1
|
Guo R, Zheng Q, Zhang L. Identification and validation of diagnostic markers and drugs for pediatric bronchopulmonary dysplasia based on integrating bioinformatics and molecular docking analysis. PLoS One 2025; 20:e0323006. [PMID: 40333951 PMCID: PMC12057968 DOI: 10.1371/journal.pone.0323006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/01/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND BPD is a prevalent chronic lung disease in infancy with lifelong impacts. Its early diagnosis and treatment are hindered by complex pathophysiology and limited mechanistic understanding. This study seeks to establish a foundation for early diagnosis and targeted therapy by identifying diagnostic markers and exploring drug-gene associations. METHODS Gene expression data were retrieved from the GEO database. Functional enrichment analyses were conducted on the differentially expressed genes (DEGs). DEGs were used to construct a PPI network. Three algorithms were applied to identify diagnostic markers. Immune cell infiltration was analyzed using the CIBERSORT tool, assessing relationships between immune cells and diagnostic markers. Molecular docking was performed to evaluate interactions between predict candidate drugs and diagnostic markers. RESULTS Six hub genes were identified as diagnostic markers. Diagnostic markers showed significant correlations with specific immune cells. Resveratrol and progesterone were found to stably bind to all six diagnostic markers in molecular docking analyses, suggesting therapeutic potential. CONCLUSION In conclusion, our results show that IL7R, CXCL10, DEFA4, PRTN3, NCAPG and CCNB1 are BPD diagnostic indicators, and revealing immunological features associated with BPD. The molecular interactions of resveratrol and progesterone with the aforementioned key targets suggest their potential as therapeutic drugs for treating BPD.
Collapse
Affiliation(s)
- Rui Guo
- Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qirui Zheng
- Department of Ultrasound, The People’s Hospital of China Medical University, The People’s Hospital of Liaoning Province, Shenyang, Liaoning Province, China
| | - Liang Zhang
- Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
2
|
Schaeffer SF, Omer B, Vachharajani A, Panchangam C. Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension: Screening and Management. Neoreviews 2025; 26:e316-e327. [PMID: 40306676 DOI: 10.1542/neo.26-5-013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/21/2025] [Indexed: 05/02/2025]
Abstract
Screening for pulmonary hypertension (PH) in infants with bronchopulmonary dysplasia (BPD) was recommended by the American Thoracic Society in 2015. However, the definition of BPD has since changed. This review summarizes the current definition of BPD, the recommendations and tools for screening for PH in infants with BPD, and the various treatment options and outcomes in infants with BPD-PH.
Collapse
Affiliation(s)
| | - Bwaar Omer
- School of Medicine, University of Missouri, Columbia, Missouri
| | | | | |
Collapse
|
3
|
Cili W, Qi Z, Zhong Q, Li Y, Huang X, Yang R, Tang S, Li Q, Yang L, Ning Y, Xie Y, Feng Y, Duan J. Proline betaine facilitates angiogenesis in bronchopulmonary dysplasia. Toxicol Appl Pharmacol 2025; 498:117301. [PMID: 40089188 DOI: 10.1016/j.taap.2025.117301] [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: 11/29/2024] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is prevalent and severe diseases in preterm infants, characterized by abnormal lung development. This study aims to investigate the therapeutic potential of proline betaine, a natural alkaloid recognized for its vasculo-protective and anti-inflammatory properties, in BPD model. METHODS Network pharmacology was utilized to predict the targets of proline betaine and BPD-related genes (BPD-RGs). In vitro, HUVECs were treated with proline betaine to evaluate its effects on proliferation and angiogenesis. In vivo, a hyperoxia-induced BPD rat model (85 % oxygen, first day to 14th day) was used to evaluate the effects of proline betaine on pulmonary injury, angiogenesis and fibrosis. RESULTS We identified a total of 100 proline-betaine targets and 825 BPD-RGs, with 20 shared targets between them. These shared targets modulated inflammation, immune response, hypoxia, and vascular homeostasis, especially the vascular phenotype. In vitro, proline betaine significantly enhanced the activity, number of tubes, and capillary length of HUVECs. The pro-angiogenic effect of proline betaine on HUVECs was dose-dependent. The hyperoxia-induced BPD rat model corroborated these findings. In vivo, proline betaine increased the radial alveolar count and reduced the mean linear intercept and collagen content in the lung. Mechanistically, proline betaine upregulated VEGF and VEGFR2 expression as well as MEK/ERK pathway activity. Notably, blocking the VEGFR2 and MEK/ERK pathways made proline betaine less effective as a medicine. CONCLUSION Proline betaine enhances angiogenesis and mitigates pulmonary injury through the MEK/ERK pathway. These findings suggest that proline betaine could serve as a novel therapeutic strategy for managing BPD in neonates.
Collapse
Affiliation(s)
- Wangdui Cili
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Zhiye Qi
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Qinghua Zhong
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Yin Li
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Xia Huang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Ruoting Yang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Si Tang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Qingyuan Li
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Li Yang
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Yue Ning
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Yunbo Xie
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Yanli Feng
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China
| | - Jiang Duan
- Department of Pediatrics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province 650032, China.
| |
Collapse
|
4
|
Masselli G, Di Bella C, Hadjidekov G, Carnelli C, Morini F, Ceccanti S, Midulla F, Cozzi DA. Pediatric Congenital Lung Malformation: Advanced Imaging Techniques in Pre- and Neonatal Evaluation. Diagnostics (Basel) 2025; 15:1112. [PMID: 40361929 PMCID: PMC12071459 DOI: 10.3390/diagnostics15091112] [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: 01/31/2025] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Pediatric congenital lung malformations (CLMs) comprise a spectrum of developmental anomalies of lung parenchyma, airways, and vasculature. CLMs are increasingly diagnosed prenatally but remain best characterized by postnatal cross-sectional imaging. During pregnancy, ultrasound (US) and fetal magnetic resonance imaging (MRI) are commonly used to monitor lung lesions. Management of CLMs, including imaging, in infants and young children depends on associated symptoms and institutional standards. Chest CT angiography (CTA) is usually the most appropriate initial postnatal imaging modality for assessing prenatally diagnosed or clinically suspected CLMs in asymptomatic infants and children. Magnetic resonance (MR) imaging/magnetic resonance angiography (MRA) may be considered as a complementary, problem-solving, imaging modality for evaluation of CLMs during fetal and neonatal periods. This article presents contemporary perspectives on the imaging approach to pediatric patients with suspected CLMs and reviews up-to-date radiologic findings and clinical characteristics of CLMs.
Collapse
Affiliation(s)
- Gabriele Masselli
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy;
| | - Chiara Di Bella
- Department of Radiological Sciences, Oncology and Pathology, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy;
| | - George Hadjidekov
- Department of Physics, Biophysics and Radiology, Medical Faculty, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria;
| | - Carlos Carnelli
- Hospital de Clínicas, Universidad de la República, Montevideo 11200, Uruguay;
| | - Francesco Morini
- Pediatric Surgery Unit, Sapienza University of Rome, Azienda Ospedaliero-Universitaria Policlinico Umberto I, 00161 Rome, Italy; (F.M.); (S.C.); (D.A.C.)
| | - Silvia Ceccanti
- Pediatric Surgery Unit, Sapienza University of Rome, Azienda Ospedaliero-Universitaria Policlinico Umberto I, 00161 Rome, Italy; (F.M.); (S.C.); (D.A.C.)
| | - Fabio Midulla
- Department of Maternal Science and Urology, Sapienza University of Rome, 00185 Rome, Italy;
| | - Denis A. Cozzi
- Pediatric Surgery Unit, Sapienza University of Rome, Azienda Ospedaliero-Universitaria Policlinico Umberto I, 00161 Rome, Italy; (F.M.); (S.C.); (D.A.C.)
| |
Collapse
|
5
|
Liu J, Bao T, Zhou Y, Ma M, Tian Z. Deficiency of Secreted Phosphoprotein 1 Alleviates Hyperoxia-induced Bronchopulmonary Dysplasia in Neonatal Mice. Inflammation 2025; 48:783-795. [PMID: 38951356 DOI: 10.1007/s10753-024-02088-1] [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: 03/22/2024] [Revised: 05/24/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung disorder characterized by impaired proximal airway and bronchoalveolar development in premature births. Secreted phosphoprotein 1 (SPP1) is involved in lung development and lung injury events, while its role was not explored in BPD. For establishing the in vivo models of BPD, a mouse model of hyperoxia-induced lung injury was generated by exposing neonatal mice to hyperoxia for 7 days after birth. Alveolar myofibroblasts (AMYFs) were treated with hyperoxia to establish the in vitro models of BPD. Based on the scRNA-seq analysis of lungs of mice housed under normoxia or hyperoxia conditions, mouse macrophages and fibroblasts were main different cell clusters between the two groups, and differentially expressed genes in fibroblasts were screened. Further GO and KEGG enrichment analysis revealed that these differentially expressed genes were mainly enriched in the pathways related to cell proliferation, apoptosis as well as the PI3K-AKT and ERK/MAPK pathways. SPP1 was found up-regulated in the lung tissues of hyperoxia mice. We also demonstrated the up-regulation of SPP1 in the BPD patients, the mouse model of hyperoxia-induced lung injury, and hyperoxia-induced cells. SPP1 deficiency was revealed to reduce the hyperoxia-induced apoptosis, oxidative stress and inflammation and increase the viability of AMYFs. In the mouse model of hyperoxia induced lung injury, SPP1 deficiency was demonstrated to reverse the hyperoxia-induced alveolar growth disruption, oxidative stress and inflammation. Overall, SPP1 exacerbates BPD progression in vitro and in vivo by regulating oxidative stress and inflammatory response via the PI3K-AKT and ERK/MAPK pathways, which might provide novel therapeutic target for BPD therapy.
