1
|
Cohen ED, Yee M, Roethlin K, Prelipcean I, Small EM, Porter GA, O'Reilly MA. Whole genome transcriptomics reveal distinct atrial versus ventricular responses to neonatal hyperoxia. Am J Physiol Heart Circ Physiol 2025; 328:H832-H845. [PMID: 40047849 DOI: 10.1152/ajpheart.00039.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/14/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
Preterm infants exposed to supplemental oxygen (hyperoxia) are at risk for developing heart failure later in life. Exposing rodents in early postnatal life to hyperoxia causes heart failure that resembles cardiac disease seen in adult humans who were born preterm. Neonatal hyperoxia exposure affects the left atrium and left ventricle differently, inhibiting the proliferation and survival of atrial cardiomyocytes while enhancing cardiomyocyte differentiation in the ventricle. In this study, whole genome transcriptomics revealed the left atria of neonatal mice are more responsive to hyperoxia than the left ventricle, with the expression of 4,285 genes affected in the atrium and 1,743 in the ventricle. Although hyperoxia activated p53 target genes in both chambers, it caused greater DNA damage, phosphorylation of the DNA damage responsive ataxia-telangiectasia mutated (ATM) kinase, mitochondrial stress, and apoptosis in the atrium. In contrast, hyperoxia induced the expression of genes involved in DNA repair and stress granules in the ventricle. Atrial cells also showed a greater loss of extracellular matrix and superoxide dismutase 3 (SOD3) expression, possibly contributing to the enlargement of the left atrium and reduced velocity of blood flow across the mitral valve seen in mice exposed to hyperoxia. Diastolic dysfunction and heart failure in hyperoxia-exposed mice may thus stem from its effects on the left atrium, suggesting chamber-specific therapies may be needed to address diastolic dysfunction and heart failure in people who were born preterm.NEW & NOTEWORTHY Preterm infants often require oxygen (hyperoxia) at birth, but early exposure increases the risk of heart failure later in life. Previously, we showed neonatal mice exposed to hyperoxia develop adult diastolic dysfunction and heart failure like preterm-born humans. In this study, RNA-sequencing reveals hyperoxia induces broader transcriptional changes in the atrium than ventricle, including upregulation of stress pathways and loss of superoxide dismutase 3 and extracellular matrix genes, highlighting the atrium's heightened vulnerability to hyperoxia.
Collapse
Grants
- HL168812 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL144776 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL133761 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL144867 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL169961 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- AG070585 HHS | NIH | National Institute on Aging (NIA)
- ES001247 HHS | NIH | National Institute of Environmental Health Sciences (DEHS)
- KL2 TR001999 NCATS NIH HHS
Collapse
Affiliation(s)
- E David Cohen
- Department of Pediatrics, Division of Cardiology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| | - Min Yee
- Department of Pediatrics, Division of Neonatology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| | - Kyle Roethlin
- Department of Pediatrics, Division of Neonatology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| | - Irina Prelipcean
- Department of Pediatrics, Division of Cardiology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| | - Eric M Small
- Department of Medicine, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| | - George A Porter
- Department of Pediatrics, Division of Cardiology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| | - Michael A O'Reilly
- Department of Pediatrics, Division of Neonatology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, United States
| |
Collapse
|
2
|
Zeng W, Deng Z, Li H, Gao S, Ju R. Purinergic P2X7 receptor mediates hyperoxia-induced injury in pulmonary microvascular endothelial cells via NLRP3-mediated pyroptotic pathway. Open Med (Wars) 2024; 19:20241097. [PMID: 39655049 PMCID: PMC11627065 DOI: 10.1515/med-2024-1097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Background Hyperoxia-induced injury is a well-recognized cause of bronchopulmonary dysplasia (BPD). Existing research studies have not well elucidated the exact mechanisms underlying hyperoxia-induced cellular damage. This study examines the involvement of the P2X7 receptor (P2X7R) in hyperoxia-induced damage to human pulmonary microvascular endothelial cells (HPMVECs) via the NOD-like receptor family, pyrin domain-containing protein 3 (NLRP3) pathway. Methods HPMVECs developing hyperoxia-induced injury were subjected to the treatment of either selective inhibitors or a P2X7R/NLRP3 agonist. Western blot analysis assisted in the quantification of the levels of P2X7R, NLRP3, caspase-1, and gasdermin D (GSDMD). Additionally, the release of TNF-α, IL-1β, and IL-18 was assessed by ELISA and qRT-PCR. Results Exposure to hyperoxia diminished cell viability and escalated the levels of P2X7R, caspase-1, NLRP3, GSDMD, and N-terminal-GSDMD. This exposure notably increased the release of TNF-α, IL-1β, and IL-18 in HPMVECs. Notably, the suppression of P2X7R using the inhibitor A438079 decreased pyroptosis and inflammatory responses. Conversely, stimulation of P2X7R by 3'-O-(4-benzoylbenzoyl) adenosine 5'-triphosphate (BzATP) triggered pyroptosis, while inhibition of NLRP3 with glibenclamide ameliorated the damage induced by BzATP. Conclusions The P2X7R/NLRP3 pathway crucially affects the hyperoxia-induced inflammation and pyroptosis in HPMVECs, hinting the potential of blocking P2X7R/NLRP3-mediated pyroptotic pathway as a valuable therapeutic strategy for BPD.
Collapse
Affiliation(s)
- Wen Zeng
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Zhuyu Deng
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Huaying Li
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Shuqiang Gao
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, Sichuan, China
| |
Collapse
|
3
|
Durlak W, Thébaud B. The vascular phenotype of BPD: new basic science insights-new precision medicine approaches. Pediatr Res 2024; 96:1162-1171. [PMID: 36550351 DOI: 10.1038/s41390-022-02428-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/27/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of preterm birth. Up to 1/3 of children with BPD develop pulmonary hypertension (PH). PH increases mortality, the risk of adverse neurodevelopmental outcome and lacks effective treatment. Current vasodilator therapies address symptoms, but not the underlying arrested vascular development. Recent insights into placental biology and novel technological advances enabling the study of normal and impaired lung development at the single cell level support the concept of a vascular phenotype of BPD. Dysregulation of growth factor pathways results in depletion and dysfunction of putative distal pulmonary endothelial progenitor cells including Cap1, Cap2, and endothelial colony-forming cells (ECFCs), a subset of vascular progenitor cells with self-renewal and de novo angiogenic capacity. Preclinical data demonstrate effectiveness of ECFCs and ECFC-derived particles including extracellular vesicles (EVs) in promoting lung vascular growth and reversing PH, but the mechanism is unknown. The lack of engraftment suggests a paracrine mode of action mediated by EVs that contain miRNA. Aberrant miRNA signaling contributes to arrested pulmonary vascular development, hence using EV- and miRNA-based therapies is a promising strategy to prevent the development of BPD-PH. More needs to be learned about disrupted pathways, timing of intervention, and mode of delivery. IMPACT: Single-cell RNA sequencing studies provide new in-depth view of developmental endothelial depletion underlying BPD-PH. Aberrant miRNA expression is a major cause of arrested pulmonary development. EV- and miRNA-based therapies are very promising therapeutic strategies to improve prognosis in BPD-PH.
Collapse
Affiliation(s)
- Wojciech Durlak
- Regenerative Medicine Program, The Ottawa Hospital Research Institute (OHRI), Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
- Jagiellonian University Medical College, Krakow, Poland
| | - Bernard Thébaud
- Regenerative Medicine Program, The Ottawa Hospital Research Institute (OHRI), Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
4
|
Cohen ED, Roethlin K, Yee M, Woeller CF, Brookes PS, Porter GA, O'Reilly MA. PPARγ drives mitochondrial stress signaling and the loss of atrial cardiomyocytes in newborn mice exposed to hyperoxia. Redox Biol 2024; 76:103351. [PMID: 39276392 PMCID: PMC11417530 DOI: 10.1016/j.redox.2024.103351] [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: 06/07/2024] [Revised: 08/22/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024] Open
Abstract
Diastolic dysfunction is increasingly common in preterm infants exposed to supplemental oxygen (hyperoxia). Previous studies in neonatal mice showed hyperoxia suppresses fatty acid synthesis genes required for proliferation and survival of atrial cardiomyocytes. The loss of atrial cardiomyocytes creates a hypoplastic left atrium that inappropriately fills the left ventricle during diastole. Here, we show that hyperoxia stimulates adenosine monophosphate-activated kinase (AMPK) and peroxisome proliferator activated receptor-gamma (PPARγ) signaling in atrial cardiomyocytes. While both pathways can regulate lipid homeostasis, PPARγ was the primary pathway by which hyperoxia inhibits fatty acid gene expression and inhibits proliferation of mouse atrial HL-1 cells. It also enhanced the toxicity of hyperoxia by increasing expression of activating transcription factor (ATF) 5 and other mitochondrial stress response genes. Silencing PPARγ signaling restored proliferation and survival of HL-1 cells as well as atrial cardiomyocytes in neonatal mice exposed to hyperoxia. Our findings reveal PPARγ enhances the toxicity of hyperoxia on atrial cardiomyocytes, thus suggesting inhibitors of PPARγ signaling may prevent diastolic dysfunction in preterm infants.
Collapse
Affiliation(s)
- E David Cohen
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA.
| | - Kyle Roethlin
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Min Yee
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Collynn F Woeller
- Department of Ophthalmology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Paul S Brookes
- Department of Anesthesiology, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - George A Porter
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
5
|
Leek C, Cantu A, Sonti S, Gutierrez MC, Eldredge L, Sajti E, Xu HN, Lingappan K. Role of sex as a biological variable in neonatal alveolar macrophages. Redox Biol 2024; 75:103296. [PMID: 39098263 PMCID: PMC11345582 DOI: 10.1016/j.redox.2024.103296] [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: 05/29/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
The lung macrophages play a crucial role in health and disease. Sexual dimorphism significantly impacts the phenotype and function of tissue-resident macrophages. The primary mechanisms responsible for sexually dimorphic outcomes in bronchopulmonary dysplasia (BPD) remain unidentified. We tested the hypothesis that biological sex plays a crucial role in the transcriptional state of alveolar macrophages, using neonatal murine hyperoxia-induced lung injury as a relevant model for human BPD. The effects of neonatal hyperoxia exposure (95 % FiO2, PND1-5: saccular stage) on the lung myeloid cells acutely after injury and during normoxic recovery were measured. Alveolar macrophages (AM) from room air- and hyperoxia exposed from male and female neonatal murine lungs were subjected to bulk-RNA Sequencing. AMs are significantly depleted in the hyperoxia-exposed lung acutely after injury, with subsequent recovery in both sexes. The transcriptome of the alveolar macrophages is impacted by neonatal hyperoxia exposure and by sex as a biological variable. Pathways related to DNA damage and interferon-signaling were positively enriched in female AMs. Metabolic pathways related to glucose and carbohydrate metabolism were positively enriched in the male AMs, while oxidative phosphorylation was negatively enriched. These pathways were shared with monocytes and airway macrophages from intubated male and female human premature neonates.
Collapse
Affiliation(s)
- Connor Leek
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Abiud Cantu
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Shilpa Sonti
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Manuel Cantu Gutierrez
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Laurie Eldredge
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Washington School of Medicine, Seattle Children's Hospital, WA, USA
| | - Eniko Sajti
- Department of Pediatrics, Division of Neonatology, University of California San Diego, San Diego, CA, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Krithika Lingappan
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA.
| |
Collapse
|
6
|
El-Atawi K, Abdul Wahab MG, Alallah J, Osman MF, Hassan M, Siwji Z, Saleh M. Beyond Bronchopulmonary Dysplasia: A Comprehensive Review of Chronic Lung Diseases in Neonates. Cureus 2024; 16:e64804. [PMID: 39156276 PMCID: PMC11329945 DOI: 10.7759/cureus.64804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
In neonates, pulmonary diseases such as bronchopulmonary dysplasia and other chronic lung diseases (CLDs) pose significant challenges due to their complexity and high degree of morbidity and mortality. This review discusses the etiology, pathophysiology, clinical presentation, and diagnostic criteria for these conditions, as well as current management strategies. The review also highlights recent advancements in understanding the pathophysiology of these diseases and evolving strategies for their management, including gene therapy and stem cell treatments. We emphasize how supportive care is useful in managing these diseases and underscore the importance of a multidisciplinary approach. Notably, we discuss the emerging role of personalized medicine, enabled by advances in genomics and precision therapeutics, in tailoring therapy according to an individual's genetic, biochemical, and lifestyle factors. We conclude with a discussion on future directions in research and treatment, emphasizing the importance of furthering our understanding of these conditions, improving diagnostic criteria, and exploring targeted treatment modalities. The review underscores the need for multicentric and longitudinal studies to improve preventative strategies and better understand long-term outcomes. Ultimately, a comprehensive, innovative, and patient-centered approach can enhance the quality of care and outcomes for neonates with CLDs.
