1
|
Maeda H, Li X, Go H, Dennery PA, Yao H. miRNA Signatures in Bronchopulmonary Dysplasia: Implications for Biomarkers, Pathogenesis, and Therapeutic Options. FRONT BIOSCI-LANDMRK 2024; 29:271. [PMID: 39082345 PMCID: PMC11799892 DOI: 10.31083/j.fbl2907271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 01/18/2025]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in premature infants characterized by alveolar dysplasia, vascular simplification and dysmorphic vascular development. Supplemental oxygen and mechanical ventilation commonly used as life-saving measures in premature infants may cause BPD. microRNAs (miRNAs), a class of small, non-coding RNAs, regulate target gene expression mainly through post-transcriptional repression. miRNAs play important roles in modulating oxidative stress, proliferation, apoptosis, senescence, inflammatory responses, and angiogenesis. These cellular processes play pivotal roles in the pathogenesis of BPD. Accumulating evidence demonstrates that miRNAs are dysregulated in the lung of premature infants with BPD, and in animal models of this disease, suggesting contributing roles of dysregulated miRNAs in the development of BPD. Therefore, miRNAs are considered promising biomarker candidates and therapeutic agents for this disease. In this review, we discuss how dysregulated miRNAs and their modulation alter cellular processes involved in BPD. We then focus on therapeutic approaches targeting miRNAs for BPD. This review provides an overview of miRNAs as biomarkers, and highlights potential pathogenic roles, and therapeutic strategies for BPD using miRNAs.
Collapse
Affiliation(s)
- Hajime Maeda
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Fukushima Medical University School of Medicine, 960-1295 Fukushima, Japan
| | - Xiaoyun Li
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Providence Veterans Affairs Medical Center, Providence, RI 02908, USA
- Department of Medicine, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
- College of Pharmacy, Jinan University, 510632 Guangzhou, Guangdong, China
| | - Hayato Go
- Department of Pediatrics, Fukushima Medical University School of Medicine, 960-1295 Fukushima, Japan
| | - Phyllis A. Dennery
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
- Providence Veterans Affairs Medical Center, Providence, RI 02908, USA
- Department of Medicine, Warren Alpert School of Medicine of Brown University, Providence, RI 02903, USA
| |
Collapse
|
2
|
Roberts JD. Nitric oxide regulation of fetal and newborn lung development and function. Nitric Oxide 2024; 147:13-25. [PMID: 38588917 PMCID: PMC11148871 DOI: 10.1016/j.niox.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/21/2024] [Accepted: 04/05/2024] [Indexed: 04/10/2024]
Abstract
In the developing lung, nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) signaling are essential in regulating lung formation and vascular tone. Animal studies have linked many anatomical and pathophysiological features of newborn lung disease to abnormalities in the NO/cGMP signaling system. They have demonstrated that driving this system with agonists and antagonists alleviates many of them. This research has spurred the rapid clinical development, testing, and application of several NO/cGMP-targeting therapies with the hope of treating and potentially preventing significant pediatric lung diseases. However, there are instances when the therapeutic effectiveness of these agents is limited. Studies indicate that injury-induced disruption of several critical components within the signaling system may hinder the promise of some of these therapies. Recent research has identified basic mechanisms that suppress NO/cGMP signaling in the injured newborn lung. They have also pinpointed biomarkers that offer insight into the activation of these pathogenic mechanisms and their influence on the NO/cGMP signaling system's integrity in vivo. Together, these will guide the development of new therapies to protect NO/cGMP signaling and safeguard newborn lung development and function. This review summarizes the important role of the NO/cGMP signaling system in regulating pulmonary development and function and our evolving understanding of how it is disrupted by newborn lung injury.
Collapse
Affiliation(s)
- Jesse D Roberts
- Cardiovascular Research Center of the General Medical Services and the Departments of Anesthesia, Critical Care and Pain Medicine, Pediatrics, and Medicine, Massachusetts General Hospital - East, 149 13th St, Boston, MA, USA; Harvard Medical School, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
3
|
Albertine KH, Rebentisch A, Dawson E, Van Boerum J, Major E, Štipka J, Foreman H, Headden D, Vordos Z, Beck E, Wang Z, Yang H, Yu B, Dahl MJ, Null DM, Bizzotto D, Veneroni C, Lavizzari A, Dellacà RL, Delavogia E, Mitsialis SA, Kourembanas S. Mesenchymal stromal cell extracellular vesicles improve lung development in mechanically ventilated preterm lambs. Am J Physiol Lung Cell Mol Physiol 2024; 326:L770-L785. [PMID: 38563994 PMCID: PMC11380989 DOI: 10.1152/ajplung.00349.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Novel therapies are needed for bronchopulmonary dysplasia (BPD) because no effective treatment exists. Mesenchymal stromal cell extracellular vesicles (MSC-sEVs) have therapeutic efficacy in a mouse pup neonatal hyperoxia BPD model. We tested the hypothesis that MSC-sEVs will improve lung functional and structural development in mechanically ventilated preterm lambs. Preterm lambs (∼129 days; equivalent to human lung development at ∼28 wk gestation) were exposed to antenatal steroids, surfactant, caffeine, and supported by mechanical ventilation for 6-7 days. Lambs were randomized to blinded treatment with either MSC-sEVs (human bone marrow MSC-derived; 2 × 1011 particles iv; n = 8; 4 F/4 M) or vehicle control (saline iv; 4 F/4 M) at 6 and 78 h post delivery. Physiological targets were pulse oximetry O2 saturation 90-94% ([Formula: see text] 60-90 mmHg), [Formula: see text] 45-60 mmHg (pH 7.25-7.35), and tidal volume 5-7 mL/kg. MSC-sEVs-treated preterm lambs tolerated enteral feedings compared with vehicle control preterm lambs. Differences in weight patterns were statistically significant. Respiratory severity score, oxygenation index, A-a gradient, distal airspace wall thickness, and smooth muscle thickness around terminal bronchioles and pulmonary arterioles were significantly lower for the MSC-sEVs group. S/F ratio, radial alveolar count, secondary septal volume density, alveolar capillary surface density, and protein abundance of VEGF-R2 were significantly higher for the MSC-sEVs group. MSC-sEVs improved respiratory system physiology and alveolar formation in mechanically ventilated preterm lambs. MSC-sEVs may be an effective and safe therapy for appropriate functional and structural development of the lung in preterm infants who require mechanical ventilation and are at risk of developing BPD.NEW & NOTEWORTHY This study focused on potential treatment of preterm infants at risk of developing bronchopulmonary dysplasia (BPD), for which no effective treatment exists. We tested treatment of mechanically ventilated preterm lambs with human mesenchymal stromal cell extracellular vesicles (MSC-sEVs). The results show improved respiratory gas exchange and parenchymal growth of capillaries and epithelium that are necessary for alveolar formation. Our study provides new mechanistic insight into potential efficacy of MSC-sEVs for preterm infants at risk of developing BPD.
Collapse
Affiliation(s)
- Kurt H Albertine
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Andrew Rebentisch
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Elaine Dawson
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Jakob Van Boerum
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Emily Major
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Juraj Štipka
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Hannah Foreman
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - David Headden
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Zoë Vordos
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Emily Beck
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Zhengming Wang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Haixia Yang
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Baifeng Yu
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Mar Janna Dahl
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Donald M Null
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, Utah, United States
| | - Davide Bizzotto
- TechRes Lab, Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano University, Milan, Italy
| | - Chiara Veneroni
- TechRes Lab, Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano University, Milan, Italy
| | - Anna Lavizzari
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Raffaele L Dellacà
- TechRes Lab, Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano University, Milan, Italy
| | - Eleni Delavogia
- Division of Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States
| | - S Alex Mitsialis
- Division of Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States
| | - Stella Kourembanas
- Division of Neonatology, Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
4
|
Jin Z, Jiang MM, Lee B. Nitric oxide is required for lung alveolarization revealed by deficiency of argininosuccinate lyase. Hum Mol Genet 2023; 33:33-37. [PMID: 37738569 PMCID: PMC12099293 DOI: 10.1093/hmg/ddad158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023] Open
Abstract
Inhaled nitric oxide (NO) therapy has been reported to improve lung growth in premature newborns. However, the underlying mechanisms by which NO regulates lung development remain largely unclear. NO is enzymatically produced by three isoforms of nitric oxide synthase (NOS) enzymes. NOS knockout mice are useful tools to investigate NO function in the lung. Each single NOS knockout mouse does not show obvious lung alveolar phenotype, likely due to compensatory mechanisms. While mice lacking all three NOS isoforms display impaired lung alveolarization, implicating NO plays a pivotal role in lung alveolarization. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing L-arginine, the sole precursor for NOS-dependent NO synthesis. ASL is also required for channeling extracellular L-arginine into a NO-synthetic complex. Thus, ASL deficiency (ASLD) is a non-redundant model for cell-autonomous, NOS-dependent NO deficiency. Here, we assessed lung alveolarization in ASL-deficient mice. Hypomorphic deletion of Asl (AslNeo/Neo) results in decreased lung alveolarization, accompanied with reduced level of S-nitrosylation in the lung. Genetic ablation of one copy of Caveolin-1, which is a negative regulator of NO production, restores total S-nitrosylation as well as lung alveolarization in AslNeo/Neo mice. Importantly, NO supplementation could partially rescue lung alveolarization in AslNeo/Neo mice. Furthermore, endothelial-specific knockout mice (VE-Cadherin Cre; Aslflox/flox) exhibit impaired lung alveolarization at 12 weeks old, supporting an essential role of endothelial-derived NO in the enhancement of lung alveolarization. Thus, we propose that ASLD is a model to study NO-mediated lung alveolarization.
Collapse
Affiliation(s)
- Zixue Jin
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| |
Collapse
|
5
|
Ivanovski N, Wang H, Tran H, Ivanovska J, Pan J, Miraglia E, Leung S, Posiewko M, Li D, Mohammadi A, Higazy R, Nagy A, Kim P, Santyr G, Belik J, Palaniyar N, Gauda EB. L-citrulline attenuates lipopolysaccharide-induced inflammatory lung injury in neonatal rats. Pediatr Res 2023; 94:1684-1695. [PMID: 37349511 DOI: 10.1038/s41390-023-02684-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/28/2023] [Accepted: 05/16/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Prenatal or postnatal lung inflammation and oxidative stress disrupt alveolo-vascular development leading to bronchopulmonary dysplasia (BPD) with and without pulmonary hypertension. L-citrulline (L-CIT), a nonessential amino acid, alleviates inflammatory and hyperoxic lung injury in preclinical models of BPD. L-CIT modulates signaling pathways mediating inflammation, oxidative stress, and mitochondrial biogenesis-processes operative in the development of BPD. We hypothesize that L-CIT will attenuate lipopolysaccharide (LPS)-induced inflammation and oxidative stress in our rat model of neonatal lung injury. METHODS Newborn rats during the saccular stage of lung development were used to investigate the effect of L-CIT on LPS-induced lung histopathology and pathways involved in inflammatory, antioxidative processes, and mitochondrial biogenesis in lungs in vivo, and in primary culture of pulmonary artery smooth muscle cells, in vitro. RESULTS L-CIT protected the newborn rat lung from LPS-induced: lung histopathology, ROS production, NFκB nuclear translocation, and upregulation of gene and protein expression of inflammatory cytokines (IL-1β, IL-8, MCP-1α, and TNF-α). L-CIT maintained mitochondrial morphology, increased protein levels of PGC-1α, NRF1, and TFAM (transcription factors involved in mitochondrial biogenesis), and induced SIRT1, SIRT3, and superoxide dismutases protein expression. CONCLUSION L-CIT may be efficacious in decreasing early lung inflammation and oxidative stress mitigating progression to BPD. IMPACT The nonessential amino acid L-citrulline (L-CIT) mitigated lipopolysaccharide (LPS)-induced lung injury in the early stage of lung development in the newborn rat. This is the first study describing the effect of L-CIT on the signaling pathways operative in bronchopulmonary dysplasia (BPD) in a preclinical inflammatory model of newborn lung injury. If our findings translate to premature infants, L-CIT could decrease inflammation, oxidative stress and preserve mitochondrial health in the lung of premature infants at risk for BPD.
