1
|
Wang MY, Yi MX, Mo XY, Wei SJ, Qiao Y, Zhang Z, Su ZL, Lu HY. Over-activation of iNKT cells aggravate lung injury in bronchopulmonary dysplasia mice. Redox Biol 2024; 77:103370. [PMID: 39342744 PMCID: PMC11470607 DOI: 10.1016/j.redox.2024.103370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe lung disease in preterm infants, the abnormal proliferate and differentiate ability of type II epithelial cells (AEC II) is the key to the pathological basis of BPD. Mechanisms regarding abnormal AEC II in BPD remain unclear. The present work investigated the role and mechanisms of invariant natural killer T (iNKT) cells in lung disorder in BPD using public datasets, clinical samples, a hyperoxia-induced BPD mouse model and AEC II-iNKT cells transwell co-culture system. Firstly, we found that the NKT cells development factor IL-15 increased over time in patients with BPD in public databases, and clinically collected peripheral blood NKT cells in patients with BPD were increased. Subsequently, the percentage of iNKT cells increased in hyperoxia group compared with normoxia group, with the highest at P7, accompanied by increased activation with abnormal lung development. The administration of anti-CD1d neutralizing antibody to inhibit iNKT cells could alleviate the abnormal lung development of hyperoxia group mice, while α-GalCer administration could aggravate lung injury in hyperoxia group mice, and adoptive transfer of iNKT cells could aggravate the abnormal lung development in hyperoxia group mice. In addition, to further verify the role of iNKT cells on AEC II, AEC II-iNKT cells co-culture system was established. The presence of iNKT cells could aggravate the abnormal expression of SP-C and T1α under hyperoxia. Meanwhile, RNA-seq analysis showed that ferroptosis-related genes were highly expressed in AEC II co-cultured with iNKT cells under hyperoxia. We further validated the effect of the presence of iNKT cells under hyperoxia environment on AEC II ferroptosis levels, suggested that iNKT cells promote AEC II ferroptosis under hyperoxia, accompanied by decreased expression of SP-C and T1α. Our study found that the recruitment of iNKT cells in the lung may be an important cause of alveolarization disorder in BPD.
Collapse
Affiliation(s)
- Ming-Yan Wang
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Meng-Xu Yi
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Xing-Yu Mo
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Shan-Jie Wei
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Yu Qiao
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China
| | - Zheng Zhang
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China
| | - Zhao-Liang Su
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, China; Institute for Medical Immunology, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China.
| | - Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, 438 Jiefang Road, Zhenjiang, 212001, China.
| |
Collapse
|
2
|
Zhu X, He S, Zhang R, Kang L, Lei X, Dong W. Protective Effect and Mechanism of Autophagy in Endothelial Cell Injury Induced by Hyperoxia. Am J Perinatol 2024; 41:e2365-e2375. [PMID: 37516120 DOI: 10.1055/s-0043-1771258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/31/2023]
Abstract
OBJECTIVE Bronchopulmonary dysplasia is a chronic lung disease in premature infants with alveolar simplification and pulmonary vascular development disorder as the main pathological feature and hyperoxia as the main etiology. Autophagy is a highly conserved cytological behavior of self-degrading cellular components and is accompanied by oxidative stress. Studies have reported that autophagy is regulated by FOXO1 posttranslational modification. However, whether autophagy can be involved in the regulation of endothelial cell injury induced by hyperoxia and its mechanism are still unclear. STUDY DESIGN We have activated and inhibited autophagy in human umbilical vein endothelial cells under hyperoxia and verified the role of autophagy in endothelial cell-related functions from both positive and negative aspects. RESULTS Our research showed that the expression level of autophagy-related proteins decreased, accompanied by decreased cell migration ability and tube formation ability and increased cell reactive oxygen species level and cell permeability under hyperoxia conditions. Using an autophagy agonist alleviated hyperoxia-induced changes and played a protective role. However, inhibition of autophagy aggravated the cell damage induced by hyperoxia. Moreover, the decrease in autophagy proteins was accompanied by the upregulation of FOXO1 phosphorylation and acetylation. CONCLUSION We concluded that autophagy was a protective mechanism against endothelial cell injury caused by hyperoxia. Autophagy might participate in this process by coregulating posttranslational modifications of FOXO1. KEY POINTS · Hyperoxia induces vascular endothelial cell injury.. · Autophagy may has a protective role under hyperoxia conditions.. · FOXO1 posttranslational modification may be involved in the regulation of autophagy..