Collapse
Affiliation(s)
- Juan Liu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huaiyin District Huaian, Jiangsu, 223300, China
| | - Tianping Bao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huaiyin District Huaian, Jiangsu, 223300, China
| | - Yajuan Zhou
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huaiyin District Huaian, Jiangsu, 223300, China
| | - Mengmeng Ma
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huaiyin District Huaian, Jiangsu, 223300, China
| | - Zhaofang Tian
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No.1 Huanghe West Road, Huaiyin District Huaian, Jiangsu, 223300, China.
| |
Collapse
|
6
|
Quitadamo PA, Comegna L, Zambianco A, Palumbo G, Gentile MA, Mondelli A. Impact of Enteral Nutrition on Clinical Outcomes in Very Low Birth Weight Infants in the NICU: A Single-Center Retrospective Cohort Study. Nutrients 2025; 17:1138. [PMID: 40218896 PMCID: PMC11990254 DOI: 10.3390/nu17071138] [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: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Maternal milk feeding in the NICU (neonatal intensive care unit) for very low birth weight (VLBW) infants mitigates the effects of preterm birth. This single-center retrospective study analyzed data from VLBW infants born between 2005 and 2019 and investigated the impact on morbidity of exposure to Mother's Own Milk (MOM), donor human milk (DHM), preterm formula (PF), during NICU hospitalization. The assessed outcomes included necrotizing enterocolitis (NEC), retinopathy of prematurity (ROP), bronchopulmonary dysplasia (BPD), and late-onset sepsis (LOS). The study also examined the impact of a human milk-based feeding protocol on these outcomes, adjusting for confounding factors. Methods: Statistical analysis involved correlation tests and odds ratios to assess associations between feeding types and outcomes. Results: Surgical NEC occurred in 10% of infants fed exclusively with PF, 1.3% of those fed with DHM, and was completely absent in infants fed exclusively or partially with MOM. ROP across all stages was observed in 24.3% of cases, with severe ROP at 4.7%, and PF feeding was associated with a higher risk of severe ROP; the incidence of LOS was lower in infants fed human milk (-22%/-66%) compared to 10% in formula-fed infants. BPD affected 25.5% of infants, with moderate-to-severe BPD in 22.2%. The association between NEC, LOS, and feeding was statistically significant, even after adjusting for covariates. The type of milk had a significant impact on the incidence of severe forms of all outcomes (p < 0.001). The rate of exclusive MOM feeding increased over time, reaching 45% in 2018-2019. Conclusions: These findings highlight the role of human milk in preventing NEC and LOS, in reducing the risk of severe ROP and BPD, and in promoting MOM feeding, with rates increasing significantly when DHM is available.
Collapse
Affiliation(s)
- Pasqua Anna Quitadamo
- NICU, Casa Sollievo della Sofferenza Institute, 71013 San Giovanni Rotondo, Italy; (L.C.); (A.M.)
| | - Laura Comegna
- NICU, Casa Sollievo della Sofferenza Institute, 71013 San Giovanni Rotondo, Italy; (L.C.); (A.M.)
| | - Alessandra Zambianco
- San Raffaele Faculty of Medicine, University of San Raffaele Vita-Salute, 20132 Milan, Italy
| | - Giuseppina Palumbo
- NICU, Casa Sollievo della Sofferenza Institute, 71013 San Giovanni Rotondo, Italy; (L.C.); (A.M.)
| | - Maria Assunta Gentile
- NICU, Casa Sollievo della Sofferenza Institute, 71013 San Giovanni Rotondo, Italy; (L.C.); (A.M.)
| | - Antonio Mondelli
- NICU, Casa Sollievo della Sofferenza Institute, 71013 San Giovanni Rotondo, Italy; (L.C.); (A.M.)
| |
Collapse
|
7
|
Menshykova AO, Dobryanskyy DO. Pathohistological Changes in the Lungs of Very Preterm Infants with Bronchopulmonary Dysplasia Depending on the Clinical Features. Am J Perinatol 2025. [PMID: 39889725 DOI: 10.1055/a-2511-8702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
OBJECTIVE Establishing clinical factors associated with histological changes in the lungs of very preterm infants with evolving or established bronchopulmonary dysplasia (BPD) is essential for the development of more effective preventive interventions. STUDY DESIGN Thirty-two infants with a gestational age (GA) of <32 weeks who died of BPD or had BPD but died due to other causes were included in the study. The associations of clinical data with histopathological changes in the lungs were assessed. RESULTS The mean (standard deviation) GA of infants was 26.7 (1.9) weeks, and the mean birth weight was 919.7 (242.9) g. We revealed significant associations of maternal smoking with vascular hypertension lesions (r s = 0.5, p < 0.05) in infants' lungs. Intrauterine growth retardation increased the risk of extensive fibroproliferation (r s = 0.4, p < 0.05). In infants with patent ductus arteriosus (PDA) requiring treatment, muscle hyperplasia (r s = 0.5, p < 0.05) was detected more often. The longer duration of mechanical ventilation (MV) correlated with diffuse interstitial fibroproliferation (r s = 0.5, p < 0.05), airway epithelial lesions (r s = 0.3, p < 0.05), and airway muscle hyperplasia (r s = 0.4, p < 0.05). In infants who needed the longer MV and/or oxygen supplementation, an increased incidence of extensive fibroproliferation was found (r s = 0.4 and r s = 0.4 respectively, p < 0.05). Antenatal steroids decreased the incidence of diffuse interstitial fibrosis (r s = - 0.4, p < 0.05). CONCLUSION In very preterm infants with a GA of less than 32 weeks, lack of antenatal steroid prophylaxis, intrauterine growth restriction, presence of hemodynamically significant PDA, and prolonged MV or oxygen supplementation are associated with the pathomorphological lung changes that are more typical for "old" BPD. Traditional preventive measures against BPD remain essential in a modern population of very preterm infants. KEY POINTS · Pathomorphological lung changes correlate with clinical data in very preterm infants who died of BPD.. · Lack of antenatal steroids prophylaxis, growth retardation, PDA, and prolonged mechanical ventilation affect lungs.. · Traditional BPD preventive measures remain essential in the modern population of preterm infants..
Collapse
Affiliation(s)
- Anna O Menshykova
- Department of Pediatrics No. 2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Dmytro O Dobryanskyy
- Department of Pediatrics No. 2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
8
|
Igawa T, Gillespie TC, Kim ES, Lee LJ, Grogan T, Chu A, Calkins KL. Prospective Cohort Study Investigating Polyunsaturated Fatty Acids and Chronic Lung Disease in Preterm Infants. Am J Perinatol 2025. [PMID: 39638326 DOI: 10.1055/a-2496-2310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
OBJECTIVE Chronic lung disease (CLD) is a complication of prematurity. Studies examining the effects of long-chain polyunsaturated fatty acids (LC-PUFAs) on CLD are conflicting. This study investigated LC-PUFAs in the red blood cell membrane (RBCM) in preterm infants. STUDY DESIGN This prospective observational study included infants with gestational age <32 weeks or birth weight <2 kg and at least one LC-PUFA measurement in the first month of life. Subjects without CLD (CON group) were compared with those with CLD (CLD group) and then by CLD severity. RESULTS Seventy infants were included (CON n = 29; CLD n = 41). Twenty-six infants had Grade 1 CLD; 12 had Grade 2 CLD; 3 had Grade 3 CLD. When the CLD group was compared with the CON group, the overall mean (95% confidence interval) RBCM% for linoleic acid (LA) was similar (CLD vs. CON 12.5% [11.7-13.4%] vs. 11.2% [10.2-12.3%], p = 0.06) but the overall mean arachidonic acid (ARA) was lower (17.6% [17.1-18.0%] vs. 18.6% [18.1-19.2%], p < 0.01). During weeks 1 to 4, LA% was similar, while ARA% was lower in weeks 2 and 3 (18.8 ± 2.2% vs. 20.0 ± 1.5%, p = 0.05, 16.8 ± 2.0% vs. 18.3 ± 1.6%, p = 0.01). A similar trend was noted when groups were compared by CLD severity. The CLD group had a higher overall mean α-linolenic acid (ALA) compared with the CON group (0.4% [0.3-0.4%] vs. 0.2% [0.2-0.3%], p < 0.01) but no difference in docosahexaenoic acid (DHA; 3.8% [3.4-4.1%] vs. 3.8% [3.4-4.3%], p = 0.80). During weeks 1 to 4, ALA% was higher during week 1 only (0.4 ± 0.3% vs. 0.2 ± 0.1%, p < 0.01), and DHA% was similar for weeks 1 to 4. Results were similar when groups were compared by CLD severity. CONCLUSION In this study, low ARA status was associated with CLD. KEY POINTS · In this study, infants with CLD had a similar RBCM% of LA, but a lower percentage of its downstream LC-PUFA, ARA, compared with infants without CLD.. · In this study, infants with CLD had a higher RBCM% of α-linolenic acid, but a similar percentage of its downstream LC-PUFA, DHA, compared with infants without CLD.. · In this study, these trends were similiar when groups were compared by CLD severity..