Collapse
Affiliation(s)
| | | | - Jubara Alallah
- Neonatology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Neonatology, King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Jeddah, SAU
| | | | | | | | - Maysa Saleh
- Pediatrics and Child Health, Al Jalila Children's Specialty Hospital, Dubai, ARE
| |
Collapse
|
7
|
Agarwal S, Fineman J, Cornfield DN, Alvira CM, Zamanian RT, Goss K, Yuan K, Bonnet S, Boucherat O, Pullamsetti S, Alcázar MA, Goncharova E, Kudryashova TV, Nicolls MR, de Jesús Pérez V. Seeing pulmonary hypertension through a paediatric lens: a viewpoint. Eur Respir J 2024; 63:2301518. [PMID: 38575157 PMCID: PMC11187317 DOI: 10.1183/13993003.01518-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/16/2024] [Indexed: 04/06/2024]
Abstract
Pulmonary hypertension (PH) is a life-threating condition associated with abnormally elevated pulmonary pressures and right heart failure. Current epidemiological data indicate that PH aetiologies are different between the adult and paediatric population. The most common forms of PH in adults are PH from left heart disease or chronic lung disease, followed by pulmonary arterial hypertension (PAH) [1]; in paediatric patients, PH is most often associated with developmental lung disorders and congenital heart disease (CHD) [2, 3]. In contrast to adults with PH, wherein patients worsen over time despite therapy, PH in children can improve with growth. For example, in infants with bronchopulmonary dysplasia (BPD) and PH morbidity and mortality are high, but with lung growth and ensuring no ongoing lung injury pulmonary vascular disease can improve as evidenced by discontinuation of vasodilator therapy in almost two-thirds of BPD-PH survivors by age 5 years [3, 4]. Paediatric pulmonary hypertension (PH) offers unique genetic and developmental insights that can help in the discovery of novel mechanisms and targets to treat adult PH https://bit.ly/3TMm6bi
Collapse
Affiliation(s)
- Stuti Agarwal
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Jeffrey Fineman
- Department of Pediatrics and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - David N Cornfield
- Division of Pediatric Pulmonary, Asthma, and Sleep Medicine, Stanford University, Palo Alto, CA, USA
| | - Cristina M Alvira
- Division of Pediatric Critical Care Medicine, Stanford University, Palo Alto, CA, USA
| | - Roham T Zamanian
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | - Kara Goss
- Department of Medicine and Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Ke Yuan
- Boston Children's Hospital, Boston, MA, USA
| | - Sebastien Bonnet
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Olivier Boucherat
- Department of Medicine, University of Laval, Quebec City, QC, Canada
| | - Soni Pullamsetti
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | | - Tatiana V Kudryashova
- University of Pittsburgh Heart, Blood, and Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Mark R Nicolls
- Division of Pulmonary and Critical Care, Stanford University, Palo Alto, CA, USA
| | | |
Collapse
|
8
|
Schumacker PT. Bronchopulmonary Dysplasia and Cell Senescence: A Case of Old Lungs in Young Infants? Am J Respir Cell Mol Biol 2024; 70:85-86. [PMID: 38109692 PMCID: PMC10848692 DOI: 10.1165/rcmb.2023-0442ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/20/2023] Open
Affiliation(s)
- Paul T Schumacker
- Department of Pediatrics Northwestern University Feinberg School of Medicine Chicago, Illinois
| |
Collapse
|
9
|
Ahmed M, Casanova NG, Zaghloul N, Gupta A, Rodriguez M, Robbins IR, Kempf CL, Sun X, Song JH, Hernon VR, Sammani S, Camp SM, Moreira A, Hsu CD, Garcia JGN. The eNAMPT/TLR4 inflammatory cascade drives the severity of intra-amniotic inflammation in pregnancy and predicts infant outcomes. Front Physiol 2023; 14:1129413. [PMID: 37415908 PMCID: PMC10319582 DOI: 10.3389/fphys.2023.1129413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction: Intra-amniotic inflammation (IAI) or chorioamnionitis is a common complication of pregnancy producing significant maternal morbidity/mortality, premature birth and neonatal risk of chronic lung diseases such as bronchopulmonary dysplasia (BPD). We examined eNAMPT (extracellular nicotinamide phosphoribosyltransferase), a critical inflammatory DAMP and TLR4 ligand, as a potential therapeutic target to reduce IAI severity and improve adverse fetal/neonatal outcomes. Methods: Blood/tissue samples were examined in: 1) women with histologically-proven chorioamnionitis, 2) very low birth weight (VLBW) neonates, and 3) a preclinical murine pregnancy model of IAI. Groups of pregnant IAI-exposed mice and pups were treated with an eNAMPT-neutralizing mAb. Results: Human placentas from women with histologically-proven chorioamnionitis exhibited dramatic NAMPT expression compared to placentas without chorioamnionitis. Increased NAMPT expression in whole blood from VLBW neonates (day 5) significantly predicted BPD development. Compared to untreated LPS-challenged murine dams (gestational day 15), pups born to eNAMPT mAb-treated dams (gestational days 15/16) exhibited a > 3-fold improved survival, reduced neonate lung eNAMPT/cytokine levels, and reduced development and severity of BPD and pulmonary hypertension (PH) following postnatal exposure to 100% hyperoxia days 1-14. Genome-wide gene expression studies of maternal uterine and neonatal cardiac tissues corroborated eNAMPT mAb-induced reductions in inflammatory pathway genes. Discussion: The eNAMPT/TLR4 inflammatory pathway is a highly druggable contributor to IAI pathobiology during pregnancy with the eNAMPT-neutralizing mAb a novel therapeutic strategy to decrease premature delivery and improve short- and long-term neonatal outcomes. eNAMPT blood expression is a potential biomarker for early prediction of chronic lung disease among premature neonates.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nancy G. Casanova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nahla Zaghloul
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Akash Gupta
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Marisela Rodriguez
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Ian R. Robbins
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L. Kempf
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Xiaoguang Sun
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Jin H. Song
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Vivian Reyes Hernon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Alvaro Moreira
- Department of Pediatrics, UT Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| |
Collapse
|
10
|
Heydarian M, Oak P, Zhang X, Kamgari N, Kindt A, Koschlig M, Pritzke T, Gonzalez-Rodriguez E, Förster K, Morty RE, Häfner F, Hübener C, Flemmer AW, Yildirim AO, Sudheendra D, Tian X, Petrera A, Kirsten H, Ahnert P, Morrell N, Desai TJ, Sucre J, Spiekerkoetter E, Hilgendorff A. Relationship between impaired BMP signalling and clinical risk factors at early-stage vascular injury in the preterm infant. Thorax 2022; 77:1176-1186. [PMID: 35580897 PMCID: PMC9685723 DOI: 10.1136/thoraxjnl-2021-218083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/11/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic lung disease, that is, bronchopulmonary dysplasia (BPD) is the most common complication in preterm infants and develops as a consequence of the misguided formation of the gas-exchange area undergoing prenatal and postnatal injury. Subsequent vascular disease and its progression into pulmonary arterial hypertension critically determines long-term outcome in the BPD infant but lacks identification of early, disease-defining changes. METHODS We link impaired bone morphogenetic protein (BMP) signalling to the earliest onset of vascular pathology in the human preterm lung and delineate the specific effects of the most prevalent prenatal and postnatal clinical risk factors for lung injury mimicking clinically relevant conditions in a multilayered animal model using wild-type and transgenic neonatal mice. RESULTS We demonstrate (1) the significant reduction in BMP receptor 2 (BMPR2) expression at the onset of vascular pathology in the lung of preterm infants, later mirrored by reduced plasma BMP protein levels in infants with developing BPD, (2) the rapid impairment (and persistent change) of BMPR2 signalling on postnatal exposure to hyperoxia and mechanical ventilation, aggravated by prenatal cigarette smoke in a preclinical mouse model and (3) a link to defective alveolar septation and matrix remodelling through platelet derived growth factor-receptor alpha deficiency. In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. CONCLUSION We identified impaired BMP signalling as a hallmark of early vascular disease in the injured neonatal lung while outlining its promising potential as a future biomarker or therapeutic target in this growing, high-risk patient population.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Prajakta Oak
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xin Zhang
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Nona Kamgari
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alida Kindt
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Markus Koschlig
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Tina Pritzke
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Erika Gonzalez-Rodriguez
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kai Förster
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Rory E Morty
- Department of Translational Pulmonology, University Hospital Heidelberg, Translational Lung Research Center campus of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Friederike Häfner
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christoph Hübener
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Andreas W Flemmer
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Ali Oender Yildirim
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Deepti Sudheendra
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Xuefei Tian
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Nick Morrell
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Tushar J Desai
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Jennifer Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee, USA
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Anne Hilgendorff
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Center for Comprehensive Developmental Care (CDeCLMU), Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| |
Collapse
|
11
|
Damianos A, Kulandavelu S, Chen P, Nwajei P, Batlahally S, Sharma M, Alvarez-Cubela S, Dominguez-Bendala J, Zambrano R, Huang J, Hare JM, Schmidt A, Wu S, Benny M, Claure N, Young K. Neonatal intermittent hypoxia persistently impairs lung vascular development and induces long-term lung mitochondrial DNA damage. J Appl Physiol (1985) 2022; 133:1031-1041. [PMID: 36135955 PMCID: PMC11918270 DOI: 10.1152/japplphysiol.00708.2021] [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: 10/11/2021] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
Abstract
Adults born preterm have an increased risk of pulmonary vascular disease. Extreme preterm infants often require supplemental oxygen but they also exhibit frequent intermittent hypoxemic episodes (IH). Here, we test the hypothesis that neonatal IH induces lung endothelial cell mitochondrial DNA (mitDNA) damage and contributes to long-term pulmonary vascular disease and pulmonary hypertension (PH). Newborn C57BL/6J mice were assigned to the following groups: 1) normoxia, 2) hyperoxia (O2 65%), 3) normoxia cycling with IH (O2 21% + O2 10%), and 4) hyperoxia cycling with IH (O2 65% + O2 10%) for 3 wk. IH episodes were initiated on postnatal day 7. Lung angiogenesis, PH, and mitDNA lesions were assessed at 3 wk and 3 mo. In vitro, the effect of IH on tubule formation and mitDNA lesions was evaluated in human pulmonary microvascular endothelial cells (HPMECs). Data were analyzed by ANOVA. In vitro, IH exposure reduced tubule formation and increased mitDNA lesions in HPMECs. This was most marked in HPMECs exposed to hyperoxia cycling with IH. In vivo, neonatal IH increased lung mitDNA lesions, impaired angiogenesis, and induced PH in 3-wk-old mice. These findings were pronounced in mice exposed to hyperoxia cycling with IH. At 3 mo follow-up, mice exposed to neonatal IH had persistently increased lung mitDNA lesions and impaired lung angiogenesis, even without concomitant hyperoxia exposure. Neonatal IH induces lung endothelial cell mitDNA damage and causes persistent impairment in lung angiogenesis. These findings provide important mechanistic insight into the pathogenesis of pulmonary vascular disease in preterm survivors.NEW & NOTEWORTHY Our current study demonstrates that neonatal intermittent hypoxia (IH) alters lung endothelial cell function, induces mitochondrial DNA lesions, and impairs lung vascular growth into adulthood. Moreover, when superimposed on hyperoxia, neonatal IH induces a severe lung vascular phenotype that is seen in preterm infants with PH. These findings suggest that neonatal IH contributes to PH in adults born preterm and importantly, that mitochondrial protection strategies may mitigate these deleterious effects.
Collapse
Affiliation(s)
- Andreas Damianos
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Shathiyah Kulandavelu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Pingping Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrick Nwajei
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Sunil Batlahally
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Silvia Alvarez-Cubela
- The Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Dominguez-Bendala
- The Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Ronald Zambrano
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jian Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Augusto Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Nelson Claure
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Karen Young
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
12
|
Tang E, Zaidi M, Lim W, Govindasamy V, Then K, Then K, Das AK, Cheong S. Headway and the remaining hurdles of mesenchymal stem cells therapy for bronchopulmonary dysplasia. THE CLINICAL RESPIRATORY JOURNAL 2022; 16:629-645. [PMID: 36055758 PMCID: PMC9527154 DOI: 10.1111/crj.13540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 12/17/2021] [Accepted: 08/15/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Preterm infants are at a high risk of developing BPD. Although progression in neonatal care has improved, BPD still causes significant morbidity and mortality, which can be attributed to the limited therapeutic choices for BPD. This review discusses the potential of MSC in treating BPD as well as their hurdles and possible solutions. DATA SOURCES The search for data was not limited to any sites but was mostly performed on all clinical trials available in ClinicalTrials.gov as well as on PubMed by applying the following keywords: lung injury, preterm, inflammation, neonatal, bronchopulmonary dysplasia and mesenchymal stem cells. STUDY SELECTIONS The articles chosen for this review were collectively determined to be relevant and appropriate in discussing MSC not only as a potential treatment strategy for curbing the incidence of BPD but also including insights on problems regarding MSC treatment for BPD. RESULTS Clinical trials regarding the use of MSC for BPD had good results but also illustrated insights on problems to be addressed in the future regarding the treatment strategy. Despite that, the clinical trials had mostly favourable reviews. CONCLUSION With BPD existing as a constant threat and there being no permanent solutions, the idea of regenerative medicine such as MSC may prove to be a breakthrough strategy when it comes to treating BPD. The success in clinical trials led to the formulation of prospective MSC-derived products such as PNEUMOSTEM®, and there is the possibility of a stem cell medication and permanent treatment for BPD in the near future.
