1
|
Yang K, He T, Sun X, Dong W. Post-translational modifications and bronchopulmonary dysplasia. Front Pediatr 2025; 12:1426030. [PMID: 39830627 PMCID: PMC11738936 DOI: 10.3389/fped.2024.1426030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Bronchopulmonary dysplasia is a prevalent respiratory disorder posing a significant threat to the quality of life in premature infants. Its pathogenesis is intricate, and therapeutic options are limited. Besides genetic coding, protein post-translational modification plays a pivotal role in regulating cellular function, contributing complexity and diversity to substrate proteins and influencing various cellular processes. Substantial evidence indicates that post-translational modifications of several substrate proteins are intricately related to the molecular mechanisms underlying bronchopulmonary dysplasia. These modifications facilitate the progression of bronchopulmonary dysplasia through a cascade of signal transduction events. This review outlines the relationships between substrate protein phosphorylation, acetylation, ubiquitination, SUMOylation, methylation, glycosylation, glycation, S-glutathionylation, S-nitrosylation and bronchopulmonary dysplasia. The aim is to provide novel insights into bronchopulmonary dysplasia's pathogenesis and potential therapeutic targets for clinical management.
Collapse
Affiliation(s)
| | | | | | - Wenbin Dong
- Department of Neonatology, Children’s Medical Center, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Tuhy T, Coursen JC, Graves T, Patatanian M, Cherry C, Niedermeyer SE, Khan SL, Rosen DT, Croglio MP, Elnashar M, Kolb TM, Mathai SC, Damico RL, Hassoun PM, Shimoda LA, Suresh K, Aldred MA, Simpson CE. Single-cell transcriptomics reveal diverging pathobiology and opportunities for precision targeting in scleroderma-associated versus idiopathic pulmonary arterial hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620225. [PMID: 39484590 PMCID: PMC11527343 DOI: 10.1101/2024.10.25.620225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Introduction Pulmonary arterial hypertension (PAH) involves progressive cellular and molecular change within the pulmonary vasculature, leading to increased vascular resistance. Current therapies targeting nitric oxide (NO), endothelin, and prostacyclin pathways yield variable treatment responses. Patients with systemic sclerosis-associated PAH (SSc-PAH) often experience worse outcomes than those with idiopathic PAH (IPAH). Methods Lung tissue samples from four SSc-PAH, four IPAH, and four failed donor specimens were obtained from the Pulmonary Hypertension Breakthrough Initiative (PHBI) lung tissue bank. Single-cell RNA sequencing (scRNAseq) was performed using the 10X Genomics Chromium Flex platform. Data normalization, clustering, and differential expression analysis were conducted using Seurat. Additional analyses included gene set enrichment analysis (GSEA), transcription factor activity analysis, and ligand-receptor signaling. Pharmacotranscriptomic screening was performed using the Connectivity Map. Results SSc-PAH samples showed a higher proportion of fibroblasts and dendritic cells/macrophages compared to IPAH and donor samples. GSEA revealed enriched pathways related to epithelial-to-mesenchymal transition (EMT), apoptosis, and vascular remodeling in SSc-PAH samples. There was pronounced differential gene expression across diverse pulmonary vascular cell types and in various epithelial cell types in both IPAH and SSc-PAH, with epithelial to endothelial cell signaling observed. Macrophage to endothelial cell signaling was particularly pronounced in SSc-PAH. Pharmacotranscriptomic screening identified TIE2, GSK-3, and PKC inhibitors, among other compounds, as potential drug candidates for reversing SSc-PAH gene expression signatures. Discussion Overlapping and distinct gene expression patterns exist in SSc-PAH versus IPAH, with significant molecular differences suggesting unique pathogenic mechanisms in SSc-PAH. These findings highlight the potential for precision-targeted therapies to improve SSc-PAH patient outcomes. Future studies should validate these targets clinically and explore their therapeutic efficacy.
Collapse
Affiliation(s)
- Tijana Tuhy
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Julie C Coursen
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Tammy Graves
- Division of Pulmonary Medicine, Indiana University, Indianapolis, IN, USA
| | - Michael Patatanian
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Christopher Cherry
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shannon E Niedermeyer
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah L Khan
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Darin T Rosen
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michael P Croglio
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Todd M Kolb
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Stephen C Mathai
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rachel L Damico
- Division of Pulmonary and Critical Care Medicine, University of Miami, Miami, FL, USA
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Micheala A Aldred
- Division of Pulmonary Medicine, Indiana University, Indianapolis, IN, USA
| | - Catherine E Simpson
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
3
|
Wu D, Bai D, Yang M, Wu B, Xu W. Role of Sox9 in BPD and its effects on the Wnt/β-catenin pathway and AEC-II differentiation. Cell Death Discov 2024; 10:20. [PMID: 38212314 PMCID: PMC10784471 DOI: 10.1038/s41420-023-01795-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/24/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024] Open
Abstract
The excessive activation of the Wnt/β-catenin signaling pathway is an important regulatory mechanism that underlies the excessive proliferation and impaired differentiation of type 2 alveolar epithelial cells (AEC-II) in bronchopulmonary dysplasia (BPD). Sox9 has been shown to be an important repressor of the Wnt/β-catenin signaling pathway and plays an important regulatory role in various pathophysiological processes. We found that the increased expression of Sox9 in the early stages of BPD could downregulate the expression of β-catenin and promote the differentiation of AEC-II cells into AEC-I, thereby alleviating the pathological changes in BPD. The expression of Sox9 in BPD is regulated by long noncoding RNA growth arrest-specific 5. These findings may provide new targets for the early intervention of BPD.
Collapse
Affiliation(s)
- Di Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Intensive Care unit, The Sixth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dongqin Bai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Miao Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bo Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Benny M, Sharma M, Kulandavelu S, Chen P, Tian R, Ballengee S, Huang J, Levine AF, Claure M, Schmidt AF, Vazquez-Padron RI, Rodrigues CO, Wu S, Velazquez OC, Young KC. Protective role of CXCR7 activation in neonatal hyperoxia-induced systemic vascular remodeling and cardiovascular dysfunction in juvenile rats. Sci Rep 2023; 13:19538. [PMID: 37945645 PMCID: PMC10636097 DOI: 10.1038/s41598-023-46422-3] [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: 05/16/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Neonatal hyperoxia induces long-term systemic vascular stiffness and cardiovascular remodeling, but the mechanisms are unclear. Chemokine receptor 7 (CXCR7) represents a key regulator of vascular homeostasis and repair by modulating TGF-β1 signaling. This study investigated whether pharmacological CXCR7 agonism prevents neonatal hyperoxia-induced systemic vascular stiffness and cardiac dysfunction in juvenile rats. Newborn Sprague Dawley rat pups assigned to room air or hyperoxia (85% oxygen), received CXCR7 agonist, TC14012 or placebo for 3 weeks. These rat pups were maintained in room air until 6 weeks when aortic pulse wave velocity doppler, cardiac echocardiography, aortic and left ventricular (LV) fibrosis were assessed. Neonatal hyperoxia induced systemic vascular stiffness and cardiac dysfunction in 6-week-old rats. This was associated with decreased aortic and LV CXCR7 expression. Early treatment with TC14012, partially protected against neonatal hyperoxia-induced systemic vascular stiffness and improved LV dysfunction and fibrosis in juvenile rats by decreasing TGF-β1 expression. In vitro, hyperoxia-exposed human umbilical arterial endothelial cells and coronary artery endothelial cells had increased TGF-β1 levels. However, treatment with TC14012 significantly reduced the TGF-β1 levels. These results suggest that dysregulation of endothelial CXCR7 signaling may contribute to neonatal hyperoxia-induced systemic vascular stiffness and cardiac dysfunction.
Collapse
Affiliation(s)
- Merline Benny
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA.
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Mayank Sharma
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shathiyah Kulandavelu
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - PingPing Chen
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Runxia Tian
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sydne Ballengee
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jiang Huang
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Amanda F Levine
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matteo Claure
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Augusto F Schmidt
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Claudia O Rodrigues
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shu Wu
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omaida C Velazquez
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karen C Young
- Department of Pediatrics, University of Miami Miller School of Medicine, 1580 NW 10Th Avenue, RM-344, Miami, FL, 33136, USA
- Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
5
|
Sun T, Yu H, Li D, Zhang H, Fu J. Emerging role of metabolic reprogramming in hyperoxia-associated neonatal diseases. Redox Biol 2023; 66:102865. [PMID: 37659187 PMCID: PMC10480540 DOI: 10.1016/j.redox.2023.102865] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/19/2023] [Accepted: 08/25/2023] [Indexed: 09/04/2023] Open
Abstract
Oxygen therapy is common during the neonatal period to improve survival, but it can increase the risk of oxygen toxicity. Hyperoxia can damage multiple organs and systems in newborns, commonly causing lung conditions such as bronchopulmonary dysplasia and pulmonary hypertension, as well as damage to other organs, including the brain, gut, and eyes. These conditions are collectively referred to as newborn oxygen radical disease to indicate the multi-system damage caused by hyperoxia. Hyperoxia can also lead to changes in metabolic pathways and the production of abnormal metabolites through a process called metabolic reprogramming. Currently, some studies have analyzed the mechanism of metabolic reprogramming induced by hyperoxia. The focus has been on mitochondrial oxidative stress, mitochondrial dynamics, and multi-organ interactions, such as the lung-gut, lung-brain, and brain-gut axes. In this article, we provide an overview of the major metabolic pathway changes reported in hyperoxia-associated neonatal diseases and explore the potential mechanisms of metabolic reprogramming. Metabolic reprogramming induced by hyperoxia can cause multi-organ metabolic disorders in newborns, including abnormal glucose, lipid, and amino acid metabolism. Moreover, abnormal metabolites may predict the occurrence of disease, suggesting their potential as therapeutic targets. Although the mechanism of metabolic reprogramming caused by hyperoxia requires further elucidation, mitochondria and the gut-lung-brain axis may play a key role in metabolic reprogramming.