Collapse
Affiliation(s)
- Teryn Igawa
- Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA and UCLA Mattel Children's Hospital, Los Angeles
| | - Tessa C Gillespie
- Department of Pediatrics, Stanford University and Stanford Medicine, Palo Alto, California
| | - Esther S Kim
- Division of Neonatology, Department of Pediatrics, UCLA - Olive View, Sylmar, California
| | - Lauren J Lee
- Department of Pediatrics, Harvard Medical School and Boston Children's Hospital, Boston, Massachusetts
| | - Tristan Grogan
- Department of Medicine Statistics Core, David Geffen School of Medicine, UCLA, Los Angeles, California
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Alison Chu
- Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA and UCLA Mattel Children's Hospital, Los Angeles
| | - Kara L Calkins
- Department of Pediatrics, Children's Discovery and Innovation Institute, David Geffen School of Medicine UCLA and UCLA Mattel Children's Hospital, Los Angeles
| |
Collapse
|
9
|
Zheng S, Gao E, Guo L, Xie L, Zhao B, Hong Q, Li J, Hu X, Tao B. LncRNA MIAT binding to GATA3 activates MAPK signaling pathway and influences bronchopulmonary dysplasia. Int J Biol Macromol 2025; 286:138280. [PMID: 39626812 DOI: 10.1016/j.ijbiomac.2024.138280] [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: 08/01/2024] [Revised: 11/18/2024] [Accepted: 11/30/2024] [Indexed: 12/19/2024]
Abstract
Bronchopulmonary dysplasia (BPD) manifests in premature neonates with aberrant pulmonary function. Numerous long non-coding RNAs (lncRNAs) have been implicated in the pathogenesis of BPD. This study aims to elucidate the impact of the lncRNA myocardial infarction-associated transcript (MIAT) on the initiation and progression of BPD. Initially, BPD murine models were established through hyperoxia induction in newborn mice. Subsequently, MIAT and GATA binding protein 3 (GATA3) expression levels were assessed, and intravenous administration of short hairpin RNAs (shRNAs) targeting MIAT and GATA3 was performed. Pulmonary histological alterations were examined through histological staining. Levels of inflammatory mediators were quantified using enzyme-linked immunosorbent assay (ELISA) kits. The interaction between MIAT and GATA3 was scrutinized through RNA immunoprecipitation, RNA pull-down, and fluorescence in situ hybridization. The downstream mechanisms of GATA3 were explored using bioinformatics analysis. In summary, lncRNA MIAT exhibited elevated expression in the lung tissues of BPD-afflicted mice. MIAT localized to the nucleus and interacted with GATA3, thereby activating the mitogen-activated protein kinase (MAPK) pathway. Knockdown of MIAT or silencing of GATA3 attenuated the inflammatory response, deactivated the MAPK pathway, and ameliorated BPD symptoms in mice, on the other hand, p-Cresyl sulfate potassium can activate the MAPK signaling pathway and attenuates the effects of si-MIAT or si-GAT3. These improvements were characterized by enhanced alveolar differentiation and reduced glycogen and collagen deposition. In conclusion, lncRNA MIAT plays a pivotal role in activating the MAPK pathway and exacerbating hyperoxia-induced BPD in mice through the binding to GATA3. It's an important discovery for the pathogenesis of BPD and may provide some new treatment for infants diagnosed with BPD.
Collapse
Affiliation(s)
- Siqiang Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Erji Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Liang Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Lei Xie
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Bin Zhao
- Department of Pulmonary Nodule Center, Shandong Public Health Clinical Center, Jinan, Shandong 250100, China
| | - Qi Hong
- Department of Thoracic Surgery, Shenyang Tenth People's Hospital, Shenyang Chest Hospital, Shenyang, Liaoning 110044, China
| | - Juanjuan Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Bo Tao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China.
| |
Collapse
|
10
|
Zhu Y, Hou H, Li Y, Zhang Y, Fang Y, Chen S, Zhang L, Jin W, Zhou Y. Hyperoxia exposure induces ferroptosis and apoptosis by downregulating PLAGL2 and repressing HIF-1α/VEGF signaling pathway in newborn alveolar typeII epithelial cell. Redox Rep 2024; 29:2387465. [PMID: 39102510 DOI: 10.1080/13510002.2024.2387465] [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] [Indexed: 08/07/2024] Open
Abstract
BACKGROUD Bronchopulmonary dysplasia (BPD) is one of the most important complications plaguing neonates and can lead to a variety of sequelae. the ability of the HIF-1α/VEGF signaling pathway to promote angiogenesis has an important role in neonatal lung development. METHOD Newborn rats were exposed to 85% oxygen. The effects of hyperoxia exposure on Pleomorphic Adenoma Gene like-2 (PLAGL2) and the HIF-1α/VEGF pathway in rats lung tissue were assessed through immunofluorescence and Western Blot analysis. In cell experiments, PLAGL2 was upregulated, and the effects of hyperoxia and PLAGL2 on cell viability were evaluated using scratch assays, CCK-8 assays, and EDU staining. The role of upregulated PLAGL2 in the HIF-1α/VEGF pathway was determined by Western Blot and RT-PCR. Apoptosis and ferroptosis effects were determined through flow cytometry and viability assays. RESULTS Compared with the control group, the expression levels of PLAGL2, HIF-1α, VEGF, and SPC in lung tissues after 3, 7, and 14 days of hyperoxia exposure were all decreased. Furthermore, hyperoxia also inhibited the proliferation and motility of type II alveolar epithelial cells (AECII) and induced apoptosis in AECII. Upregulation of PLAGL2 restored the proliferation and motility of AECII and suppressed cell apoptosis and ferroptosis, while the HIF-1α/VEGF signaling pathway was also revived. CONCLUSIONS We confirmed the positive role of PLAGL2 and HIF-1α/VEGF signaling pathway in promoting BPD in hyperoxia conditions, and provided a promising therapeutic targets.
Collapse
Affiliation(s)
- Yuting Zhu
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Hongmei Hou
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Yawen Li
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Yanyu Zhang
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Yuanyuan Fang
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Si Chen
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Le Zhang
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Weilai Jin
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| | - Yahui Zhou
- Department of Neonatology, The Affiliated Children's Hospital of Jiangnan University, Wuxi, People's Republic of China
| |
Collapse
|
11
|
Yang X, Wang X, Dong W. Aryl hydrocarbon receptor (AhR) is regulated by hyperoxia in premature infants. J Matern Fetal Neonatal Med 2024; 37:2349179. [PMID: 38816997 DOI: 10.1080/14767058.2024.2349179] [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: 01/22/2024] [Accepted: 04/24/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE To investigate whether aryl hydrocarbon receptor (AhR) is involved in hyperoxia-mediated oxidative stress by observing the relationship between AhR and reactive oxygen species (ROS) in peripheral blood mononuclear cells (PBMCs) after oxygen exposure in premature infants. METHODS After 48 h of oxygen inhalation at different concentrations, discarded peripheral blood was collected to separate PBMCs and plasma. ROS were labeled with MitoSOXTM Red and detected by fluorescence microscopy in PBMCs. The level of MDA in plasma was detected by thiobarbituric acid colorimetry, the level of MCP-1 in plasma was detected by enzyme-linked immunosorbent assay (ELISA), the localization of AhR was detected by immunofluorescence, and the level of AhR expression in PBMCs was detected by Western blotting. RESULTS As the volume fraction of inspired oxygen increased, compared with those in the air control group, the levels of ROS, MDA in plasma, and MCP-1 in plasma increased gradually in the low concentration oxygen group, medium concentration oxygen group and high concentration oxygen group. The cytoplasm-nuclear translocation rate of AhR gradually increased, and the expression level of AhR gradually decreased. The levels of ROS in PBMCs, MDA in the plasma and MCP-1 in the plasma of premature infants were positively correlated with the cytoplasm-nuclear translocation rate of AhR but negatively correlated with the level of AhR expression. CONCLUSION Aryl hydrocarbon receptor (AhR) is regulated by hyperoxia in premature infants.
Collapse
Affiliation(s)
- Xi Yang
- 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
| | - Xia Wang
- 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
|
12
|
刘 怡, 颜 崇, 张 媛, 翁 博, 蔡 成. [Risk factors for bronchopulmonary dysplasia in preterm infants and establishment of a prediction model]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:1148-1154. [PMID: 39587742 PMCID: PMC11601104 DOI: 10.7499/j.issn.1008-8830.2404065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/23/2024] [Indexed: 11/27/2024]
Abstract
OBJECTIVES To investigate the risk factors for bronchopulmonary dysplasia (BPD) in preterm infants, and to establish a risk prediction model. METHODS A total of 120 preterm infants who were admitted to the neonatal intensive care unit of Shanghai Children's Hospital from January to December 2022 were included. According to the diagnostic criteria for BPD released by the National Institute of Child Health and Human Development in 2018, they were divided into a non-BPD group (84 infants) and a BPD group (36 infants). The clinical data of the infants and their mothers were compared between the two groups. The univariate analysis and the stepwise multivariate regression analysis were used to identify the risk factors for BPD and establish a risk prediction model. RESULTS The results showed that a gestational age of <28 weeks, duration of noninvasive respiratory support, comorbidity with infectious pneumonia, and chorioamnionitis in the mother were independent risk factors for BPD in preterm infants (P<0.05). A nomogram model for predicting the development of BPD was established based on the risk factors, with an area under the receiver operating characteristic curve of 0.93, and the calibration curve of this nomogram had a slope of about 1. The goodness-of-fit test indicated the model fitted well (χ2=8.287, P=0.406). CONCLUSIONS A gestational age of <28 weeks, duration of noninvasive respiratory support, comorbidity with infectious pneumonia, and chorioamnionitis in the mother are independent risk factors for BPD in preterm infants.
Collapse
|
13
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024; 239:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [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: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
14
|
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
|
15
|
Lok IM, Wever KE, Vliegenthart RJS, Onland W, van Kaam AH, van Tuyl M. Effects of postnatal corticosteroids on lung development in newborn animals. A systematic review. Pediatr Res 2024; 96:1141-1152. [PMID: 38493255 PMCID: PMC11522003 DOI: 10.1038/s41390-024-03114-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/18/2024] [Accepted: 02/05/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Postnatal systemic corticosteroids reduce the risk of bronchopulmonary dysplasia but the effect depends on timing, dosing, and type of corticosteroids. Animal studies may provide valuable information on these variable effects. This systematic review summarizes the effects of postnatal systemic corticosteroids on lung development in newborn animals. METHODS A systematic search was performed in PubMed and Embase in December 2022. The protocol was published on PROSPERO (CRD42021177701). RESULTS Of the 202 eligible studies, 51 were included. Only newborn rodent studies met the inclusion criteria. Most studies used dexamethasone (98%). There was huge heterogeneity in study outcome measures and corticosteroid treatment regimens. Reporting of study quality indicators was mediocre and risk of bias was unclear due to poor reporting of study methodology. Meta-analysis showed that postnatal corticosteroids caused a decrease in body weight as well as persistent alveolar simplification. Subgroup analyses revealed that healthy animals were most affected. CONCLUSION In newborn rodents, postnatal systemic corticosteroids have a persistent negative effect on body weight and lung development. There was huge heterogeneity in experimental models, mediocre study quality, unclear risk of bias, and very small subgroups for meta-analysis which limited firm conclusions. IMPACT Postnatal corticosteroids reduce the risk of bronchopulmonary dysplasia but the effect depends on timing, dosing, and type of corticosteroids while the underlying mechanism of this variable effect is unknown. This is the first systematic review and meta-analysis of preclinical newborn animal studies reviewing the effect of postnatal systemic corticosteroids on lung development. In newborn rodent models, postnatal corticosteroids have a persistent negative effect on body weight and lung alveolarization, especially in healthy animals.