Collapse
Affiliation(s)
- Xiaodan Zhu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Shasha He
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Rong Zhang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Lan Kang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Xiaoping Lei
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| |
Collapse
|
3
|
Dennery PA, Yao H. Emerging role of cellular senescence in normal lung development and perinatal lung injury. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:10-16. [PMID: 38567372 PMCID: PMC10987039 DOI: 10.1016/j.pccm.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cellular senescence is a status of irreversible growth arrest, which can be triggered by the p53/p21cip1 and p16INK4/Rb pathways via intrinsic and external factors. Senescent cells are typically enlarged and flattened, and characterized by numerous molecular features. The latter consists of increased surfaceome, increased residual lysosomal activity at pH 6.0 (manifested by increased activity of senescence-associated beta-galactosidase [SA-β-gal]), senescence-associated mitochondrial dysfunction, cytoplasmic chromatin fragment, nuclear lamin b1 exclusion, telomere-associated foci, and the senescence-associated secretory phenotype. These features vary depending on the stressor leading to senescence and the type of senescence. Cellular senescence plays pivotal roles in organismal aging and in the pathogenesis of aging-related diseases. Interestingly, senescence can also both promote and inhibit wound healing processes. We recently report that senescence as a programmed process contributes to normal lung development. Lung senescence is also observed in Down Syndrome, as well as in premature infants with bronchopulmonary dysplasia and in a hyperoxia-induced rodent model of this disease. Furthermore, this senescence results in neonatal lung injury. In this review, we briefly discuss the molecular features of senescence. We then focus on the emerging role of senescence in normal lung development and in the pathogenesis of bronchopulmonary dysplasia as well as putative signaling pathways driving senescence. Finally, we discuss potential therapeutic approaches targeting senescent cells to prevent perinatal lung diseases.
Collapse
Affiliation(s)
- Phyllis A. Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
| |
Collapse
|
4
|
Zhou Y, Zhu Y, Jin W, Yan R, Fang Y, Zhang F, Tang T, Chen S, Chen J, Zhang F, Yu Z, Zang L, Yu Z. Tat-P combined with GAPR1 releases Beclin1 to promote autophagy and improve Bronchopulmonary dysplasia model. iScience 2023; 26:107509. [PMID: 37636035 PMCID: PMC10448080 DOI: 10.1016/j.isci.2023.107509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/25/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Long-term exposure to hyperoxia can leading to the bronchopulmonary dysplasia (BPD). The progression of BPD is primarily driven by the apoptosis of alveolar epithelial cells, and the regulation of autophagy has an impact on apoptosis. This study aims to investigate the therapeutic potential and underlying mechanism of an autophagy-promoting peptide (Tat-P) in ameliorating BPD. In vitro experiments demonstrated that Tat-P promoted autophagy and partially prevented apoptosis caused by exposure to hyperoxia. Further investigation into the mechanism revealed that Tat-P competitively binds to GAPR1, displacing the Beclin1 protein and thereby inhibiting the apoptosis. In vivo experiments conducted on Sprague-Dawley pups exposed to high oxygen levels demonstrated that Tat-P promoted autophagy and reduced apoptosis in lung tissues and ameliorated BPD-related phenotypes. Our findings elucidate the underlying mechanisms and effects of Tat-P in enhancing autophagy and preventing apoptosis. This study presents an approach for the prevention and treatment of BPD.