Collapse
Affiliation(s)
- Nikola Ivanovski
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Huanhuan Wang
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Harvard Tran
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Julijana Ivanovska
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jingyi Pan
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Emily Miraglia
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Sharon Leung
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Melanie Posiewko
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Daniel Li
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Atefeh Mohammadi
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Anita Nagy
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Division of Anatomical Pathology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Peter Kim
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Giles Santyr
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jaques Belik
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Nades Palaniyar
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Estelle B Gauda
- Translational Medicine and Cell Biology Programs, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Division of Neonatology, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
6
|
Fike CD, Aschner JL. Pharmacotherapy for Pulmonary Hypertension in Infants with Bronchopulmonary Dysplasia: Past, Present, and Future. Pharmaceuticals (Basel) 2023; 16:503. [PMID: 37111262 PMCID: PMC10141152 DOI: 10.3390/ph16040503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
Approximately 8-42% of premature infants with chronic lung disease of prematurity, bronchopulmonary dysplasia (BPD), develop pulmonary hypertension (PH). Infants with BPD-PH carry alarmingly high mortality rates of up to 47%. Effective PH-targeted pharmacotherapies are desperately needed for these infants. Although many PH-targeted pharmacotherapies are commonly used to treat BPD-PH, all current use is off-label. Moreover, all current recommendations for the use of any PH-targeted therapy in infants with BPD-PH are based on expert opinion and consensus statements. Randomized Control Trials (RCTs) are needed to determine the efficacy of PH-targeted treatments in premature infants with or at risk of BPD-PH. Prior to performing efficacy RCTs, studies need to be conducted to obtain pharmacokinetic, pharmacodynamic, and safety data for any pharmacotherapy used in this understudied and fragile patient population. This review will discuss current and needed treatment strategies, identify knowledge deficits, and delineate both challenges to be overcome and approaches to be taken to develop effective PH-targeted pharmacotherapies that will improve outcomes for premature infants with or at risk of developing BPD-PH.
Collapse
Affiliation(s)
- Candice D. Fike
- Department of Pediatrics, University of Utah Health, Salt Lake City, UT 84108, USA
| | - Judy L. Aschner
- Department of Pediatrics, Joseph M. Sanzari Children’s Hospital at Hackensack University Medical Center, Hackensack, NJ 07601, USA
- Department of Pediatrics, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| |
Collapse
|
7
|
Stankiewicz B, Mierzewska-Schmidt M, Górczyńska K, Pałko KJ, Baranowski A, Kozarski M, Darowski M. Ventilation inhomogeneity in CDH infants – A new attitude within a simulation study. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
8
|
Roberts K, Stepanovich G, Bhatt-Mehta V, Donn SM. New Pharmacologic Approaches to Bronchopulmonary Dysplasia. J Exp Pharmacol 2021; 13:377-396. [PMID: 33790663 PMCID: PMC8006962 DOI: 10.2147/jep.s262350] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary Dysplasia is the most common long-term respiratory morbidity of preterm infants, with the risk of development proportional to the degree of prematurity. While its pathophysiologic and histologic features have changed over time as neonatal demographics and respiratory therapies have evolved, it is now thought to be characterized by impaired distal lung growth and abnormal pulmonary microvascular development. Though the exact sequence of events leading to the development of BPD has not been fully elucidated and likely varies among patients, it is thought to result from inflammatory and mechanical/oxidative injury from chronic ventilatory support in fragile, premature lungs susceptible to injury from surfactant deficiency, structural abnormalities, inadequate antioxidant defenses, and a chest wall that is more compliant than the lung. In addition, non-pulmonary issues may adversely affect lung development, including systemic infections and insufficient nutrition. Once BPD has developed, its management focuses on providing adequate gas exchange while promoting optimal lung growth. Pharmacologic strategies to ameliorate or prevent BPD continue to be investigated. A variety of agents, to be reviewed henceforth, have been developed or re-purposed to target different points in the pathways that lead to BPD, including anti-inflammatories, diuretics, steroids, pulmonary vasodilators, antioxidants, and a number of molecules involved in the cell signaling cascade thought to be involved in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Katelyn Roberts
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gretchen Stepanovich
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Varsha Bhatt-Mehta
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
- College of Pharmacy, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Steven M Donn
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
9
|
Liu M, Iosef C, Rao S, Domingo-Gonzalez R, Fu S, Snider P, Conway SJ, Umbach GS, Heilshorn SC, Dewi RE, Dahl MJ, Null DM, Albertine KH, Alvira CM. Transforming Growth Factor-induced Protein Promotes NF-κB-mediated Angiogenesis during Postnatal Lung Development. Am J Respir Cell Mol Biol 2021; 64:318-330. [PMID: 33264084 PMCID: PMC7909333 DOI: 10.1165/rcmb.2020-0153oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-β-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvβ3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.
Collapse
Affiliation(s)
- Min Liu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Cristiana Iosef
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Shailaja Rao
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | | | - Sha Fu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- Liuyang People’s Hospital, Hunan, China
| | - Paige Snider
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Simon J. Conway
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Gray S. Umbach
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- University of Texas Southwestern Medical School, Dallas, Texas; and
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Mar J. Dahl
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Donald M. Null
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kurt H. Albertine
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Cristina M. Alvira
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| |
Collapse
|
10
|
Vieira F, Makoni M, Szyld E, Sekar K. The Controversy Persists: Is There a Qualification Criterion to Utilize Inhaled Nitric Oxide in Pre-term Newborns? Front Pediatr 2021; 9:631765. [PMID: 33869113 PMCID: PMC8044816 DOI: 10.3389/fped.2021.631765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/01/2021] [Indexed: 11/22/2022] Open
Abstract
Inhaled nitric oxide (iNO) use in premature newborns remains controversial among clinicians. In 2014, the American Academy of Pediatrics, Committee on Fetus and Newborn released a statement that the available data do not support routine iNO use in pre-term newborns. Despite the absence of significant benefits, 2016 California data showed that clinicians continue to utilize iNO in pre-term infants. With studies as recent as January 2017, the Cochrane review confirmed no major advantages of iNO in pre-term newborns. Still, it recognized that a subset of pre-term infants with pulmonary hypertension (PHTN) had not been separately investigated. Furthermore, recent non-randomized controlled trials have suggested that iNO may benefit specific subgroups of pre-term newborns, especially those with PHTN, prolonged rupture of membranes, and antenatal steroid exposure. Those pre-term infants who showed a clinical response to iNO had increased survival without disability. These findings underscore the need for future studies in pre-term newborns with hypoxemic respiratory failure and PHTN. This review will discuss the rationale for using iNO, controversies regarding the diagnosis of PHTN, and additional novel approaches of iNO treatment in perinatal asphyxia and neonatal resuscitation in the pre-term population < 34 weeks gestation.
Collapse
Affiliation(s)
- Frederico Vieira
- Neonatal Perinatal Section, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marjorie Makoni
- Neonatal Perinatal Section, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Edgardo Szyld
- Neonatal Perinatal Section, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Krishnamurthy Sekar
- Neonatal Perinatal Section, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
11
|
Gentle SJ, Tipple TE, Patel R. Neonatal comorbidities and gasotransmitters. Nitric Oxide 2020; 97:27-32. [PMID: 32014495 DOI: 10.1016/j.niox.2020.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/10/2019] [Accepted: 01/29/2020] [Indexed: 01/28/2023]
Abstract
Hydrogen sulfide, nitric oxide, and carbon monoxide are endogenously produced gases that regulate various signaling pathways. The role of these transmitters is complex as constitutive production of these molecules may have anti-inflammatory, anti-microbial, and/or vasodilatory effects whereas induced production or formation of secondary metabolites may lead to cellular death. Given this fine line between friend and foe, therapeutic attenuation of these molecules' production has involved both inhibition of endogenous formation and therapeutic supplementation. All three gases have been implicated as regulators of critical aspects of neonatal physiology, and in turn, comorbidities including necrotizing enterocolitis, hypoxic ischemic encephalopathy, and pulmonary hypertension. In this review, we present current perspectives on these associations, highlight areas where insights remain sparse, and identify areas for potential for future investigations.
Collapse
Affiliation(s)
- Samuel J Gentle
- Department of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Trent E Tipple
- Section of Neonatal-Perinatal Medicine, University of Oklahoma College of Medicine, Oklahoma City, OK, USA
| | - Rakesh Patel
- Department of Pathology and Center for Free Radical Biology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
12
|
Sherlock LG, Wright CJ, Kinsella JP, Delaney C. Inhaled nitric oxide use in neonates: Balancing what is evidence-based and what is physiologically sound. Nitric Oxide 2019; 95:12-16. [PMID: 31866361 DOI: 10.1016/j.niox.2019.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 12/10/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Inhaled nitric oxide is a powerful therapeutic used in neonatology. Its use is evidenced-based for term and near-term infants with persistent pulmonary hypertension; however, it is frequently used off-label both in term and preterm babies. This article reviews the off-label uses of iNO in infants. Rationale is discussed for a selective application of iNO based on physiologically guided principles, and new research avenues are considered.
Collapse
Affiliation(s)
- Laurie G Sherlock
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - John P Kinsella
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA
| | - Cassidy Delaney
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
13
|
Abstract
In the absence of effective interventions to prevent preterm births, improved survival of infants who are born at the biological limits of viability has relied on advances in perinatal care over the past 50 years. Except for extremely preterm infants with suboptimal perinatal care or major antenatal events that cause severe respiratory failure at birth, most extremely preterm infants now survive, but they often develop chronic lung dysfunction termed bronchopulmonary dysplasia (BPD; also known as chronic lung disease). Despite major efforts to minimize injurious but often life-saving postnatal interventions (such as oxygen, mechanical ventilation and corticosteroids), BPD remains the most frequent complication of extreme preterm birth. BPD is now recognized as the result of an aberrant reparative response to both antenatal injury and repetitive postnatal injury to the developing lungs. Consequently, lung development is markedly impaired, which leads to persistent airway and pulmonary vascular disease that can affect adult lung function. Greater insights into the pathobiology of BPD will provide a better understanding of disease mechanisms and lung repair and regeneration, which will enable the discovery of novel therapeutic targets. In parallel, clinical and translational studies that improve the classification of disease phenotypes and enable early identification of at-risk preterm infants should improve trial design and individualized care to enhance outcomes in preterm infants.