Collapse
Affiliation(s)
- Eireen Tang
- CryoCord Sdn Bhd, Bio‐X CentreCyberjayaMalaysia
| | | | | | | | - Kong‐Yong Then
- Brighton Healthcare (Bio‐X Healthcare Sdn Bhd), Bio‐X CentreCyberjayaMalaysia
| | | | | | - Soon‐Keng Cheong
- Faculty of Medicine & Health Sciences, Universiti Tunku Abdul Rahman (UTAR)KajangMalaysia
| |
Collapse
|
13
|
Prematurity and Long-Term Respiratory Morbidity—What Is the Critical Gestational Age Threshold? J Clin Med 2022; 11:jcm11030751. [PMID: 35160203 PMCID: PMC8836586 DOI: 10.3390/jcm11030751] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/23/2022] [Accepted: 01/28/2022] [Indexed: 12/04/2022] Open
Abstract
Respiratory morbidity is a hallmark complication of prematurity. Children born preterm are exposed to both short- and long-term respiratory morbidity. This study aimed to investigate whether a critical gestational age threshold exists for significant long-term respiratory morbidity. A 23-year, population-based cohort analysis was performed comparing singleton deliveries at a single tertiary medical center. A comparison of four gestational age groups was performed according to the WHO classification: term (≥37.0 weeks, reference group), moderate to late preterm (32.0–36.6 weeks), very preterm (28.0–31.6 weeks) and extremely preterm (24.0–27.6 weeks). Hospitalizations of the offspring up to the age of 18 years involving respiratory morbidities were evaluated. A Kaplan–Meier survival curve was used to compare cumulative hospitalization incidence between the groups. A Cox proportional hazards model was used to control for confounders and time to event. Overall, 220,563 singleton deliveries were included: 93.6% term deliveries, 6% moderate to late preterm, 0.4% very preterm and 0.1% extremely preterm. Hospitalizations involving respiratory morbidity were significantly higher in children born preterm (12.7% in extremely preterm children, 11.7% in very preterm, 7.0% in late preterm vs. 4.7% in term, p < 0.001). The Kaplan–Meier survival curve demonstrated a significantly higher cumulative incidence of respiratory-related hospitalizations in the preterm groups (log-rank, p < 0.001). In the Cox regression model, delivery before 32 weeks had twice the risk of long-term respiratory morbidity. Searching for a specific gestational age threshold, the slope for hospitalization rate was attenuated beyond 30 weeks’ gestation. In our population, it seems that 30 weeks’ gestation may be the critical threshold for long-term respiratory morbidity of the offspring, as the risk for long-term respiratory-related hospitalization seems to be attenuated beyond this point until term.
Collapse
|
14
|
Cho HY, Miller-DeGraff L, Perrow LA, Gladwell W, Panduri V, Lih FB, Kleeberger SR. Murine Neonatal Oxidant Lung Injury: NRF2-Dependent Predisposition to Adulthood Respiratory Viral Infection and Protection by Maternal Antioxidant. Antioxidants (Basel) 2021; 10:antiox10121874. [PMID: 34942977 PMCID: PMC8698620 DOI: 10.3390/antiox10121874] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 12/04/2022] Open
Abstract
NRF2 protects against oxidant-associated airway disorders via cytoprotective gene induction. To examine if NRF2 is an important determinant of respiratory syncytial virus (RSV) susceptibility after neonate lung injury, Nrf2-deficient (Nrf2−/−) and wild-type (Nrf2+/+) mice neonatally exposed to hyperoxia were infected with RSV. To investigate the prenatal antioxidant effect on neonatal oxidative lung injury, time-pregnant Nrf2−/− and Nrf2+/+ mice were given an oral NRF2 agonist (sulforaphane) on embryonic days 11.5–17.5, and offspring were exposed to hyperoxia. Bronchoalveolar lavage and histopathologic analyses determined lung injury. cDNA microarray analyses were performed on placenta and neonatal lungs. RSV-induced pulmonary inflammation, injury, oxidation, and virus load were heightened in hyperoxia-exposed mice, and injury was more severe in hyperoxia-susceptible Nrf2−/− mice than in Nrf2+/+ mice. Maternal sulforaphane significantly alleviated hyperoxic lung injury in both neonate genotypes with more marked attenuation of severe neutrophilia, edema, oxidation, and alveolarization arrest in Nrf2−/− mice. Prenatal sulforaphane altered different genes with similar defensive functions (e.g., inhibition of cell/perinatal death and inflammation, potentiation of angiogenesis/organ development) in both strains, indicating compensatory transcriptome changes in Nrf2−/− mice. Conclusively, oxidative injury in underdeveloped lungs NRF2-dependently predisposed RSV susceptibility. In utero sulforaphane intervention suggested NRF2-dependent and -independent pulmonary protection mechanisms against early-life oxidant injury.
Collapse
Affiliation(s)
- Hye-Youn Cho
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA; (L.M.-D.); (L.A.P.); (W.G.); (S.R.K.)
- Correspondence: ; Tel.: +1-984-287-4088
| | - Laura Miller-DeGraff
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA; (L.M.-D.); (L.A.P.); (W.G.); (S.R.K.)
| | - Ligon A. Perrow
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA; (L.M.-D.); (L.A.P.); (W.G.); (S.R.K.)
| | - Wesley Gladwell
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA; (L.M.-D.); (L.A.P.); (W.G.); (S.R.K.)
| | - Vijayalakshmi Panduri
- Epigenetic and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA;
| | - Fred B. Lih
- Mass Spectrometry Research and Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA;
| | - Steven R. Kleeberger
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC 27709, USA; (L.M.-D.); (L.A.P.); (W.G.); (S.R.K.)
| |
Collapse
|
15
|
Yallapragada SG, Savani RC, Goss KN. Cardiovascular impact and sequelae of bronchopulmonary dysplasia. Pediatr Pulmonol 2021; 56:3453-3463. [PMID: 33756045 DOI: 10.1002/ppul.25370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
The development, growth, and function of the cardiac, pulmonary, and vascular systems are closely intertwined during both fetal and postnatal life. In utero, placental, environmental, and genetic insults may contribute to abnormal pulmonary alveolarization and vascularization that increase susceptibility to the development of bronchopulmonary dysplasia (BPD) in preterm infants. However, the shared milieu of stressors may also contribute to abnormal cardiac or vascular development in the fetus and neonate, leading to the potential for cardiovascular dysfunction. Further, cardiac or pulmonary maladaptation can potentiate dysfunction in the other organ, amplify the risk for BPD in the neonate, and increase the trajectory for overall neonatal morbidity. Beyond infancy, there is an increased risk for systemic and pulmonary vascular disease including hypertension, as well as potential cardiac dysfunction, particularly within the right ventricle. This review will focus on the cardiovascular antecedents of BPD in the fetus, cardiovascular consequences of preterm birth in the neonate including associations with BPD, and cardiovascular impact of prematurity and BPD throughout the lifespan.
Collapse
Affiliation(s)
- Sushmita G Yallapragada
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kara N Goss
- Division of Pulmonary and Critical Care, Departments of Medicine and Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
Chitin-Derived AVR-48 Prevents Experimental Bronchopulmonary Dysplasia (BPD) and BPD-Associated Pulmonary Hypertension in Newborn Mice. Int J Mol Sci 2021; 22:ijms22168547. [PMID: 34445253 PMCID: PMC8395179 DOI: 10.3390/ijms22168547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/03/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of prematurity and a key contributor to the large health care burden associated with prematurity, longer hospital stays, higher hospital costs, and frequent re-hospitalizations of affected patients through the first year of life and increased resource utilization throughout childhood. This disease is associated with abnormal pulmonary function that may lead to BPD-associated pulmonary hypertension (PH), a major contributor to neonatal mortality and morbidity. In the absence of any definitive treatment options, this life-threatening disease is associated with high resource utilization during and after neonatal intensive care unit (NICU) stay. The goal of this study was to test the safety and efficacy of a small molecule derivative of chitin, AVR-48, as prophylactic therapy for preventing experimental BPD in a mouse model. Two doses of AVR-48 were delivered either intranasally (0.11 mg/kg), intraperitoneally (10 mg/kg), or intravenously (IV) (10 mg/kg) to newborn mouse pups on postnatal day (P)2 and P4. The outcomes were assessed by measuring total inflammatory cells in the broncho-alveolar lavage fluid (BALF), chord length, septal thickness, and radial alveolar counts of the alveoli, Fulton’s Index (for PH), cell proliferation and cell death by immunostaining, and markers of inflammation by Western blotting and ELISA. The bioavailability and safety of the drug were assessed by pharmacokinetic and toxicity studies in both neonatal mice and rat pups (P3-P5). Following AVR-48 treatment, alveolar simplification was improved, as evident from chord length, septal thickness, and radial alveolar counts; total inflammatory cells were decreased in the BALF; Fulton’s Index was decreased and lung inflammation and cell death were decreased, while angiogenesis and cell proliferation were increased. AVR-48 was found to be safe and the no-observed-adverse-effect level (NOAEL) in rat pups was determined to be 100 mg/kg when delivered via IV dosing with a 20-fold safety margin. With no reported toxicity and with a shorter half-life, AVR-48 is able to reverse the worsening cardiopulmonary phenotype of experimental BPD and BPD-PH, compared to controls, thus positioning it as a future drug candidate.
Collapse
|
17
|
Dylag AM, Haak J, Warren R, Yee M, Pryhuber GS, O'Reilly MA. Low Dose Hyperoxia Primes Airways for Fibrosis in Mice after Influenza A Infection. Am J Physiol Lung Cell Mol Physiol 2021; 321:L750-L763. [PMID: 34323115 DOI: 10.1152/ajplung.00289.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
It is well known that supplemental oxygen used to treat preterm infants in respiratory distress is associated with permanently disrupting lung development and the host response to influenza A virus (IAV). However, many infants who go home with normally functioning lungs are also at risk for hyperreactivity after a respiratory viral infection. We recently reported a new, low-dose hyperoxia mouse model (40% for 8 days; 40x8) that causes a transient change in lung function that resolves, rendering 40x8 adult animals functionally indistinguishable from room air controls. Here we reported that when infected with IAV, 40x8 mice display an early transient activation of TGFβ signaling and later airway hyperreactivity associated with peribronchial inflammation (profibrotic macrophages) and fibrosis compared to infected room air controls, suggesting neonatal oxygen induced hidden molecular changes that prime the lung for hyperreactive airways disease. While searching for potential activators of TGFβ signaling, we discovered that thrombospondin-1 (TSP-1) is elevated in naïve 40x8 mice compared to controls and localized to lung megakaryocytes and platelets before and during IAV infection. Elevated TSP-1 was also identified in human autopsy samples of former preterm infants with bronchopulmonary dysplasia. These findings reveal how low doses of oxygen that do not durably change lung function may prime it for hyperreactive airways disease by changing expression of genes, such as TSP-1, thus helping to explain why former preterm infants who have normal lung function are susceptible to airway obstruction and increased morbidity after viral infection.
Collapse
Affiliation(s)
- Andrew M Dylag
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Jeannie Haak
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Rachel Warren
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY, United States
| | - Min Yee
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Gloria S Pryhuber
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Michael A O'Reilly
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| |
Collapse
|
18
|
Mukherjee D, Konduri GG. Pediatric Pulmonary Hypertension: Definitions, Mechanisms, Diagnosis, and Treatment. Compr Physiol 2021; 11:2135-2190. [PMID: 34190343 PMCID: PMC8289457 DOI: 10.1002/cphy.c200023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pediatric pulmonary hypertension (PPH) is a multifactorial disease with diverse etiologies and presenting features. Pulmonary hypertension (PH), defined as elevated pulmonary artery pressure, is the presenting feature for several pulmonary vascular diseases. It is often a hidden component of other lung diseases, such as cystic fibrosis and bronchopulmonary dysplasia. Alterations in lung development and genetic conditions are an important contributor to pediatric pulmonary hypertensive disease, which is a distinct entity from adult PH. Many of the causes of pediatric PH have prenatal onset with altered lung development due to maternal and fetal conditions. Since lung growth is altered in several conditions that lead to PPH, therapy for PPH includes both pulmonary vasodilators and strategies to restore lung growth. These strategies include optimal alveolar recruitment, maintaining physiologic blood gas tension, nutritional support, and addressing contributing factors, such as airway disease and gastroesophageal reflux. The outcome for infants and children with PH is highly variable and largely dependent on the underlying cause. The best outcomes are for neonates with persistent pulmonary hypertension (PPHN) and reversible lung diseases, while some genetic conditions such as alveolar capillary dysplasia are lethal. © 2021 American Physiological Society. Compr Physiol 11:2135-2190, 2021.