Collapse
Affiliation(s)
- Yahui Zhou
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Yuting Zhu
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Weilai Jin
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Ru Yan
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Yuanyuan Fang
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Fan Zhang
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Tonghui Tang
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Si Chen
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Jing Chen
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Fan Zhang
- Department of Pediatrics, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, China
| | - Zhangbin Yu
- Department of Neonatology, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, Guangdong, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Le Zang
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| | - Zhiwei Yu
- Department of Neonatology, Wuxi Children’s Hospital affiliated to Jiangnan University, Wuxi, China
| |
Collapse
|
5
|
Plataki M, Choi AMK. AMPK Activation: Respiratory Panacea? Am J Respir Cell Mol Biol 2023; 68:237-238. [PMID: 36383980 PMCID: PMC9989480 DOI: 10.1165/rcmb.2022-0428ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Maria Plataki
- Pulmonary and Critical Care Medicine Weill Cornell Medicine New York, New York
- New York-Presbyterian Hospital/Weill Cornell Medicine New York, New York
| | - Augustine M K Choi
- Pulmonary and Critical Care Medicine Weill Cornell Medicine New York, New York
- New York-Presbyterian Hospital/Weill Cornell Medicine New York, New York
| |
Collapse
|
6
|
Soni S, Jiang Y, Zhang L, Thakur A, Cataltepe S. AMPK-driven Macrophage Responses Are Autophagy Dependent in Experimental Bronchopulmonary Dysplasia. Am J Respir Cell Mol Biol 2023; 68:279-287. [PMID: 36306501 PMCID: PMC9989474 DOI: 10.1165/rcmb.2022-0282oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/28/2022] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of bronchopulmonary dysplasia (BPD) remains incompletely understood. Recent studies suggest insufficient AMP-activated protein kinase (AMPK) activation as a potential cause of impaired autophagy in rodent and nonhuman primate models of BPD. Impaired autophagy is associated with enhanced inflammatory signaling in alveolar macrophages (AMs) and increased severity of murine BPD induced by neonatal hyperoxia exposure. The goal of this study was to determine the role of autophagy and AMPK activation in macrophage responses in murine BPD. C57BL/6J mice were exposed to neonatal hyperoxia starting on postnatal day (P)1 and treated with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) between P3 and P6. Mice were euthanized on P7, and markers of AMPK activation and autophagy were assessed by immunoblotting. Alveolarization was assessed using radial alveolar counts, mean linear intercept measurements, and quantification of alveolar septal myofibroblasts. Relative mRNA expression of M1-like and M2-like genes was assessed in AMs isolated from BAL fluid from wild-type, LysMCre--Becn1fl/fl, and LysMCre+-Becn1fl/fl mice after neonatal hyperoxia exposure. AICAR treatment resulted in AMPK activation and induction of autophagic activity in whole-lung and BAL cell lysates and attenuated hyperoxia-induced alveolar simplification in neonatal lungs. AICAR-treated control but not Beclin1-deficient AMs demonstrated significantly decreased expression of M1-like markers and significantly increased expression of M2-like markers. In conclusion, pharmacologic activation of AMPK by AICAR resulted in induction of autophagy and played a protective role, at least in part, through attenuation of proinflammatory signaling in AMs via autophagy-dependent mechanisms in a murine model of BPD.
Collapse
Affiliation(s)
- Sourabh Soni
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yujie Jiang
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Pulmonary and Critical Care Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China; and
| | - Liang Zhang
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Abhijeet Thakur
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sule Cataltepe
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
7
|
Abstract
Bronchopulmonary dysplasia (BPD) in neonates is the most common pulmonary disease that causes neonatal mortality, has complex pathogenesis, and lacks effective treatment. It is associated with chronic obstructive pulmonary disease, pulmonary hypertension, and right ventricular hypertrophy. The occurrence and development of BPD involve various factors, of which premature birth is the most crucial reason for BPD. Under the premise of abnormal lung structure and functional product, newborns are susceptible to damage to oxides, free radicals, hypoxia, infections and so on. The most influential is oxidative stress, which induces cell death in different ways when the oxidative stress balance in the body is disrupted. Increasing evidence has shown that programmed cell death (PCD), including apoptosis, necrosis, autophagy, and ferroptosis, plays a significant role in the molecular and biological mechanisms of BPD and the further development of the disease. Understanding the mode of PCD and its signaling pathways can provide new therapeutic approaches and targets for the clinical treatment of BPD. This review elucidates the mechanism of BPD, focusing on the multiple types of PCD in BPD and their molecular mechanisms, which are mainly based on experimental results obtained in rodents.