Collapse
|
14
|
McPherson C. Pharmacotherapy for the Prevention of Bronchopulmonary Dysplasia: Can Anything Compete with Caffeine and Corticosteroids? Neonatal Netw 2019; 38:242-249. [PMID: 31470395 DOI: 10.1891/0730-0832.38.4.242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a morbidity of prematurity with implications for respiratory and neurologic health into adulthood. Multiple risk factors contribute to the development of BPD leading to examination of various prevention strategies. The roles of systemic corticosteroids and caffeine have been addressed by the American Academy of Pediatrics. The place in therapy of other agents commonly utilized in clinical practice remains unclear. Inhaled nitric oxide has been the subject of numerous large, randomized controlled trials in preterm infants. Despite sound rationale, these trials have largely failed to document benefit, suggesting a limited role for inhaled nitric oxide therapy in the preterm population. In contrast, intramuscular vitamin A has been documented to reduce the incidence of BPD in randomized trials. However, the invasiveness and the sporadic availability of this therapy have led to decreased utilization. All macrolide antibiotics do not appear to have a similar impact on the incidence of BPD; however, azithromycin administered to infants colonized with Ureaplasma may have impact. Questions remain about the optimal dosing approach and long-term safety of this intervention. Finally, diuretic therapy is widely used in clinical practice despite significant toxicities and limited data supporting a role in BPD prevention. Taken together, available data suggest that caffeine and selective use of corticosteroids remain the mainstays of pharmacologic BPD prevention.
Collapse
|
15
|
Shiraishi J, Kusuda S, Cho K, Nakao A, Hiroma T, Sugiura H, Suzuki S, Oshiro M, Yoshimoto S, Watabe S. Standardization of nitric oxide inhalation in extremely preterm infants in Japan. Pediatr Int 2019; 61:152-157. [PMID: 30523661 DOI: 10.1111/ped.13746] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/31/2018] [Accepted: 11/13/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND In perinatal medicine, inhaled nitric oxide (iNO) has been an important tool for the treatment of full-term and late-preterm infants with persistent pulmonary hypertension of the newborn (PPHN) and hypoxemic respiratory failure (HRF). Its use in more premature infants, however, is controversial. To evaluate the current clinical practices regarding use of acute iNO in extremely preterm infants, a nationwide survey was conducted in Japan. METHODS A questionnaire survey was conducted from May to September, 2015. Questionnaires about PPHN and iNO treatment were sent to the doctor in charge of the neonatal care unit in 213 perinatal medical centers (PMC) that possessed iNO equipment in Japan. RESULTS A total of 143 of the 213 PMC provided responses (67.1%). A diagnosis of PPHN was made exclusively on echocardiography in all PMC. On definitive PPHN diagnosis, iNO was selected in the majority of the PMC (72%) and started from ≤10 p.p.m. in most PMC (49.7%) for extremely preterm infants. During iNO therapy, cardiac function was checked on echocardiography by a neonatologist every ≤8 h. iNO weaning was started when differential peripheral oxygen saturation (SpO2 ) disappeared, or when SpO2 reached 100% and so on. After iNO concentration reached 5 p.p.m., it was decreased gradually and carefully in five steps, taking 12-24 h to go from 5 to 0 p.p.m. CONCLUSIONS Inhaled nitric oxide was predominantly used in extremely preterm infants as early rescue therapy for PPHN based on echocardiography performed by a neonatologist.
Collapse
Affiliation(s)
- Jun Shiraishi
- Department of Neonatal Medicine, Osaka Women's and Children's Hospital, Izumi, Osaka, Japan
| | - Satoshi Kusuda
- Neonatal Research Network of Japan, Shinjuku, Tokyo, Japan
| | - Kazutoshi Cho
- Maternity and Perinatal Center, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Atsushi Nakao
- Neonatology, Japanese Red Cross Medical Center, Shibuya, Tokyo, Japan
| | - Takehiko Hiroma
- Neonatology, Nagano Children's Hospital, Azumino, Nagano, Japan
| | - Hiroshi Sugiura
- Neonatology, Seirei Hamamatsu General Hospital, Hamamatsu, Shizuoka, Japan
| | - Satoshi Suzuki
- Pediatrics, Nagoya City West Medical Center, Nagoya, Aichi, Japan
| | - Makoto Oshiro
- Neonatology, Japanese Red Cross Nagoya Daiichi Hospital, Nagoya, Aichi, Japan
| | - Seiji Yoshimoto
- Neonatology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Hyogo, Japan
| | - Shinichi Watabe
- Neonatal Intensive Care, Kurashiki Central Hospital, Kurashiki, Okayama, Japan
| |
Collapse
|
16
|
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
|
17
|
Sowder JC, Dahl MJ, Zuspan KR, Albertine KH, Null DM, Barneck MD, Grimmer JF. Effect of Heliox on Respiratory Outcomes during Rigid Bronchoscopy in Term Lambs. Otolaryngol Head Neck Surg 2018; 159:35-41. [PMID: 29512419 DOI: 10.1177/0194599818763067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective To (1) compare physiologic changes during rigid bronchoscopy during spontaneous and mechanical ventilation and (2) evaluate the efficacy of a helium-oxygen (heliox) gas mixture as compared with room air during rigid bronchoscopy. Study Design Crossover animal study evaluating physiologic parameters during rigid bronchoscopy. Outcomes were compared with predicted computational fluid analysis. Setting Simulated ventilation via computational fluid dynamics analysis and term lambs undergoing rigid bronchoscopy. Methods Respiratory and physiologic outcomes were analyzed in a lamb model simulating bronchoscopy during foreign body aspiration to compare heliox with room air. The main outcome measures were blood oxygen saturation, heart rate, blood pressure, partial pressure of oxygen, and partial pressure of carbon dioxide. Computational fluid dynamics analysis was performed with SOLIDWORKS within a rigid pediatric bronchoscope during simulated ventilation comparing heliox with room air. Results For room air, lambs desaturated within 3 minutes during mechanical ventilation versus normal oxygen saturation during spontaneous ventilation ( P = .01). No improvement in respiratory outcomes was seen between heliox and room air during mechanical ventilation. Computational fluid dynamics analysis demonstrates increased turbulence within size 3.5 bronchoscopes when comparing heliox and room air. Meaningful comparisons could not be made due to the intolerance of the lambs to heliox in vivo. Conclusion During mechanical ventilation on room air, lambs desaturate more quickly during rigid bronchoscopy on settings that should be adequate. Heliox does not improve ventilation during rigid bronchoscopy.
Collapse
Affiliation(s)
- Justin C Sowder
- 1 Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Mar Janna Dahl
- 2 Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Kaitlin R Zuspan
- 2 Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Kurt H Albertine
- 2 Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Donald M Null
- 3 Division of Neonatology, University of California-Davis School of Medicine, Sacramento, California, USA
| | - Mitchell D Barneck
- 4 School of Medicine, Oregon Health and Sciences University, Portland, Oregon, USA
| | - J Fredrik Grimmer
- 1 Division of Otolaryngology-Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
18
|
Askie LM, Davies LC, Schreiber MD, Hibbs AM, Ballard PL, Ballard RA. Race Effects of Inhaled Nitric Oxide in Preterm Infants: An Individual Participant Data Meta-Analysis. J Pediatr 2018; 193:34-39.e2. [PMID: 29241680 DOI: 10.1016/j.jpeds.2017.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/12/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To assess whether inhaled nitric oxide (iNO) improves survival without bronchopulmonary dysplasia (BPD) for preterm African American infants. STUDY DESIGN An individual participant data meta-analysis was conducted, including 3 randomized, placebo-controlled trials that enrolled infants born at <34 weeks of gestation receiving respiratory support, had at least 15% (or a minimum of 10 infants in each trial arm) of African American race, and used a starting iNO of >5 parts per million with the intention to treat for 7 days minimum. The primary outcome was a composite of death or BPD. Secondary outcomes included death before discharge, postnatal steroid use, gross pulmonary air leak, pulmonary hemorrhage, measures of respiratory support, and duration of hospital stay. RESULTS Compared with other races, African American infants had a significant reduction in the composite outcome of death or BPD with iNO treatment: 49% treated vs 63% controls (relative risk, 0.77; 95% CI, 0.65-0.91; P = .003; interaction P = .016). There were no differences between racial groups for death. There was also a significant difference between races (interaction P = .023) of iNO treatment for BPD in survivors, with the greatest effect in African American infants (P = .005). There was no difference between racial groups in the use of postnatal steroids, pulmonary air leak, pulmonary hemorrhage, or other measures of respiratory support. CONCLUSION iNO therapy should be considered for preterm African American infants at high risk for BPD. iNO to prevent BPD in African Americans may represent an example of a racially customized therapy for infants.
Collapse
Affiliation(s)
- Lisa M Askie
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | - Lucy C Davies
- National Health and Medical Research Council Clinical Trials Centre, University of Sydney, Sydney, Australia
| | | | - Anna Maria Hibbs
- Department of Pediatrics, Case Western Reserve University, and Rainbow Babies and Children's Hospital, Cleveland, OH
| | | | | |
Collapse
|
19
|
Hasan SU, Potenziano J, Konduri GG, Perez JA, Van Meurs KP, Walker MW, Yoder BA. Effect of Inhaled Nitric Oxide on Survival Without Bronchopulmonary Dysplasia in Preterm Infants: A Randomized Clinical Trial. JAMA Pediatr 2017; 171:1081-1089. [PMID: 28973344 PMCID: PMC5710365 DOI: 10.1001/jamapediatrics.2017.2618] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IMPORTANCE Bronchopulmonary dysplasia (BPD) occurs in approximately 40% of infants born at younger than 30 weeks' gestation and is associated with adverse pulmonary and neurodevelopmental outcomes. OBJECTIVE To test whether administration of inhaled nitric oxide to preterm infants requiring positive pressure respiratory support on postnatal days 5 to 14 improves the rate of survival without BPD. DESIGN, SETTING, AND PARTICIPANTS This intent-to-treat study was a randomized clinical trial performed at 33 US and Canadian neonatal intensive care units. Participants included 451 neonates younger than 30 weeks' gestation with birth weight less than 1250 g receiving mechanical ventilation or positive pressure respiratory support on postnatal days 5 to 14. Enrollment spanned from December 23, 2009, to April 23, 2012, and neurodevelopmental outcome studies were completed by April 4, 2014. INTERVENTIONS Placebo (nitrogen) or inhaled nitric oxide initiated at 20 ppm was decreased to 10 ppm between 72 and 96 hours after starting treatment and then to 5 ppm on day 10 or 11. Infants remained on the 5-ppm dose until completion of therapy (24 days). MAIN OUTCOMES AND MEASURES The primary outcome was the rate of survival without BPD at 36 weeks' postmenstrual age (PMA). Secondary outcomes included BPD severity, postnatal corticosteroid use, respiratory support, survival, and neurodevelopmental outcomes at 18 to 24 months' PMA. RESULTS In total, 222 infants (52.3% male [n = 116]) received placebo, and 229 infants (50.2% male [n = 115]) received inhaled nitric oxide. Their mean (SD) gestation was 25.6 (1.5) vs 25.6 (1.4) weeks, and their mean (SD) birth weight was 750 (164) vs 724 (160) g. Survival without BPD at 36 weeks' PMA was similar between the placebo and inhaled nitric oxide groups (31.5% [n = 70] vs 34.9% [n = 80]) (odds ratio, 1.17; 95% CI, 0.79-1.73). Rates for severe BPD (26.6% [55 of 207] vs 20.5% [43 of 210]) and postnatal corticosteroid use for BPD (41.0% [91 of 222] vs 41.5% [95 of 229]) and the mean (SD) days of positive pressure respiratory support (55 [40] vs 54 [42]), oxygen therapy (88 [41] vs 91 [59]), and hospitalization (105 [37] vs 108 [54]) were equivalent between the 2 groups. No differences in the incidence of common morbidities were observed. Respiratory outcomes on discharge to home, at 1 year, and at age 18 to 24 months' PMA and neurodevelopmental assessments at 18 to 24 months' PMA did not differ between groups. CONCLUSIONS AND RELEVANCE Inhaled nitric oxide, initiated at 20 ppm on postnatal days 5 to 14 to high-risk preterm infants and continued for 24 days, appears to be safe but did not improve survival without BPD at 36 weeks' PMA or respiratory and neurodevelopmental outcomes at 18 to 24 months' PMA. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00931632.