Collapse
Affiliation(s)
- Devashis Mukherjee
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| | - Girija G. Konduri
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Children’s Research Institute, Children’s Wisconsin, Milwaukee, Wisconsin, 53226 USA
| |
Collapse
|
19
|
Sucre J, Haist L, Bolton CE, Hilgendorff A. Early Changes and Indicators Characterizing Lung Aging in Neonatal Chronic Lung Disease. Front Med (Lausanne) 2021; 8:665152. [PMID: 34136503 PMCID: PMC8200413 DOI: 10.3389/fmed.2021.665152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 05/04/2021] [Indexed: 12/16/2022] Open
Abstract
Infants suffering from neonatal chronic lung disease, i.e., bronchopulmonary dysplasia, are facing long-term consequences determined by individual genetic background, presence of infections, and postnatal treatment strategies such as mechanical ventilation and oxygen toxicity. The adverse effects provoked by these measures include inflammatory processes, oxidative stress, altered growth factor signaling, and remodeling of the extracellular matrix. Both, acute and long-term consequences are determined by the capacity of the immature lung to respond to the challenges outlined above. The subsequent impairment of lung growth translates into an altered trajectory of lung function later in life. Here, knowledge about second and third hit events provoked through environmental insults are of specific importance when advocating lifestyle recommendations to this patient population. A profound exchange between the different health care professionals involved is urgently needed and needs to consider disease origin while future monitoring and treatment strategies are developed.
Collapse
Affiliation(s)
- Jennifer Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, TN, United States
| | - Lena Haist
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center With the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany.,Center for Comprehensive Developmental Care (CDeCLMU), University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Charlotte E Bolton
- Division of Respiratory Medicine, NIHR Nottingham Biomedical Research Centre, School of Medicine, University of Nottingham, City Hospital NUH Campus, Nottingham, United Kingdom
| | - Anne Hilgendorff
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center With the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany.,Center for Comprehensive Developmental Care (CDeCLMU), University Hospital Ludwig-Maximilian University, Munich, Germany
| |
Collapse
|
20
|
Woeller CF, Lim SA, Roztocil E, Yee M, Beier EE, Puzas JE, O'Reilly MA. Neonatal hyperoxia impairs adipogenesis of bone marrow-derived mesenchymal stem cells and fat accumulation in adult mice. Free Radic Biol Med 2021; 167:287-298. [PMID: 33757863 PMCID: PMC8096722 DOI: 10.1016/j.freeradbiomed.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 10/21/2022]
Abstract
Preterm birth is a risk factor for growth failure and development of respiratory disease in children and young adults. Their early exposure to oxygen may contribute to lung disease because adult mice exposed to hyperoxia as neonates display reduced lung function, changes in the host response to respiratory viral infections, and develop pulmonary hypertension and heart failure that shortens their lifespan. Here, we provide new evidence that neonatal hyperoxia also impairs growth by inhibiting fat accumulation. Failure to accumulate fat may reflect a systemic defect in adipogenic potential of stem cells because bone marrow-derived mesenchymal cells (BMSCs) isolated from the mice grew slower and were more oxidized compared to controls. They also displayed reduced capacity to accumulate lipid and differentiate into adipocytes. BMSCs from adult mice exposed to neonatal hyperoxia express lower levels of peroxisome proliferator-activated receptor gamma (PPARγ), a transcription factor that drives adipocyte differentiation. The defect in adipogenesis was rescued by expressing PPARγ in these cells. These findings reveal early life exposure to high levels of oxygen may suppresses fat accumulation and impair adipogenic differentiation upstream of PPARγ signaling, thus potentially contributing to growth failure seen in people born preterm.
Collapse
Affiliation(s)
- Collynn F Woeller
- Departments of Ophthalmology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| | - Sydney A Lim
- Departments of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Elisa Roztocil
- Departments of Ophthalmology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Min Yee
- Departments of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Eric E Beier
- Departments of Orthopaedics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - J Edward Puzas
- Departments of Orthopaedics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Michael A O'Reilly
- Departments of Ophthalmology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA; Departments of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| |
Collapse
|
21
|
van Vliet T, Casciaro F, Demaria M. To breathe or not to breathe: Understanding how oxygen sensing contributes to age-related phenotypes. Ageing Res Rev 2021; 67:101267. [PMID: 33556549 DOI: 10.1016/j.arr.2021.101267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
Aging is characterized by a progressive loss of tissue integrity and functionality due to disrupted homeostasis. Molecular oxygen is pivotal to maintain tissue functions, and aerobic species have evolved a sophisticated sensing system to ensure proper oxygen supply and demand. It is not surprising that aberrations in oxygen and oxygen-associated pathways subvert health and promote different aspects of aging. In this review, we discuss emerging findings on how oxygen-sensing mechanisms regulate different cellular and molecular processes during normal physiology, and how dysregulation of oxygen availability lead to disease and aging. We describe various clinical manifestations associated with deregulation of oxygen balance, and how oxygen-modulating therapies and natural oxygen oscillations influence longevity. We conclude by discussing how a better understanding of oxygen-related mechanisms that orchestrate aging processes may lead to the development of new therapeutic strategies to extend healthy aging.
Collapse
|
22
|
Kamat SM, Mendelsohn AR, Larrick JW. Rejuvenation Through Oxygen, More or Less. Rejuvenation Res 2021; 24:158-163. [PMID: 33784834 DOI: 10.1089/rej.2021.0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Modest modulation of oxygen intake, either by inducing mild intermittent hypoxia or hyperoxia appears to induce modest rejuvenative changes in mammals, in part, by activating key regulator hypoxia-induced factor 1a (HIF-1a). Interestingly both lower oxygen and transient higher oxygen levels induce this hypoxia regulator. Hyperbaric oxygen induces HIF-1a by the hyperoxic-hypoxic paradox that results from an overinduction of protective factors under intermittent hyperoxic conditions, leading to a state somewhat similar to that induced by hypoxia. A key difference being that SIRT1 is induced by hyperoxia, whereas it is reduced during hypoxia by the activity of HIF-1a. In a recent report, a small clinical trial employing 60 sessions of intermittent hyperbaric oxygen therapy (HBOT) studying old humans resulted in increased mean telomere length of immune cells including B cells, natural killer cells, T helper, and cytotoxic T lymphocytes. Moreover, there was a reduction in CD28null senescent T helper and cytotoxic T cells. In a parallel report, HBOT has been reported to enhance cognition in older adults, especially attention and information processing speed through increased cerebral blood flow (CBF) in brain regions where CBF tends to decline with age. The durability of these beneficial changes is yet to be determined. These preliminary results require follow-up, including more extensive characterization of changes in aging-associated biomarkers. An interesting avenue of potential work is to elucidate potential connections between hypoxia and epigenetics, especially the induction of the master pluripotent regulatory factors, which when expressed transiently have been reported to ameliorate some aging biomarkers and pathologies.
Collapse
|
23
|
Cohen ED, Yee M, Porter GA, Ritzer E, McDavid AN, Brookes PS, Pryhuber GS, O’Reilly MA. Neonatal hyperoxia inhibits proliferation and survival of atrial cardiomyocytes by suppressing fatty acid synthesis. JCI Insight 2021; 6:140785. [PMID: 33507880 PMCID: PMC8021108 DOI: 10.1172/jci.insight.140785] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
Preterm birth increases the risk for pulmonary hypertension and heart failure in adulthood. Oxygen therapy can damage the immature cardiopulmonary system and may be partially responsible for the cardiovascular disease in adults born preterm. We previously showed that exposing newborn mice to hyperoxia causes pulmonary hypertension by 1 year of age that is preceded by a poorly understood loss of pulmonary vein cardiomyocyte proliferation. We now show that hyperoxia also reduces cardiomyocyte proliferation and survival in the left atrium and causes diastolic heart failure by disrupting its filling of the left ventricle. Transcriptomic profiling showed that neonatal hyperoxia permanently suppressed fatty acid synthase (Fasn), stearoyl-CoA desaturase 1 (Scd1), and other fatty acid synthesis genes in the atria of mice, the HL-1 line of mouse atrial cardiomyocytes, and left atrial tissue explanted from human infants. Suppressing Fasn or Scd1 reduced HL-1 cell proliferation and increased cell death, while overexpressing these genes maintained their expansion in hyperoxia, suggesting that oxygen directly inhibits atrial cardiomyocyte proliferation and survival by repressing Fasn and Scd1. Pharmacologic interventions that restore Fasn, Scd1, and other fatty acid synthesis genes in atrial cardiomyocytes may, thus, provide a way of ameliorating the adverse effects of supplemental oxygen on preterm infants.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul S. Brookes
- Department of Anesthesiology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York, USA
| | | | | |
Collapse
|
24
|
Issah Y, Naik A, Tang SY, Forrest K, Brooks TG, Lahens N, Theken KN, Mermigos M, Sehgal A, Worthen GS, FitzGerald GA, Sengupta S. Loss of circadian protection against influenza infection in adult mice exposed to hyperoxia as neonates. eLife 2021; 10:e61241. [PMID: 33650487 PMCID: PMC7924938 DOI: 10.7554/elife.61241] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/14/2021] [Indexed: 12/13/2022] Open
Abstract
Adverse early-life exposures have a lasting negative impact on health. Neonatal hyperoxia that is a risk factor for bronchopulmonary dysplasia confers susceptibility to influenza A virus (IAV) infection later in life. Given our previous findings that the circadian clock protects against IAV, we asked if the long-term impact of neonatal hyperoxia vis-à-vis IAV infection includes circadian disruption. Here, we show that neonatal hyperoxia abolishes the clock-mediated time of day protection from IAV in mice, independent of viral burden through host tolerance pathways. We discovered that the lung intrinsic clock (and not the central or immune clocks) mediated this dysregulation. Loss of circadian protein, Bmal1, in alveolar type 2 (AT2) cells recapitulates the increased mortality, loss of temporal gating, and other key features of hyperoxia-exposed animals. Our data suggest a novel role for the circadian clock in AT2 cells in mediating long-term effects of early-life exposures to the lungs.
Collapse
Affiliation(s)
- Yasmine Issah
- The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Amruta Naik
- The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Soon Y Tang
- Institute of Translational Medicine and Therapeutics (ITMAT), University of PennsylvaniaPhiladelphiaUnited States
| | - Kaitlyn Forrest
- The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Thomas G Brooks
- Institute of Translational Medicine and Therapeutics (ITMAT), University of PennsylvaniaPhiladelphiaUnited States
| | - Nicholas Lahens
- Institute of Translational Medicine and Therapeutics (ITMAT), University of PennsylvaniaPhiladelphiaUnited States
| | - Katherine N Theken
- Institute of Translational Medicine and Therapeutics (ITMAT), University of PennsylvaniaPhiladelphiaUnited States
- Systems Pharmacology University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Mara Mermigos
- The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Amita Sehgal
- Chronobiology and Sleep Institute, University of PennsylvaniaPhiladelphiaUnited States
- Department of Neuroscience, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - George S Worthen
- The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pediatrics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Garret A FitzGerald
- Institute of Translational Medicine and Therapeutics (ITMAT), University of PennsylvaniaPhiladelphiaUnited States
- Systems Pharmacology University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
- Chronobiology and Sleep Institute, University of PennsylvaniaPhiladelphiaUnited States
| | - Shaon Sengupta
- The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Institute of Translational Medicine and Therapeutics (ITMAT), University of PennsylvaniaPhiladelphiaUnited States
- Chronobiology and Sleep Institute, University of PennsylvaniaPhiladelphiaUnited States
- Department of Pediatrics, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| |
Collapse
|
25
|
Yee M, David Cohen E, Haak J, Dylag AM, O'Reilly MA. Neonatal hyperoxia enhances age-dependent expression of SARS-CoV-2 receptors in mice. Sci Rep 2020; 10:22401. [PMID: 33372179 PMCID: PMC7769981 DOI: 10.1038/s41598-020-79595-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/06/2020] [Indexed: 12/28/2022] Open
Abstract
The severity of COVID-19 lung disease is higher in the elderly and people with pre-existing co-morbidities. People who were born preterm may be at greater risk for COVID-19 because their early exposure to oxygen (hyperoxia) at birth increases the severity of respiratory viral infections. Hyperoxia at birth increases the severity of influenza A virus infections in adult mice by reducing the number of alveolar epithelial type 2 (AT2) cells. Since AT2 cells express the SARS-CoV-2 receptors angiotensin converting enzyme (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2), their expression should decline as AT2 cells are depleted by hyperoxia. Instead, ACE2 was detected in airway Club cells and endothelial cells at birth, and then AT2 cells at one year of age. Neonatal hyperoxia stimulated expression of ACE2 in Club cells and in AT2 cells by 2 months of age. It also stimulated expression of TMPRSS2 in the lung. Increased expression of SARS-CoV-2 receptors was blocked by mitoTEMPO, a mitochondrial superoxide scavenger that reduced oxidative stress and DNA damage seen in oxygen-exposed mice. Our finding that hyperoxia enhances the age-dependent expression of SARS-CoV-2 receptors in mice helps explain why COVID-19 lung disease is greater in the elderly and people with pre-existing co-morbidities.