Collapse
Affiliation(s)
- Irene M Lok
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Kimberley E Wever
- Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Wes Onland
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Anton H van Kaam
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - Minke van Tuyl
- Department of Neonatology, Emma Children's Hospital Amsterdam UMC, location University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Reproduction & Development (AR&D) Research Institute, Amsterdam, The Netherlands.
| |
Collapse
|
16
|
Tenero L, Piazza M, Sandri M, Ferrante G, Giacomello E, Ficial B, Zaffanello M, Biban P, Piacentini G. Early Diagnosis of Bronchopulmonary Dysplasia with E-Nose: A Pilot Study in Preterm Infants. SENSORS (BASEL, SWITZERLAND) 2024; 24:6282. [PMID: 39409322 PMCID: PMC11479126 DOI: 10.3390/s24196282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common respiratory disease in preterm and is still associated with increased mortality and morbidity. The great interest lies in identifying early biomarkers that can predict the development of BPD. This pilot study explores the potential of e-nose for the early identification of BPD risk in premature infants by analyzing volatile organic compounds (VOCs) in the exhaled breath condensate (EBC). Fourteen mechanically ventilated very preterm infants were included in this study. The clinical parameters and EBC were collected within the first 24 h of life. The discriminative ability of breath prints between preterms who did and did not develop BPD was investigated using pattern recognition, a machine learning algorithm, and standard statistical methods. We found that e-nose probes can significantly predict the outcome of "no-BPD" vs. "BPD". Specifically, a subset of probes (S18, S24, S14, and S6) were found to be significantly predictive, with an AUC of 0.87, 0.89, 0.82, 0.8, and p = 0.019, 0.009, 0.043, 0.047, respectively. The e-nose is an easy-to-use, handheld, non-invasive electronic device that quickly samples breath. Our preliminary study has shown that it has the potential for early prediction of BPD in preterms.
Collapse
Affiliation(s)
- Laura Tenero
- Pediatric Section, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy;
| | - Michele Piazza
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (M.P.); (G.F.); (E.G.); (G.P.)
| | - Marco Sandri
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (M.P.); (G.F.); (E.G.); (G.P.)
| | - Giuliana Ferrante
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (M.P.); (G.F.); (E.G.); (G.P.)
| | - Elisabetta Giacomello
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (M.P.); (G.F.); (E.G.); (G.P.)
| | - Benjamim Ficial
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy; (B.F.); (P.B.)
| | - Marco Zaffanello
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (M.P.); (G.F.); (E.G.); (G.P.)
| | - Paolo Biban
- Neonatal Intensive Care Unit, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy; (B.F.); (P.B.)
| | - Giorgio Piacentini
- Department of Surgery, Dentistry, Paediatrics and Gynaecology, University of Verona, 37129 Verona, Italy; (M.P.); (G.F.); (E.G.); (G.P.)
| |
Collapse
|
17
|
Long Y, Luo Y, Hu L, Liao H, Liu J. Targeting miR-146b-5p to Regulate KDM6B Expression Aggravates Bronchopulmonary Dysplasia. Mol Biotechnol 2024; 66:2078-2086. [PMID: 37584827 DOI: 10.1007/s12033-023-00849-1] [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: 03/09/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
miR-146b-5p has been studied to be highly expressed in bronchopulmonary dysplasia (BPD), but whether it is involved in regulating the process of BPD in premature infants remains unclear. This study was to explore miR-146b-5p in premature BPD and reveal its molecular mechanism. BPD mouse model and high-oxygen MLE-12 cell model were established. HE staining, TUNEL staining, and IF staining were conducted to evaluate the pathological injury and protein expression in mouse lung tissue. LDH assay, MMT assay, and flow cytometry were achieved to evaluate cytotoxicity, cell viability, and apoptosis. ELISA and immunoblotting were performed to evaluate inflammatory cytokines and Wnt pathway proteins in lung tissues and cells. Dual-luciferase reporter assay and RIP assay were needed to examine the targeting relationship between miR-146b-5p and KDM6B. miR-146b-5p was abundantly expressed in BPD and KDM6B was lowly expressed. miR-146b-5p knockdown improved hyperoxia-induced lung epithelial cell inflammation and apoptosis in both models. miR-146b-6p upregulation or KDM6B downregulation aggravated hyperoxia-induced inflammation and apoptosis of lung epithelial cells. This effect of overexpressing miR-146b-5p was rescued by forcing KDM6B. MiR-146b-5p activated Wnt signaling by regulating KDM6B. miR-146b-5p activates the Wnt pathway through targeted regulation of KDM6B, thereby aggravating hyperoxia-induced inflammation and apoptosis of lung epithelial cells.
Collapse
Affiliation(s)
- YunFeng Long
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Yong Luo
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Liu Hu
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Hong Liao
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China
| | - Jin Liu
- Department of Neonatology, The First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang City, 422000, Hunan Province, China.
| |
Collapse
|
18
|
Urru SA, Geist M, Carlinger R, Bodrero E, Bruschettini M. Strategies for cessation of caffeine administration in preterm infants. Cochrane Database Syst Rev 2024; 7:CD015802. [PMID: 39045901 PMCID: PMC11267609 DOI: 10.1002/14651858.cd015802.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
BACKGROUND Apnea and intermittent hypoxemia (IH) are common developmental disorders in infants born earlier than 37 weeks' gestation. Caffeine administration has been shown to lower the incidence of these disorders in preterm infants. Cessation of caffeine treatment is based on different post-menstrual ages (PMA) and resolution of symptoms. There is uncertainty about the best timing for caffeine discontinuation. OBJECTIVES To evaluate the effects of early versus late discontinuation of caffeine administration in preterm infants. SEARCH METHODS We searched CENTRAL, PubMed, Embase, and three trial registries in August 2023; we applied no date limits. We checked the references of included studies and related systematic reviews. SELECTION CRITERIA We included randomized controlled trials (RCTs) in preterm infants born earlier than 37 weeks' gestation, up to a PMA of 44 weeks and 0 days, who received caffeine for any indication for at least seven days. We compared three different strategies for caffeine cessation: 1. at different PMAs, 2. before or after five days without symptoms, and 3. at a predetermined PMA versus at the resolution of symptoms. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Primary outcomes were: restarting caffeine therapy, intubation within one week of treatment discontinuation, and the need for non-invasive respiratory support within one week of treatment discontinuation. Secondary outcomes were: number of episodes of apnea in the seven days after treatment discontinuation, number of infants with at least one episode of apnea in the seven days after treatment discontinuation, number of episodes of intermittent hypoxemia (IH) within seven days of treatment discontinuation, number of infants with at least one episode of IH in the seven days after of treatment discontinuation, all-cause mortality prior to hospital discharge, major neurodevelopmental disability, number of days of respiratory support after treatment discontinuation, duration of hospital stay, and cost of neonatal care. We used GRADE to assess the certainty of evidence for each outcome. MAIN RESULTS We included three RCTs (392 preterm infants). Discontinuation of caffeine at PMA less than 35 weeks' gestation versus PMA equal to or longer than 35 weeks' gestation This comparison included one single completed RCT with 98 premature infants with a gestational age between 25 + 0 and 32 + 0 weeks at birth. All infants had discontinued caffeine treatment for five days at randomization. The infants received either an oral loading dose of caffeine citrate (20 mg/kg) at randomization followed by oral maintenance dosage (6 mg/kg/day) until 40 weeks PMA, or usual care (controls), during which caffeine was stopped before 37 weeks PMA. Early cessation of caffeine administration in preterm infants at PMA less than 35 weeks' gestation may result in an increase in the number of IH episodes in the seven days after discontinuation of treatment, compared to prolonged caffeine treatment beyond 35 weeks' gestation (mean difference [MD] 4.80, 95% confidence interval [CI] 2.21 to 7.39; 1 RCT, 98 infants; low-certainty evidence). Early cessation may result in little to no difference in all-cause mortality prior to hospital discharge compared to late discontinuation after 35 weeks PMA (risk ratio [RR] not estimable; 98 infants; low-certainty evidence). No data were available for the following outcomes: restarting caffeine therapy, intubation within one week of treatment discontinuation, need for non-invasive respiratory support within one week of treatment discontinuation, number of episodes of apnea, number of infants with at least one episode of apnea in the seven days after discontinuation of treatment, or number of infants with at least one episode of IH in the seven days after discontinuation of treatment. Discontinuation based on PMA versus resolution of symptoms This comparison included two RCTs with a total of 294 preterm infants. Discontinuing caffeine at the resolution of symptoms compared to discontinuing treatment at a predetermined PMA may result in little to no difference in all-cause mortality prior to hospital discharge (RR 1.00, 95% CI 0.14 to 7.03; 2 studies, 294 participants; low-certainty evidence), or in the number of infants with at least one episode of apnea within the seven days after discontinuing treatment (RR 0.60, 95% CI 0.31 to 1.18; 2 studies; 294 infants; low-certainty evidence). Discontinuing caffeine based on the resolution of symptoms probably results in more infants with IH in the seven days after discontinuation of treatment (RR 0.38, 95% CI 0.20 to 0.75; 1 study; 174 participants; moderate-certainty evidence). No data were available for the following outcomes: restarting caffeine therapy, intubation within one week of treatment discontinuation, need for non-invasive respiratory support within one week of treatment discontinuation, or number of episodes of IH in the seven days after treatment discontinuation. Adverse effects In the Rhein 2014 study, five of the infants randomized to caffeine had the caffeine treatment discontinued at the discretion of the clinical team, because of tachycardia. The Pradhap 2023 study reported adverse events, including recurrence of apnea of prematurity (15% in the short and 13% in the regular course caffeine therapy group), varying severities of bronchopulmonary dysplasia, hyperglycemia, extrauterine growth restriction, retinopathy of prematurity requiring laser treatment, feeding intolerance, osteopenia, and tachycardia, with no significant differences between the groups. The Prakash 2021 study reported that adverse effects of caffeine therapy for apnea of prematurity included tachycardia, feeding intolerance, and potential neurodevelopmental impacts, though most were mild and transient. We identified three ongoing studies. AUTHORS' CONCLUSIONS There may be little or no difference in the incidence of all-cause mortality and apnea in infants who were randomized to later discontinuation of caffeine treatment. However, the number of infants with at least one episode of IH was probably reduced with later cessation. No data were found to evaluate the benefits and harms of later caffeine discontinuation for: restarting caffeine therapy, intubation within one week of treatment discontinuation, or need for non-invasive respiratory support within one week of treatment discontinuation. Further studies are needed to evaluate the short-term and long-term effects of different caffeine cessation strategies in premature infants.