Collapse
Affiliation(s)
- Shabih U. Hasan
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta Children’s Hospital Research Institute, Calgary, Alberta, Canada
| | | | | | - Jose A. Perez
- Department of Neonatology, Winnie Palmer Hospital, Pediatrix Medical Group, Orlando, Florida
| | - Krisa P. Van Meurs
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California,Lucile Packard Children’s Hospital Stanford, Palo Alto, California
| | - M. Whit Walker
- Division of Neonatology, Department of Pediatrics, University of South Carolina School of Medicine, Greenville
| | - Bradley A. Yoder
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City
| | | |
Collapse
|
20
|
Laube M, Amann E, Uhlig U, Yang Y, Fuchs HW, Zemlin M, Mercier JC, Maier RF, Hummler HD, Uhlig S, Thome UH. Inflammatory Mediators in Tracheal Aspirates of Preterm Infants Participating in a Randomized Trial of Inhaled Nitric Oxide. PLoS One 2017; 12:e0169352. [PMID: 28046032 PMCID: PMC5207654 DOI: 10.1371/journal.pone.0169352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 12/15/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Ventilated preterm infants frequently develop bronchopulmonary dysplasia (BPD) which is associated with elevated inflammatory mediators in their tracheal aspirates (TA). In animal models of BPD, inhaled nitric oxide (iNO) has been shown to reduce lung inflammation, but data for human preterm infants is missing. METHODS Within a European multicenter trial of NO inhalation for preterm infants to prevent BPD (EUNO), TA was collected to determine the effects of iNO on pulmonary inflammation. TA was collected from 43 premature infants randomly assigned to receive either iNO or placebo gas (birth weight 530-1230 g, median 800 g, gestational age 24 to 28 2/7 weeks, median 26 weeks). Interleukin (IL)-1β, IL-6, IL-8, transforming growth factor (TGF)-β1, interferon γ-induced protein 10 (IP-10), macrophage inflammatory protein (MIP)-1α, acid sphingomyelinase (ASM), neuropeptide Y and leukotriene B4 were measured in serial TA samples from postnatal day 2 to 14. Furthermore, TA levels of nitrotyrosine and nitrite were determined under iNO therapy. RESULTS The TA levels of IP-10, IL-6, IL-8, MIP-1α, IL-1β, ASM and albumin increased with advancing postnatal age in critically ill preterm infants, whereas nitrotyrosine TA levels declined in both, iNO-treated and placebo-treated infants. The iNO treatment generally increased nitrite TA levels, whereas nitrotyrosine TA levels were not affected by iNO treatment. Furthermore, iNO treatment transiently reduced early inflammatory and fibrotic markers associated with BPD development including TGF-β1, IP-10 and IL-8, but induced a delayed increase of ASM TA levels. CONCLUSION Treatment with iNO may have played a role in reducing several inflammatory and fibrotic mediators in TA of preterm infants compared to placebo-treated infants. However, survival without BPD was not affected in the main EUNO trial. TRIAL REGISTRATION NCT00551642.
Collapse
Affiliation(s)
- Mandy Laube
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany
- * E-mail:
| | - Elena Amann
- Division of Neonatology and Pediatric Critical Care, Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Ulrike Uhlig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Yang Yang
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Hans W. Fuchs
- Department of Pediatrics, University Medical Center Freiburg, Freiburg, Germany
| | - Michael Zemlin
- Department of Pediatrics, University of Marburg, Marburg, Germany
- Department of Pediatrics, University of Saarland, Homburg, Germany
| | | | - Rolf F. Maier
- Department of Pediatrics, University of Marburg, Marburg, Germany
| | - Helmut D. Hummler
- Division of Neonatology and Pediatric Critical Care, Department of Pediatrics, University of Ulm, Ulm, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Ulrich H. Thome
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
21
|
Barrington KJ, Finer N, Pennaforte T, Cochrane Neonatal Group. Inhaled nitric oxide for respiratory failure in preterm infants. Cochrane Database Syst Rev 2017; 1:CD000509. [PMID: 28045472 PMCID: PMC6464861 DOI: 10.1002/14651858.cd000509.pub5] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Inhaled nitric oxide (iNO) is effective in term infants with hypoxic respiratory failure. The pathophysiology of respiratory failure and the potential risks of iNO differ substantially in preterm infants, necessitating specific study in this population. OBJECTIVES To determine effects of treatment with inhaled nitric oxide (iNO) on death, bronchopulmonary dysplasia (BPD), intraventricular haemorrhage (IVH) or other serious brain injury and on adverse long-term neurodevelopmental outcomes in preterm newborn infants with hypoxic respiratory failure.Owing to substantial variation in study eligibility criteria, which decreases the utility of an overall analysis, we divided participants post hoc into three groups: (1) infants treated over the first three days of life because of defects in oxygenation, (2) preterm infants with evidence of pulmonary disease treated routinely with iNO and (3) infants treated later (after three days of age) because of elevated risk of BPD. SEARCH METHODS We used standard methods of the Cochrane Neonatal Review Group. We searched MEDLINE, Embase, Healthstar and the Cochrane Central Register of Controlled Trials in the Cochrane Library through January 2016. We also searched the abstracts of the Pediatric Academic Societies. SELECTION CRITERIA Eligible for inclusion were randomised and quasi-randomised studies in preterm infants with respiratory disease that compared effects of iNO gas versus control, with or without placebo. DATA COLLECTION AND ANALYSIS We used standard methods of the Cochrane Neonatal Review Group and applied the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach to assess the quality of evidence. MAIN RESULTS We found 17 randomised controlled trials of iNO therapy in preterm infants. We grouped these trials post hoc into three categories on the basis of entry criteria: treatment during the first three days of life for impaired oxygenation, routine use in preterm babies along with respiratory support and later treatment for infants at increased risk for bronchopulmonary dysplasia (BPD). We performed no overall analyses.Eight trials providing early rescue treatment for infants on the basis of oxygenation criteria demonstrated no significant effect of iNO on mortality or BPD (typical risk ratio (RR) 0.94, 95% confidence interval (CI) 0.87 to 1.01; 958 infants). Four studies examining routine use of iNO in infants with pulmonary disease reported no significant reduction in death or BPD (typical RR 0.94, 95% CI 0.87 to 1.02; 1924 infants), although this small effect approached significance. Later treatment with iNO based on risk of BPD (three trials) revealed no significant benefit for this outcome in analyses of summary data (typical RR 0.92, 95% CI 0.85 to 1.01; 1075 infants).Investigators found no clear effect of iNO on the frequency of all grades of IVH nor severe IVH. Early rescue treatment was associated with a non-significant 20% increase in severe IVH.We found no effect on the incidence of neurodevelopmental impairment. AUTHORS' CONCLUSIONS iNO does not appear to be effective as rescue therapy for the very ill preterm infant. Early routine use of iNO in preterm infants with respiratory disease does not prevent serious brain injury or improve survival without BPD. Later use of iNO to prevent BPD could be effective, but current 95% confidence intervals include no effect; the effect size is likely small (RR 0.92) and requires further study.
Collapse
Affiliation(s)
- Keith J Barrington
- CHU Ste‐JustineDepartment of Pediatrics3175 Cote Ste CatherineMontrealQCCanadaH3T 1C5
| | - Neil Finer
- University of California San DiegoDepartment of Pediatrics200 W Arbor DrSan DiegoCaliforniaUSA92103‐8774
| | | | | |
Collapse
|
22
|
Gien J, Tseng N, Seedorf G, Kuhn K, Abman SH. Endothelin-1-Rho kinase interactions impair lung structure and cause pulmonary hypertension after bleomycin exposure in neonatal rat pups. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1090-L1100. [PMID: 27760762 PMCID: PMC5206397 DOI: 10.1152/ajplung.00066.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 10/10/2016] [Indexed: 02/08/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the chronic lung disease associated with premature birth, characterized by impaired vascular and alveolar growth. In neonatal rats bleomycin decreases lung growth and causes pulmonary hypertension (PH), which is poorly responsive to nitric oxide. In the developing lung, through Rho kinase (ROCK) activation, ET-1 impairs endothelial cell function; however, whether ET-1-ROCK interactions contribute to impaired vascular and alveolar growth in experimental BPD is unknown. Neonatal rats were treated daily with intraperitoneal bleomycin with and without selective ETA (BQ123/BQ610) and ETB (BQ788) receptor blockers, nonselective ET receptor blocker (ETRB) (bosentan), or fasudil (ROCK inhibitor). At day 14, lungs were harvested for morphometrics, and measurements of Fulton's index (RV/LV+S), medial wall thickness (MWT), and vessel density. Lung ET-1 protein and ROCK activity (phospho-MYPT-1:total MYPT-1 ratio) were also measured by Western blot analysis. Bleomycin increased lung ET-1 protein expression by 65%, RV/LV+S by 60%, mean linear intercept (MLI) by 212%, and MWT by 140% and decreased radial alveolar count (RAC) and vessel density by 40 and 44%, respectively (P < 0.01 for each comparison). After bleomycin treatment, fasudil and bosentan partially restored RAC and vessel density and decreased MLI, RV/LV+S, and MWT to normal values. Bleomycin increased ROCK activity by 120%, which was restored to normal values by bosentan but not selective ETRB. We conclude that ET-1-ROCK interactions contribute to decreased alveolar and vascular growth and PH in experimental BPD. We speculate that nonselective ETRB and ROCK inhibitors may be effective in the treatment of infants with BPD and PH.
Collapse
Affiliation(s)
- Jason Gien
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado;
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Nancy Tseng
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Gregory Seedorf
- Section of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; and
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine Kuhn
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Steven H Abman
- Section of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; and
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
23
|
Early inhaled nitric oxide in preterm infants <34 weeks with evolving bronchopulmonary dysplasia. J Perinatol 2016; 36:883-9. [PMID: 27442155 DOI: 10.1038/jp.2016.112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/08/2016] [Accepted: 04/18/2016] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To investigate whether early treatment with inhaled nitric oxide (iNO) could prevent bronchopulmonary dysplasia (BPD) in very preterm infants. STUDY DESIGN A non-randomized, controlled trial was conducted prospectively in 27 neonatal intensive care units over 12 months. Preterm infants with gestational age <34 weeks and after 7 days of life, who received invasive mechanical ventilation (MV) or nasal continuous positive airway pressure for >2 days, were treated either with low-dose iNO (from 5 as initial dose to 2 parts per million as maintenance dose for ⩾7 days, n=162) or as non-placebo control (n=240). Primary outcome was the incidence of moderate-to-severe BPD at 36 weeks postmenstrual age and/or death before discharge. Secondary outcomes were major complications. RESULTS iNO was started on average on day 19 of life (median duration 18 days, range 7 to 55 days). Rate of survival without BPD was significantly lower in the iNO than in the control group, whereas overall rates of BPD, death and major complications were similar between the two groups. Infants who started MV and iNO on postnatal days 15 to 21 had significantly increased survival without BPD (47.6% vs 17.1%, P=0.03, relative risk 2.7, 95% confidence interval 1.1 to 6.5). Additionally, pooled data from both groups showed that rates of perinatal co-morbidities and postnatal complications were higher in BPD infants than in non-BPD infants. The overall incidence of BPD was 55.6% and 75.9% for birth weight <1500 and <1000 g, respectively, or 1.6% for the total population <34 weeks of gestation admitted through the network. CONCLUSION Treatment with low-dose iNO did not decrease the overall risk of BPD and death nor showed adverse effects in short-term morbidities among very preterm infants. The benefit of delayed iNO treatment on BPD warrants further studies.