Collapse
Affiliation(s)
- Min Yee
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - E David Cohen
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Jeannie Haak
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Andrew M Dylag
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Michael A O'Reilly
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.
| |
Collapse
|
26
|
Kumar VHS, Wang H, Nielsen L. Short-term perinatal oxygen exposure may impair lung development in adult mice. Biol Res 2020; 53:51. [PMID: 33168088 PMCID: PMC7654066 DOI: 10.1186/s40659-020-00318-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 10/29/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Hyperoxia at resuscitation increases oxidative stress, and even brief exposure to high oxygen concentrations during stabilization may trigger organ injury with adverse long-term outcomes in premature infants. We studied the long-term effects of short-term perinatal oxygen exposure on cell cycle gene expression and lung growth in adult mice. METHODS We randomized mice litters at birth to 21, 40, or 100%O2 for 30 min and recovered in room air for 4 or 12 weeks. Cell cycle gene expression, protein analysis, and lung morphometry were assessed at 4 and 12 weeks. RESULTS The principal component analysis demonstrated a high degree of correlation for cell cycle gene expression among the three oxygen groups. Lung elastin was significantly lower in the 100%O2 groups at 4 weeks. On lung morphometry, radial alveolar count, alveolar number, and septal count were similar. However, the mean linear intercept (MLI) and septal length significantly correlated among the oxygen groups. The MLI was markedly higher in the 100%O2 groups at 4 and 12 weeks of age, and the septal length was significantly lower in the 100%O2 groups at 12 weeks. CONCLUSION Short-term exposure to high oxygen concentrations lead to subtle changes in lung development that may affect alveolarization. The changes are related explicitly to secondary crest formation that may result in alteration in lung elastin. Resuscitation with high oxygen concentrations may have a significant impact on lung development and long-term outcomes such as BPD in premature infants.
Collapse
Affiliation(s)
- Vasantha H S Kumar
- Division of Neonatology, Department of Pediatrics, University At Buffalo, 1001 fifth Floor Main Street Buffalo, Buffalo, NY, 14203, USA.
| | - Huamei Wang
- Division of Neonatology, Department of Pediatrics, University At Buffalo, 1001 fifth Floor Main Street Buffalo, Buffalo, NY, 14203, USA
| | - Lori Nielsen
- Division of Neonatology, Department of Pediatrics, University At Buffalo, 1001 fifth Floor Main Street Buffalo, Buffalo, NY, 14203, USA
| |
Collapse
|
27
|
Gong J, Feng Z, Peterson AL, Carr JF, Vang A, Braza J, Choudhary G, Dennery PA, Yao H. Endothelial to mesenchymal transition during neonatal hyperoxia-induced pulmonary hypertension. J Pathol 2020; 252:411-422. [PMID: 32815166 DOI: 10.1002/path.5534] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD), a chronic lung disease in premature infants, results from mechanical ventilation and hyperoxia, amongst other factors. Although most BPD survivors can be weaned from supplemental oxygen, many show evidence of cardiovascular sequelae in adulthood, including pulmonary hypertension and pulmonary vascular remodeling. Endothelial-mesenchymal transition (EndoMT) plays an important role in mediating vascular remodeling in idiopathic pulmonary arterial hypertension. Whether hyperoxic exposure, a known mediator of BPD in rodent models, causes EndoMT resulting in vascular remodeling and pulmonary hypertension remains unclear. We hypothesized that neonatal hyperoxic exposure causes EndoMT, leading to the development of pulmonary hypertension in adulthood. To test this hypothesis, newborn mice were exposed to hyperoxia and then allowed to recover in room air until adulthood. Neonatal hyperoxic exposure gradually caused pulmonary vascular and right ventricle remodeling as well as pulmonary hypertension. Male mice were more susceptible to developing pulmonary hypertension compared to female mice, when exposed to hyperoxia as newborns. Hyperoxic exposure induced EndoMT in mouse lungs as well as in cultured lung microvascular endothelial cells (LMVECs) isolated from neonatal mice and human fetal donors. This was augmented in cultured LMVECs from male donors compared to those from female donors. Using primary mouse LMVECs, hyperoxic exposure increased phosphorylation of both Smad2 and Smad3, but reduced Smad7 protein levels. Treatment with a selective TGF-β inhibitor SB431542 blocked hyperoxia-induced EndoMT in vitro. Altogether, we show that neonatal hyperoxic exposure caused vascular remodeling and pulmonary hypertension in adulthood. This was associated with increased EndoMT. These novel observations provide mechanisms underlying hyperoxia-induced vascular remodeling and potential approaches to prevent BPD-associated pulmonary hypertension by targeting EndoMT. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jiannan Gong
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA.,Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, PR China
| | - Zihang Feng
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA
| | - Abigail L Peterson
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA
| | - Jennifer F Carr
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA
| | - Alexander Vang
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA
| | - Julie Braza
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI, USA.,Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Phyllis A Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA.,Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, USA
| |
Collapse
|
28
|
Lewandowski AJ, Levy PT, Bates ML, McNamara PJ, Nuyt AM, Goss KN. Impact of the Vulnerable Preterm Heart and Circulation on Adult Cardiovascular Disease Risk. Hypertension 2020; 76:1028-1037. [PMID: 32816574 PMCID: PMC7480939 DOI: 10.1161/hypertensionaha.120.15574] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Preterm birth accounts for over 15 million global births per year. Perinatal interventions introduced since the early 1980s, such as antenatal glucocorticoids, surfactant, and invasive ventilation strategies, have dramatically improved survival of even the smallest, most vulnerable neonates. As a result, a new generation of preterm-born individuals has now reached early adulthood, and they are at increased risk of cardiovascular diseases. To better understand the sequelae of preterm birth, cardiovascular follow-up studies in adolescents and young adults born preterm have focused on characterizing changes in cardiac, vascular, and pulmonary structure and function. Being born preterm associates with a reduced cardiac reserve and smaller left and right ventricular volumes, as well as decreased vascularity, increased vascular stiffness, and higher pressure of both the pulmonary and systemic vasculature. The purpose of this review is to present major epidemiological evidence linking preterm birth with cardiovascular disease; to discuss findings from clinical studies showing a long-term impact of preterm birth on cardiac remodeling, as well as the systemic and pulmonary vascular systems; to discuss differences across gestational ages; and to consider possible driving mechanisms and therapeutic approaches for reducing cardiovascular burden in individuals born preterm.
Collapse
Affiliation(s)
- Adam J Lewandowski
- From the Oxford Cardiovascular Clinical Research Facility, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, United Kingdom (A.J.L.)
| | - Philip T Levy
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Harvard University, MA (P.T.L.)
| | - Melissa L Bates
- Department of Health and Human Physiology (M.L.B.), University of Iowa.,Division of Neonatology (M.L.B., P.J.M.), University of Iowa
| | - Patrick J McNamara
- Division of Neonatology (M.L.B., P.J.M.), University of Iowa.,Division of Cardiology (P.J.M.), University of Iowa
| | - Anne Monique Nuyt
- Department of Pediatrics, Division of Neonatology, CHU Sainte-Justine, Faculty of Medicine, Université de Montréal, QC, Canada (A.M.N.)
| | - Kara N Goss
- Departments of Pediatrics (K.N.G.), School of Medicine and Public Health, University of Wisconsin-Madison.,Medicine (K.N.G.), School of Medicine and Public Health, University of Wisconsin-Madison
| |
Collapse
|
29
|
Chao CM, Chong L, Chu X, Shrestha A, Behnke J, Ehrhardt H, Zhang J, Chen C, Bellusci S. Targeting Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension (BPD-PH): Potential Role of the FGF Signaling Pathway in the Development of the Pulmonary Vascular System. Cells 2020; 9:cells9081875. [PMID: 32796770 PMCID: PMC7464452 DOI: 10.3390/cells9081875] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/11/2022] Open
Abstract
More than 50 years after the first description of Bronchopulmonary dysplasia (BPD) by Northway, this chronic lung disease affecting many preterm infants is still poorly understood. Additonally, approximately 40% of preterm infants suffering from severe BPD also suffer from Bronchopulmonary dysplasia-associated pulmonary hypertension (BPD-PH), leading to a significant increase in total morbidity and mortality. Until today, there is no curative therapy for both BPD and BPD-PH available. It has become increasingly evident that growth factors are playing a central role in normal and pathologic development of the pulmonary vasculature. Thus, this review aims to summarize the recent evidence in our understanding of BPD-PH from a basic scientific point of view, focusing on the potential role of Fibroblast Growth Factor (FGF)/FGF10 signaling pathway contributing to disease development, progression and resolution.
Collapse
Affiliation(s)
- Cho-Ming Chao
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center, German Center for Lung Research, 35392 Giessen, Germany; (J.B.); (H.E.)
- Correspondence: (C.-M.C.); (S.B.)
| | - Lei Chong
- Institute of Pediatrics, National Key Clinical Specialty of Pediatric Respiratory Medicine, Discipline of Pediatric Respiratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China;
| | - Xuran Chu
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Amit Shrestha
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
| | - Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center, German Center for Lung Research, 35392 Giessen, Germany; (J.B.); (H.E.)
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Feulgenstrasse 12, D-35392 Gießen, Universities of Gießen and Marburg Lung Center, German Center for Lung Research, 35392 Giessen, Germany; (J.B.); (H.E.)
| | - Jinsan Zhang
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- International Collaborative Center on Growth Factor Research, Life Science Institute, Wenzhou University, Wenzhou 325035, China
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
| | - Saverio Bellusci
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (J.Z.); (C.C.)
- Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (X.C.); (A.S.)
- Correspondence: (C.-M.C.); (S.B.)
| |
Collapse
|
30
|
Yee M, Cohen ED, Haak J, Dylag AM, O'Reilly MA. Neonatal hyperoxia enhances age-dependent expression of SARS-CoV-2 receptors in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32743585 PMCID: PMC7386505 DOI: 10.1101/2020.07.22.215962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The severity of COVID-19 lung disease is higher in the elderly and people with pre-existing co-morbidities. People who were born preterm may be at greater risk for COVID-19 because their early exposure to oxygen at birth increases their risk of being hospitalized when infected with RSV and other respiratory viruses. Our prior studies in mice showed how high levels of oxygen (hyperoxia) between postnatal days 0-4 increases the severity of influenza A virus infections by reducing the number of alveolar epithelial type 2 (AT2) cells. Because AT2 cells express the SARS-CoV-2 receptors angiotensin converting enzyme (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2), we expected their expression would decline as AT2 cells were depleted by hyperoxia. Instead, we made the surprising discovery that expression of Ace2 and Tmprss2 mRNA increases as mice age and is accelerated by exposing mice to neonatal hyperoxia. ACE2 is primarily expressed at birth by airway Club cells and becomes detectable in AT2 cells by one year of life. Neonatal hyperoxia increases ACE2 expression in Club cells and makes it detectable in 2-month-old AT2 cells. This early and increased expression of SARS-CoV-2 receptors was not seen in adult mice who had been administered the mitochondrial superoxide scavenger mitoTEMPO during hyperoxia. Our finding that early life insults such as hyperoxia enhances the age-dependent expression of SARS-CoV-2 receptors in the respiratory epithelium helps explain why COVID-19 lung disease is greater in the elderly and people with pre-existing co-morbidities.