Collapse
Affiliation(s)
- Silvana Am Urru
- Hospital Pharmacy Unit, Santa Chiara Hospital, Azienda Provinciale per i Servizi Sanitari (APSS), Trento, Italy
- Department of Chemistry and Pharmacy, School of Hospital Pharmacy, University of Sassari, Sassari, Italy
| | - Milena Geist
- Institute for Medical Information Processing, Biometry, and Epidemiology - IBE, LMU Munich, Munich, Germany
- Pettenkofer School of Public Health, Munich, Germany
| | | | - Enrico Bodrero
- Neonatal Intensive Care Unit, Ospedale S. Croce e Carle, Cuneo, Italy
| | - Matteo Bruschettini
- Paediatrics, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Department of Research and Education, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
19
|
Purcell E, Shah J, Powell C, Nguyen T, Zhou L, McDonald CA, Allison BJ, Malhotra A. Umbilical cord blood-derived therapy for preterm lung injury: a systematic review and meta-analysis. Stem Cells Transl Med 2024; 13:606-624. [PMID: 38819251 PMCID: PMC11227974 DOI: 10.1093/stcltm/szae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/14/2024] [Indexed: 06/01/2024] Open
Abstract
INTRODUCTION Lung injuries, such as bronchopulmonary dysplasia (BPD), remain a major complication of preterm birth, with limited therapeutic options. One potential emerging therapy is umbilical cord blood (UCB)-derived therapy. OBJECTIVES To systematically assess the safety and efficacy of UCB-derived therapy for preterm lung injury in preclinical and clinical studies. METHODS A systematic search of MEDLINE, Embase, CENTRAL, ClinicalTrials.gov, and WHO International Trials Registry Platform was performed. A meta-analysis was conducted with Review Manager (5.4.1) using a random effects model. Data was expressed as standardized mean difference (SMD) for preclinical data and pooled relative risk (RR) for clinical data, with 95% confidence intervals (CI). Potential effect modifiers were investigated via subgroup analysis. Certainty of evidence was assessed using the GRADE system. RESULTS Twenty-three preclinical studies and six clinical studies met eligibility criteria. Statistically significant improvements were seen across several preclinical outcomes, including alveolarization (SMD, 1.32, 95%CI [0.99, 1.65]), angiogenesis (SMD, 1.53, 95%CI [0.87, 2.18]), and anti-inflammatory cytokines (SMD, 1.68, 95%CI [1.03, 2.34]). In clinical studies, 103 preterm infants have received UCB-derived therapy for preterm lung injury and no significant difference was observed in the development of BPD (RR, 0.93, 95%CI [0.73, 1.18]). Across both preclinical and clinical studies, administration of UCB-derived therapy appeared safe. Certainty of evidence was assessed as "low." CONCLUSIONS Administration of UCB-derived therapy was associated with statistically significant improvements across several lung injury markers in preclinical studies. Early clinical studies demonstrated the administration of UCB-derived therapy as safe and feasible but lacked data regarding efficacy.
Collapse
Affiliation(s)
- Elisha Purcell
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Jainam Shah
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Cameron Powell
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Timothy Nguyen
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Monash Newborn, Monash Children's Hospital, VIC 3168, Melbourne, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, VIC 3168, Melbourne, Australia
| | - Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, VIC 3168, Melbourne, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Monash Newborn, Monash Children's Hospital, VIC 3168, Melbourne, Australia
| |
Collapse
|
20
|
樊 雨, 张 伊, 温 和, 晏 红, 沈 蔚, 丁 月, 龙 运, 张 志, 李 桂, 姜 泓, 饶 红, 邱 建, 魏 贤, 张 亚, 曾 纪, 赵 常, 许 伟, 王 凡, 员 丽, 杨 秀, 李 薇, 林 霓, 陈 倩, 夏 昌, 钟 鑫, 崔 其. [Risk factors for bronchopulmonary dysplasia in twin preterm infants: a multicenter study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:611-618. [PMID: 38926378 PMCID: PMC11562066 DOI: 10.7499/j.issn.1008-8830.2312005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/08/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVES To investigate the risk factors for bronchopulmonary dysplasia (BPD) in twin preterm infants with a gestational age of <34 weeks, and to provide a basis for early identification of BPD in twin preterm infants in clinical practice. METHODS A retrospective analysis was performed for the twin preterm infants with a gestational age of <34 weeks who were admitted to 22 hospitals nationwide from January 2018 to December 2020. According to their conditions, they were divided into group A (both twins had BPD), group B (only one twin had BPD), and group C (neither twin had BPD). The risk factors for BPD in twin preterm infants were analyzed. Further analysis was conducted on group B to investigate the postnatal risk factors for BPD within twins. RESULTS A total of 904 pairs of twins with a gestational age of <34 weeks were included in this study. The multivariate logistic regression analysis showed that compared with group C, birth weight discordance of >25% between the twins was an independent risk factor for BPD in one of the twins (OR=3.370, 95%CI: 1.500-7.568, P<0.05), and high gestational age at birth was a protective factor against BPD (P<0.05). The conditional logistic regression analysis of group B showed that small-for-gestational-age (SGA) birth was an independent risk factor for BPD in individual twins (OR=5.017, 95%CI: 1.040-24.190, P<0.05). CONCLUSIONS The development of BPD in twin preterm infants is associated with gestational age, birth weight discordance between the twins, and SGA birth.
Collapse
Affiliation(s)
| | | | - 和梅 温
- 四川锦欣妇女儿童医院新生儿科,四川成都610011
| | - 红 晏
- 贵州医科大学附属医院新生儿科,贵州贵阳550001
| | - 蔚 沈
- 厦门大学附属妇女儿童医院新生儿科,福建厦门361003
| | - 月琴 丁
- 南方医科大学附属东莞医院新生儿科,广东东莞523000
| | - 运峰 龙
- 邵阳学院附属第一医院新生儿科,湖南邵阳422000
| | | | | | | | | | - 建武 邱
- 汕头大学医学院附属粤北人民医院新生儿科,广东韶关512026
| | - 贤 魏
- 武汉科技大学附属孝感医院新生儿科,湖北孝感432000
| | - 亚昱 张
- 内蒙古医科大学附属医院新生儿科,内蒙古呼和浩特010050
| | - 纪斌 曾
- 汕头大学医学院第二附属医院新生儿科,广东汕头515041
| | - 常亮 赵
- 包钢集团第三;职工医院新生儿科,内蒙古包头014010
| | - 伟鹏 许
- 暨南大学附属第一医院新生儿科,广东广州510630
| | | | | | | | - 薇 李
- 东莞市滨海湾中心医院新生儿科,广东东莞523808
| | - 霓阳 林
- 汕头大学;医学院第一附属医院新生儿科,广东汕头515041
| | | | | | | | | |
Collapse
|
21
|
Luo Y, Zhang Z, Xi S, Li T. Bioinformatics analyses and experimental validation of ferroptosis-related genes in bronchopulmonary dysplasia pathogenesis. PLoS One 2024; 19:e0291583. [PMID: 38875180 PMCID: PMC11178182 DOI: 10.1371/journal.pone.0291583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 05/18/2024] [Indexed: 06/16/2024] Open
Abstract
OBJECTIVE We aimed to study the involvement of ferroptosis in the pathogenesis of bronchopulmonary dysplasia (BPD) by conducting bioinformatics analyses and identifying and validating the associated ferroptosis-related genes to explore new directions for treating BPD. METHODS The dataset GSE32472 on BPD was downloaded from the public genome database. Using R language, differentially expressed genes (DEGs) between the BPD and normal group were screened. In the present study, we adopted weighted gene correlation network analysis (WGCNA) for identifying BPD-related gene modules and ferroptosis-related genes were extracted from FerrDb. Their results were intersected to obtain the hub genes. After that, to explore the hub gene-related signaling pathways, the hub genes were exposed to gene ontology enrichment analysis. With the purpose of verifying the mRNA expression of the hub genes, a single-gene gene set enrichment analysis and quantitative reverse transcription polymerase chain reaction were conducted. Immune cell infiltration in BPD was analyzed using the CIBERSORT inverse fold product algorithm. RESULTS A total of 606 DEGs were screened. WGCNA provided the BPD-related gene module darkgreen4. The intersection of DEGs, intramodular genes, and ferroptosis-related genes revealed six ferroptosis-associated hub genes (ACSL1, GALNT14, WIPI1, MAPK14, PROK2, and CREB5). Receiver operating characteristic curve analysis demonstrated that the hub genes screened for BPD were of good diagnostic significance. According to the results of immune infiltration analysis, the proportions of CD8, CD4 naive, and memory resting T cells and M2 macrophage were elevated in the normal group, and the proportions of M0 macrophage, resting mast cell, and neutrophils were increased in the BPD group. CONCLUSIONS A total of six ferroptosis-associated hub genes in BPD were identified in this study, and they may be potential new therapeutic targets for BPD.
Collapse
Affiliation(s)
- Yifan Luo
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Zongli Zhang
- Institute of Pediatric Diseases, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Shibing Xi
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| | - Tao Li
- Department of Pediatrics, Affiliated Taihe Hospital of Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
22
|
Boel L, Gallacher DJ, Marchesi JR, Kotecha S. The Role of the Airway and Gut Microbiome in the Development of Chronic Lung Disease of Prematurity. Pathogens 2024; 13:472. [PMID: 38921770 PMCID: PMC11206380 DOI: 10.3390/pathogens13060472] [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: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/27/2024] Open
Abstract
Chronic lung disease (CLD) of prematurity, a common cause of morbidity and mortality in preterm-born infants, has a multifactorial aetiology. This review summarizes the current evidence for the effect of the gut and airway microbiota on the development of CLD, highlighting the differences in the early colonisation patterns in preterm-born infants compared to term-born infants. Stool samples from preterm-born infants who develop CLD have less diversity than those who do not develop CLD. Pulmonary inflammation, which is a hallmark in the development of CLD, may potentially be influenced by gut bacteria. The respiratory microbiota is less abundant than the stool microbiota in preterm-born infants. There is a lack of clear evidence for the role of the respiratory microbiota in the development of CLD, with results from individual studies not replicated. A common finding is the presence of a single predominant bacterial genus in the lungs of preterm-born infants who develop CLD. Probiotic preparations have been proposed as a potential therapeutic strategy to modify the gut or lung microbiota with the aim of reducing rates of CLD but additional robust evidence is required before this treatment is introduced into routine clinical practice.