Collapse
Affiliation(s)
- Tong Sun
- Department of Pediatics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Danni Li
- Department of Pediatics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Jianhua Fu
- Department of Pediatics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Gong W, Li M, Zhao L, Wang P, Wang X, Wang B, Liu X, Tu X. Sustained release of a highly specific GSK3β inhibitor SB216763 in the PCL scaffold creates an osteogenic niche for osteogenesis, anti-adipogenesis, and potential angiogenesis. Front Bioeng Biotechnol 2023; 11:1215233. [PMID: 37576993 PMCID: PMC10419179 DOI: 10.3389/fbioe.2023.1215233] [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: 05/01/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
The safe and effective use of Wnt signaling is a hot topic in developing osteogenic drugs. SB216763 (S33) is a widely used highly specific GSK3β inhibitor. Here, we show that S33 initiates canonical Wnt signaling by inhibiting GSK3β activity in the bone marrow stromal cell line ST2 and increases osteoblast marker alkaline phosphatase activity, osteoblast marker gene expression including Alpl, Col1α1, and Runx2, promoting osteogenic differentiation and mineralization of ST2 cells. In addition, S33 suppressed the expression of adipogenic transcription factors Pparg and Cebpa in ST2 cells to suppress adipogenesis. ICRT-14, a specific transcriptional inhibitor of Wnt signaling, reversed the effects of S33 on the differentiation of ST2 cells. S33 also increased the expression of osteoclast cytokines RANKL and Opg but decreased the RANKL/Opg ratio and had the potential to inhibit osteoclast differentiation. In addition, we printed the PSCI3D (polycaprolactone, S33, cell-integrated 3D) scaffolds using a newly established integrated 3D printing system for hard materials and cells. S33 sustained release in the hydrogel of the scaffold with 25.4% release on day 1% and 81.7% release over 7 days. Cells in the scaffolds had good cell viability. The ratio of live/dead cells remained above 94% for 7 days, while the cells in the scaffolds proliferated linearly, and the proliferative activity of the PSCI3D scaffold group increased 1.4-fold and 1.7-fold on days 4 and 7, respectively. Similarly, in PSCI3D scaffolds, osteogenic differentiation of st2 cells was increased. The alkaline phosphatase activity increased 1.4- and 4.0-fold on days 7 and 14, respectively, and mineralization increased 1.7-fold at 21 days. In addition, PSCI3D conditioned medium promoted migration and tubulogenesis of HUVECs, and S33 upregulated the expression of Vegfa, a key factor in angiogenesis. In conclusion, our study suggests that S33 functions in osteogenesis, anti-adipogenesis, and potential inhibition of osteoclast differentiation. And the sustained release of S33 in PSCI3D scaffolds creates a safe osteogenic niche, which promotes cell proliferation, osteogenesis, and angiogenesis and has application prospects.
Collapse
Affiliation(s)
- Weimin Gong
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Molin Li
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Lizhou Zhao
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Pengtao Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xiaofang Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Bo Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xing Liu
- Department of Orthopedics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Zou W, Ye D, Liu S, Hu J, Zhu T, He F, Ran P. GSK-3β Inhibitors Attenuate the PM2.5-Induced Inflammatory Response in Bronchial Epithelial Cells. Int J Chron Obstruct Pulmon Dis 2021; 16:2845-2856. [PMID: 34703220 PMCID: PMC8523522 DOI: 10.2147/copd.s327887] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose PM2.5-associated airway inflammation has recently been recognized as pivotal to the development of COPD. Aberrant glycogen synthase kinase (GSK)-3β signaling is linked to the inflammatory response. Therefore, we investigated the effects of GSK-3β inhibitors on the PM2.5-induced inflammatory response in bronchial epithelial cells. Methods The production of phosphorylated GSK-3β (p-GSK-3β) was analyzed by immunohistochemistry with PM2.5-induced mice. HBECs were treated with various inhibitors targeting GSK-3β or JNK before PM2.5 stimulation. The production of GSK-3β signaling was analyzed by Western blotting. Inflammatory cytokine production was detected by qRT–PCR and ELISA. Results PM2.5 exposure caused lung inflammation, upregulated serum concentrations of HMGB1 and IL-6, decreased IL-10 expression, and significantly attenuated p-GSK-3β production in mice. HBECs exposed to PM2.5 showed significantly reduced p-GSK-3β production, an increased ratio of p-JNK/JNK, increased NF-κB activation and IκB degradation, and upregulated the inflammatory cytokines HMGB1 and IL-6. Intervention with GSK-3β inhibitors TDZD-8 and SB216763 significantly suppressed PM2.5-induced outcomes. Moreover, the JNK inhibitor SP600125 also reduced the level of NF-κB phosphorylation induced by PM2.5. The differences in the levels of inflammation-related cytokines in the TDZD-8 groups were greater than those in the SB216763 groups. Conclusion Inhibition of GSK-3β weakens the PM2.5-induced inflammatory response by regulating the JNK/NF-κB signaling pathway in bronchial epithelial cells.
Collapse
Affiliation(s)
- Weifeng Zou
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Dong Ye
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Sha Liu
- The Second Hospital, University of South China, Hengyang, Hunan, People's Republic of China
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Tao Zhu
- State Key Laboratory of Respiratory Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Fang He
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Pixin Ran
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
8
|
Ali A, Zambrano R, Duncan MR, Chen S, Luo S, Yuan H, Chen P, Benny M, Schmidt A, Young K, Kerr N, de Rivero Vaccari JP, Keane RW, Dietrich WD, Wu S. Hyperoxia-activated circulating extracellular vesicles induce lung and brain injury in neonatal rats. Sci Rep 2021; 11:8791. [PMID: 33888735 PMCID: PMC8062626 DOI: 10.1038/s41598-021-87706-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/16/2021] [Indexed: 01/02/2023] Open
Abstract
Hyperoxia-induced lung injury plays a key role in the development of bronchopulmonary dysplasia (BPD), characterized by inflammatory injury and impaired lung development in preterm infants. Although BPD is a predictor of poor neurodevelopmental outcomes, currently it is uncertain how lung injury contributes to brain injury in preterm infants. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membranous structures that regulate intercellular and inter-organ communications. Gasdermin D (GSDMD) has emerged as a key executor of inflammasome-mediated cell death and inflammation. In this study, we utilized a neonatal rat model of BPD to assess if hyperoxia stimulates lung release of circulating EVs and if these EVs induce lung and brain injury. We found that hyperoxia-exposed rats had elevated numbers of plasma-derived EVs compared to rats maintained in room air. These EVs also had increased cargos of surfactant protein C, a marker of type II alveolar epithelial cells (AEC), and the active (p30) form of GSDMD. When these EVs were adoptively transferred into normal newborn rats via intravenous injection, they were taken up both by lung and brain tissues. Moreover, EVs from hyperoxic animals induced not only the pathological hallmarks of BPD, but also brain inflammatory injury in recipient rats, as well as inducing cell death in cultured pulmonary vascular endothelial cells and neural stem cells (NSC). Similarly, hyperoxia-exposed cultured AEC-like cells released EVs that also contained increased GSDMD-p30 and these EVs induced pyroptotic cell death in NSC. Overall, these data indicate that hyperoxia-activated circulating EVs mediate a lung to brain crosstalk resulting in brain injury and suggest a mechanism that links lung injury and neurodevelopmental impairment in BPD infants.
Collapse
Affiliation(s)
- Anum Ali
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Ronald Zambrano
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Matthew R Duncan
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Shaoyi Chen
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Shihua Luo
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Huijun Yuan
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Pingping Chen
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Merline Benny
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Augusto Schmidt
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Karen Young
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Nadine Kerr
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Shu Wu
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA.
| |
Collapse
|
9
|
Zhang H, Lu H, Yu L, Yuan J, Qin S, Li C, Ge RS, Chen H, Ye L. Effects of gestational exposure to perfluorooctane sulfonate on the lung development of offspring rats. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 272:115535. [PMID: 33223333 DOI: 10.1016/j.envpol.2020.115535] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 06/11/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a man-made fluorosurfactant widely used in industry and consumer products. Previous studies with rats suggested that gestational exposure to PFOS may affect the lung development in the offspring. The mechanism, however, is still unknown. In the present study, we have exposed 24 pregnant SD rats from gestational day 12-18 to different doses of PFOS (0, 1 or 5 mg/kg BW/day). The lungs of the offspring were analyzed at postnatal days 1, 3, 7 and 14. PFOS treatment appeared to reduce the alveolar numbers, resulting in simplified alveolar structure and thickened alveolar septa. Also, PFOS treated animals had increased lung inflammation with up-regulated inflammasome associated proteins NLRP3, ASC, Caspase-1 and GSDMD and increased inflammatory cytokines IL-18 and IL-1β. At the same time, HIF-1α and VEGFA were significantly down-regulated. Since HIF-1α and VEGFA are critical factors promoting alveolar development and pulmonary angiogenesis, these results suggested that PFOS may also affect lung development by inhibiting HIF-1α and VEGFA expression. Our results here indicate that gestational exposure to PFOS may affect lung development in the offspring with pathological characteristics similar to bronchopulmonary dysplasia (BPD), a severe lung developmental defect. The results also suggest that environmental factors such as PFOS may contribute to the increasing incidence of developmental lung diseases, such as BPD, by elevating lung inflammation and inhibiting lung development.