Collapse
Affiliation(s)
- Min Yee
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642
| | - E David Cohen
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642
| | - Jeannie Haak
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642
| | - Andrew M Dylag
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642
| | - Michael A O'Reilly
- The Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, NY 14642
| |
Collapse
|
31
|
Gilfillan M, Das P, Shah D, Alam MA, Bhandari V. Inhibition of microRNA-451 is associated with increased expression of Macrophage Migration Inhibitory Factor and mitgation of the cardio-pulmonary phenotype in a murine model of Bronchopulmonary Dysplasia. Respir Res 2020; 21:92. [PMID: 32321512 PMCID: PMC7178994 DOI: 10.1186/s12931-020-01353-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Macrophage migration inhibitory factor (MIF) has been implicated as a protective factor in the development of bronchopulmonary dysplasia (BPD) and is known to be regulated by MicroRNA-451 (miR-451). The aim of this study was to evaluate the role of miR-451 and the MIF signaling pathway in in vitro and in vivo models of BPD. Methods Studies were conducted in mouse lung endothelial cells (MLECs) exposed to hyperoxia and in a newborn mouse model of hyperoxia-induced BPD. Lung and cardiac morphometry as well as vascular markers were evaluated. Results Increased expression of miR-451 was noted in MLECs exposed to hyperoxia and in lungs of BPD mice. Administration of a miR-451 inhibitor to MLECs exposed to hyperoxia was associated with increased expression of MIF and decreased expression of angiopoietin (Ang) 2. Treatment with the miR-451 inhibitor was associated with improved lung morphometry indices, significant reduction in right ventricular hypertrophy, decreased mean arterial wall thickness and improvement in vascular density in BPD mice. Western blot analysis demonstrated preservation of MIF expression in BPD animals treated with a miR-451 inhibitor and increased expression of vascular endothelial growth factor-A (VEGF-A), Ang1, Ang2 and the Ang receptor, Tie2. Conclusion We demonstrated that inhibition of miR-451 is associated with mitigation of the cardio-pulmonary phenotype, preservation of MIF expression and increased expression of several vascular growth factors.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,St Christopher's Hospital for Children, Philadelphia, PA, 19134, USA
| | - Pragnya Das
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA
| | - Dilip Shah
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA
| | - Mohammad Afaque Alam
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Temple University, Philadelphia, PA, 19140, USA
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA. .,St Christopher's Hospital for Children, Philadelphia, PA, 19134, USA. .,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA. .,Temple University, Philadelphia, PA, 19140, USA. .,Pediatrics, Obstetrics and Gynecology and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, USA. .,Neonatology, The Children's Regional Hospital at Cooper, One Cooper Plaza, Camden, NJ, 08103, USA.
| |
Collapse
|
32
|
Menon RT, Shrestha AK, Reynolds CL, Barrios R, Caron KM, Shivanna B. Adrenomedullin Is Necessary to Resolve Hyperoxia-Induced Experimental Bronchopulmonary Dysplasia and Pulmonary Hypertension in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:711-722. [PMID: 32093901 DOI: 10.1016/j.ajpath.2019.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 10/29/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
Abstract
Bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH) is an infantile lung disease characterized by aberrant angiogenesis and impaired resolution of lung injury. Adrenomedullin (AM) signals through calcitonin receptor-like receptor and receptor activity-modifying protein 2 and modulates lung injury initiation. However, its role in lung injury resolution and the mechanisms by which it regulates angiogenesis remain unclear. Consequently, we hypothesized that AM resolves hyperoxia-induced BPD and PH via endothelial nitric oxide synthase (NOS3). AM-sufficient (ADM+/+) or -deficient (ADM+/-) mice were exposed to normoxia or hyperoxia through postnatal days (PNDs) 1 to 14, and the hyperoxia-exposed mice were allowed to recover in normoxia for an additional 56 days. Lung injury and development and PH were quantified at different time points. Human pulmonary microvascular endothelial cells were also used to examine the effects of AM signaling on the NOS3 pathway and angiogenesis. Lung blood vessels and NOS3 expression decreased and the extent of hyperoxia-induced BPD and PH increased in ADM+/- mice compared with ADM+/+ mice. Hyperoxia-induced apoptosis and PH resolved by PND14 and PND70, respectively, in ADM+/+ mice but not in ADM+/- mice. Knockdown of ADM, calcitonin receptor-like receptor, and receptor activity-modifying protein 2 in vitro decreased NOS3 expression, nitric oxide generation, and angiogenesis. Furthermore, NOS3 knockdown abrogated the angiogenic effects of AM. Collectively, these results indicate that AM resolves hyperoxic lung injury via NOS3.
Collapse
Affiliation(s)
- Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Corey L Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, Texas
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
33
|
Yao H, Gong J, Peterson AL, Lu X, Zhang P, Dennery PA. Fatty Acid Oxidation Protects against Hyperoxia-induced Endothelial Cell Apoptosis and Lung Injury in Neonatal Mice. Am J Respir Cell Mol Biol 2020; 60:667-677. [PMID: 30571144 DOI: 10.1165/rcmb.2018-0335oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In neonates, hyperoxia or positive pressure ventilation causes continued lung injury characterized by simplified vascularization and alveolarization, which are the hallmarks of bronchopulmonary dysplasia. Although endothelial cells (ECs) have metabolic flexibility to maintain cell function under stress, it is unknown whether hyperoxia causes metabolic dysregulation in ECs, leading to lung injury. We hypothesized that hyperoxia alters EC metabolism, which causes EC dysfunction and lung injury. To test this hypothesis, we exposed lung ECs to hyperoxia (95% O2/5% CO2) followed by air recovery (O2/rec). We found that O2/rec reduced mitochondrial oxidative phosphorylation without affecting mitochondrial DNA copy number or mitochondrial mass and that it specifically decreased fatty acid oxidation (FAO) in ECs. This was associated with increased ceramide synthesis and apoptosis. Genetic deletion of carnitine palmitoyltransferase 1a (Cpt1a), a rate-limiting enzyme for carnitine shuttle, further augmented O2/rec-induced apoptosis. O2/rec-induced ceramide synthesis and apoptosis were attenuated when the FAO was enhanced by l-carnitine. Newborn mice were exposed to hyperoxia (>95% O2) between Postnatal Days 1 and 4 and were administered l-carnitine (150 and 300 mg/kg, i.p.) or etomoxir, a specific Cpt1 inhibitor (30 mg/kg, i.p.), daily between Postnatal Days 10 and 14. Etomoxir aggravated O2/rec-induced apoptosis and simplified alveolarization and vascularization in mouse lungs. Similarly, arrested alveolarization and reduced vessel numbers were further augmented in EC-specific Cpt1a-knockout mice compared with wild-type littermates in response to O2/rec. Treatment with l-carnitine (300 mg/kg) attenuated O2/rec-induced lung injury, including simplified alveolarization and decreased vessel numbers. Altogether, enhancing FAO protects against hyperoxia-induced EC apoptosis and lung injury in neonates.
Collapse
Affiliation(s)
- Hongwei Yao
- 1 Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, and
| | - Jiannan Gong
- 1 Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, and.,2 Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China; and
| | - Abigail L Peterson
- 1 Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, and
| | - Xuexin Lu
- 1 Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, and
| | - Peng Zhang
- 3 Cardiology Division, Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
| | - Phyllis A Dennery
- 1 Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, and.,4 Department of Pediatrics, Warren Alpert Medical School, Brown University, Providence, Rhode Island
| |
Collapse
|
34
|
Abstract
Significance: Redox homeostasis is finely tuned and governed by distinct intracellular mechanisms. The dysregulation of this either by external or internal events is a fundamental pathophysiologic base for many pulmonary diseases. Recent Advances: Based on recent discoveries, it is increasingly clear that cellular redox state and oxidation of signaling molecules are critical modulators of lung disease and represent a final common pathway that leads to poor respiratory outcomes. Critical Issues: Based on the wide variety of stimuli that alter specific redox signaling pathways, improved understanding of the disease and patient-specific alterations are needed for the development of therapeutic targets. Further Directions: For the full comprehension of redox signaling in pulmonary disease, it is essential to recognize the role of reactive oxygen intermediates in modulating biological responses. This review summarizes current knowledge of redox signaling in pulmonary development and pulmonary vascular disease.
Collapse
Affiliation(s)
- Gaston Ofman
- Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Trent E Tipple
- Redox Biology Laboratory, Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
35
|
Zhang X, Lu A, Li Z, Sun J, Dai D, Qian L. Exosomes secreted by endothelial progenitor cells improve the bioactivity of pulmonary microvascular endothelial cells exposed to hyperoxia in vitro. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:254. [PMID: 31355221 DOI: 10.21037/atm.2019.05.10] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Paracrine factors secreted by endothelial progenitor cells (EPCs) are suggested to be responsible, in part, for the improved microvascular development in bronchopulmonary dysplasia (BPD) models. This study aims to investigate the potential role of exosomes derived from EPCs (EPC-EXOs), a component of paracrine secretion, in angiogenesis by mediating the activity of PMVECs exposed to hyperoxia. Methods EPCs were isolated from bone marrow of rats. EPC-EXOs were isolated by ExoQuick-TC kits from the conditioned media of EPCs. The PMVECs were divided into three groups, including the normal group, the hyperoxia group (exposed to 85% O2) and the EPC-EXOs treatment group (exposed to 85% O2 and EPC-EXOs with the concentration of 100 µg/mL). The activities of proliferation, migration and tube formation of PMVECs were detected at the endpoint. The mRNA and protein expression levels of VEGF, VEGFR2 and eNOS in different groups were detected by real-time quantitative PCR and western blot. Results We found EPC-EXOs exhibited a cup or biconcave morphology, with the size ranging from 30 to 150 nm, and positive for the characteristic exosomal surface marker proteins, CD63 and TSG101. Comparing to the control group, Hyperoxic stress impaired the proliferation, migration, and tubule formation of PMVECs, and decreased the expression of endothelial nitric oxide synthase (eNOS), vascular endothelial growth factor (VEGF), and vascular endothelial growth factor receptor 2 (VEGFR-2) of PMVECs. Comparing to the hyperoxia group, EPC-EXOs treatment enhanced the bioactivity of PMVECs in vitro, and increased the expression of eNOS, VEGF and VEGFR2. Conclusions Our data demonstrate EPCs secrete exosomes that have independent angiogenic activity in vitro. This may help explain in part the protective effects of EPCs on hyperoxic injury in the developing lung vasculature and may represent a promising therapeutic strategy for BPD.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Respiratory Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Aizhen Lu
- Respiratory Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Zhi Li
- The Children's Hospital of Zhejiang University, School of Medicine, Hangzhou 310052, China
| | - Jiali Sun
- Respiratory Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Dan Dai
- Respiratory Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Liling Qian
- Respiratory Department, Children's Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
36
|
Will JP, Hirani D, Thielen F, Klein F, Vohlen C, Dinger K, Dötsch J, Alejandre Alcázar MA. Strain-dependent effects on lung structure, matrix remodeling, and Stat3/Smad2 signaling in C57BL/6N and C57BL/6J mice after neonatal hyperoxia. Am J Physiol Regul Integr Comp Physiol 2019; 317:R169-R181. [PMID: 31067073 DOI: 10.1152/ajpregu.00286.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of preterm infants, characterized by lung growth arrest and matrix remodeling. Various animal models provide mechanistic insights in the pathogenesis of BPD. Since there is increasing evidence that genetic susceptibility modifies the response to lung injury, we investigated strain-dependent effects in hyperoxia (HYX)-induced lung injury of newborn mice. To this end, we exposed newborn C57BL/6N and C57BL/6J mice to 85% O2 (HYX) or normoxia (NOX; 21% O2) for 28 days, followed by lung excision for histological and molecular measurements. BL/6J-NOX mice exhibited a lower body and lung weight than BL/6N-NOX mice; hyperoxia reduced body weight in both strains and increased lung weight only in BL/6J-HYX mice. Quantitative histomorphometric analyses revealed reduced alveolar formation in lungs of both strains after HYX, but the effect was greater in BL/6J-HYX mice than BL/6N-HYX mice. Septal thickness was lower in BL/6J-NOX mice than BL/6N-NOX mice but increased in both strains after HYX. Elastic fiber density was significantly greater in BL/6J-HYX mice than BL/6N-HYX mice. Lungs of BL/6J-HYX mice were protected from changes in gene expression of fibrillin-1, fibrillin-2, fibulin-4, fibulin-5, and surfactant proteins seen in BL/6N-HYX mice. Finally, Stat3 was activated by HYX in both strains; in contrast, activation of Smad2 was markedly greater in lungs of BL/6N mice than BL/6J mice after HYX. In summary, we demonstrate strain-dependent differences in lung structure and matrix, alveolar epithelial cell markers, and Smad2 (transforming growth factor β) signaling in neonatal HYX-induced lung injury. Strain-dependent effects and genetic susceptibility need be taken into consideration for reproducibility and reliability of results in animal models.