Collapse
|
8
|
Wang Y, Wang X, Xu Q, Yin J, Wang H, Zhang L. CircRNA, lncRNA, and mRNA profiles of umbilical cord blood exosomes from preterm newborns showing bronchopulmonary dysplasia. Eur J Pediatr 2022; 181:3345-3365. [PMID: 35790551 PMCID: PMC9395505 DOI: 10.1007/s00431-022-04544-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/14/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023]
Abstract
Bronchopulmonary dysplasia (BPD) represents a multifactorial chronic pulmonary pathology and a major factor causing premature illness and death. The therapeutic role of exosomes in BPD has been feverishly investigated. Meanwhile, the potential roles of exosomal circRNAs, lncRNAs, and mRNAs in umbilical cord blood (UCB) serum have not been studied. This study aimed to detect the expression profiles of circRNAs, lncRNAs, and mRNAs in UCB-derived exosomes of infants with BPD. Microarray analysis was performed to compare the RNA profiles of UCB-derived exosomes of a preterm newborn with (BPD group) and without (non-BPD, NBPD group) BPD. Then, circRNA/lncRNA-miRNA-mRNA co-expression networks were built to determine their association with BPD. In addition, cell counting kit-8 (CCK-8) assay was used to evaluate the proliferation of lipopolysaccharide (LPS)-induced human bronchial epithelial cells (BEAS-2B cells) and human umbilical vein endothelial cells (HUVECs). The levels of tumor necrosis factor (TNF)-α and interleukin (IL)-1β in LPS-induced BEAS-2B cells and HUVECs were assessed through Western blot analysis. Then, quantitative reverse transcription-polymerase chain reaction assay was used to evaluate the expression levels of four differentially expressed circRNAs (hsa_circ_0086913, hsa_circ_0049170, hsa_circ_0087059, and hsa_circ_0065188) and two lncRNAs (small nucleolar RNA host gene 20 (SNHG20) and LINC00582) detected in LPS-induced BEAS-2B cells or HUVECs. A total of 317 circRNAs, 104 lncRNAs, and 135 mRNAs showed significant differential expression in UCB-derived exosomes of preterm infants with BPD compared with those with NBPD. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted to examine differentially expressed exosomal circRNAs, lncRNAs, and mRNAs. The results showed that the GO terms and KEGG pathways mostly involving differentially expressed exosomal RNAs were closely associated with endothelial or epithelial cell development. In vitro, CCK-8 and Western blot assays revealed that LPS remarkably inhibited the viability and promoted inflammatory responses (TNF-α and IL-1β) of BEAS-2B cells or HUVECs. The expression levels of circRNAs hsa_circ_0049170 and hsa_circ_0087059 were upregulated in LPS-induced BEAS-2B cells; the expression level of hsa_circ_0086913 was upregulated and that of hsa_circ_0065188 was downregulated in LPS-induced HUVECs. Moreover, the expression level of lncRNA SNHG20 was upregulated and that of LINC00582 was downregulated in LPS-induced BEAS-2B cells. Further, 455 circRNA/lncRNA-miRNA-mRNA interaction networks were predicted, including hsa_circ_0086913/hsa-miR-103a-3p/transmembrane 4 L six family member 1 (TM4SF1) and lncRNA-SNHG20/hsa-miR-6720-5p/spermine synthase (SMS) networks, which may take part in BPD. CONCLUSION This study provided a systematic perspective on UCB-derived exosomal circRNAs and lncRNAs and laid an important foundation for further investigating the potential biological functions of exosomal circRNAs and lncRNAs in BPD. WHAT IS KNOWN • BPD represents a multifactorial chronic pulmonary pathology and a major factor causing premature illness and death. • The therapeutic role of exosomes in BPD has been feverishly investigated, and exosomal RNAs were ignored. WHAT IS NEW • The profiles of UCB-derived exosomal circRNAs, lncRNAs, and mRNAs were performed. • Several differentially expressed circRNAs and lncRNAs were identified in LPS-induced BEAS-2B cells and HUVECs.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neonatology, Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Xuan Wang
- Department of Neonatology, Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Qiushi Xu
- Department of Neonatology, Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Jiao Yin
- Department of Neonatology, Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Huaiyan Wang
- Department of Neonatology, Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| | - Lin Zhang
- Department of Neonatology, Changzhou Maternal and Child Health Care Hospital, Changzhou, China
| |
Collapse
|
9
|
Yuan Y, Yang Y, Lei X, Dong W. Caffeine and bronchopulmonary dysplasia: Clinical benefits and the mechanisms involved. Pediatr Pulmonol 2022; 57:1392-1400. [PMID: 35318830 DOI: 10.1002/ppul.25898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/05/2022] [Accepted: 03/16/2022] [Indexed: 11/06/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease that occurs during the neonatal period and is commonly associated with prematurity. This condition results in a severe economic burden on society and the families involved. Caffeine is used not only for the treatment of apnea in prematurity, but also for the prevention of BPD. There are multiple clinical benefits of caffeine treatment, including improved extubation success, a reduced duration of mechanical ventilation, improved lung function, and a reduction of patent ductus arteriosus requiring treatment. These clinical benefits of caffeine for the treatment of BPD are supported by both clinical trials and evidence from animal models. However, the mechanism by which caffeine protects against BPD remains unclear. Here, we review the clinical value of caffeine in the prevention of BPD and its potential mechanisms of action, including anti-inflammatory, antioxidant, antifibrotic, and antiapoptotic properties, the regulation of angiogenesis, and diuretic effects. Our aim is to provide a new theoretical basis for the clinical treatment of BPD.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yang Yang