Collapse
Affiliation(s)
- Huishan Zhang
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China; Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hemin Lu
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Lin Yu
- Department of Pediatrics, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, China
| | - Jiexin Yuan
- Department of Pediatric Cardiothoracic Surgery, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shan Qin
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Cong Li
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China
| | - Ren-Shan Ge
- Department of Anesthesiology, Perioperative Medicine, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Haolin Chen
- Department of Anesthesiology, Perioperative Medicine, Zhejiang Province Key Lab of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Leping Ye
- Department of Pediatrics, Peking University First Hospital, No.1 Xi'an Men Street, West District, Beijing, 100034, China.
| |
Collapse
|
10
|
Benny M, Hernandez DR, Sharma M, Yousefi K, Kulandavelu S, Batlahally S, Zambrano R, Chen P, Martinez EC, Schmidt AF, Shehadeh LA, Vasquez-Padron RI, Wu S, Velazquez OC, Young KC. Neonatal hyperoxia exposure induces aortic biomechanical alterations and cardiac dysfunction in juvenile rats. Physiol Rep 2020; 8:e14334. [PMID: 31925922 PMCID: PMC6954121 DOI: 10.14814/phy2.14334] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Supplemental oxygen (O2) therapy in preterm infants impairs lung development, but the impact of O2 on long‐term systemic vascular structure and function has not been well‐explored. The present study tested the hypothesis that neonatal O2 therapy induces long‐term structural and functional alterations in the systemic vasculature, resulting in vascular stiffness observed in children and young adults born preterm. Newborn Sprague‐Dawley rats were exposed to normoxia (21% O2) or hyperoxia (85% O2) for 1 and 3 weeks. A subgroup exposed to 3 weeks hyperoxia was recovered in normoxia for an additional 3 weeks. Aortic stiffness was assessed by pulse wave velocity (PWV) using Doppler ultrasound and pressure myography. Aorta remodeling was assessed by collagen deposition and expression. Left ventricular (LV) function was assessed by echocardiography. We found that neonatal hyperoxia exposure increased vascular stiffness at 3 weeks, which persisted after normoxic recovery at 6 weeks of age. These findings were accompanied by increased PWV, aortic remodeling, and altered LV function as evidenced by decreased ejection fraction, cardiac output, and stroke volume. Importantly, these functional changes were associated with increased collagen deposition in the aorta. Together, these findings demonstrate that neonatal hyperoxia induces early and sustained biomechanical alterations in the systemic vasculature and impairs LV function. Early identification of preterm infants who are at risk of developing systemic vascular dysfunction will be crucial in developing targeted prevention strategies that may improve the long‐term cardiovascular outcomes in this vulnerable population.
Collapse
Affiliation(s)
- 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
| | - Diana R Hernandez
- Department of Surgery, 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
| | - Keyvan Yousefi
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida.,Division of Cardiology, Department of Medicine, 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
| | - 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
| | - 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
| | - 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
| | - Eliana C Martinez
- 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
| | - Augusto F 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
| | - Lina A Shehadeh
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.,Division of Cardiology, Department of Medicine, 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
| | - Omaida C Velazquez
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Karen C 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
| |
Collapse
|
11
|
Nischarin downregulation attenuates cell injury induced by oxidative stress via Wnt signaling. Neuroreport 2020; 31:1199-1207. [PMID: 33075003 DOI: 10.1097/wnr.0000000000001536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Nischarin (NISCH) is a key protein functioning as a molecular scaffold and thereby hosting interactions with several protein partners. Here, we aimed to investigate whether NISCH downregulation could protect rat pheochromocytoma (PC12) cells against oxidative stress-induced injury using a model of cell injury induced by hydrogen peroxide (H2O2). Cell viability was evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Cell apoptosis rate was evaluated using flow cytometry. The expressions of apoptosis-related proteins Bax, Bcl-2, caspase-3 and NISCH were examined via Western blot analysis and immunofluorescence staining analyses. The expressions of NISCH, glycogen synthase kinase-3β (GSK-3β) and T-cell factor-1 (TCF-1) were examined using Western blot analysis. The results showed that incubation of H2O2 for 48 h significantly decreased the cell viability, increased the cell apoptosis rate and the NISCH expression in PC12 cells, whereas NISCH downregulation blocked the effects of H2O2 on cells. In addition, the expression of Bcl-2 was significantly reduced, and the expression of Bax and caspase-3 were significantly increased by H2O2 treatment. However, these effects were partially inhibited by the downregulation of NISCH. Furthermore, H2O2 significantly weakened the transduction of Wnt signaling, including the increases of GSK-3β and TCF-1 expressions and the decrease of β-catenin expression, while NISCH downregulation attenuated the effect of H2O2 on Wnt signaling. Moreover, inhibition of the Wnt pathway further decreased the cell viability and promoted the cell apoptosis induced by H2O2 in PC12 cells. Our results suggest that NISCH downregulation may protect cells against oxidative stress-induced injury through regulating the transduction of Wnt signaling.
Collapse
|
12
|
Lingappan K, Savani RC. The Wnt Signaling Pathway and the Development of Bronchopulmonary Dysplasia. Am J Respir Crit Care Med 2020; 201:1174-1176. [PMID: 32101467 PMCID: PMC7233338 DOI: 10.1164/rccm.202002-0277ed] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Krithika Lingappan
- Division of Neonatal-Perinatal MedicineBaylor College of MedicineHouston, Texasand
| | - Rashmin C Savani
- Division of Neonatal-Perinatal MedicineUniversity of Texas Southwestern Medical CenterDallas, Texas
| |
Collapse
|
13
|
Jeffers A, Qin W, Owens S, Koenig KB, Komatsu S, Giles FJ, Schmitt DM, Idell S, Tucker TA. Glycogen Synthase Kinase-3β Inhibition with 9-ING-41 Attenuates the Progression of Pulmonary Fibrosis. Sci Rep 2019; 9:18925. [PMID: 31831767 PMCID: PMC6908609 DOI: 10.1038/s41598-019-55176-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/25/2019] [Indexed: 12/11/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease with a median survival of 3 years after diagnosis. Although the etiology of IPF is unknown, it is characterized by extensive alveolar epithelial cell apoptosis and proliferation of myofibroblasts in the lungs. While the origins of these myofibroblast appear to be diverse, fibroblast differentiation contributes to expansion of myofibroblasts and to disease progression. We found that agents that contribute to neomatrix formation and remodeling in pulmonary fibrosis (PF); TGF-β, Factor Xa, thrombin, plasmin and uPA all induced fibroblast/myofibroblast differentiation. These same mediators enhanced GSK-3β activation via phosphorylation of tyrosine-216 (p-Y216). Inhibition of GSK-3β signaling with the novel inhibitor 9-ING-41 blocked the induction of myofibroblast markers; α-SMA and Col-1 and reduced morphological changes of myofibroblast differentiation. In in vivo studies, the progression of TGF-β and bleomycin mediated PF was significantly attenuated by 9-ING-41 administered at 7 and 14 days respectively after the establishment of injury. Specifically, 9-ING-41 treatment significantly improved lung function (compliance and lung volumes; p < 0.05) of TGF-β adenovirus treated mice compared to controls. Similar results were found in mice with bleomycin-induced PF. These studies clearly show that activation of the GSK-3β signaling pathway is critical for the induction of myofibroblast differentiation in lung fibroblasts ex vivo and pulmonary fibrosis in vivo. The results offer a strong premise supporting the continued investigation of the GSK-3β signaling pathway in the control of fibroblast-myofibroblast differentiation and fibrosing lung injury. These data provide a strong rationale for extension of clinical trials of 9-ING-41 to patients with IPF.