Collapse
Affiliation(s)
- Johannes P Will
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Dharmesh Hirani
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Center of Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Thielen
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Fabian Klein
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Christina Vohlen
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Katharina Dinger
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Jörg Dötsch
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Miguel A Alejandre Alcázar
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Center of Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
37
|
Loering S, Cameron GJM, Starkey MR, Hansbro PM. Lung development and emerging roles for type 2 immunity. J Pathol 2019; 247:686-696. [PMID: 30506724 DOI: 10.1002/path.5211] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Lung development is a complex process mediated through the interaction of multiple cell types, factors and mediators. In mice, it starts as early as embryonic day 9 and continues into early adulthood. The process can be separated into five different developmental stages: embryonic, pseudoglandular, canalicular, saccular, and alveolar. Whilst lung bud formation and branching morphogenesis have been studied extensively, the mechanisms of alveolarisation are incompletely understood. Aberrant lung development can lead to deleterious consequences for respiratory health such as bronchopulmonary dysplasia (BPD), a disease primarily affecting preterm neonates, which is characterised by increased pulmonary inflammation and disturbed alveolarisation. While the deleterious effects of type 1-mediated inflammatory responses on lung development have been well established, the role of type 2 responses in postnatal lung development remains poorly understood. Recent studies indicate that type 2-associated immune cells, such as group 2 innate lymphoid cells and alveolar macrophages, are increased in number during postnatal alveolarisation. Here, we present the current state of understanding of the postnatal stages of lung development and the key cell types and mediators known to be involved. We also provide an overview of how stem cells are involved in lung development and regeneration, and the negative influences of respiratory infections. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Svenja Loering
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Guy J M Cameron
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Center's GrowUpWell and Healthy Lungs, School of Biomedical Sciences and Pharmacy, The University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Center for Inflammation, Centenary Institute and The School of Life Sciences, University of Technology, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
DNA Repair Protein OGG1 in Pulmonary Infection and Other Inflammatory Lung Diseases. OXIDATIVE STRESS IN LUNG DISEASES 2019. [PMCID: PMC7121726 DOI: 10.1007/978-981-13-8413-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
In the last decades, extensive research has uncovered functional roles and underlying mechanisms of DNA repair enzyme 8-oxoguanine DNA glycosylase (OGG1) in the pathogenesis of inflammatory response in infection and other diseases in the lung. OGG1 excises 8-oxo-7,8-dihydroguanine (8-oxo-dG) lesion on DNA that is often induced by generation of reactive oxygen species (ROS) and has been linked to mutations, cancer development, and tissue damage. Most, if not all, environmental toxic agents and mammalian cellular metabolites elicit the generation of ROS, either directly, indirectly, or both, which is among the first cellular responses. ROS in combination with other oxidative molecules/moieties are recognized as a major factor for killing invading pathogens but meanwhile can cause tissue damage. ROS potentially modify proteins, lipids, and DNA due to the strong molecular reactivity. While oxidative stress causes increased levels of all types of oxidatively modified DNA bases, accumulation of 8-oxo-dG in the DNA has been singled out to be a main culprit linking to various inflammatory disease processes. Oxidatively damaged DNA bases such as 8-oxo-dG are primarily repaired by the base excision repair (BER) mechanism, in which OGG1, as the lesion recognition enzyme, plays a fundamental role in fixing this DNA damage. In this chapter, we summarize the roles and potential mechanistic analyses of OGG1 in lung infection and other inflammatory diseases.
Collapse
|
39
|
Tang X. Interleukin-33 (IL-33) Increases Hyperoxia-Induced Bronchopulmonary Dysplasia in Newborn Mice by Regulation of Inflammatory Mediators. Med Sci Monit 2018; 24:6717-6728. [PMID: 30244258 PMCID: PMC6266634 DOI: 10.12659/msm.910851] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background Interleukin-33 (IL-33) has been reported to affect chronic inflammation of the lungs, but its impact on hyperoxia-injured lungs in newborns remains obscure. This study aimed to investigate the role of IL-33 in the lungs of neonatal mice with hyperoxia-induced bronchopulmonary dysplasia (BPD). Material/Methods Twenty-four C57BL/6 baby mice were randomly separated into three groups: the on-air group (N=16); the O2 group (N=8); and the O2 + anti-IL-33 group (N=8). Forced mechanical ventilation with oxygen-rich air (MV-O2) was used in 16 mouse pups. The mouse pups were incubated in containers with either air or 85% O2 for 1, 3, 7, 14, 21, and 28 days after birth. At the end of the treatment period, the mouse lungs were studied by histology, Western blot, and quantitative real-time polymerase chain reaction (qRT-PCR) to examine the expression of the pro-inflammatory mediators, including interleukin (IL)-1β, chemokine (CC motif) ligand 1 (CXCL-1), and monocyte chemoattractant protein-1 (MCP-1). Results Following forced MV-O2, increased levels of IL-33 in whole mouse lungs were associated with impaired alveolar growth and with changes consistent with BPD, including reduced numbers of enlarged alveoli, increased apoptosis, and increased expression of IL-1β, CXCL-1, and MCP-1. IL-33 inhibition improved alveolar development in hyperoxia-impaired lungs and suppressed IL-1β and MCP-1 expression and was associated with increased transforming growth factor-β (TGF-β) signaling, reduced pulmonary NF-κB activity and decreased expression of the TGF-β inhibitor SMAD-7 in forced MV-O2 exposed mouse pups. Conclusions IL-33 increased hyperoxia-induced BPD in newborn mice by regulation of the expression of inflammatory mediators.
Collapse
Affiliation(s)
- Xiqin Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China (mainland)
| |
Collapse
|
40
|
Urs R, Kotecha S, Hall GL, Simpson SJ. Persistent and progressive long-term lung disease in survivors of preterm birth. Paediatr Respir Rev 2018; 28:87-94. [PMID: 29752125 DOI: 10.1016/j.prrv.2018.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 02/05/2023]
Abstract
Preterm birth accounts for approximately 11% of births globally, with rates increasing across many countries. Concurrent advances in neonatal care have led to increased survival of infants of lower gestational age (GA). However, infants born <32 weeks of GA experience adverse respiratory outcomes, manifesting with increased respiratory symptoms, hospitalisation and health care utilisation into early childhood. The development of bronchopulmonary dysplasia (BPD) - the chronic lung disease of prematurity - further increases the risk of poor respiratory outcomes throughout childhood, into adolescence and adulthood. Indeed, survivors of preterm birth have shown increased respiratory symptoms, altered lung structure, persistent and even declining lung function throughout childhood. The mechanisms behind this persistent and sometimes progressive lung disease are unclear, and the implications place those born preterm at increased risk of respiratory morbidity into adulthood. This review aims to summarise what is known about the long-term pulmonary outcomes of contemporary preterm birth, examine the possible mechanisms of long-term respiratory morbidity in those born preterm and discuss addressing the unknowns and potentials for targeted treatments.
Collapse
Affiliation(s)
- Rhea Urs
- Telethon Kids Institute, Perth, Australia; School of Physiotherapy and Exercise Science, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Sailesh Kotecha
- Department of Child Health, School of Medicine, Cardiff University, Cardiff, UK
| | - Graham L Hall
- Telethon Kids Institute, Perth, Australia; School of Physiotherapy and Exercise Science, Faculty of Health Sciences, Curtin University, Perth, Australia
| | | |
Collapse
|
41
|
Goss K. Long-term pulmonary vascular consequences of perinatal insults. J Physiol 2018; 597:1175-1184. [PMID: 30067297 DOI: 10.1113/jp275859] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/10/2018] [Indexed: 01/01/2023] Open
Abstract
Development of the pulmonary circulation is a critical component of fetal lung development, and continues throughout infancy and childhood, marking an extended window of susceptibility to vascular maldevelopment and maladaptation. Perinatal vascular insults may result in abnormal vascular structure or function, including decreased angiogenic signaling and vascular endowment, impaired vasoreactivity through increased pulmonary artery endothelial dysfunction and remodeling, or enhanced genetic susceptibility to pulmonary vascular disease through epigenetic modifications or germline mutations. Although some infants develop early onset pulmonary hypertension, due to the unique adaptive capabilities of the immature host many do not have clinically evident early pulmonary vascular dysfunction. These individuals remain at increased risk for development of late-onset pulmonary hypertension, and may be particularly susceptible to secondary insults. This review will address the role of perinatal vascular insults in the development of late pulmonary vascular dysfunction with an effort to highlight areas of critical research need.
Collapse
Affiliation(s)
- Kara Goss
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
42
|
Yee M, Cohen ED, Domm W, Porter GA, McDavid AN, O’Reilly MA. Neonatal hyperoxia depletes pulmonary vein cardiomyocytes in adult mice via mitochondrial oxidation. Am J Physiol Lung Cell Mol Physiol 2018; 314:L846-L859. [PMID: 29345197 PMCID: PMC6008126 DOI: 10.1152/ajplung.00409.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Supplemental oxygen given to preterm infants has been associated with permanently altering postnatal lung development. Now that these individuals are reaching adulthood, there is growing concern that early life oxygen exposure may also promote cardiovascular disease through poorly understood mechanisms. We previously reported that adult mice exposed to 100% oxygen between postnatal days 0 and 4 develop pulmonary hypertension, defined pathologically by capillary rarefaction, dilation of arterioles and veins, cardiac failure, and a reduced lifespan. Here, Affymetrix Gene Arrays are used to identify early transcriptional changes that take place in the lung before pulmonary capillary rarefaction. We discovered neonatal hyperoxia reduced expression of cardiac muscle genes, including those involved in contraction, calcium signaling, mitochondrial respiration, and vasodilation. Quantitative RT-PCR, immunohistochemistry, and genetic lineage mapping using Myh6CreER; Rosa26RmT/mG mice revealed this reflected loss of pulmonary vein cardiomyocytes. The greatest loss of cadiomyocytes was seen within the lung followed by a graded loss beginning at the hilum and extending into the left atrium. Loss of these cells was seen by 2 wk of age in mice exposed to ≥80% oxygen and was attributed, in part, to reduced proliferation. Administering mitoTEMPO, a scavenger of mitochondrial superoxide during neonatal hyperoxia prevented loss of these cells. Since pulmonary vein cardiomyocytes help pump oxygen-rich blood out of the lung, their early loss following neonatal hyperoxia may contribute to cardiovascular disease seen in these mice, and perhaps in people who were born preterm.
Collapse
Affiliation(s)
- Min Yee
- 1Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - Ethan David Cohen
- 2Department of Medicine, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - William Domm
- 1Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - George A. Porter
- 1Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - Andrew N. McDavid
- 3Biostatistics and Computational Biology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | | |
Collapse
|
43
|
Costa J, Zhu Y, Cox T, Fawcett P, Shaffer T, Alapati D. Inflammatory Response of Pulmonary Artery Smooth Muscle Cells Exposed to Oxidative and Biophysical Stress. Inflammation 2018; 41:1250-1258. [DOI: 10.1007/s10753-018-0772-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Tetri LH, Diffee GM, Barton GP, Braun RK, Yoder HE, Haraldsdottir K, Eldridge MW, Goss KN. Sex-Specific Skeletal Muscle Fatigability and Decreased Mitochondrial Oxidative Capacity in Adult Rats Exposed to Postnatal Hyperoxia. Front Physiol 2018; 9:326. [PMID: 29651255 PMCID: PMC5884929 DOI: 10.3389/fphys.2018.00326] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/15/2018] [Indexed: 01/17/2023] Open
Abstract
Premature birth affects more than 10% of live births, and is characterized by relative hyperoxia exposure in an immature host. Long-term consequences of preterm birth include decreased aerobic capacity, decreased muscular strength and endurance, and increased prevalence of metabolic diseases such as type 2 diabetes mellitus. Postnatal hyperoxia exposure in rodents is a well-established model of chronic lung disease of prematurity, and also recapitulates the pulmonary vascular, cardiovascular, and renal phenotype of premature birth. The objective of this study was to evaluate whether postnatal hyperoxia exposure in rats could recapitulate the skeletal and metabolic phenotype of premature birth, and to characterize the subcellular metabolic changes associated with postnatal hyperoxia exposure, with a secondary aim to evaluate sex differences in this model. Compared to control rats, male rats exposed to 14 days of postnatal hyperoxia then aged to 1 year demonstrated higher skeletal muscle fatigability, lower muscle mitochondrial oxidative capacity, more mitochondrial damage, and higher glycolytic enzyme expression. These differences were not present in female rats with the same postnatal hyperoxia exposure. This study demonstrates detrimental mitochondrial and muscular outcomes in the adult male rat exposed to postnatal hyperoxia. Given that young adults born premature also demonstrate skeletal muscle dysfunction, future studies are merited to determine whether this dysfunction as well as reduced aerobic capacity is due to reduced mitochondrial oxidative capacity and metabolic dysfunction.