Collapse
Affiliation(s)
- Lieve Boel
- Neonatal Unit, University Hospital of Wales, Cardiff CF14 4XW, UK; (L.B.); (D.J.G.)
| | - David J. Gallacher
- Neonatal Unit, University Hospital of Wales, Cardiff CF14 4XW, UK; (L.B.); (D.J.G.)
| | - Julian R. Marchesi
- Division of Digestive Diseases, Faculty of Medicine, Imperial College, London W2 1NY, UK;
| | - Sailesh Kotecha
- Department of Child Health, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
23
|
Saenz de Pipaon M, Nelin LD, Gehred A, Rossholt ME, Moltu S, Van den Akker C, van Kaam AH, Sánchez A, Khashu M, Roehr CC, Carnielli V. The role of nutritional interventions in the prevention and treatment of chronic lung disease of prematurity. Pediatr Res 2024:10.1038/s41390-024-03133-3. [PMID: 38565917 DOI: 10.1038/s41390-024-03133-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/20/2024] [Indexed: 04/04/2024]
Abstract
Chronic lung disease of prematurity or bronchopulmonary dysplasia (BPD) is a common complication of preterm birth. Nutrition may affect incidence and severity of BPD. In this context, the Section on Nutrition, Gastroenterology and Metabolism, the Pulmonary Section of the European Society for Paediatric Research (ESPR) and SPR have joined forces to review the current knowledge on nutritional issues related to BPD. The aim of this narrative review is to discuss the clinical implications for nutritional practice. Nutrient deficiencies may influence pathogenesis of BPD. Adequate nutrition and growth can play a crucial role in the prevention of and recovery from BPD. Optimal nutrition strategy is an important principle, especially in the early postnatal period. As optimal energy intake in infants at risk of BPD or with evolving BPD is not yet defined, further research with well-designed studies on nutritional strategies for preterm infants with BPD is urgently needed. IMPACT: Based on current evidence it seems reasonable to recommend that BPD diagnosed infants should receive an energy supply ranging from 120 to 150 Kcal/kg/d. Exclusive MOM feed with adequate fortification should be encouraged as this is associated with a significant reduction in the risk of BPD. Suboptimal nutritional delivery is often seen in preterm infants with BPD compared to controls.
Collapse
Affiliation(s)
- Miguel Saenz de Pipaon
- Department of Neonatology, Instituto de Investigación Sanitaria, La Paz University Hospital-IdiPAZ (Universidad Autonoma), Madrid, Spain.
| | - Leif D Nelin
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Alison Gehred
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Madelaine E Rossholt
- Division of Paediatric and Adolescent Medicine, Department of Neonatal Intensive Care, Ullevål, Oslo University Hospital, Oslo, Norway
| | - Sissel Moltu
- Division of Paediatric and Adolescent Medicine, Department of Neonatal Intensive Care, Ullevål, Oslo University Hospital, Oslo, Norway.
| | - Chris Van den Akker
- Department of Pediatrics, Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam, Netherlands
| | - Anton H van Kaam
- Department of Pediatrics, Amsterdam UMC, Vrije Universiteit, University of Amsterdam, Emma Children's Hospital, Amsterdam, Netherlands
| | - Ana Sánchez
- Department of Neonatology, Instituto de Investigación Sanitaria, La Paz University Hospital-IdiPAZ (Universidad Autonoma), Madrid, Spain
| | | | - Charles C Roehr
- National Perinatal Epidemiology Unit, Oxford Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
- Faculty of Health Sciences, University of Bristol, Bristol, UK
| | - Virgilio Carnielli
- Division of Neonatology, Mother and Child Department, G. Salesi Children Hospital, Azienda Ospedaliero Universitaria delle, Ancona, Marche, Italy
| |
Collapse
|
24
|
Abushahin A, Hamad SG, Sabouni A, Alomar S, Sudarsanan A, Kammouh H, Chandra P. Incidence and Predictors of Bronchopulmonary Dysplasia Development and Severity Among Preterm Infants Born at 32 Weeks of Gestation or Less. Cureus 2024; 16:e59425. [PMID: 38694676 PMCID: PMC11061581 DOI: 10.7759/cureus.59425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 05/04/2024] Open
Abstract
Background As the most common chronic lung disease (CLD) related to premature birth, bronchopulmonary dysplasia (BPD) is associated with long-term lung disease along with cardiovascular and neurodevelopmental disorders. However, data on the incidence and predictors of BPD in Qatar are lacking. Objectives In this study, we aimed to determine the incidence of BPD among infants born at ≤ 32 weeks gestational age (GA) at our neonatal unit, and identify risk factors for the development of BPD and moderate-severe BPD. Methods This was a retrospective observational cohort study conducted at a single site: a level-III neonatal intensive care unit (NICU) in Qatar. We included 1539 neonates born at ≤ 32 weeks of gestation with birth weights of ≤ 1500 grams who were admitted to the NICU between 2017 and 2020. Univariate and multivariate logistic regression analyses were performed to identify potential factors and predictors and their possible associations with the development of BPD and moderate-severe BPD. We also applied BPD classifications to determine the variability in the incidence of BPD in our cohort according to various definitions (2001 National Institute of Child Health and Human Development (NICHD) Diagnostic Criteria, 2016 Revisions of NICHD Criteria, and 2019 Neonatal Research Network Jensen Grading). Results A total of 451 infants (29.3%) had BPD (BPD group) while 1088 (70.7%) did not (non-BPD group), and the overall incidence of BPD was 29.3%. The most relevant risk factors associated with a higher risk of developing BPD identified in the multivariate logistic regression analysis were appropriate weight for gestational age (adjusted OR (aOR) 3.67, 95%CI 2.02-6.67, P < 0.001), presence of patent ductus arteriosus (PDA) (aOR 2.61, 95%CI 1.86-3.66, P < 0.001), late-onset sepsis (aOR 2.16; 95%CI 1.29-3.62; P = 0.003), and use of invasive ventilation (aOR 1.90; 95%CI 1.35-2.69; P < 0.001). The most relevant independent risk factors associated with a higher risk for developing moderate-severe BDP were postnatal steroids (aOR 7.12, 95%CI 3.77-13.44, P < 0.001), use of inhaled nitric oxide (aOR 3.65, 95%CI 1.48-9.01, P = 0.005), use of invasive ventilation (aOR 2.13, 95%CI 1.13-4.00, P = 0.019), late-onset sepsis (aOR 2.07, 95%CI 1.10-3.91, P = 0.025), and male sex (aOR 2.04, 95%CI 1.24-3.36, P = 0.005). The difference in the distribution of BPD severity across the three different definitions of NICHD was significant (P < 0.001). Conclusion The results of this study showed that the incidence of BPD remained high in infants born at ≤ 32 weeks of gestational age and birth weight <1500 g with appropriate weight for gestational age. The presence of PDA at birth or first echocardiography, late-onset sepsis, and use of invasive ventilation were significant risk factors for the incidence of BPD. The identification of risk factors will contribute to the implementation of lung-protective strategies for at-risk infants who may benefit from potential preventive therapy.
Collapse
Affiliation(s)
| | - Sara G Hamad
- Pediatric Pulmonology, Hamad Medical Corporation, Doha, QAT
| | - Amal Sabouni
- Neonatology, Hamad Medical Corporation, Doha, QAT
| | - Sufwan Alomar
- Pediatric Pulmonology, Hamad Medical Corporation, Doha, QAT
| | | | - Hiba Kammouh
- Pediatric Pulmonology, Hamad Medical Corporation, Doha, QAT
| | - Prem Chandra
- Medical Research Center, Hamad Medical Corporation, Doha, QAT
| |
Collapse
|
25
|
Perdijk O, Azzoni R, Marsland BJ. The microbiome: an integral player in immune homeostasis and inflammation in the respiratory tract. Physiol Rev 2024; 104:835-879. [PMID: 38059886 DOI: 10.1152/physrev.00020.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/07/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
The last decade of microbiome research has highlighted its fundamental role in systemic immune and metabolic homeostasis. The microbiome plays a prominent role during gestation and into early life, when maternal lifestyle factors shape immune development of the newborn. Breast milk further shapes gut colonization, supporting the development of tolerance to commensal bacteria and harmless antigens while preventing outgrowth of pathogens. Environmental microbial and lifestyle factors that disrupt this process can dysregulate immune homeostasis, predisposing infants to atopic disease and childhood asthma. In health, the low-biomass lung microbiome, together with inhaled environmental microbial constituents, establishes the immunological set point that is necessary to maintain pulmonary immune defense. However, in disease perturbations to immunological and physiological processes allow the upper respiratory tract to act as a reservoir of pathogenic bacteria, which can colonize the diseased lung and cause severe inflammation. Studying these host-microbe interactions in respiratory diseases holds great promise to stratify patients for suitable treatment regimens and biomarker discovery to predict disease progression. Preclinical studies show that commensal gut microbes are in a constant flux of cell division and death, releasing microbial constituents, metabolic by-products, and vesicles that shape the immune system and can protect against respiratory diseases. The next major advances may come from testing and utilizing these microbial factors for clinical benefit and exploiting the predictive power of the microbiome by employing multiomics analysis approaches.