Collapse
Affiliation(s)
- Ann Jeffers
- The Texas Lung Injury Institute, Tyler, TX, USA.,Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Wenyi Qin
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Shuzi Owens
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Kathleen B Koenig
- The Texas Lung Injury Institute, Tyler, TX, USA.,Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Satoshi Komatsu
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | | | | | - Steven Idell
- The Texas Lung Injury Institute, Tyler, TX, USA.,Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA
| | - Torry A Tucker
- The Texas Lung Injury Institute, Tyler, TX, USA. .,Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, USA.
| |
Collapse
|
14
|
Dapaah-Siakwan F, Zambrano R, Luo S, Duncan MR, Kerr N, Donda K, Vaccari JPDR, Keane RW, Dietrich WD, Benny M, Young K, Wu S. Caspase-1 Inhibition Attenuates Hyperoxia-induced Lung and Brain Injury in Neonatal Mice. Am J Respir Cell Mol Biol 2019; 61:341-354. [DOI: 10.1165/rcmb.2018-0192oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Fredrick Dapaah-Siakwan
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Ronald Zambrano
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Shihua Luo
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Matthew R. Duncan
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Nadine Kerr
- Miami Project to Cure Paralysis
- Department of Physiology and Biophysics, and
| | - Keyur Donda
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Juan Pablo de Rivero Vaccari
- Miami Project to Cure Paralysis
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Robert W. Keane
- Miami Project to Cure Paralysis
- Department of Physiology and Biophysics, and
| | - W. Dalton Dietrich
- Miami Project to Cure Paralysis
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Merline Benny
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Karen Young
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| | - Shu Wu
- Division of Neonatology and
- Batchelor Children’s Research Institute, Department of Pediatrics
| |
Collapse
|
15
|
Sun Y, Chen C, Di T, Yang J, Wang K, Zhu Y, Zhu R, Zhou A, Qian Y. Human β-Defensin-2 Improves Hyperoxia-Induced Lung Structural and Functional Injury in Neonatal Rats. Med Sci Monit 2019; 25:6074-6084. [PMID: 31411185 PMCID: PMC6705181 DOI: 10.12659/msm.915814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is a major complication of extreme prematurity, characterized by alveolar simplification and pulmonary malfunction. Hyperoxia-induced lung injury in neonatal rats has been used as a model of BPD, as indicated by lung architectural change and alveolar simplification that resembles clinical feature of BPD. β-defensin-2 (BD2) plays an important role in lung diseases by inhibiting inflammation response. However, little is known about its role in BPD. The aim of this study was to determine the effect of human BD2 (hBD2) gene on hyperoxia-induced animal model of BPD. Material/Methods The neonatal rats were exposed to 90% oxygen (O2) continuously for 14 days to mimic the BPD-like lung injury. These rats were then randomly assigned to the following four groups: in room air (air), in 90% O2, in 90% O2 with null adenovirus vector infection (O2+Ad), and in 90% O2 with gene therapy through adenovirus transfected hBD2 (O2+Ad-hBD2). Morphology of lungs, pulmonary function and expression of inflammatory cytokines on P7, P10, P14, and P21 were documented and compared across the 4 groups. Results The overexpression of hBD2 mediated by the adenovirus vector was successfully constructed. hBD2 gene therapy increased hBD2 mRNA expression, increased radial alveolar count (RAC), lung volume and compliance, decreased mean linear intercept (MLI), tissue damping, and elastance. Furthermore, pro-inflammatory cytokines IL-1β, IL-6, and TNF-α were inhibited and anti-inflammatory cytokines IL-10 was increased in the lungs of rats in O2+Ad-hBD2 group. Conclusions In hyperoxia-induced rat models of BPD, hBD2 promotes alveolarization and improves pulmonary function. The mechanism may contribute in alleviating inflammation response and inhibiting pro-inflammatory factors including IL-1β, IL-6, and TNF-α.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Cuie Chen
- Department of Pediatrics, Yiwu Maternity and Children Health Care Hospital, Jinhua, Zhejiang, China (mainland)
| | - Tianwei Di
- Department of Pediatrics, Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Jiaojiao Yang
- Department of Pediatrics, Cangnan People's Hospital, Wenzhou, Zhejiang, China (mainland)
| | - Kai Wang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Yanke Zhu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Ronghe Zhu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Aihua Zhou
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Yan Qian
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| |
Collapse
|
16
|
Li J, Shi J, Li P, Guo X, Wang T, Liu A. Genipin attenuates hyperoxia-induced lung injury and pulmonary hypertension via targeting glycogen synthase kinase-3 β in neonatal rats. Nutrition 2019; 57:237-244. [DOI: 10.1016/j.nut.2018.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/06/2018] [Accepted: 05/25/2018] [Indexed: 12/18/2022]
|
17
|
Donda K, Zambrano R, Moon Y, Percival J, Vaidya R, Dapaah-Siakwan F, Luo S, Duncan MR, Bao Y, Wang L, Qin L, Benny M, Young K, Wu S. Riociguat prevents hyperoxia-induced lung injury and pulmonary hypertension in neonatal rats without effects on long bone growth. PLoS One 2018; 13:e0199927. [PMID: 29990355 PMCID: PMC6038999 DOI: 10.1371/journal.pone.0199927] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains the most common and serious chronic lung disease of premature infants. Severe BPD complicated with pulmonary hypertension (PH) increases the mortality of these infants. Riociguat is an allosteric soluble guanylate cyclase stimulator and is approved by the FDA for treating PH in adults. However, it has not been approved for use in neonates due to concern for adverse effects on long bone growth. To address this concern we investigated if administration of riociguat is beneficial in preventing hyperoxia-induced lung injury and PH without side effects on long bone growth in newborn rats. Newborn rats were randomized to normoxia (21% O2) or hyperoxia (85% O2) exposure groups within 24 hours of birth, and received riociguat or placebo by once daily intraperitoneal injections during continuous normoxia or hyperoxia exposure for 9 days. In the hyperoxia control group, radial alveolar count, mean linear intercept and vascular density were significantly decreased, the pathological hallmarks of BPD, and these were accompanied by an increased inflammatory response. There was also significantly elevated vascular muscularization of peripheral pulmonary vessels, right ventricular systolic pressure and right ventricular hypertrophy indicating PH. However, administration of riociguat significantly decreased lung inflammation, improved alveolar and vascular development, and decreased PH during hyperoxia by inducing cGMP production. Additionally, riociguat did not affect long bone growth or structure. These data indicate that riociguat is beneficial in preventing hyperoxia-induced lung injury and PH without affecting long bone growth and structure and hence, suggests riociguat may be a potential novel agent for preventing BPD and PH in neonates.
Collapse
Affiliation(s)
- Keyur Donda
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Ronald Zambrano
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Younghye Moon
- Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Justin Percival
- Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Ruben Vaidya
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Fredrick Dapaah-Siakwan
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Shihua Luo
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Matthew R. Duncan
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Yong Bao
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Luqing Wang
- Department of Orthopedic Surgery, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ling Qin
- Department of Orthopedic Surgery, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Merline Benny
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Karen Young
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Shu Wu
- Pediatrics and Batchelor Children’s Research Institute, University of Miami School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
18
|
Hussain M, Xu C, Lu M, Wu X, Tang L, Wu X. Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3226-3242. [PMID: 28866134 DOI: 10.1016/j.bbadis.2017.08.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/29/2017] [Indexed: 12/23/2022]
Abstract
Embryonic lung development requires reciprocal endodermal-mesodermal interactions; mediated by various signaling proteins. Wnt/β-catenin is a signaling protein that exhibits the pivotal role in lung development, injury and repair while aberrant expression of Wnt/β-catenin signaling leads to asthmatic airway remodeling: characterized by hyperplasia and hypertrophy of airway smooth muscle cells, alveolar and vascular damage goblet cells metaplasia, and deposition of extracellular matrix; resulting in decreased lung compliance and increased airway resistance. The substantial evidence suggests that Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Here, we summarized the recent advances related to the mechanistic role of Wnt/β-catenin signaling in lung development, consequences of aberrant expression or deletion of Wnt/β-catenin signaling in expansion and progression of asthmatic airway remodeling, and linking early-impaired pulmonary development and airway remodeling later in life. Finally, we emphasized all possible recent potential therapeutic significance and future prospectives, that are adaptable for therapeutic intervention to treat asthmatic airway remodeling.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| | - Chengyun Xu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China.
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| |
Collapse
|
19
|
Vnukov VV, Gutsenko OI, Milyutina NP, Kornienko IV, Ananyan AA, Plotnikov AA, Panina SB. SkQ1 regulates expression of Nrf2, ARE-controlled genes encoding antioxidant enzymes, and their activity in cerebral cortex under oxidative stress. BIOCHEMISTRY (MOSCOW) 2017; 82:942-952. [DOI: 10.1134/s0006297917080090] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Tantray MA, Khan I, Hamid H, Alam MS, Dhulap A, Ganai AA. Oxazolo[4,5-b]pyridine-Based Piperazinamides as GSK-3β Inhibitors with Potential for Attenuating Inflammation and Suppression of Pro-Inflammatory Mediators. Arch Pharm (Weinheim) 2017; 350. [PMID: 28543747 DOI: 10.1002/ardp.201700022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/28/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022]
Abstract
Recent studies reveal that glycogen synthase kinase-3β (GSK-3β) acts as a pro-inflammatory enzyme, and by inhibiting this kinase, inflammation can be controlled. In this regard, a series of 17 piperazine-linked oxazolo[4,5-b]pyridine-based derivatives was synthesized and evaluated for in vitro GSK-3β inhibitory and in vivo anti-inflammatory activity. The compounds 7d, 7e, 7g, and 7c displayed the best GSK-3β inhibitory activity among all the synthesized compounds, with corresponding IC50 values of 0.34, 0.39, 0.47, and 0.53 µM. Among the compounds 7d, 7e, 7g, and 7c examined for in vivo anti-inflammatory activity in the rat paw edema model, compound 7d exhibited maximum inhibition, reducing the paw volume by 62.79 and 65.91% at 3 and 5 h post-carrageenan administration, respectively, in comparison to indomethacin (76.74% at 3 h and 79.54% at 5 h after carrageenan administration). Furthermore, these compounds (7d, 7e, 7g, and 7c) were also found to substantially inhibit pro-inflammatory mediators, i.e., TNF-α, IL-1β, and IL-6, ex vivo in comparison to indomethacin and did not pose any gastric ulceration risk, indicating the potential of this oxazolopyridine scaffold for the development of GSK-3β inhibitors and their application as anti-inflammatory agents.