Collapse
Affiliation(s)
- Laura H Tetri
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Gary M Diffee
- Department of Kinesiology, University of Wisconsin, Madison, WI, United States
| | - Gregory P Barton
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Rudolf K Braun
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Hannah E Yoder
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Kristin Haraldsdottir
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States.,Department of Kinesiology, University of Wisconsin, Madison, WI, United States
| | - Marlowe W Eldridge
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States.,Department of Kinesiology, University of Wisconsin, Madison, WI, United States
| | - Kara N Goss
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States.,Department of Medicine, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
45
|
Perez VADJ. Long-Term Right Ventricular Adaptation to Postnatal Hyperoxia: Too Much of a Good Thing? Am J Respir Cell Mol Biol 2018; 56:559-560. [PMID: 28459383 DOI: 10.1165/rcmb.2016-0429ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
46
|
Menon RT, Shrestha AK, Reynolds CL, Barrios R, Shivanna B. Long-term pulmonary and cardiovascular morbidities of neonatal hyperoxia exposure in mice. Int J Biochem Cell Biol 2018; 94:119-124. [PMID: 29223466 PMCID: PMC5745292 DOI: 10.1016/j.biocel.2017.12.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/14/2017] [Accepted: 12/01/2017] [Indexed: 12/14/2022]
Abstract
Pulmonary hypertension (PH) frequently occurs in infants with bronchopulmonary dysplasia (BPD), causing increased mortality and right ventricular (RV) dysfunction that persists into adulthood. A first step in developing better therapeutic options is identifying and characterizing an appropriate animal model. Previously, we characterized the short-term morbidities of a model in which C57BL/6J wild-type (WT) mice were exposed to 70% O2 (hyperoxia) during the neonatal period. Here, we aimed to determine the long-term morbidities using lung morphometry, echocardiography (Echo), and cardiac magnetic resonance imaging (cMRI). The major highlight of this study is the use of the state-of-the art imaging technique, cMRI, in mice to characterize the long-term cardiac effects of neonatal hyperoxia exposure. To this end, WT mice were exposed to 21% O2 (normoxia) or hyperoxia for two weeks of life, followed by recovery in normoxia for six weeks. Alveolarization, pulmonary vascularization, pulmonary hypertension, and RV function were quantified at eight weeks. We found that hyperoxia exposure resulted in persistent alveolar and pulmonary vascular simplification. Furthermore, the Echo and cMRI studies demonstrated that hyperoxia-exposed mice had signs of PH and RV dysfunction as indicated by increased RV pressure, mass, and end-systolic and -diastolic volumes, and decreased RV stroke volume and ejection fractions. Taken together, our results demonstrate that neonatal hyperoxia exposure in mice cause cardiopulmonary morbidities that persists into adulthood and provides evidence for the use of this model to develop novel therapies for BPD infants with PH.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Atmosphere Exposure Chambers
- Bronchopulmonary Dysplasia/physiopathology
- Disease Models, Animal
- Echocardiography
- Feasibility Studies
- Female
- Heart/diagnostic imaging
- Heart/physiopathology
- Hyperoxia/physiopathology
- Hypertension, Pulmonary/diagnostic imaging
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/pathology
- Lung/blood supply
- Lung/diagnostic imaging
- Lung/pathology
- Magnetic Resonance Imaging
- Male
- Mice, Inbred C57BL
- Myocardium/pathology
- Organ Size
- Pulmonary Circulation
- Stroke Volume
- Time Factors
- Ultrasonography, Doppler, Pulsed
- Ventricular Dysfunction, Right/diagnostic imaging
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/pathology
Collapse
Affiliation(s)
- Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Corey L Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, TX, USA
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
47
|
Firsova AB, Bird AD, Abebe D, Ng J, Mollard R, Cole TJ. Fresh Noncultured Endothelial Progenitor Cells Improve Neonatal Lung Hyperoxia-Induced Alveolar Injury. Stem Cells Transl Med 2017; 6:2094-2105. [PMID: 29027762 PMCID: PMC5702522 DOI: 10.1002/sctm.17-0093] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023] Open
Abstract
Treatment of preterm human infants with high oxygen can result in disrupted lung alveolar and vascular development. Local or systemic administration of endothelial progenitor cells (EPCs) is reported to remedy such disruption in animal models. In this study, the effects of both fresh (enriched for KDR) and cultured bone marrow (BM)-derived cell populations with EPC characteristics were examined following hyperoxia in neonatal mouse lungs. Intraperitoneal injection of fresh EPCs into five-day-old mice treated with 90% oxygen resulted in full recovery of hyperoxia-induced alveolar disruption by 56 days of age. Partial recovery in septal number following hyperoxia was observed following injection of short-term cultured EPCs, yet aberrant tissue growths appeared following injection of long-term cultured cells. Fresh and long-term cultured cells had no impact on blood vessel development. Short-term cultured cells increased blood vessel number in normoxic and hyperoxic mice by 28 days but had no impact on day 56. Injection of fresh EPCs into normoxic mice significantly reduced alveolarization compared with phosphate buffered saline-injected normoxic controls. These results indicate that fresh BM EPCs have a higher and safer corrective profile in a hyperoxia-induced lung injury model compared with cultured BM EPCs but may be detrimental to the normoxic lung. The appearance of aberrant tissue growths and other side effects following injection of cultured EPCs warrants further investigation. Stem Cells Translational Medicine 2017;6:2094-2105.
Collapse
Affiliation(s)
- Alexandra B Firsova
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - A Daniel Bird
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Degu Abebe
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Judy Ng
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Richard Mollard
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Department of Veterinary and Agricultural Science, University of Melbourne, Parkville, Victoria, Australia
| | - Timothy J Cole
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
48
|
Perez M, Lee KJ, Cardona HJ, Taylor JM, Robbins ME, Waypa GB, Berkelhamer SK, Farrow KN. Aberrant cGMP signaling persists during recovery in mice with oxygen-induced pulmonary hypertension. PLoS One 2017; 12:e0180957. [PMID: 28792962 PMCID: PMC5549891 DOI: 10.1371/journal.pone.0180957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 06/23/2017] [Indexed: 12/25/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD), a common complication of preterm birth, is associated with pulmonary hypertension (PH) in 25% of infants with moderate to severe BPD. Neonatal mice exposed to hyperoxia for 14d develop lung disease similar to BPD, with evidence of associated PH. The cyclic guanosine monophosphate (cGMP) signaling pathway has not been well studied in BPD-associated PH. In addition, there is little data about the natural history of hyperoxia-induced PH in mice or the utility of phosphodiesterase-5 (PDE5) inhibition in established disease. C57BL/6 mice were placed in room air or 75% O2 within 24h of birth for 14d, followed by recovery in room air for an additional 7 days (21d). Additional pups were treated with either vehicle or sildenafil for 7d during room air recovery. Mean alveolar area, pulmonary artery (PA) medial wall thickness (MWT), RVH, and vessel density were evaluated at 21d. PA protein from 21d animals was analyzed for soluble guanylate cyclase (sGC) activity, PDE5 activity, and cGMP levels. Neonatal hyperoxia exposure results in persistent alveolar simplification, RVH, decreased vessel density, increased MWT, and disrupted cGMP signaling despite a period of room air recovery. Delayed treatment with sildenafil during room air recovery is associated with improved RVH and decreased PA PDE5 activity, but does not have significant effects on alveolar simplification, PA remodeling, or vessel density. These data are consistent with clinical studies suggesting inconsistent effects of sildenafil treatment in infants with BPD-associated PH.
Collapse
Affiliation(s)
- Marta Perez
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
- * E-mail:
| | - Keng Jin Lee
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
| | - Herminio J. Cardona
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
| | - Joann M. Taylor
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
| | - Mary E. Robbins
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
| | - Gregory B. Waypa
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
| | - Sara K. Berkelhamer
- Department of Pediatrics, University at Buffalo, Buffalo, NY, United States of America
| | - Kathryn N. Farrow
- Department of Pediatrics, Northwestern University, Chicago, IL, United States of America
| |
Collapse
|
49
|
Goss KN, Kumari S, Tetri LH, Barton G, Braun RK, Hacker TA, Eldridge MW. Postnatal Hyperoxia Exposure Durably Impairs Right Ventricular Function and Mitochondrial Biogenesis. Am J Respir Cell Mol Biol 2017; 56:609-619. [PMID: 28129517 PMCID: PMC5449491 DOI: 10.1165/rcmb.2016-0256oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/22/2016] [Indexed: 12/17/2022] Open
Abstract
Prematurity complicates 12% of births, and young adults with a history of prematurity are at risk to develop right ventricular (RV) hypertrophy and impairment. The long-term risk for pulmonary vascular disease, as well as mechanisms of RV dysfunction and ventricular-vascular uncoupling after prematurity, remain poorly defined. Using an established model of prematurity-related lung disease, pups from timed-pregnant Sprague Dawley rats were randomized to normoxia or hyperoxia (fraction of inspired oxygen, 0.85) exposure for the first 14 days of life. After aging to 1 year in standard conditions, rats underwent hemodynamic assessment followed by tissue harvest for biochemical and histological evaluation. Aged hyperoxia-exposed rats developed significantly greater RV hypertrophy, associated with a 40% increase in RV systolic pressures. Although cardiac index was similar, hyperoxia-exposed rats demonstrated a reduced RV ejection fraction and significant RV-pulmonary vascular uncoupling. Hyperoxia-exposed RV cardiomyocytes demonstrated evidence of mitochondrial dysregulation and mitochondrial DNA damage, suggesting potential mitochondrial dysfunction as a cause of RV dysfunction. Aged rats exposed to postnatal hyperoxia recapitulate many features of young adults born prematurely, including increased RV hypertrophy and decreased RV ejection fraction. Our data suggest that postnatal hyperoxia exposure results in mitochondrial dysregulation that persists into adulthood with eventual RV dysfunction. Further evaluation of long-term mitochondrial function is warranted in both animal models of premature lung disease and in human adults who were born preterm.
Collapse
MESH Headings
- Aging/pathology
- Animals
- Animals, Newborn
- Autophagy
- Body Weight
- DNA Damage
- DNA, Mitochondrial/metabolism
- Female
- Fibrosis
- Gene Expression Profiling
- Hemodynamics
- Hyperoxia/complications
- Hyperoxia/diagnostic imaging
- Hyperoxia/metabolism
- Hyperoxia/physiopathology
- Hypertrophy, Right Ventricular/diagnostic imaging
- Hypertrophy, Right Ventricular/etiology
- Hypertrophy, Right Ventricular/genetics
- Hypertrophy, Right Ventricular/physiopathology
- Male
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Organ Size
- Organelle Biogenesis
- Rats, Sprague-Dawley
- Ventricular Function, Right
Collapse
Affiliation(s)
- Kara N. Goss
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Rankin Laboratory of Pulmonary Medicine, and
| | - Santosh Kumari
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Rankin Laboratory of Pulmonary Medicine, and
| | - Laura H. Tetri
- Division of Pediatric Critical Care, Department of Pediatrics
- Rankin Laboratory of Pulmonary Medicine, and
| | - Greg Barton
- Division of Pediatric Critical Care, Department of Pediatrics
- Rankin Laboratory of Pulmonary Medicine, and
| | - Rudolf K. Braun
- Division of Pediatric Critical Care, Department of Pediatrics
- Rankin Laboratory of Pulmonary Medicine, and
| | - Timothy A. Hacker
- Cardiovascular Research Center, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Marlowe W. Eldridge
- Division of Pediatric Critical Care, Department of Pediatrics
- Rankin Laboratory of Pulmonary Medicine, and
| |
Collapse
|
50
|
Cox AM, Gao Y, Perl AKT, Tepper RS, Ahlfeld SK. Cumulative effects of neonatal hyperoxia on murine alveolar structure and function. Pediatr Pulmonol 2017; 52:616-624. [PMID: 28186703 PMCID: PMC5621136 DOI: 10.1002/ppul.23654] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/07/2016] [Accepted: 11/23/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) results from alveolar simplification and abnormal development of alveolar and capillary structure. Survivors of BPD display persistent deficits in airflow and membrane and vascular components of alveolar gas diffusion. Despite being the defining feature of BPD, various neonatal hyperoxia models of BPD have not routinely assessed pulmonary gas diffusion. METHODS To simulate the most commonly-utilized neonatal hyperoxia models, we exposed neonatal mice to room air or ≥90% hyperoxia during key stages of distal lung development: through the first 4 (saccular), 7 (early alveolar), or 14 (bulk alveolar) postnatal days, followed by a period of recovery in room air until 8 weeks of age when alveolar septation is essentially complete. We systematically assessed and correlated the effects of neonatal hyperoxia on the degree of alveolar-capillary structural and functional impairment. We hypothesized that the degree of alveolar-capillary simplification would correlate strongly with worsening diffusion impairment. RESULTS Neonatal hyperoxia exposure, of any duration, resulted in alveolar simplification and impaired pulmonary gas diffusion. Mean Linear Intercept increased in proportion to the length of hyperoxia exposure while alveolar and total lung volume increased markedly only with prolonged exposure. Surprisingly, despite having a similar effect on alveolar surface area, only prolonged hyperoxia for 14 days resulted in reduced pulmonary microvascular volume. Estimates of alveolar and capillary structure, in general, correlated poorly with assessment of gas diffusion. CONCLUSION Our results help define the physiological and structural consequences of commonly-employed neonatal hyperoxia models of BPD and inform their clinical utility. Pediatr Pulmonol. 2017;52:616-624. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Angela M. Cox
- Program in Developmental Biology and Neonatal Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, Indiana
- Division of Neonatology, James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yong Gao
- Program in Developmental Biology and Neonatal Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, Indiana
- Program in Pulmonary Inflammation, Asthma and Allergic Diseases, Herman B Wells Center for Pediatric Research, Indianapolis, Indiana
| | - Anne-Karina T. Perl
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Robert S. Tepper
- Program in Pulmonary Inflammation, Asthma and Allergic Diseases, Herman B Wells Center for Pediatric Research, Indianapolis, Indiana
- Division of Pulmonary Medicine, Department of Pediatrics, James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana
| | - Shawn K. Ahlfeld
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Correspondence to: Shawn K. Ahlfeld, MD, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, MLC 7009, Cincinnati, OH 45229.
| |
Collapse
|