Collapse
Affiliation(s)
- Olaf Perdijk
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Rossana Azzoni
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Yaremenko AV, Pechnikova NA, Porpodis K, Damdoumis S, Aggeli A, Theodora P, Domvri K. Association of Fetal Lung Development Disorders with Adult Diseases: A Comprehensive Review. J Pers Med 2024; 14:368. [PMID: 38672994 PMCID: PMC11051200 DOI: 10.3390/jpm14040368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Fetal lung development is a crucial and complex process that lays the groundwork for postnatal respiratory health. However, disruptions in this delicate developmental journey can lead to fetal lung development disorders, impacting neonatal outcomes and potentially influencing health outcomes well into adulthood. Recent research has shed light on the intriguing association between fetal lung development disorders and the development of adult diseases. Understanding these links can provide valuable insights into the developmental origins of health and disease, paving the way for targeted preventive measures and clinical interventions. This review article aims to comprehensively explore the association of fetal lung development disorders with adult diseases. We delve into the stages of fetal lung development, examining key factors influencing fetal lung maturation. Subsequently, we investigate specific fetal lung development disorders, such as respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), congenital diaphragmatic hernia (CDH), and other abnormalities. Furthermore, we explore the potential mechanisms underlying these associations, considering the role of epigenetic modifications, transgenerational effects, and intrauterine environmental factors. Additionally, we examine the epidemiological evidence and clinical findings linking fetal lung development disorders to adult respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and other respiratory ailments. This review provides valuable insights for healthcare professionals and researchers, guiding future investigations and shaping strategies for preventive interventions and long-term care.
Collapse
Affiliation(s)
- Alexey V. Yaremenko
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
| | - Nadezhda A. Pechnikova
- Laboratory of Chemical Engineering A’, School of Chemical Engineering, Faculty of Engineering, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (N.A.P.); (A.A.)
- Saint Petersburg Pasteur Institute, Saint Petersburg 197101, Russia
| | - Konstantinos Porpodis
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
| | - Savvas Damdoumis
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
| | - Amalia Aggeli
- Laboratory of Chemical Engineering A’, School of Chemical Engineering, Faculty of Engineering, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (N.A.P.); (A.A.)
| | - Papamitsou Theodora
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Kalliopi Domvri
- Oncology Unit, Pulmonary Department, George Papanikolaou Hospital, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (K.P.); (S.D.)
- Laboratory of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
- Pathology Department, George Papanikolaou Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| |
Collapse
|
27
|
Li S, Liang S, Xie S, Chen H, Huang H, He Q, Zhang H, Wang X. Investigation of the miRNA-mRNA Regulatory Circuits and Immune Signatures Associated with Bronchopulmonary Dysplasia. J Inflamm Res 2024; 17:1467-1480. [PMID: 38476468 PMCID: PMC10929271 DOI: 10.2147/jir.s448394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) has become a major cause of morbidity and mortality in preterm infants worldwide, yet its pathogenesis and underlying mechanisms remain poorly understood. The present study sought to explore microRNA-mRNA regulatory networks and immune cells involvement in BPD through a combination of bioinformatic analysis and experimental validation. Methods MicroRNA and mRNA microarray datasets were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed microRNAs (DEMs) were identified in BPD patients compared to control subjects, and their target genes were predicted using miRWalk, miRNet, miRDB, and TargetScan databases. Subsequently, protein-protein interaction (PPI) and functional enrichment analyses were conducted on the target genes. 30 hub genes were screened using the Cytohubba plugin of the Cytoscape software. Additionally, mRNA microarray data was utilized to validate the expression of hub genes and to perform immune infiltration analysis. Finally, real-time PCR (RT-PCR), immunohistochemistry (IHC), and flow cytometry were conducted using a mouse model of BPD to confirm the bioinformatics findings. Results Two DEMs (miR-15b-5p and miR-20a-5p) targeting genes primarily involved in the regulation of cell cycle phase transition, ubiquitin ligase complex, protein serine/threonine kinase activity, and MAPK signaling pathway were identified. APP and four autophagy-related genes (DLC1, PARP1, NLRC4, and NRG1) were differentially expressed in the mRNA microarray dataset. Analysis of immune infiltration revealed significant differences in levels of neutrophils and naive B cells between BPD patients and control subjects. RT-PCR and IHC confirmed reduced expression of APP in a mouse model of BPD. Although the proportion of total neutrophils did not change appreciably, the activation of neutrophils, marked by loss of CD62L, was significantly increased in BPD mice. Conclusion Downregulation of APP mediated by miR-15b-5p and miR-20a-5p may be associated with the development of BPD. Additionally, increased CD62L- neutrophil subset might be important for the immune-mediated injury in BPD.
Collapse
Affiliation(s)
- Sen Li
- Guangzhou Women and Children’s Medical Center, State Key Laboratory of Respiratory Disease and Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Shuling Liang
- Guangdong Provincial Research Center for Child Health, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Shunyu Xie
- Guangzhou Women and Children’s Medical Center, State Key Laboratory of Respiratory Disease and Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Haixia Chen
- Guangzhou Women and Children’s Medical Center, State Key Laboratory of Respiratory Disease and Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Haoying Huang
- Guangzhou Women and Children’s Medical Center, State Key Laboratory of Respiratory Disease and Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Qixin He
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, People’s Republic of China
| | - Huayan Zhang
- Division of Neonatology and Center for Newborn Care, Guangzhou Women and Children’s Medical Center, Guangzhou, Guangdong Province, People’s Republic of China
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Xiaohui Wang
- Guangzhou Women and Children’s Medical Center, State Key Laboratory of Respiratory Disease and Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| |
Collapse
|
28
|
Goryunov K, Ivanov M, Kulikov A, Shevtsova Y, Burov A, Podurovskaya Y, Zubkov V, Degtyarev D, Sukhikh G, Silachev D. A Review of the Use of Extracellular Vesicles in the Treatment of Neonatal Diseases: Current State and Problems with Translation to the Clinic. Int J Mol Sci 2024; 25:2879. [PMID: 38474125 PMCID: PMC10932115 DOI: 10.3390/ijms25052879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Neonatal disorders, particularly those resulting from prematurity, pose a major challenge in health care and have a significant impact on infant mortality and long-term child health. The limitations of current therapeutic strategies emphasize the need for innovative treatments. New cell-free technologies utilizing extracellular vesicles (EVs) offer a compelling opportunity for neonatal therapy by harnessing the inherent regenerative capabilities of EVs. These nanoscale particles, secreted by a variety of organisms including animals, bacteria, fungi and plants, contain a repertoire of bioactive molecules with therapeutic potential. This review aims to provide a comprehensive assessment of the therapeutic effects of EVs and mechanistic insights into EVs from stem cells, biological fluids and non-animal sources, with a focus on common neonatal conditions such as hypoxic-ischemic encephalopathy, respiratory distress syndrome, bronchopulmonary dysplasia and necrotizing enterocolitis. This review summarizes evidence for the therapeutic potential of EVs, analyzes evidence of their mechanisms of action and discusses the challenges associated with the implementation of EV-based therapies in neonatal clinical practice.
Collapse
Affiliation(s)
- Kirill Goryunov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Mikhail Ivanov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Andrey Kulikov
- Medical Institute, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Moscow 117198, Russia;
| | - Yulia Shevtsova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Artem Burov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Yulia Podurovskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Victor Zubkov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Dmitry Degtyarev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Gennady Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Denis Silachev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|
29
|
Sullivan BA, Beam K, Vesoulis ZA, Aziz KB, Husain AN, Knake LA, Moreira AG, Hooven TA, Weiss EM, Carr NR, El-Ferzli GT, Patel RM, Simek KA, Hernandez AJ, Barry JS, McAdams RM. Transforming neonatal care with artificial intelligence: challenges, ethical consideration, and opportunities. J Perinatol 2024; 44:1-11. [PMID: 38097685 PMCID: PMC10872325 DOI: 10.1038/s41372-023-01848-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Artificial intelligence (AI) offers tremendous potential to transform neonatology through improved diagnostics, personalized treatments, and earlier prevention of complications. However, there are many challenges to address before AI is ready for clinical practice. This review defines key AI concepts and discusses ethical considerations and implicit biases associated with AI. Next we will review literature examples of AI already being explored in neonatology research and we will suggest future potentials for AI work. Examples discussed in this article include predicting outcomes such as sepsis, optimizing oxygen therapy, and image analysis to detect brain injury and retinopathy of prematurity. Realizing AI's potential necessitates collaboration between diverse stakeholders across the entire process of incorporating AI tools in the NICU to address testability, usability, bias, and transparency. With multi-center and multi-disciplinary collaboration, AI holds tremendous potential to transform the future of neonatology.
Collapse
Affiliation(s)
- Brynne A Sullivan
- Division of Neonatology, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Kristyn Beam
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Zachary A Vesoulis
- Division of Newborn Medicine, Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA
| | - Khyzer B Aziz
- Division of Neonatology, Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Ameena N Husain
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Lindsey A Knake
- Division of Neonatology, Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Alvaro G Moreira
- Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Thomas A Hooven
- Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Elliott M Weiss
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Treuman Katz Center for Pediatric Bioethics and Palliative Care, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nicholas R Carr
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - George T El-Ferzli
- Division of Neonatology, Department of Pediatrics, Ohio State University, Nationwide Children's Hospital, Columbus, OH, USA
| | - Ravi M Patel
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Kelsey A Simek
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Antonio J Hernandez
- Division of Neonatology, Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - James S Barry
- Division of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ryan M McAdams
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
30
|
Sun Z, Lu H, Yang B, Li M, Ren Y, Shi H, Gao X, Chen X. Montelukast Sodium to Prevent and Treat Bronchopulmonary Dysplasia in Very Preterm Infants: A Quasi-Randomized Controlled Trial. J Clin Med 2023; 12:7745. [PMID: 38137814 PMCID: PMC10744034 DOI: 10.3390/jcm12247745] [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: 10/08/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease in preterm infants and lacks effective methods for prevention and treatment. The aim of this study is to explore the efficacy and safety of montelukast in preventing or treating BPD in preterm infants. The preterm infants with BPD risk factors were divided randomly into a montelukast group and a control group. In the montelukast group, preterm infants were given 1 mg/kg of montelukast sodium daily. There was no placebo in the control group. There was no significant difference in the incidence of moderate or severe BPD between the two groups (31.8% vs. 35%). The duration of respiratory support in the montelukast group was shorter than that in the control group (36.4 ± 12.8 d vs. 43.1 ± 15.9 d, p = 0.037). The pulmonary severity score (PSS) at 21 days of life in the montelukast group was significantly lower than that in the control group (0.56 ± 0.13 vs. 0.62 ± 0.14, p = 0.048). There were no significant differences in the duration of mechanical ventilation, length of stay, hospitalization expenses, or incidence of adverse events. Although montelukast cannot alleviate the severity of BPD, it may shorten the duration of respiratory support and decrease the PSS in very preterm infants. There were no significant adverse drug events associated with montelukast treatment.