Collapse
Affiliation(s)
- Mushtaq A Tantray
- Faculty of Science, Department of Chemistry, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Imran Khan
- Faculty of Science, Department of Chemistry, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Hinna Hamid
- Faculty of Science, Department of Chemistry, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Mohammad Sarwar Alam
- Faculty of Science, Department of Chemistry, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Abhijeet Dhulap
- CSIR - Unit for Research and Development of Information Products (URDIP), Pune, India
| | - Ajaz Ahmad Ganai
- Molecular Virology Lab., Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
21
|
Hoffman O, Burns N, Vadász I, Eltzschig HK, Edwards MG, Vohwinkel CU. Detrimental ELAVL-1/HuR-dependent GSK3β mRNA stabilization impairs resolution in acute respiratory distress syndrome. PLoS One 2017; 12:e0172116. [PMID: 28196122 PMCID: PMC5308835 DOI: 10.1371/journal.pone.0172116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/31/2017] [Indexed: 12/20/2022] Open
Abstract
A hallmark of acute respiratory distress syndrome (ARDS) is accumulation of protein-rich edema in the distal airspaces and its removal is critical for patient survival. Previous studies have shown a detrimental role of Glycogen Synthase Kinase (GSK) 3β during ARDS via inhibition of alveolar epithelial protein transport. We hypothesized that post-transcriptional regulation of GSK3β could play a functional role in ARDS resolution. To address this hypothesis, we performed an in silico analysis to identify regulatory genes whose expression correlation to GSK3β messenger RNA utilizing two lung cancer cell line array datasets. Among potential regulatory partners of GSK3β, these studies identified the RNA-binding protein ELAVL-1/HuR (Embryonic Lethal, Abnormal Vision, Drosophila-Like) as a central component in a likely GSK3β signaling network. ELAVL-1/HuR is a RNA-binding protein that selectively binds to AU-rich elements of mRNA and enhances its stability thereby increasing target gene expression. Subsequent studies with siRNA suppression of ELAVL-1/HuR demonstrated deceased GSK3β mRNA and protein expression and improved clearance of FITC-albumin in A549 cells. Conversely, stabilization of ELAVL-1/HuR with the proteasome inhibitor MG-132 resulted in induction of GSK3β at mRNA and protein level and attenuated FITC-albumin clearance. Utilizing ventilator-induced lung injury or intra-tracheal installation of hydrochloric acid to induce ARDS in mice, we observed increased mRNA and protein expression of ELAVL-1/HuR and GSK3β. Together, our findings indicate a previously unknown interaction between GSK3β and ELAV-1 during ARDS, and suggest the inhibition of the ELAV-1- GSK3β pathways as a novel ARDS treatment approach.
Collapse
Affiliation(s)
- Olivia Hoffman
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
| | - Nana Burns
- Developmental Lung Biology, Cardio Vascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatric Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Giessen, Germany
| | - Holger K. Eltzschig
- Organ Protection Program, Department of Anesthesiology, University of Colorado, School of Medicine, Aurora, Colorado, United States of America
| | - Michael G. Edwards
- Developmental Lung Biology, Cardio Vascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatric Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Christine U. Vohwinkel
- Department of Pediatrics, University of Colorado at Denver, Aurora, Colorado, United States of America
- Developmental Lung Biology, Cardio Vascular Pulmonary Research Laboratories, Division of Pulmonary Sciences and Critical Care Medicine, Division of Pediatric Critical Care, Departments of Medicine and Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
22
|
Hummler JK, Dapaah-Siakwan F, Vaidya R, Zambrano R, Luo S, Chen S, Kerr N, de Rivero Vaccari JP, Keane RW, Dietrich WD, Bancalari E, Young KC, Wu S. Inhibition of Rac1 Signaling Downregulates Inflammasome Activation and Attenuates Lung Injury in Neonatal Rats Exposed to Hyperoxia. Neonatology 2017; 111:280-288. [PMID: 28013306 DOI: 10.1159/000450918] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 09/20/2016] [Indexed: 01/28/2023]
Abstract
BACKGROUND Inflammatory injury, particularly the production of active interleukin (IL)-1β plays a major role in the pathogenesis of bronchopulmonary dysplasia (BPD) in preterm infants. The release of active IL-1β is controlled by posttranscriptional modifications of its proform (pro-IL-1β) through the inflammasome. Rac1 is a member of the Rho family of GTPases that regulate the inflammatory process. OBJECTIVE This study tested the hypothesis that Rac1 signaling increases inflammasome activation that results in damaging inflammation, and that the inhibition of Rac1 signaling prevents lung injury, by inhibiting inflammasome activation in a newborn rat model of BPD induced by hyperoxia. METHODS Newborn rat pups were exposed to room air or hyperoxia (85% O2) and received daily intraperitoneal injections of placebo (normal saline) or NSC23766, a specific Rac1 inhibitor, for 10 days. The effects on lung inflammation, alveolarization, vascular development, vascular remodeling, right ventricular systolic pressure, and right ventricular hypertrophy (RVH) were then assessed. RESULTS Hyperoxia exposure upregulated Rac1 and increased the production of active IL-1β, which was accompanied by increasing expression of the inflammasome. In addition, hyperoxia induced the pathological hallmarks of BPD. However, treatment with NSC23766 significantly decreased inflammasome activation and macrophage infiltration, improved alveolar and vascular development, and reduced pulmonary vascular remodeling and RVH. CONCLUSION These results indicate that Rac1 signaling regulates the expression of the inflammasome and plays a pivotal role in the pathogenesis of hyperoxia-induced neonatal lung injury. Therefore, targeting Rac1 signaling may provide a novel strategy to prevent and treat BPD in preterm infants.
Collapse
Affiliation(s)
- Julia K Hummler
- Division of Neonatology, Department of Pediatrics, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Nardiello C, Mižíková I, Morty RE. Looking ahead: where to next for animal models of bronchopulmonary dysplasia? Cell Tissue Res 2016; 367:457-468. [PMID: 27917436 PMCID: PMC5320021 DOI: 10.1007/s00441-016-2534-3] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/01/2016] [Indexed: 11/16/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of preterm birth, with appreciable morbidity and mortality in a neonatal intensive care setting. Much interest has been shown in the identification of pathogenic pathways that are amenable to pharmacological manipulation (1) to facilitate the development of novel therapeutic and medical management strategies and (2) to identify the basic mechanisms of late lung development, which remains poorly understood. A number of animal models have therefore been developed and continue to be refined with the aim of recapitulating pathological pulmonary hallmarks noted in lungs from neonates with BPD. These animal models rely on several injurious stimuli, such as mechanical ventilation or oxygen toxicity and infection and sterile inflammation, as applied in mice, rats, rabbits, pigs, lambs and nonhuman primates. This review addresses recent developments in modeling BPD in experimental animals and highlights important neglected areas that demand attention. Additionally, recent progress in the quantitative microscopic analysis of pathology tissue is described, together with new in vitro approaches of value for the study of normal and aberrant alveolarization. The need to examine long-term sequelae of damage to the developing neonatal lung is also considered, as is the need to move beyond the study of the lungs alone in experimental animal models of BPD.
Collapse
Affiliation(s)
- Claudio Nardiello
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Ivana Mižíková
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Parkstrasse 1, 61231, Bad Nauheim, Germany. .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
| |
Collapse
|
24
|
Ota C, Baarsma HA, Wagner DE, Hilgendorff A, Königshoff M. Linking bronchopulmonary dysplasia to adult chronic lung diseases: role of WNT signaling. Mol Cell Pediatr 2016; 3:34. [PMID: 27718180 PMCID: PMC5055515 DOI: 10.1186/s40348-016-0062-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most common chronic lung diseases in infants caused by pre- and/or postnatal lung injury. BPD is characterized by arrested alveolarization and vascularization due to extracellular matrix remodeling, inflammation, and impaired growth factor signaling. WNT signaling is a critical pathway for normal lung development, and its altered signaling has been shown to be involved in the onset and progression of incurable chronic lung diseases in adulthood, such as chronic obstructive pulmonary disease (COPD) or idiopathic pulmonary fibrosis (IPF). In this review, we summarize the impact of WNT signaling on different stages of lung development and its potential contribution to developmental lung diseases, especially BPD, and chronic lung diseases in adulthood.
