Bronchopulmonary Dysplasia. Am J Perinatol 2017; 34:155-163. [PMID: 27355979 PMCID: PMC5199631 DOI: 10.1055/s-0036-1584897] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background The clinical needs of infants with severe bronchopulmonary dysplasia (BPD) that remain ventilator-dependent are complex, and management strategies that optimize survival and long-term outcomes controversial. We hypothesized that an interdisciplinary ventilator care program (VCP), committed to the care of this population will improve survival through standardized approaches to cardiopulmonary care and related comorbidities, enhanced communication, and continuity of care. Methods Retrospective chart reviews were performed on patients at Children's Hospital Colorado's neonatal intensive care unit, who underwent tracheostomy placement between 2000 and 2013. Data were collected for two time periods: 2000 to 2005 and 2006 to 2013, before and after initiation of the VCP. Collected data on infants with ventilator-dependent BPD included: gestational age (GA), age at tracheostomy, respiratory severity score (RSS; mean airway pressure [MAP] × fraction of inspired oxygen [FiO2]), comorbidities, medication use, and the age of death. Tracheostomy patients without severe BPD were excluded. Results Despite no difference in GA, birth weight, or cardiorespiratory comorbidities, survival to discharge increased from 50 to 85% after implementation of the VCP (p < 0.05). Between period 1 and 2, there were differences in systemic and inhaled steroid use and mucolytic use. Conclusion These findings suggest an interdisciplinary approach to the care of infants with ventilator-dependent BPD can improve survival.
Collapse
Affiliation(s)
- Jason Gien
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado,Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - John Kinsella
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado,Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Jodi Thrasher
- Division of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Alicia Grenolds
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado,Division of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Steven H. Abman
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado,Division of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Christopher D. Baker
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado,Division of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
250
|
Jung E, Choi CW, Kim SY, Sung TJ, Kim H, Park KU, Kim HS, Kim BI, Choi JH. Coexistence of Ureaplasma and chorioamnionitis is associated with prolonged mechanical ventilation. Pediatr Int 2017; 59:34-40. [PMID: 27337221 DOI: 10.1111/ped.13072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 04/27/2016] [Accepted: 06/07/2016] [Indexed: 11/27/2022]
Abstract
BACKGROUND Both histologic chorioamnionitis (HCAM) and Ureaplasma infection are considered important contributors to perinatal lung injury. We tested the hypothesis that coexistence of maternal HCAM and perinatal Ureaplasma exposure increases the risk of prolonged mechanical ventilation in extremely low-birthweight (ELBW) infants. METHODS A retrospective cohort study was carried out of all ELBW infants born between January 2008 and December 2013 at a single academic center. Placental pathology and gastric fluid Ureaplasma data were available for all infants. Culture and polymerase chain reaction were used to detect Ureaplasma in gastric fluid. Prolonged mechanical ventilation was defined as mechanical ventilation that began within 28 days after birth and continued. RESULTS Of 111 ELBW infants enrolled, 84 survived beyond 36 weeks of postmenstrual age (PMA) and were included in the analysis. Eighteen infants (21.4%) had both HCAM and Ureaplasma exposure. Seven infants (8.3%) required mechanical ventilation beyond 36 weeks of PMA. Coexistence of HCAM and Ureaplasma in gastric fluid was significantly associated with prolonged mechanical ventilation after adjustment for gestational age, sex, mode of delivery, and use of macrolide antibiotics (OR, 8.7; 95%CI: 1.1-67.2). CONCLUSIONS Coexistence of maternal HCAM and perinatal Ureaplasma exposure significantly increases the risk of prolonged mechanical ventilation in ELBW infants.
Collapse
Affiliation(s)
- Euiseok Jung
- Department of Pediatrics, Asan Medical Center Children's Hospital, Seoul, Korea
| | - Chang Won Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Su Yeong Kim
- Department of Pediatrics, Hallym University Kangdong Sacred Heart Hospital, Seoul, Korea
| | - Tae-Jung Sung
- Department of Pediatrics, Hallym University Kangnam Sacred Heart Hospital, Seoul, Korea
| | - Haeryoung Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyoung Un Park
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Han-Suk Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Beyong Il Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea.,Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jung-Hwan Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|