Collapse
|
24
|
Sokol GM, Konduri GG, Van Meurs KP. Inhaled nitric oxide therapy for pulmonary disorders of the term and preterm infant. Semin Perinatol 2016; 40:356-369. [PMID: 27480246 PMCID: PMC5065760 DOI: 10.1053/j.semperi.2016.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The 21st century began with the FDA approval of inhaled nitric oxide therapy for the treatment of neonatal hypoxic respiratory failure associated with pulmonary hypertension in recognition of the 2 randomized clinical trials demostrating a significant reduction in the need for extracorporeal support in the term and near-term infant. Inhaled nitric oxide is one of only a few therapeutic agents approved for use through clinical investigations primarily in the neonate. This article provides an overview of the pertinent biology and chemistry of nitric oxide, discusses potential toxicities, and reviews the results of pertinent clinical investigations and large randomized clinical trials including neurodevelopmental follow-up in term and preterm neonates. The clinical investigations conducted by the Eunice Kennedy Shriver NICHD Neonatal Research Network will be discussed and placed in context with other pertinent clinical investigations exploring the efficacy of inhaled nitric oxide therapy in neonatal hypoxic respiratory failure.
Collapse
Affiliation(s)
- Gregory M. Sokol
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46254
| | - G. Ganesh Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Krisa P. Van Meurs
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA 94304
| |
Collapse
|
25
|
Hajian B, De Backer J, Vos W, Van Holsbeke C, Ferreira F, Quinn DA, Hufkens A, Claes R, De Backer W. Pulmonary vascular effects of pulsed inhaled nitric oxide in COPD patients with pulmonary hypertension. Int J Chron Obstruct Pulmon Dis 2016; 11:1533-41. [PMID: 27462149 PMCID: PMC4940019 DOI: 10.2147/copd.s106480] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Severe chronic obstructive pulmonary disease (COPD) is often associated with secondary pulmonary hypertension (PH), which worsens prognosis. PH can be lowered by oxygen, but also by inhaled nitric oxide (NO), which has the potential to improve the health status of these patients. NO is an important mediator in vascular reactions in the pulmonary circulation. Oral compounds can act through NO-mediated pathways, but delivering pulsed inhaled NO (iNO) directly to the airways and pulmonary vasculature could equally benefit patients. Therefore, a proof-of-concept study was performed to quantify pulmonary blood vessel caliber changes after iNO administration using computed tomography (CT)-based functional respiratory imaging (FRI). Methods Six patients with secondary PH due to COPD received “pulsed” iNO in combination with oxygen for 20 minutes via a nasal cannula. Patients underwent a high-resolution CT scan with contrast before and after iNO. Using FRI, changes in volumes of blood vessels and associated lobes were quantified. Oxygen saturation and blood pressure were monitored and patients were asked about their subjective feelings. Results Pulmonary blood vessel volume increased by 7.06%±5.37% after iNO. A strong correlation (Ω20=0.32, P=0.002) was obtained between ventilation and observed vasodilation, suggesting that using the pulsed system, iNO is directed toward the ventilated zones, which consequently experience more vasodilation. Patients did not develop oxygen desaturation, remained normotensive, and perceived an improvement in their dyspnea sensation. Conclusion Inhalation of pulsed NO with oxygen causes vasodilation in the pulmonary circulation of COPD patients, mainly in the well-ventilated areas. A high degree of heterogeneity was found in the level of vasodilation. Patients tend to feel better after the treatment. Chronic use trials are warranted.
Collapse
Affiliation(s)
- Bita Hajian
- Department of Respiratory Medicine, University Hospital Antwerp, Edegem
| | | | - Wim Vos
- FluidDA nv, Antwerp, Belgium
| | | | | | | | - Annemie Hufkens
- Department of Respiratory Medicine, University Hospital Antwerp, Edegem
| | - Rita Claes
- Department of Respiratory Medicine, University Hospital Antwerp, Edegem
| | | |
Collapse
|
26
|
Seedorf G, Metoxen AJ, Rock R, Markham N, Ryan S, Vu T, Abman SH. Hepatocyte growth factor as a downstream mediator of vascular endothelial growth factor-dependent preservation of growth in the developing lung. Am J Physiol Lung Cell Mol Physiol 2016; 310:L1098-110. [PMID: 27036872 PMCID: PMC4935471 DOI: 10.1152/ajplung.00423.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 03/29/2016] [Indexed: 01/18/2023] Open
Abstract
Impaired vascular endothelial growth factor (VEGF) signaling contributes to the pathogenesis of bronchopulmonary dysplasia (BPD). We hypothesized that the effects of VEGF on lung structure during development may be mediated through its downstream effects on both endothelial nitric oxide synthase (eNOS) and hepatocyte growth factor (HGF) activity, and that, in the absence of eNOS, trophic effects of VEGF would be mediated through HGF signaling. To test this hypothesis, we performed an integrative series of in vitro (fetal rat lung explants and isolated fetal alveolar and endothelial cells) and in vivo studies with normal rat pups and eNOS(-/-) mice. Compared with controls, fetal lung explants from eNOS(-/-) mice had decreased terminal lung bud formation, which was restored with recombinant human VEGF (rhVEGF) treatment. Neonatal eNOS(-/-) mice were more susceptible to hyperoxia-induced inhibition of lung growth than controls, which was prevented with rhVEGF treatment. Fetal alveolar type II (AT2) cell proliferation was increased with rhVEGF treatment only with mesenchymal cell (MC) coculture, and these effects were attenuated with anti-HGF antibody treatment. Unlike VEGF, HGF directly stimulated isolated AT2 cells even without MC coculture. HGF directly stimulates fetal pulmonary artery endothelial cell growth and tube formation, which is attenuated by treatment with JNJ-38877605, a c-Met inhibitor. rHGF treatment preserves alveolar and vascular growth after postnatal exposure to SU-5416, a VEGF receptor inhibitor. We conclude that the effects of VEGF on AT2 and endothelial cells during lung development are partly mediated through HGF-c-Met signaling and speculate that reciprocal VEGF-HGF signaling between epithelia and endothelia is disrupted in infants who develop BPD.
Collapse
Affiliation(s)
- Gregory Seedorf
- Pediatric Heart Lung Center and Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; and
| | - Alexander J Metoxen
- Pediatric Heart Lung Center and Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; and
| | - Robert Rock
- Pediatric Heart Lung Center and Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; and
| | - Neil Markham
- Pediatric Heart Lung Center and Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; and
| | - Sharon Ryan
- Pediatric Heart Lung Center and Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; and
| | - Thiennu Vu
- Department of Medicine, University of California, San Francisco, California
| | - Steven H Abman
- Pediatric Heart Lung Center and Department of Pediatrics, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado; and
| |
Collapse
|
27
|
Randomized Trial of Late Surfactant Treatment in Ventilated Preterm Infants Receiving Inhaled Nitric Oxide. J Pediatr 2016; 168:23-29.e4. [PMID: 26500107 PMCID: PMC4698022 DOI: 10.1016/j.jpeds.2015.09.031] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 08/12/2015] [Accepted: 09/09/2015] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess whether late surfactant treatment in extremely low gestational age (GA) newborn infants requiring ventilation at 7-14 days, who often have surfactant deficiency and dysfunction, safely improves survival without bronchopulmonary dysplasia (BPD). STUDY DESIGN Extremely low GA newborn infants (GA ≤28 0/7 weeks) who required mechanical ventilation at 7-14 days were enrolled in a randomized, masked controlled trial at 25 US centers. All infants received inhaled nitric oxide and either surfactant (calfactant/Infasurf) or sham instillation every 1-3 days to a maximum of 5 doses while intubated. The primary outcome was survival at 36 weeks postmenstrual age (PMA) without BPD, as evaluated by physiological oxygen/flow reduction. RESULTS A total of 511 infants were enrolled between January 2010 and September 2013. There were no differences between the treated and control groups in mean birth weight (701 ± 164 g), GA (25.2 ± 1.2 weeks), percentage born at GA <26 weeks (70.6%), race, sex, severity of lung disease at enrollment, or comorbidities of prematurity. Survival without BPD did not differ between the treated and control groups at 36 weeks PMA (31.3% vs 31.7%; relative benefit, 0.98; 95% CI, 0.75-1.28; P = .89) or 40 weeks PMA (58.7% vs 54.1%; relative benefit, 1.08; 95% CI, 0.92-1.27; P = .33). There were no between-group differences in serious adverse events, comorbidities of prematurity, or severity of lung disease to 36 weeks. CONCLUSION Late treatment with up to 5 doses of surfactant in ventilated premature infants receiving inhaled nitric oxide was well tolerated, but did not improve survival without BPD at 36 or 40 weeks. Pulmonary and neurodevelopmental assessments are ongoing. TRIAL REGISTRATION ClinicalTrials.gov: NCT01022580.
Collapse
|
28
|
Jobe AH. Animal Models, Learning Lessons to Prevent and Treat Neonatal Chronic Lung Disease. Front Med (Lausanne) 2015; 2:49. [PMID: 26301222 PMCID: PMC4528292 DOI: 10.3389/fmed.2015.00049] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/14/2015] [Indexed: 11/23/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a unique injury syndrome caused by prolonged injury and repair imposed on an immature and developing lung. The decreased septation and decreased microvascular development phenotype of BPD can be reproduced in newborn rodents with increased chronic oxygen exposure and in premature primates and sheep with oxygen and/or mechanical ventilation. The inflammation caused by oxidants, inflammatory agonists, and/or stretch injury from mechanical ventilation seems to promote the anatomic abnormalities. Multiple interventions targeted to specific inflammatory cells or pathways or targeted to decreasing ventilation-mediated injury can substantially prevent the anatomic changes associated with BPD in term rodents and in preterm sheep or primate models. Most of the anti-inflammatory therapies with benefit in animal models have not been tested clinically. None of the interventions that have been tested clinically are as effective as anticipated from the animal models. These inconsistencies in responses likely are explained by the antenatal differences in lung exposures of the developing animals relative to very preterm humans. The animals generally have normal lungs while the lungs of preterm infants are exposed variably to intrauterine inflammation, growth abnormalities, antenatal corticosteroids, and poorly understood effects from the causes of preterm delivery. The animal models have been essential for the definition of the mediators that can cause a BPD phenotype. These models will be necessary to develop and test future-targeted interventions to prevent and treat BPD.
Collapse
Affiliation(s)
- Alan H Jobe
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati , Cincinnati, OH , USA
| |
Collapse
|
29
|
Abstract
Bronchopulmonary dysplasia (BPD), the chronic lung disease associated with preterm birth, results from the disruption of normal pulmonary vascular and alveolar growth. Though BPD was once described as primarily due to postnatal injury from mechanical ventilation and oxygen therapy after preterm birth, it is increasingly appreciated that BPD results from antenatal and perinatal factors that interrupt lung development in infants born at the extremes of prematurity. The lung in BPD consists of a simplified parenchymal architecture that limits gas exchange and leads to increased cardiopulmonary morbidity and mortality. This review outlines recent advances in the understanding of pulmonary vascular development and describes how the disruption of these mechanisms results in BPD. We point to future therapies that may augment postnatal vascular growth to prevent and treat this severe chronic lung disease.