Collapse
Affiliation(s)
- Chiharu Ota
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.
| | - Hoeke A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.,The Perinatal Center, Campus Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
25
|
Xu W, Zhao Y, Zhang B, Xu B, Yang Y, Wang Y, Liu C. Resveratrol attenuates hyperoxia-induced oxidative stress, inflammation and fibrosis and suppresses Wnt/β-catenin signalling in lungs of neonatal rats. Clin Exp Pharmacol Physiol 2016; 42:1075-83. [PMID: 26174235 DOI: 10.1111/1440-1681.12459] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/28/2015] [Accepted: 07/04/2015] [Indexed: 11/27/2022]
Abstract
Although survival rate of infants born prematurely has been raised by supplemental oxygen treatment, it is followed by high morbidity of hyperoxia-induced bronchopulmonary dysplasia. In this study, the effect of resveratrol on the lung injury was evaluated in hyperoxia-exposed rats of preterm birth. The results demonstrated that hyperoxia led to thickened alveolar wall, simplified alveolar architecture and fibrosis. In addition, elevated methane dicarboxylic aldehyde level, decreased glutathione level and superoxide dismutase activity were also found in hyperoxic lungs, as well as the increased tumor necrosis factor-α, interleukin-1β and interleukin-6 in the bronchoalveolar lavage fluid. Fibrotic-associated proteins transforming growth factor-β1, α-smooth muscle actin, collagen I and fibronectin deposition were also found in interstitial substance of lungs. Furthermore, Wnt/β-catenin signalling was found to be active in hyperoxia-induced lungs. In addition, expression of SP-C was increased and T1α was decreased in hyperoxia-exposed lungs. Resveratrol intraperitoneal administration alleviated hyperoxia-induced histological injury of lungs, regulated redox balance, decreased pro-inflammatory cytokine release, and down-regulated expression of fibrotic-associated proteins. Furthermore, Wnt/β-catenin signalling was also suppressed by resveratrol, as represented by diminished expression of lymphoid enhancer factor-1, Wnt induced signalling protein-1 and cyclin D1. In addition, the increase of SP-C and decrease of T1α expression was prevented as well. The present study showed that resveratrol could protect lungs from hyperoxia-induced injury through its antioxidant, anti-inflammatory and anti-fibrotic effects. The transdifferentiation of alveolar epithelial type II cells to alveolar epithelial type I cells promotion and Wnt/β-catenin signalling suppression are also involved in the protective effect.
Collapse
Affiliation(s)
- Wei Xu
- Department of Paediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Zhao
- Department of Paediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Binglun Zhang
- Department of Paediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bo Xu
- Department of Ophthalmology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yang Yang
- Department of Paediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yujing Wang
- Department of Paediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chunfeng Liu
- Department of Paediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Jagarapu J, Kelchtermans J, Rong M, Chen S, Hehre D, Hummler S, Faridi MH, Gupta V, Wu S. Efficacy of Leukadherin-1 in the Prevention of Hyperoxia-Induced Lung Injury in Neonatal Rats. Am J Respir Cell Mol Biol 2016; 53:793-801. [PMID: 25909334 DOI: 10.1165/rcmb.2014-0422oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lung inflammation plays a key role in the pathogenesis of bronchopulmonary dysplasia (BPD), a chronic lung disease of premature infants. The challenge in BPD management is the lack of effective and safe antiinflammatory agents. Leukadherin-1 (LA1) is a novel agonist of the leukocyte surface integrin CD11b/CD18 that enhances leukocyte adhesion to ligands and vascular endothelium and thus reduces leukocyte transendothelial migration and influx to the injury sites. Its functional significance in preventing hyperoxia-induced neonatal lung injury is unknown. We tested the hypothesis that administration of LA1 is beneficial in preventing hyperoxia-induced neonatal lung injury, an experimental model of BPD. Newborn rats were exposed to normoxia (21% O2) or hyperoxia (85% O2) and received twice-daily intraperitoneal injection of LA1 or placebo for 14 days. Hyperoxia exposure in the presence of the placebo resulted in a drastic increase in the influx of neutrophils and macrophages into the alveolar airspaces. This increased leukocyte influx was accompanied by decreased alveolarization and angiogenesis and increased pulmonary vascular remodeling and pulmonary hypertension (PH), the pathological hallmarks of BPD. However, administration of LA1 decreased macrophage infiltration in the lungs during hyperoxia. Furthermore, treatment with LA1 improved alveolarization and angiogenesis and decreased pulmonary vascular remodeling and PH. These data indicate that leukocyte recruitment plays an important role in the experimental model of BPD induced by hyperoxia. Targeting leukocyte trafficking using LA1, an integrin agonist, is beneficial in preventing lung inflammation and protecting alveolar and vascular structures during hyperoxia. Thus, targeting integrin-mediated leukocyte recruitment and inflammation may provide a novel strategy in preventing and treating BPD in preterm infants.
Collapse
Affiliation(s)
- Jawahar Jagarapu
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Jelte Kelchtermans
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Min Rong
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Shaoyi Chen
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Dorothy Hehre
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Stefanie Hummler
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| | - Mohd Hafeez Faridi
- 2 Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Vineet Gupta
- 2 Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois
| | - Shu Wu
- 1 Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida; and
| |
Collapse
|
27
|
Li L, McBride DW, Doycheva D, Dixon BJ, Krafft PR, Zhang JH, Tang J. G-CSF attenuates neuroinflammation and stabilizes the blood-brain barrier via the PI3K/Akt/GSK-3β signaling pathway following neonatal hypoxia-ischemia in rats. Exp Neurol 2015; 272:135-44. [PMID: 25585014 PMCID: PMC4499024 DOI: 10.1016/j.expneurol.2014.12.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/22/2014] [Accepted: 12/25/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Neonatal hypoxia occurs in approximately 60% of premature births and is associated with a multitude of neurological disorders. While various treatments have been developed, translating them from bench to bedside has been limited. We previously showed G-CSF administration was neuroprotective in a neonatal hypoxia-ischemia rat pup model, leading us to hypothesize that G-CSF inactivation of GSK-3β via the PI3K/Akt pathway may attenuate neuroinflammation and stabilize the blood-brain barrier (BBB). METHODS P10 Sprague-Dawley rat pups were subjected to unilateral carotid artery ligation followed by hypoxia for 2.5h. We assessed inflammation by measuring expression levels of IKKβ, NF-κB, TNF-α, IL-1β, IL-10, and IL-12 as well as neutrophil infiltration. BBB stabilization was evaluated by measuring Evans blue extravasation, and Western blot analysis of Claudin-3, Claudin-5, ICAM-1, and VCAM-1. MEASUREMENTS AND MAIN RESULTS First, the time course study showed that p-β-catenin/β-catenin, IKKβ, and NF-κB expression levels peaked at 48h post-HI. The knockdown of GSK-3β with siRNA prevented the HI-induced increase of p-β-catenin/β-catenin, IKKβ, and NF-κB expression levels 48h after HI. G-CSF treatment reduced brain water content and neuroinflammation by downregulating IKKβ, NF-κB, TNF-α, IL-1β, and IL-12 and upregulating IL-10, thereby reducing neutrophil infiltration. Additionally, G-CSF stabilizes the BBB by downregulating VCAM-1 and ICAM-1, as well as upregulating Claudins 3 and 5 in endothelial cells. G-CSFR knockdown by siRNA and Akt inhibition by Wortmannin reversed G-CSF's neuroprotective effects. CONCLUSIONS We demonstrate G-CSF plays a pivotal role in attenuating neuroinflammation and BBB disruption following HI by inactivating GSK-3β through the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Li Li
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Devin W McBride
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Desislava Doycheva
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Brandon J Dixon
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Paul R Krafft
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA; Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Departments of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
28
|
Mühlfeld C, Hegermann J, Wrede C, Ochs M. A review of recent developments and applications of morphometry/stereology in lung research. Am J Physiol Lung Cell Mol Physiol 2015; 309:L526-36. [DOI: 10.1152/ajplung.00047.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/09/2015] [Indexed: 11/22/2022] Open
Abstract
Design-based stereology is the gold standard of morphometry in lung research. Here, we analyze the current use of morphometric and stereological methods in lung research and provide an overview on recent methodological developments and biological observations made by the use of stereology. Based on this analysis we hope to provide useful recommendations for a good stereological practice to further the use of advanced and unbiased stereological methods.
Collapse
Affiliation(s)
- Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany; and
- Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany; and
- Cluster of Excellence REBIRTH (From Regenerative Biology to Reconstructive Therapy), Hannover, Germany
| |
Collapse
|
29
|
Silva DMG, Nardiello C, Pozarska A, Morty RE. Recent advances in the mechanisms of lung alveolarization and the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2015; 309:L1239-72. [PMID: 26361876 DOI: 10.1152/ajplung.00268.2015] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 09/09/2015] [Indexed: 02/08/2023] Open
Abstract
Alveolarization is the process by which the alveoli, the principal gas exchange units of the lung, are formed. Along with the maturation of the pulmonary vasculature, alveolarization is the objective of late lung development. The terminal airspaces that were formed during early lung development are divided by the process of secondary septation, progressively generating an increasing number of alveoli that are of smaller size, which substantially increases the surface area over which gas exchange can take place. Disturbances to alveolarization occur in bronchopulmonary dysplasia (BPD), which can be complicated by perturbations to the pulmonary vasculature that are associated with the development of pulmonary hypertension. Disturbances to lung development may also occur in persistent pulmonary hypertension of the newborn in term newborn infants, as well as in patients with congenital diaphragmatic hernia. These disturbances can lead to the formation of lungs with fewer and larger alveoli and a dysmorphic pulmonary vasculature. Consequently, affected lungs exhibit a reduced capacity for gas exchange, with important implications for morbidity and mortality in the immediate postnatal period and respiratory health consequences that may persist into adulthood. It is the objective of this Perspectives article to update the reader about recent developments in our understanding of the molecular mechanisms of alveolarization and the pathogenesis of BPD.