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoping Lei
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Pediatrics, Division of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Forno E, Abman SH, Singh J, Robbins ME, Selvadurai H, Schumacker PT, Robinson PD. Update in Pediatrics 2020. Am J Respir Crit Care Med 2021; 204:274-284. [PMID: 34126039 DOI: 10.1164/rccm.202103-0605up] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Erick Forno
- Division of Pediatric Pulmonary Medicine, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Steven H Abman
- Department of Pediatrics, Children's Hospital Colorado, Denver, Colorado.,University of Colorado Anschutz School of Medicine, Denver, Colorado
| | - Jagdev Singh
- Department of Respiratory Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| | - Mary E Robbins
- Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois; and.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Hiran Selvadurai
- Department of Respiratory Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| | - Paul T Schumacker
- Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, Chicago, Illinois; and.,Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Paul D Robinson
- Department of Respiratory Medicine, Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Discipline of Pediatrics and Child Health, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
11
|
Khemani RG, Lee JT, Wu D, Schenck EJ, Hayes MM, Kritek PA, Mutlu GM, Gershengorn HB, Coudroy R. Update in Critical Care 2020. Am J Respir Crit Care Med 2021; 203:1088-1098. [PMID: 33734938 DOI: 10.1164/rccm.202102-0336up] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Robinder G Khemani
- Pediatric ICU, Department of Anesthesiology and Critical Care Medicine, Children's Hospital Los Angeles, Los Angeles, California.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jessica T Lee
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Wu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Edward J Schenck
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York.,NewYork-Presbyterian Hospital, Weill Cornell Medical Center, New York, New York
| | - Margaret M Hayes
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Patricia A Kritek
- Division of Pulmonary, Critical Care and Sleep Medicine, School of Medicine, University of Washington Seattle, Washington
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Hayley B Gershengorn
- Division of Pulmonary, Critical Care, and Sleep Medicine, Miller School of Medicine, University of Miami, Miami, Florida.,Division of Critical Care Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Rémi Coudroy
- Institut National de la Santé et de la Recherche Médicale, Poitiers, France; and.,Médecine Intensive Réanimation, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| |
Collapse
|
12
|
Woeller CF, Lim SA, Roztocil E, Yee M, Beier EE, Puzas JE, O'Reilly MA. Neonatal hyperoxia impairs adipogenesis of bone marrow-derived mesenchymal stem cells and fat accumulation in adult mice. Free Radic Biol Med 2021; 167:287-298. [PMID: 33757863 PMCID: PMC8096722 DOI: 10.1016/j.freeradbiomed.2021.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 10/21/2022]
Abstract
Preterm birth is a risk factor for growth failure and development of respiratory disease in children and young adults. Their early exposure to oxygen may contribute to lung disease because adult mice exposed to hyperoxia as neonates display reduced lung function, changes in the host response to respiratory viral infections, and develop pulmonary hypertension and heart failure that shortens their lifespan. Here, we provide new evidence that neonatal hyperoxia also impairs growth by inhibiting fat accumulation. Failure to accumulate fat may reflect a systemic defect in adipogenic potential of stem cells because bone marrow-derived mesenchymal cells (BMSCs) isolated from the mice grew slower and were more oxidized compared to controls. They also displayed reduced capacity to accumulate lipid and differentiate into adipocytes. BMSCs from adult mice exposed to neonatal hyperoxia express lower levels of peroxisome proliferator-activated receptor gamma (PPARγ), a transcription factor that drives adipocyte differentiation. The defect in adipogenesis was rescued by expressing PPARγ in these cells. These findings reveal early life exposure to high levels of oxygen may suppresses fat accumulation and impair adipogenic differentiation upstream of PPARγ signaling, thus potentially contributing to growth failure seen in people born preterm.