Collapse
Affiliation(s)
- Christopher D Baker
- Pediatric Heart Lung Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo., USA
| | | |
Collapse
|
30
|
Albertine KH. Utility of large-animal models of BPD: chronically ventilated preterm lambs. Am J Physiol Lung Cell Mol Physiol 2015; 308:L983-L1001. [PMID: 25770179 PMCID: PMC4437012 DOI: 10.1152/ajplung.00178.2014] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 02/19/2015] [Indexed: 11/22/2022] Open
Abstract
This paper is focused on unique insights provided by the preterm lamb physiological model of bronchopulmonary dysplasia (BPD). Connections are also made to insights provided by the former preterm baboon model of BPD, as well as to rodent models of lung injury to the immature, postnatal lung. The preterm lamb and baboon models recapitulate the clinical setting of preterm birth and respiratory failure that require prolonged ventilation support for days or weeks with oxygen-rich gas. An advantage of the preterm lamb model is the large size of preterm lambs, which facilitates physiological studies for days or weeks during the evolution of neonatal chronic lung disease (CLD). To this advantage is linked an integrated array of morphological, biochemical, and molecular analyses that are identifying the role of individual genes in the pathogenesis of neonatal CLD. Results indicate that the mode of ventilation, invasive mechanical ventilation vs. less invasive high-frequency nasal ventilation, is related to outcomes. Our approach also includes pharmacological interventions that test causality of specific molecular players, such as vitamin A supplementation in the pathogenesis of neonatal CLD. The new insights that are being gained from our preterm lamb model may have important translational implications about the pathogenesis and treatment of BPD in preterm human infants.
Collapse
Affiliation(s)
- Kurt H Albertine
- Department of Pediatrics, University of Utah, School of Medicine, Salt Lake City, Utah; Department of Medicine, University of Utah, School of Medicine, Salt Lake City, Utah; and Department of Neurobiology and Anatomy, University of Utah, School of Medicine, Salt Lake City, Utah
| |
Collapse
|
31
|
Farrow KN, Steinhorn RH. Pulmonary hypertension in premature infants. Sharpening the tools of detection. Am J Respir Crit Care Med 2015; 191:12-4. [PMID: 25551345 DOI: 10.1164/rccm.201411-2112ed] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kathryn N Farrow
- 1 Department of Pediatrics Northwestern University Feinberg School of Medicine Chicago, Illinois
| | | |
Collapse
|
32
|
Ballard PL, Keller RL, Black DM, Durand DJ, Merrill JD, Eichenwald EC, Truog WE, Mammel MC, Steinhorn R, Ryan RM, Courtney SE, Horneman H, Ballard RA, Investigators of TOLSURF Pilot and TOLSURF. Inhaled nitric oxide increases urinary nitric oxide metabolites and cyclic guanosine monophosphate in premature infants: relationship to pulmonary outcome. Am J Perinatol 2015; 32:225-32. [PMID: 24968129 PMCID: PMC5032843 DOI: 10.1055/s-0034-1382255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Inhaled nitric oxide (iNO) has been tested to prevent bronchopulmonary dysplasia (BPD) in premature infants, however, the role of cyclic guanosine monophosphate (cGMP) is not known. We hypothesized that levels of NO metabolites (NOx) and cGMP in urine, as a noninvasive source for biospecimen collection, would reflect the dose of iNO and relate to pulmonary outcome. STUDY DESIGN Studies were performed on 125 infants who required mechanical ventilation at 7 to 14 days and received 24 days of iNO at 20-2 ppm. A control group of 19 infants did not receive iNO. RESULTS In NO-treated infants there was a dose-dependent increase of both NOx and cGMP per creatinine (maximal 3.1- and 2-fold, respectively, at 10-20 ppm iNO) compared with off iNO. NOx and cGMP concentrations at both 2 ppm and off iNO were inversely related to severity of lung disease during the 1st month, and the NOx levels were lower in infants who died or developed BPD at term. NOx was higher in Caucasian compared with other infants at all iNO doses. CONCLUSION Urinary NOx and cGMP are biomarkers of endogenous NO production and lung uptake of iNO, and some levels reflect the severity of lung disease. These results support a role of the NO-cGMP pathway in lung development.
Collapse
Affiliation(s)
- Philip L. Ballard
- Department of Pediatrics, University of California, San Francisco, California
| | - Roberta L. Keller
- Department of Pediatrics, University of California, San Francisco, California
| | - Dennis M. Black
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - David J. Durand
- Division of Neonatology, Children’s Hospital and Research Center Oakland, Oakland, California
| | - Jeffrey D. Merrill
- Division of Neonatology, Children’s Hospital and Research Center Oakland, Oakland, California
| | - Eric C. Eichenwald
- Department of Pediatrics, University of Texas Medical School, Houston, Texas
| | - William E. Truog
- Department of Pediatrics, Children’s Mercy Hospitals and Clinics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Mark C. Mammel
- Department of Pediatrics, Children’s Hospital and Clinics of Minnesota, St. Paul, Minnesota
| | - Robin Steinhorn
- Department of Pediatrics, University of California Davis Children’s Hospital, Sacramento, California
| | - Rita M. Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Sherry E. Courtney
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Hart Horneman
- Department of Pediatrics, University of California, San Francisco, California
| | - Roberta A. Ballard
- Department of Pediatrics, University of California, San Francisco, California
| | | |
Collapse
|
33
|
Bronchopulmonary dysplasia: NHLBI Workshop on the Primary Prevention of Chronic Lung Diseases. Ann Am Thorac Soc 2015; 11 Suppl 3:S146-53. [PMID: 24754823 DOI: 10.1513/annalsats.201312-424ld] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of extreme preterm birth. Infants who develop BPD manifest aberrant or arrested pulmonary development and can experience lifelong alterations in cardiopulmonary function. Despite decades of promising research, primary prevention of BPD has proven elusive. This workshop report identifies current barriers to the conduct of primary prevention studies for BPD and causal pathways implicated in BPD pathogenesis. Throughout, we highlight promising areas for research to improve understanding of normal and aberrant lung development, distinguish BPD endotypes, and ascertain biomarkers for more targeted therapeutic approaches to prevention. We conclude with research recommendations and priorities to accelerate discovery and promote lung health in infants born preterm.
Collapse
|
34
|
Jensen EA, Kirpalani H. Inhaled nitric oxide to prevent bronchopulmonary dysplasia in preterm infants--less than a silver bullet. J Pediatr 2014; 165:1079-81. [PMID: 25266344 DOI: 10.1016/j.jpeds.2014.08.040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Accepted: 08/20/2014] [Indexed: 11/16/2022]
Affiliation(s)
- Erik A Jensen
- Division of Neonatology Department of Pediatrics, The Children's Hospital of Philadelphia The University of Pennsylvania, Philadelphia, Pennsylvania
| | - Haresh Kirpalani
- Division of Neonatology Department of Pediatrics, The Children's Hospital of Philadelphia The University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
35
|
Hilgendorff A, Reiss I, Ehrhardt H, Eickelberg O, Alvira CM. Chronic lung disease in the preterm infant. Lessons learned from animal models. Am J Respir Cell Mol Biol 2014; 50:233-45. [PMID: 24024524 DOI: 10.1165/rcmb.2013-0014tr] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neonatal chronic lung disease, also known as bronchopulmonary dysplasia (BPD), is the most common complication of premature birth, affecting up to 30% of very low birth weight infants. Improved medical care has allowed for the survival of the most premature infants and has significantly changed the pathology of BPD from a disease marked by severe lung injury to the "new" form characterized by alveolar hypoplasia and impaired vascular development. However, increased patient survival has led to a paucity of pathologic specimens available from infants with BPD. This, combined with the lack of a system to model alveolarization in vitro, has resulted in a great need for animal models that mimic key features of the disease. To this end, a number of animal models have been created by exposing the immature lung to injuries induced by hyperoxia, mechanical stretch, and inflammation and most recently by the genetic modification of mice. These animal studies have 1) allowed insight into the mechanisms that determine alveolar growth, 2) delineated factors central to the pathogenesis of neonatal chronic lung disease, and 3) informed the development of new therapies. In this review, we summarize the key findings and limitations of the most common animal models of BPD and discuss how knowledge obtained from these studies has informed clinical care. Future studies should aim to provide a more complete understanding of the pathways that preserve and repair alveolar growth during injury, which might be translated into novel strategies to treat lung diseases in infants and adults.
Collapse
Affiliation(s)
- Anne Hilgendorff
- 1 Department of Perinatology Grosshadern, Ludwig-Maximilian-University, Munich, Germany
| | | | | | | | | |
Collapse
|
36
|
Manzano RM, Mascaretti RS, Carrer V, Haddad LB, Fernandes AR, Reyes AMA, Rebello CM. A hyperoxic lung injury model in premature rabbits: the influence of different gestational ages and oxygen concentrations. PLoS One 2014; 9:e95844. [PMID: 24755658 PMCID: PMC3995887 DOI: 10.1371/journal.pone.0095844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 03/31/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Many animal models have been developed to study bronchopulmonary dysplasia (BPD). The preterm rabbit is a low-cost, easy-to-handle model, but it has a high mortality rate in response to the high oxygen concentrations used to induce lung injury. The aim of this study was to compare the mortality rates of two models of hyperoxia-induced lung injury in preterm rabbits. METHODS Pregnant New Zealand white rabbits were subjected to caesarean section on gestational day 28 or 29 (full term = 31 days). The premature rabbits in the 28-day gestation group were exposed to room air or FiO₂ ≥95%, and the rabbits in the 29-day gestation group were exposed to room air or FiO₂ = 80% for 11 days. The mean linear intercept (Lm), internal surface area (ISA), number of alveoli, septal thickness and proportion of elastic and collagen fibers were quantified. RESULTS The survival rates in the 29-day groups were improved compared with the 28-day groups. Hyperoxia impaired the normal development of the lung, as demonstrated by an increase in the Lm, the septal thickness and the proportion of elastic fibers. Hyperoxia also decreased the ISA, the number of alveoli and the proportion of collagen fibers in the 28-day oxygen-exposed group compared with the control 28-day group. A reduced number of alveoli was found in the 29-day oxygen exposed animals compared with the control 29-day group. CONCLUSIONS The 29-day preterm rabbits had a reduced mortality rate compared with the 28-day preterm rabbits and maintained a reduction in the alveoli number, which is comparable to BPD in humans.