Collapse
Affiliation(s)
- Diogo M G Silva
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Claudio Nardiello
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Agnieszka Pozarska
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rory E Morty
- Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany; Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
30
|
Xu Y, Zhang J, Jiang W, Zhang S. Astaxanthin induces angiogenesis through Wnt/β-catenin signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2015; 22:744-751. [PMID: 26141761 DOI: 10.1016/j.phymed.2015.05.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/21/2015] [Accepted: 05/24/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE In the present study, we sought to elucidate whether astaxanthin contributes to induce angiogenesis and its mechanisms. MATERIALS AND METHODS To this end, we examined the role of astaxanthin on human brain microvascular endothelial cell line (HBMEC) and rat aortic smooth muscle cell (RASMC) proliferation, invasion and tube formation in vitro. For study of mechanism, the Wnt/β-catenin signaling pathway inhibitor IWR-1-endo was used. HMBECs and RASMCs proliferation were tested by cell counting. Scratch adhesion test was used to assess the ability of invasion. A matrigel tube formation assay was performed to test capillary tube formation ability. The Wnt/β-catenin pathway activation in HMBECs and RASMCs were tested by Western blot. RESULTS Our data suggested that astaxanthin induces angiogenesis by increasing proliferation, invasion and tube formation in vitro. Wnt and β-catenin expression were increased by astaxanthin and counteracted by IWR-1-endo in HMBECs and RASMCs. Tube formation was increased by astaxanthin and counteracted by IWR-1-endo. CONCLUSIONS It may be suggested that astaxanthin induces angiogenesis in vitro via a programmed Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Yangyang Xu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China
| | - Jie Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China
| | - Wanglin Jiang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China.
| | - Shuping Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, PR. China.
| |
Collapse
|
31
|
Drummond S, Ramachandran S, Torres E, Huang J, Hehre D, Suguihara C, Young KC. CXCR4 blockade attenuates hyperoxia-induced lung injury in neonatal rats. Neonatology 2015; 107:304-11. [PMID: 25825119 PMCID: PMC4458217 DOI: 10.1159/000371835] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 01/02/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Lung inflammation is a key factor in the pathogenesis of bronchopulmonary dysplasia (BPD). Stromal-derived factor-1 (SDF-1) and its receptor chemokine receptor 4 (CXCR4) modulate the inflammatory response. It is not known if antagonism of CXCR4 alleviates lung inflammation in neonatal hyperoxia-induced lung injury. OBJECTIVE We aimed to determine whether CXCR4 antagonism would attenuate lung injury in rodents with experimental BPD by decreasing pulmonary inflammation. METHODS Newborn rats exposed to normoxia (room air, RA) or hyperoxia (FiO2 = 0.9) from postnatal day 2 (P2) to P16 were randomized to receive the CXCR4 antagonist, AMD3100 or placebo (PL) from P5 to P15. Lung alveolarization, angiogenesis and inflammation were evaluated at P16. RESULTS Compared to the RA pups, hyperoxic PL pups had a decrease in alveolarization, reduced lung vascular density and increased lung inflammation. In contrast, AMD3100-treated hyperoxic pups had improved alveolarization and increased angiogenesis. This improvement in lung structure was accompanied by a decrease in the macrophage and neutrophil counts in the bronchoalveolar lavage fluid and reduced lung myeloperoxidase activity. CONCLUSION CXCR4 antagonism decreases lung inflammation and improves alveolar and vascular structure in neonatal rats with experimental BPD. These findings suggest a novel therapeutic strategy to alleviate lung injury in preterm infants with BPD.
Collapse
Affiliation(s)
- Shelley Drummond
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Fla., USA
| | | | | | | | | | | | | |
Collapse
|
32
|
O'Reilly M, Thébaud B. Animal models of bronchopulmonary dysplasia. The term rat models. Am J Physiol Lung Cell Mol Physiol 2014; 307:L948-58. [PMID: 25305248 DOI: 10.1152/ajplung.00160.2014] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the chronic lung disease of prematurity that affects very preterm infants. Although advances in perinatal care have enabled the survival of infants born as early as 23-24 wk of gestation, the challenge of promoting lung growth while protecting the ever more immature lung from injury is now bigger. Consequently, BPD remains one of the most common complications of extreme prematurity and still lacks specific treatments. Progress in our understanding of BPD and the potential of developing therapeutic strategies have arisen from large (baboons, sheep, and pigs) and small (rabbits, rats, and mice) animal models. This review focuses specifically on the use of the rat to model BPD and summarizes how the model is used in various research studies and the advantages and limitations of this particular model, and it highlights recent therapeutic advances in BPD by using this rat model.
Collapse
Affiliation(s)
- Megan O'Reilly
- Department of Pediatrics and Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada; and
| | - Bernard Thébaud
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research, Regenerative Medicine Program and Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Alapati D, Rong M, Chen S, Hehre D, Hummler SC, Wu S. Inhibition of β-catenin signaling improves alveolarization and reduces pulmonary hypertension in experimental bronchopulmonary dysplasia. Am J Respir Cell Mol Biol 2014; 51:104-13. [PMID: 24484510 DOI: 10.1165/rcmb.2013-0346oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common and serious chronic lung disease of preterm infants. The development of pulmonary hypertension (PH) significantly increases the mortality and morbidity of this disease. β-Catenin signaling plays an important role in tissue development and remodeling. Aberrant β-catenin signaling is associated with clinical and experiment models of BPD. To test the hypothesis that inhibition of β-catenin signaling is beneficial in promoting alveolar and vascular development and preventing PH in experimental BPD, we examined the effects of ICG001, a newly developed pharmacological inhibitor of β-catenin, in preventing hyperoxia-induced BPD in neonatal rats. Newborn rat pups were randomized at postnatal day (P)2 to room air (RA) + DMSO (placebo), RA + ICG001, 90% FiO2 (O2) + DMSO, or O2 + ICG001. ICG001 (10 mg/kg) or DMSO was given by daily intraperitoneal injection for 14 days during continuous exposure to RA or hyperoxia. Primary human pulmonary arterial smooth muscle cells (PASMCs) were cultured in RA or hyperoxia (95% O2) in the presence of DMSO or ICG001 for 24 to 72 hours. Treatment with ICG001 significantly increased alveolarization and reduced pulmonary vascular remodeling and PH during hyperoxia. Furthermore, administering ICG001 decreased PASMC proliferation and expression of extracellular matrix remodeling molecules in vitro under hyperoxia. Finally, these structural, cellular, and molecular effects of ICG001 were associated with down-regulation of multiple β-catenin target genes. These data indicate that β-catenin signaling mediates hyperoxia-induced alveolar impairment and PH in neonatal animals. Targeting β-catenin may provide a novel strategy to alleviate BPD in preterm infants.
Collapse
Affiliation(s)
- Deepthi Alapati
- Department of Pediatrics, Division of Neonatology, Batchelor Children's Research Institute, University of Miami Miller School of Medicine, Miami, Florida
| | | | | | | | | | | |
Collapse
|
34
|
McKenna S, Michaelis KA, Agboke F, Liu T, Han K, Yang G, Dennery PA, Wright CJ. Sustained hyperoxia-induced NF-κB activation improves survival and preserves lung development in neonatal mice. Am J Physiol Lung Cell Mol Physiol 2014; 306:L1078-89. [PMID: 24748603 DOI: 10.1152/ajplung.00001.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Oxygen toxicity contributes to the pathogenesis of bronchopulmonary dysplasia (BPD). Neonatal mice exposed to hyperoxia develop a simplified lung structure that resembles BPD. Sustained activation of the transcription factor NF-κB and increased expression of protective target genes attenuate hyperoxia-induced mortality in adults. However, the effect of enhancing hyperoxia-induced NF-κB activity on lung injury and development in neonatal animals is unknown. We performed this study to determine whether sustained NF-κB activation, mediated through IκBβ overexpression, preserves lung development in neonatal animals exposed to hyperoxia. Newborn wild-type (WT) and IκBβ-overexpressing (AKBI) mice were exposed to hyperoxia (>95%) or room air from day of life (DOL) 0-14, after which all animals were kept in room air. Survival curves were generated through DOL 14. Lung development was assessed using radial alveolar count (RAC) and mean linear intercept (MLI) at DOL 3 and 28 and pulmonary vessel density at DOL 28. Lung tissue was collected, and NF-κB activity was assessed using Western blot for IκB degradation and NF-κB nuclear translocation. WT mice demonstrated 80% mortality through 14 days of exposure. In contrast, AKBI mice demonstrated 60% survival. Decreased RAC, increased MLI, and pulmonary vessel density caused by hyperoxia in WT mice were significantly attenuated in AKBI mice. These findings were associated with early and sustained NF-κB activation and expression of cytoprotective target genes, including vascular endothelial growth factor receptor 2. We conclude that sustained hyperoxia-induced NF-κB activation improves neonatal survival and preserves lung development. Potentiating early NF-κB activity after hyperoxic exposure may represent a therapeutic intervention to prevent BPD.
Collapse
Affiliation(s)
- Sarah McKenna
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Katherine A Michaelis
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Fadeke Agboke
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Thanh Liu
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Kristie Han
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Guang Yang
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Phyllis A Dennery
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado;
| |
Collapse
|
35
|
Bhandari V. Postnatal inflammation in the pathogenesis of bronchopulmonary dysplasia. ACTA ACUST UNITED AC 2014; 100:189-201. [PMID: 24578018 DOI: 10.1002/bdra.23220] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/02/2014] [Accepted: 01/05/2014] [Indexed: 12/18/2022]
Abstract
Exposure to hyperoxia, invasive mechanical ventilation, and systemic/local sepsis are important antecedents of postnatal inflammation in the pathogenesis of bronchopulmonary dysplasia (BPD). This review will summarize information obtained from animal (baboon, lamb/sheep, rat and mouse) models that pertain to the specific inflammatory agents and signaling molecules that predispose a premature infant to BPD.