Collapse
Affiliation(s)
- Collynn F Woeller
- Departments of Ophthalmology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| | - Sydney A Lim
- Departments of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Elisa Roztocil
- Departments of Ophthalmology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Min Yee
- Departments of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Eric E Beier
- Departments of Orthopaedics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - J Edward Puzas
- Departments of Orthopaedics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Michael A O'Reilly
- Departments of Ophthalmology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA; Departments of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| |
Collapse
|
13
|
Comparative analysis of ACE2 protein expression in rodent, non-human primate, and human respiratory tract at baseline and after injury: A conundrum for COVID-19 pathogenesis. PLoS One 2021; 16:e0247510. [PMID: 33626084 PMCID: PMC7904186 DOI: 10.1371/journal.pone.0247510] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/08/2021] [Indexed: 02/08/2023] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is the putative functional receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Current literature on the abundance and distribution of ACE2 protein in the human respiratory tract is controversial. We examined the effect of age and lung injury on ACE2 protein expression in rodent and non-human primate (NHP) models. We also examined ACE2 expression in human tissues with and without coronavirus disease 19 (COVID-19). ACE2 expression was detected at very low levels in preterm, but was absent in full-term and adult NHP lung homogenates. This pattern of ACE2 expression contrasted with that of transmembrane protease serine type 2 (TMPRSS2), which was significantly increased in full-term newborn and adult NHP lungs compared to preterm NHP lungs. ACE2 expression was not detected in NHP lungs with cigarette smoke-induced airway disease or bronchopulmonary dysplasia. Murine lungs lacked basal ACE2 immunoreactivity, but responded to hyperoxia, bacterial infection, and allergen exposure with new ACE2 expression in bronchial epithelial cells. In human specimens, robust ACE2 immunoreactivity was detected in ciliated epithelial cells in paranasal sinus specimens, while ACE2 expression was detected only in rare type 2 alveolar epithelial cells in control lungs. In autopsy specimens from patients with COVID-19 pneumonia, ACE2 was detected in rare ciliated epithelial and endothelial cells in the trachea, but not in the lung. There was robust expression of ACE2 expression in F344/N rat nasal mucosa and lung specimens, which authentically recapitulated the ACE2 expression pattern in human paranasal sinus specimens. Thus, ACE2 protein expression demonstrates a significant gradient between upper and lower respiratory tract in humans and is scarce in the lung. This pattern of ACE2 expression supports the notion of sinonasal epithelium being the main entry site for SARS-CoV-2 but raises further questions on the pathogenesis and cellular targets of SARS-CoV-2 in COVID-19 pneumonia.
Collapse
|
14
|
Yang K, Dong W. SIRT1-Related Signaling Pathways and Their Association With Bronchopulmonary Dysplasia. Front Med (Lausanne) 2021; 8:595634. [PMID: 33693011 PMCID: PMC7937618 DOI: 10.3389/fmed.2021.595634] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic and debilitating disease that can exert serious and overwhelming effects on the physical and mental health of premature infants, predominantly due to intractable short- and long-term complications. Oxidative stress is one of the most predominant causes of BPD. Hyperoxia activates a cascade of hazardous events, including mitochondrial dysfunction, uncontrolled inflammation, reduced autophagy, increased apoptosis, and the induction of fibrosis. These events may involve, to varying degrees, alterations in SIRT1 and its associated targets. In the present review, we describe SIRT1-related signaling pathways and their association with BPD. Our intention is to provide new insights into the molecular mechanisms that regulate BPD and identify potential therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Kun Yang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|