Collapse
Affiliation(s)
| | | | - Valéria Carrer
- Albert Einstein Jewish Hospital, São Paulo, São Paulo, Brazil
| | | | | | | | - Celso Moura Rebello
- Albert Einstein Jewish Hospital, São Paulo, São Paulo, Brazil
- School of Medicine - Clinics Hospital University of Sao Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
37
|
Gadhia MM, Cutter GR, Abman SH, Kinsella JP. Effects of early inhaled nitric oxide therapy and vitamin A supplementation on the risk for bronchopulmonary dysplasia in premature newborns with respiratory failure. J Pediatr 2014; 164:744-8. [PMID: 24388327 PMCID: PMC3962699 DOI: 10.1016/j.jpeds.2013.11.040] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 10/02/2013] [Accepted: 11/18/2013] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To assess whether the combination of early inhaled nitric oxide (iNO) therapy and vitamin A supplementation lowers the incidence of bronchopulmonary dysplasia (BPD) in premature newborns with respiratory failure. STUDY DESIGN A total of 793 mechanically ventilated infants (birth weight 500-1250 g) were randomized (after stratification by birth weight) to receive placebo or iNO (5 ppm) for 21 days or until extubation (500-749, 750-999, or 1000-1250 g). A total of 398 newborns received iNO, and of these, 118 (30%) received vitamin A according to their enrollment center. We compared patients who received iNO + vitamin A with those who received iNO alone. The primary outcome was a composite of death or BPD at 36 weeks postconceptual age. RESULTS BPD was reduced in infants who received iNO + vitamin A for the 750-999 g birth weight group compared with iNO alone (P = .01). This group also showed a reduction in the combined outcome of BPD + death compared with iNO alone (P = .01). The use of vitamin A did not change the risk for BPD in the placebo group. Overall, the use of vitamin A was low (229 of 793 patients, or 29%). Combined therapy improved Bayley Scales of Infant Development II Mental and Psychomotor Developmental Index scores at 1 year compared with infants treated solely with iNO for the 500-749 g birth weight group. CONCLUSIONS In this retrospective analysis of the nonrandomized use of vitamin A, combined iNO + vitamin A therapy in preterm infants with birth weight 750-999 g reduced the incidence of BPD and BPD + death and improved neurocognitive outcomes at 1 year in the 500-749 g birth weight group.
Collapse
Affiliation(s)
- Monika M Gadhia
- Division of Neonatology, Department of Pediatrics, Weill Cornell Medical College, New York, NY.
| | - Gary R Cutter
- Department of Biostatistics, University of Alabama, Birmingham, AL
| | - Steven H Abman
- Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO; Section of Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - John P Kinsella
- Pediatric Heart Lung Center, University of Colorado School of Medicine, Aurora, CO; Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
38
|
Vadivel A, Alphonse RS, Ionescu L, Machado DS, O’Reilly M, Eaton F, Haromy A, Michelakis ED, Thébaud B. Exogenous hydrogen sulfide (H2S) protects alveolar growth in experimental O2-induced neonatal lung injury. PLoS One 2014; 9:e90965. [PMID: 24603989 PMCID: PMC3946270 DOI: 10.1371/journal.pone.0090965] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/05/2014] [Indexed: 01/02/2023] Open
Abstract
Background Bronchopulmonary dysplasia (BPD), the chronic lung disease of prematurity, remains a major health problem. BPD is characterized by impaired alveolar development and complicated by pulmonary hypertension (PHT). Currently there is no specific treatment for BPD. Hydrogen sulfide (H2S), carbon monoxide and nitric oxide (NO), belong to a class of endogenously synthesized gaseous molecules referred to as gasotransmitters. While inhaled NO is already used for the treatment of neonatal PHT and currently tested for the prevention of BPD, H2S has until recently been regarded exclusively as a toxic gas. Recent evidence suggests that endogenous H2S exerts beneficial biological effects, including cytoprotection and vasodilatation. We hypothesized that H2S preserves normal alveolar development and prevents PHT in experimental BPD. Methods We took advantage of a recently described slow-releasing H2S donor, GYY4137 (morpholin-4-ium-4-methoxyphenyl(morpholino) phosphinodithioate) to study its lung protective potential in vitro and in vivo. Results In vitro, GYY4137 promoted capillary-like network formation, viability and reduced reactive oxygen species in hyperoxia-exposed human pulmonary artery endothelial cells. GYY4137 also protected mitochondrial function in alveolar epithelial cells. In vivo, GYY4137 preserved and restored normal alveolar growth in rat pups exposed from birth for 2 weeks to hyperoxia. GYY4137 also attenuated PHT as determined by improved pulmonary arterial acceleration time on echo-Doppler, pulmonary artery remodeling and right ventricular hypertrophy. GYY4137 also prevented pulmonary artery smooth muscle cell proliferation. Conclusions H2S protects from impaired alveolar growth and PHT in experimental O2-induced lung injury. H2S warrants further investigation as a new therapeutic target for alveolar damage and PHT.
Collapse
Affiliation(s)
- Arul Vadivel
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research, Regenerative Medicine Program and Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
| | - Rajesh S. Alphonse
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Lavinia Ionescu
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Desiree S. Machado
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Megan O’Reilly
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Farah Eaton
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Al Haromy
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Evangelos D. Michelakis
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Cardiovascular Research Center and Pulmonary Research Group, University of Alberta, Edmonton, Canada
| | - Bernard Thébaud
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research, Regenerative Medicine Program and Children’s Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail:
| |
Collapse
|
39
|
Ahlfeld SK, Conway SJ. Assessment of inhibited alveolar-capillary membrane structural development and function in bronchopulmonary dysplasia. ACTA ACUST UNITED AC 2014; 100:168-79. [PMID: 24604816 DOI: 10.1002/bdra.23226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 01/17/2014] [Accepted: 01/19/2014] [Indexed: 12/20/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of extreme prematurity and is defined clinically by dependence on supplemental oxygen due to impaired gas exchange. Optimal gas exchange is dependent on the development of a sufficient surface area for diffusion. In the mammalian lung, rapid acquisition of distal lung surface area is accomplished in neonatal and early adult life by means of vascularization and secondary septation of distal lung airspaces. Extreme preterm birth interrupts secondary septation and pulmonary capillary development and ultimately reduces the efficiency of the alveolar-capillary membrane. Although pulmonary health in BPD infants rapidly improves over the first few years, persistent alveolar-capillary membrane dysfunction continues into adolescence and adulthood. Preventative therapies have been largely ineffective, and therapies aimed at promoting normal development of the air-blood barrier in infants with established BPD remain largely unexplored. The purpose of this review will be: (1) to summarize the histological evidence of aberrant alveolar-capillary membrane development associated with extreme preterm birth and BPD, (2) to review the clinical evidence assessing the long-term impact of BPD on alveolar-capillary membrane function, and (3) to discuss the need to develop and incorporate direct measurements of functional gas exchange into clinically relevant animal models of inhibited alveolar development.
Collapse
Affiliation(s)
- Shawn K Ahlfeld
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | | |
Collapse
|
40
|
Baker CD, Abman SH, Mourani PM. Pulmonary Hypertension in Preterm Infants with Bronchopulmonary Dysplasia. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2014; 27:8-16. [PMID: 24669351 DOI: 10.1089/ped.2013.0323] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/16/2014] [Indexed: 01/02/2023]
Abstract
Bronchopulmonary dysplasia (BPD), the chronic lung disease of prematurity, is a significant contributor to perinatal morbidity and mortality. Premature birth disrupts pulmonary vascular growth and initiates a cascade of events that result in impaired gas exchange, abnormal vasoreactivity, and pulmonary vascular remodeling that may ultimately lead to pulmonary hypertension (PH). Even infants who appear to have mild BPD suffer from varying degrees of pulmonary vascular disease (PVD). Although recent studies have enhanced our understanding of the pathobiology of PVD and PH in BPD, much remains unknown with respect to how PH should be properly defined, as well as the most accurate methods for the diagnosis and treatment of PH in infants with BPD. This article will provide neonatologists and primary care providers, as well as pediatric cardiologists and pulmonologists, with a review of the pathophysiology of PH in preterm infants with BPD and a summary of current clinical recommendations for managing PH in this population.
Collapse
Affiliation(s)
- Christopher D Baker
- Section of Pulmonary Medicine, University of Colorado School of Medicine , Aurora, Colarado. ; Pediatric Heart Lung Center, University of Colorado School of Medicine , Aurora, Colarado
| | - Steven H Abman
- Section of Pulmonary Medicine, University of Colorado School of Medicine , Aurora, Colarado. ; Pediatric Heart Lung Center, University of Colorado School of Medicine , Aurora, Colarado
| | - Peter M Mourani
- Pediatric Heart Lung Center, University of Colorado School of Medicine , Aurora, Colarado. ; Section of Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine , Aurora, Colarado
| |
Collapse
|
41
|
Generation of purified nitric oxide from liquid N2O4 for the treatment of pulmonary hypertension in hypoxemic swine. Nitric Oxide 2014; 37:66-72. [PMID: 24513304 DOI: 10.1016/j.niox.2014.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/13/2014] [Accepted: 02/02/2014] [Indexed: 12/23/2022]
Abstract
Inhaled nitric oxide (NO) selectively dilates pulmonary blood vessels, reduces pulmonary vascular resistance (PVR), and enhances ventilation-perfusion matching. However, existing modes of delivery for the treatment of chronic pulmonary hypertension are limited due to the bulk and heft of large tanks of compressed gas. We present a novel system for the generation of inhaled NO that is based on the initial heat-induced evaporation of liquid N2O4 into gas phase NO2 followed by the room temperature reduction to NO by an antioxidant, ascorbic acid cartridge just prior to inhalation. The biologic effects of NO generated from liquid N2O4 were compared with the effects of NO gas, on increased mean pulmonary artery pressure (mPAP) and PVR in a hypoxemic (FiO2 15%) swine model of pulmonary hypertension. We showed that NO concentration varied directly with the fixed cross sectional flow of the outflow aperture when studied at temperatures of 45, 47.5 and 50°C and was independent of the rate of heating. Liquid N2O4-sourced NO at 1, 5, and 20 ppm significantly reduced the elevated mPAP and PVR induced by experimental hypoxemia and was biologically indistinguishable from gas source NO in this model. These experiments show that it is feasible to generate highly purified NO gas from small volumes of liquid N2O4 at concentrations sufficient to lower mPAP and PVR in hypoxemic swine, and suggest that a miniaturized ambulatory system designed to generate biologically active NO from liquid N2O4 is achievable.
Collapse
|
42
|
Abstract
Nitric oxide, an important signaling molecule with multiple regulatory effects throughout the body, is an important tool for the treatment of full-term and late-preterm infants with persistent pulmonary hypertension of the newborn and hypoxemic respiratory failure. Several randomized controlled trials have evaluated its role in the management of preterm infants ≤ 34 weeks' gestational age with varying results. The purpose of this clinical report is to summarize the existing evidence for the use of inhaled nitric oxide in preterm infants and provide guidance regarding its use in this population.
Collapse
|
43
|
Poon CY, Edwards MO, Kotecha S. Long term cardiovascular consequences of chronic lung disease of prematurity. Paediatr Respir Rev 2013; 14:242-9. [PMID: 24209460 DOI: 10.1016/j.prrv.2012.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pulmonary arterial (PA) hypertension in preterm infant is an important consequence of chronic lung disease of prematurity (CLD) arising mainly due to impaired alveolar development and dysregulated angiogenesis of the pulmonary circulation. Although PA pressure and resistance in these children normalise by school age, their pulmonary vasculature remains hyper-reactive to hypoxia until early childhood. Furthermore, there is evidence that systemic blood pressure in preterm born children with or without CLD is mildly increased at school age and in young adulthood when compared to term-born children. Arterial stiffness may be increased in CLD survivors due to increased smooth muscle tone of the pre-resistance and resistance vessels rather than the loss of elasticity in the large arteries. This review explores the long term effects of CLD on the pulmonary and systemic circulations along with their clinical correlates and therapeutic approaches.