Collapse
Affiliation(s)
- Vineet Bhandari
- Division of Perinatal Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
36
|
Jain D, Bancalari E. Bronchopulmonary dysplasia: clinical perspective. ACTA ACUST UNITED AC 2014; 100:134-44. [PMID: 24578124 DOI: 10.1002/bdra.23229] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/17/2014] [Accepted: 01/23/2014] [Indexed: 11/11/2022]
Abstract
Since Northway's original description of BPD almost 45 years ago, the clinical presentation of BPD has evolved into a disease process, which mostly involves extremely premature infants. This new form of BPD is the result of multiple antenatal and postnatal factors that can cause injury to the developing lung leading to altered alveolar and vascular development. Over the years, there has been considerable increase in knowledge of factors that contribute to the development of BPD. This has led to different strategies for prevention as well as management of BPD. Some of these strategies have been successful and have withstood the test of clinical trials, such as vitamin A supplementation, post-natal steroids, caffeine, and volume targeted ventilation. The evidence for other interventions has been weak or negative. With better understanding of the complex and multifactorial pathogenesis of BPD, it is quite clear that any single therapy is very unlikely to eliminate this problem unless it reduces prematurity. Further development in prevention and treatment of BPD will likely need a multi-pronged strategy with novel therapeutic agents acting at various stages of the disease process.
Collapse
|
37
|
Altered canonical Wingless-Int signaling in the ovine fetal lung after exposure to intra-amniotic lipopolysaccharide and antenatal betamethasone. Pediatr Res 2014; 75:281-7. [PMID: 24232635 DOI: 10.1038/pr.2013.226] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/16/2013] [Indexed: 01/18/2023]
Abstract
BACKGROUND Antenatal inflammation and maternal corticosteroids induce fetal lung maturation but interfere with late lung development. Canonical Wingless-Int (Wnt) signaling directs lung development and repair. We showed that intra-amniotic (IA) lipopolysaccharide (LPS) exposure disrupted developmental signaling pathways in the preterm lamb lungs. Therefore, we hypothesized that pulmonary Wnt signaling was altered by exposure to IA LPS and/or antenatal corticosteroids. METHODS Ovine fetuses were exposed to IA LPS, maternal intramuscular betamethasone, a control saline injection, or a combination thereof at 107 and/or 114 d gestational age (term = 150 d gestational age) before delivery at 121 d gestational age. RESULTS IA LPS exposure decreased the lung expression of lymphoid enhancer-binding factor 1 (LEF1), a major Wnt pathway effector. WNT1, WNT4, and downstream messenger β-catenin decreased after LPS exposure. WNT7b mRNA increased fourfold 14 d post-LPS exposure. Betamethasone treatment 7 d before LPS exposure prevented the reduction in LEF1 expression, whereas betamethasone administration after LPS normalized the LPS-induced increase in Wnt7b mRNA. CONCLUSION IA LPS exposure decreased canonical Wnt signaling in the developing lung. Antenatal corticosteroids before or after IA inflammation had different effects on pulmonary Wnt signaling. This study provides new insights into possible mechanisms by which prenatal inflammation affects lung development and how corticosteroid can be beneficial in this setting.
Collapse
|
38
|
Richter J, Toelen J, Vanoirbeek J, Kakigano A, Dekoninck P, Verbeken E, Deprest J. Functional assessment of hyperoxia-induced lung injury after preterm birth in the rabbit. Am J Physiol Lung Cell Mol Physiol 2013; 306:L277-83. [PMID: 24375793 DOI: 10.1152/ajplung.00315.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The objective of this study was to document early neonatal (7 days) pulmonary outcome in the rabbit model for preterm birth and hyperoxia-induced lung injury. Preterm pups were delivered at 28 days (term = 31 days; early saccular phase of lung development) by cesarean section, housed in an incubator, and gavage fed for 7 days. Pups were divided into the following groups: 1) normoxia (21% O2; normoxia group) and 2) and hyperoxia (>95% O2; hyperoxia group). Controls were pups born at term who were housed in normoxic conditions (control group). Outcome measures were survival, pulmonary function tests using the whole body plethysmograph and forced oscillation technique, and lung morphometry. There was a significant difference in survival of preterm pups whether they were exposed to normoxia (83.3%) or hyperoxia (55.9%). Hyperoxic exposure was associated with increased tissue damping and elasticity and decreased static compliance compared with normoxic controls (P < 0.01). Morphometry revealed an increased linear intercept and increased mean wall transection length, which translates to larger alveoli with septal thickening in hyperoxia compared with normoxia (P < 0.01). In conclusion, the current experimental hyperoxic conditions to which preterm pups are exposed induce the typical clinical features of bronchopulmonary dysplasia. This model will be used to study novel preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Jute Richter
- Clinical Dept. of Obstetrics and Gynaecology and Academic Dept. of Development and Regeneration, Organ System Cluster, Univ. Hospitals of Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
39
|
Baarsma HA, Bos S, Meurs H, Visser KH, Smit M, Schols AMWJ, Langen RC, Kerstjens HAM, Gosens R. Pharmacological inhibition of GSK-3 in a guinea pig model of LPS-induced pulmonary inflammation: I. Effects on lung remodeling and pathology. Respir Res 2013; 14:113. [PMID: 24152196 PMCID: PMC4015129 DOI: 10.1186/1465-9921-14-113] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 10/14/2013] [Indexed: 12/13/2022] Open
Abstract
Background Glycogen synthase kinase-3 (GSK-3) is a constitutively active kinase that regulates multiple signalling proteins and transcription factors involved in a myriad of cellular processes. The kinase acts as a negative regulator in β-catenin signalling and is critically involved in the smad pathway. Activation of both pathways may contribute to pulmonary features of chronic obstructive pulmonary disease (COPD). Methods In the present study, we investigated the effect of the selective GSK-3 inhibitor SB216763 on pulmonary pathology in a guinea pig model of lipopolysaccharide (LPS)-induced COPD. Guinea pigs were instilled intranasally with LPS or saline twice weekly for 12 weeks and pre-treated with either intranasally instilled SB216763 or corresponding vehicle 30 min prior to each LPS/saline challenge. Results Repeated LPS exposures activated β-catenin signalling, primarily in the airway epithelium and submucosa. LPS also induced pulmonary inflammation and tissue remodelling as indicated by inflammatory cell influx, increased pulmonary fibronectin expression and enhanced small airway collagen content. Inhibition of GSK-3 by SB216763 did not affect LPS-induced inflammatory cell influx, but prevented the small airway remodelling and, unexpectedly, inhibited the activation of β-catenin in vivo. LPS or SB216763 treatment had no effect on the airway smooth muscle content and alveolar airspace size. However, GSK-3 inhibition prevented LPS-induced right ventricle hypertrophy. Conclusions Our findings indicate that GSK-3 inhibition prevents LPS-induced pulmonary pathology in guinea pigs, and that locally reduced LPS-induced β-catenin activation appears in part to underlie this effect.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
40
|
Panguluri SK, Tur J, Fukumoto J, Deng W, Sneed KB, Kolliputi N, Bennett ES, Tipparaju SM. Hyperoxia-induced hypertrophy and ion channel remodeling in left ventricle. Am J Physiol Heart Circ Physiol 2013; 304:H1651-61. [PMID: 23585127 DOI: 10.1152/ajpheart.00474.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ventricular arrhythmias account for high mortality in cardiopulmonary patients in intensive care units. Cardiovascular alterations and molecular-level changes in response to the commonly used oxygen treatment remains unknown. In the present study we investigated cardiac hypertrophy and cardiac complications in mice subjected to hyperoxia. Results demonstrate that there is a significant increase in average heart weight to tibia length (22%) in mice subjected to hyperoxia treatment vs. normoxia. Functional assessment was performed in mice subjected to hyperoxic treatment, and results demonstrate impaired cardiac function with decreased cardiac output and heart rate. Staining of transverse cardiac sections clearly demonstrates an increase in the cross-sectional area from hyperoxic hearts compared with control hearts. Quantitative real-time RT-PCR and Western blot analysis indicated differential mRNA and protein expression levels between hyperoxia-treated and control left ventricles for ion channels including Kv4.2 (-2 ± 0.08), Kv2.1 (2.54 ± 0.48), and Scn5a (1.4 ± 0.07); chaperone KChIP2 (-1.7 ± 0.06); transcriptional factors such as GATA4 (-1.5 ± 0.05), Irx5 (5.6 ± 1.74), NFκB1 (4.17 ± 0.43); hypertrophy markers including MHC-6 (2.17 ± 0.36) and MHC-7 (4.62 ± 0.76); gap junction protein Gja1 (4.4 ± 0.8); and microRNA processing enzyme Drosha (4.6 ± 0.58). Taken together, the data presented here clearly indicate that hyperoxia induces left ventricular remodeling and hypertrophy and alters the expression of Kv4.2 and MHC6/7 in the heart.
Collapse
Affiliation(s)
- Siva K Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | |
Collapse
|