Collapse
Affiliation(s)
- Chuen Yeow Poon
- Department of Child Health, School of Medicine, Cardiff University, University Hospital of Wales, Heath Park, Cardiff CF14 4XN, UK.
| | | | | |
Collapse
|
44
|
Bachiller PR, Cornog KH, Kato R, Buys ES, Roberts JD. Soluble guanylate cyclase modulates alveolarization in the newborn lung. Am J Physiol Lung Cell Mol Physiol 2013; 305:L569-81. [PMID: 23934926 DOI: 10.1152/ajplung.00401.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nitric oxide (NO) regulates lung development through incompletely understood mechanisms. NO controls pulmonary vascular smooth muscle cell (SMC) differentiation largely through stimulating soluble guanylate cyclase (sGC) to produce cGMP and increase cGMP-mediated signaling. To examine the role of sGC in regulating pulmonary development, we tested whether decreased sGC activity reduces alveolarization in the normal and injured newborn lung. For these studies, mouse pups with gene-targeted sGC-α1 subunit truncation were used because we determined that they have decreased pulmonary sGC enzyme activity. sGC-α1 knockout (KO) mouse pups were observed to have decreased numbers of small airway structures and lung volume compared with wild-type (WT) mice although lung septation and body weights were not different. However, following mild lung injury caused by breathing 70% O2, the sGC-α1 KO mouse pups had pronounced inhibition of alveolarization, as evidenced by an increase in airway mean linear intercept, reduction in terminal airway units, and decrease in lung septation and alveolar openings, as well as reduced somatic growth. Because cGMP regulates SMC phenotype, we also tested whether decreased sGC activity reduces lung myofibroblast differentiation. Cellular markers revealed that vascular SMC differentiation decreased, whereas myofibroblast activation increased in the hyperoxic sGC-α1 KO pup lung. These results indicate that lung development, particularly during hyperoxic injury, is impaired in mouse pups with diminished sGC activity. These studies support the investigation of sGC-targeting agents as therapies directed at improving development in the newborn lung exposed to injury.
Collapse
Affiliation(s)
- Patricia R Bachiller
- Jr., Cardiovascular Research Center, Massachusetts General Hospital - East, 149 13 St., Charlestown, MA 02129.
| | | | | | | | | |
Collapse
|
45
|
Kato S, Zhang R, Roberts JD. Proprotein convertases play an important role in regulating PKGI endoproteolytic cleavage and nuclear transport. Am J Physiol Lung Cell Mol Physiol 2013; 305:L130-40. [PMID: 23686857 PMCID: PMC3726948 DOI: 10.1152/ajplung.00391.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 05/15/2013] [Indexed: 12/27/2022] Open
Abstract
Nitric oxide and cGMP modulate vascular smooth muscle cell (SMC) phenotype by regulating cell differentiation and proliferation. Recent studies suggest that cGMP-dependent protein kinase I (PKGI) cleavage and the nuclear translocation of a constitutively active kinase fragment, PKGIγ, are required for nuclear cGMP signaling in SMC. However, the mechanisms that control PKGI proteolysis are unknown. Inspection of the amino acid sequence of a PKGI cleavage site that yields PKGIγ and a protease database revealed a putative minimum consensus sequence for proprotein convertases (PCs). Therefore we investigated the role of PCs in regulating PKGI proteolysis. We observed that overexpression of PCs, furin and PC5, but not PC7, which are all expressed in SMC, increase PKGI cleavage in a dose-dependent manner in human embryonic kidney (HEK) 293 cells. Moreover, furin-induced proteolysis of mutant PKGI, in which alanines were substituted into the putative PC consensus sequence, was decreased in these cells. In addition, overexpression of furin increased PKGI proteolysis in LoVo cells, which is an adenocarcinoma cell line expressing defective furin without PC activity. Also, expression of α1-PDX, an engineered serpin-like PC inhibitor, reduced PC activity and decreased PKGI proteolysis in HEK293 cells. Last, treatment of low-passage rat aortic SMC with membrane-permeable PC inhibitor peptides decreased cGMP-stimulated nuclear PKGIγ translocation. These data indicate for the first time that PCs have a role in regulating PKGI proteolysis and the nuclear localization of its active cleavage product, which are important for cGMP-mediated SMC phenotype.
Collapse
Affiliation(s)
- Shin Kato
- Cardiovascular Research Center of the General Medical Services, Massachusetts General Hospital, Boston, MA, USA
| | | | | |
Collapse
|
46
|
Abstract
Bronchopulmonary dysplasia (BPD) is among the most common chronic lung diseases in infants in the US. Improved survival of preterm infants who developed BPD is becoming increasingly important because of the high risk for persistent pulmonary morbidities such as poor respiratory gas exchange, pulmonary hypertension, and excess airway expiratory resistance later in life. This review focuses on unique insights provided by the two large-animal, physiological models of neonatal chronic lung disease: preterm baboons and preterm lambs. The models' are valuable because they contribute to better understanding of the underlying molecular pathogenic mechanisms. An epigenetic hypothesis is proposed as a pathogenic mechanism for BPD and its persistent pulmonary morbidities.
Collapse
Affiliation(s)
- Kurt H. Albertine
- Departments of Pediatrics, Medicine, Neurobiology & Anatomy, University of Utah, School of Medicine, Salt Lake City, Utah 84158-1289
| |
Collapse
|
47
|
Dani C, Pratesi S. Nitric oxide for the treatment of preterm infants with respiratory distress syndrome. Expert Opin Pharmacother 2012. [PMID: 23194109 DOI: 10.1517/14656566.2013.746662] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Inhaled Nitric oxide (iNO) has been proposed as effective treatment for improving oxygenation in preterm infants with respiratory distress syndrome (RDS), and for preventing the development of bronchopulmonary dysplasia (BPD). AREAS COVERED This drug evaluation mainly reviews the results of clinical studies on the effects of iNO in preterm infants with RDS which have provided contradictory results probably due to their different designs. Three recent meta-analyses of these studies have concluded that iNO therapy is not effective in decreasing the risk of death and BPD and cannot be recommended as routine treatment. The same meta-analyses suggest that some strategy of iNO treatment and some subgroups of patients, such as infants with persistent pulmonary hypertension of the newborn (PPHN), should be further studied. EXPERT OPINION At present, the available evidence does not support the use of iNO in preterm infants with RDS, and iNO therapy cannot be recommended for the routine treatment of respiratory failure in premature neonates. In the future, further studies in selected populations using adequate doses and investigating the effectiveness of other drugs, such as sildenafil, might affect the use and diffusion of iNO.
Collapse
Affiliation(s)
- Carlo Dani
- Careggi University Hospital of Florence, Department of Surgical and Medical Critical Care, Section of Neonatology, Viale Morgagni, 85, 50134 Florence, Italy.
| | | |
Collapse
|
48
|
Alphonse RS, Rajabali S, Thébaud B. Lung injury in preterm neonates: the role and therapeutic potential of stem cells. Antioxid Redox Signal 2012; 17:1013-40. [PMID: 22400813 DOI: 10.1089/ars.2011.4267] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Continuous improvements in perinatal care have allowed the survival of ever more premature infants, making the task of protecting the extremely immature lung from injury increasingly challenging. Premature infants at risk of developing chronic lung disease or bronchopulmonary dysplasia (BPD) are now born at the late canalicular stage of lung development, just when the airways become juxtaposed to the lung vasculature and when gas-exchange becomes possible. Readily available strategies, including improved antenatal management (education, regionalization, steroids, and antibiotics), together with exogenous surfactant and exclusive/early noninvasive ventilatory support, will likely decrease the incidence/severity of BPD over the next few years. Nonetheless, because of the extreme immaturity of the developing lung, the extent to which disruption of lung growth after prematurity and neonatal management lead to an earlier or more aggravated decline in respiratory function in later life is a matter of concern. Consequently, much more needs to be learned about the mechanisms of lung development, injury, and repair. Recent insight into stem cell biology has sparked interest for stem cells to repair damaged organs. This review summarizes the exciting potential of stem cell-based therapies for lung diseases in general and BPD in particular.
Collapse
|
49
|
Raffay TM, Martin RJ, Reynolds JD. Can nitric oxide-based therapy prevent bronchopulmonary dysplasia? Clin Perinatol 2012; 39:613-38. [PMID: 22954273 PMCID: PMC3437658 DOI: 10.1016/j.clp.2012.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A growing understanding of endogenous nitric oxide (NO) biology is helping to explain how and when exogenous NO may confer benefit or harm; this knowledge is also helping to identify new better-targeted NO-based therapies. In this review, results of the bronchopulmonary dysplasia clinical trials that used inhaled NO in the preterm population are placed in context, the biologic basis for novel NO therapeutics is considered, and possible future directions for NO-focused clinical and basic research in developmental lung disease are identified.
Collapse
Affiliation(s)
- Thomas M. Raffay
- Division of Neonatology, Department of Pediatrics Rainbow Babies & Children’s Hospital, Case Medical Center/University Hospitals, Cleveland, Ohio
| | - Richard J. Martin
- Division of Neonatology, Department of Pediatrics Rainbow Babies & Children’s Hospital, Case Medical Center/University Hospitals, Cleveland, Ohio
| | - James D. Reynolds
- Department of Anesthesia and Perioperative Medicine, Case Medical Center/University Hospitals, Cleveland, Ohio
,Institute for Transformative Molecular Medicine, Case Medical Center/University Hospitals, Cleveland, Ohio
| |
Collapse
|
50
|
Postolow F, Fediuk J, Nolette N, Hinton M, Dakshinamurti S. Hypoxia and nitric oxide exposure promote apoptotic signaling in contractile pulmonary arterial smooth muscle but not in pulmonary epithelium. Pediatr Pulmonol 2011; 46:1194-208. [PMID: 21618721 DOI: 10.1002/ppul.21491] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 04/08/2011] [Accepted: 04/11/2011] [Indexed: 11/11/2022]
Abstract
RATIONALE Neonatal pulmonary hypertension is characterized by hypoxia, abnormal vascular remodeling, and impaired alveolarization. Nitric oxide (NO) regulates cell replication and activation of apoptosis. Our objective was to examine cell phenotype-specific effects of hypoxia and NO exposure on cumulative apoptotic signal in neonatal pulmonary epithelial cells and arterial smooth muscle. DESIGN/METHODS Primary cultured newborn porcine pulmonary arterial myocytes and epithelial cells were grown in normoxic (21% O2) or hypoxic conditions (10% O2). Myocyte phenotype was predetermined by serum-supplementation or -deprivation. Cells were exposed to sodium nitroprusside (10(-7) -10(-4) M) or diluent for 3 days. Cell survival was estimated by MTT assay; BAX, Bcl-2, and cleaved caspase-3 by Western blot; cell cycle entry by laser scanning cytometry. RESULTS Hypoxic epithelial cells exhibited a small increase in anti-apoptotic Bcl2, and decrease in BAX. Cell survival and active caspase-3 were unchanged. Exposure to NO had no impact on epithelial apoptosis, but initiated necrosis. In contractile myocytes, pro-apoptotic BAX abundance and caspase-3 activation were increased by hypoxia, augmented by NO exposure promoting apoptosis. Hypoxia decreased BAX/Bcl-2 ratio and promoted survival of synthetic myocytes; NO increased apoptosis of normoxic synthetic myocytes, but decreased apoptosis of hypoxic synthetic myocytes. CONCLUSION The effect of NO on pulmonary apoptosis is phenotype-dependent. A cumulative apoptotic effect of hypoxia and NO in vitro exerted on contractile myocytes may lead to contraction of this subpopulation, while synthetic myocyte survival and proliferation is enhanced by hypoxia and NO. Epithelial survival is unaffected. We speculate that alveolar rarefaction reported after neonatal hypoxia may arise from growth arrest in the vascular rather than the epithelial compartment.
Collapse
Affiliation(s)
- F Postolow
- Department of Pediatrics, University of Manitoba, 715 McDermot Avenue, Winnipeg, Manitoba R3E 3P4, Canada
| | | | | | | | | |
Collapse
|