Collapse
Affiliation(s)
- Zhongyi Sun
- Department of Neonatology, Xuzhou Central Hospital, Xuzhou Clinical School, Xuzhou Medical University, Xuzhou 221009, China; (Z.S.)
- Department of Pediatrics, The First Affiliation Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hongyan Lu
- Department of Pediatrics, Affiliation Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Bo Yang
- Department of Neonatology, Xuzhou Central Hospital, Xuzhou Clinical School, Xuzhou Medical University, Xuzhou 221009, China; (Z.S.)
| | - Min Li
- Department of Neonatology, Xuzhou Central Hospital, Xuzhou Clinical School, Xuzhou Medical University, Xuzhou 221009, China; (Z.S.)
| | - Yi Ren
- Department of Neonatology, Xuzhou Central Hospital, Xuzhou Clinical School, Xuzhou Medical University, Xuzhou 221009, China; (Z.S.)
| | - Hongshan Shi
- Department of Neonatology, Xuzhou Central Hospital, Xuzhou Clinical School, Xuzhou Medical University, Xuzhou 221009, China; (Z.S.)
| | - Xiangyu Gao
- Department of Neonatology, Xuzhou Central Hospital, Xuzhou Clinical School, Xuzhou Medical University, Xuzhou 221009, China; (Z.S.)
| | - Xiaoqing Chen
- Department of Pediatrics, The First Affiliation Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
31
|
Bollaboina SKY, Urakurva AK, Kamsetti S, Kotha R. A Systematic Review: Is Early Fluid Restriction in Preterm Neonates Going to Prevent Bronchopulmonary Dysplasia? Cureus 2023; 15:e50805. [PMID: 38249238 PMCID: PMC10798906 DOI: 10.7759/cureus.50805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
Preterm birth causes constant challenges, with bronchopulmonary dysplasia (BPD) being a major concern. Immediately after birth, it takes time to establish feeding between the mother and the premature baby. During this time, the telological shifting of fluid from extracellular space to intracellular space will help the baby; this transition should be smooth. Both normal physiologic changes and pathophysiologic events are capable of disrupting this delicate fluid shifting that occurs in very low-birth-weight infants during the first week of life. The immaturity of the renal system and evaporative losses complicate this process. This lack of fluid displacement can be associated with an increased amount of water in the lungs and reduced lung compliance. This can lead to the need for more ventilatory support and a higher oxygen requirement, which, in turn, leads to lung damage. The fluid restriction is also associated with complications such as severe dehydration, intracranial hemorrhage, and bilirubin toxicity. However, the administration of large amounts of fluid and salt is associated with an increased incidence of patent ductus arteriosus, BPD, necrotizing enterocolitis, and intraventricular hemorrhage. There were studies conducted in both the pre-surfactant and surfactant eras that were inconclusive regarding fluid restriction in BPD. We only included very recent studies. This systematic review attempts to summarize the current evidence, focusing on the efficacy and safety of early fluid management in preterm infants. This reduces the risk of BPD and improves outcomes for premature infants. As we know, intact survival is very important. Our review supported the early fluid restriction.
Collapse
Affiliation(s)
| | | | - Saritha Kamsetti
- Pediatrics, Government Medical College Vikarabad, Vikarabad, IND
| | - Rakesh Kotha
- Neonatology, Osmania Medical College, Hyderabad, IND
| |
Collapse
|
32
|
Wang M, Zhang F, Ning X, Wu C, Zhou Y, Gou Z, Fan Y, Duan R, Li Z, Shao C, Lu L. Regulating NLRP3 Inflammasome-Induced Pyroptosis via Nrf2: TBHQ Limits Hyperoxia-Induced Lung Injury in a Mouse Model of Bronchopulmonary Dysplasia. Inflammation 2023; 46:2386-2401. [PMID: 37556072 PMCID: PMC10673969 DOI: 10.1007/s10753-023-01885-4] [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: 03/22/2023] [Revised: 07/08/2023] [Accepted: 07/31/2023] [Indexed: 08/10/2023]
Abstract
Nuclear factor e2-related factor 2 (Nrf2) plays a key role in cellular resistance to oxidative stress injury. Oxidative stress injury, caused by Nrf2 imbalance, results in increased pyroptosis, DNA damage, and inflammatory activation, which may lead to the arrest of alveolar development and bronchopulmonary dysplasia (BPD) in premature infants under hyperoxic conditions. We established a BPD mouse model to investigate the effects of tert-butylhydroquinone (TBHQ), an Nrf2 activator, on oxidative stress injury, pyroptosis, NLRP3 inflammasome activation, and alveolar development. TBHQ reduced abnormal cell death in the lung tissue of BPD mice and restored the number and normal structure of the alveoli. TBHQ administration activated the Nrf2/heme oxygenase-1 (HO-1) signaling pathway, resulting in the decrease in the following: reactive oxygen species (ROS), activation of the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, and IL-18 and IL-1β expression and activation, as well as inhibition of pyroptosis. In contrast, after Nrf2 gene knockout in BPD mice, there was more severe oxidative stress injury and cell death in the lungs, there were TUNEL + and NLRP3 + co-positive cells in the alveoli, the pyroptosis was significantly increased, and the development of alveoli was significantly blocked. We demonstrated that TBHQ may promote alveolar development by enhancing Nrf2-induced antioxidation in the lung tissue of BPD mice and that the decrease in the NLRP3 inflammasome and pyroptosis caused by Nrf2 activation may be the underlying mechanism. These results suggest that TBHQ is a promising treatment for lung injury in premature infants with hyperoxia.
Collapse
Affiliation(s)
- Minrong Wang
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Feng Zhang
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Xuemei Ning
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Chan Wu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Yue Zhou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Zhixian Gou
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Yang Fan
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Rongrong Duan
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Zhongni Li
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Chunyan Shao
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China
| | - Liqun Lu
- Department of Pediatrics, The First Affiliated Hospital of Chengdu Medical College, No. 278, Middle Section of Baoguang Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China.
- Clinic Medical College, Chengdu Medical College, No. 783 Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, People's Republic of China.
| |
Collapse
|
33
|
Rodrigues I, Soares H, Rocha G, Azevedo I. Impact of advanced maternal age on neonatal morbidity: a systematic review. J Matern Fetal Neonatal Med 2023; 36:2287981. [PMID: 38016703 DOI: 10.1080/14767058.2023.2287981] [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: 04/07/2022] [Accepted: 11/21/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVE This systematic review aimed to understand the impact of advanced maternal age (AMA) on the neonatal morbidity, based on the available scientific evidence. METHODS A systematic search was conducted on 22 November 2021, using the PubMed and Scopus databases to identify studies that compared the morbidity of neonates delivered to AMA mothers with that of neonates delivered to non-AMA mothers. RESULTS Sixteen studies that evaluated the effect of AMA on the neonatal morbidity were included in this review. Nine of these studies found some association between AMA and increased neonatal morbidity (with two of them only reporting an increase in asymptomatic hypoglycemia, and one only reporting an association in twins), six found no association between AMA and neonatal morbidity and one study found a decrease in morbidity in preterm neonates. The studies that found an increase in overall neonatal morbidity with AMA considered older ages for the definition of AMA, particularly ≥40 and ≥45 years. CONCLUSION The current evidence seems to support a lack of association between AMA and the neonatal morbidity of the delivered neonates. However, more studies focusing on the neonatal outcomes of AMA pregnancies are needed to better understand this topic.
Collapse
Affiliation(s)
- Inês Rodrigues
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Henrique Soares
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Neonatology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Gustavo Rocha
- Department of Neonatology, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Inês Azevedo
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Neonatology, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
34
|
Lan J, Chen X, Xu F, Tao F, Liu L, Cheng R, Li N, Pan Y. Self-assembled miR-134-5p inhibitor nanoparticles ameliorate experimental bronchopulmonary dysplasia (BPD) via suppressing ferroptosis. Mikrochim Acta 2023; 190:491. [PMID: 38030848 PMCID: PMC10687138 DOI: 10.1007/s00604-023-06069-3] [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: 07/21/2023] [Accepted: 10/22/2023] [Indexed: 12/01/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in premature infants with increased levels of reactive oxygen species (ROS) and ferroptosis. Herein, we designed a peptide-based nanoparticle to deliver therapeutic molecules to pulmonary, thereby ameliorating BPD. The BPD-induced damages of lung tissues were detected by H&E and immunohistochemistry staining. Inflammatory cytokines, Fe2+, and ROS levels were quantified by the indicated kits, respectively. The targeting relationship was verified by luciferase reporter assay and pull-down assay. Subsequently, self-assembled miR-134-5p inhibitor nanoparticles with pulmonary epithelial cell-targeting were synthesized. The characteristics were detected by transmission electron microscopy, luminescence imaging, and dynamic light scattering. A significant ferroptosis was observed in the BPD mice. The protein level of GPX4 was decreased significantly compared to the control group. Constantly, miR-134-5p showed positive regulation on ferroptosis by targeting GPX4. The designed nanoparticles were mainly accumulated in the lung region. Besides, it ameliorated experimental bronchopulmonary dysplasia via suppressing ferroptosis, in vivo and in vitro. Our findings provided a miR-134-5p/GPX4 axis in regulating ferroptosis of BPD and prompted the potential of applying the peptide-based nanoparticle to BPD treatment.
Collapse
Affiliation(s)
- Jiang Lan
- Shenzhen Longhua Maternity and Child Health Care Hospital, Shenzhen, 518000, China
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xu Chen
- Children's Hospital Affiliated to Nanjing Medical University (Nanjing Children's Hospital), Nanjing, 210008, China
| | - Fengdan Xu
- Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, 523325, China
| | - Fangfei Tao
- Children's Hospital Affiliated to Nanjing Medical University (Nanjing Children's Hospital), Nanjing, 210008, China
| | - Liyuan Liu
- Shenzhen Longhua Maternity and Child Health Care Hospital, Shenzhen, 518000, China
| | - Rui Cheng
- Children's Hospital Affiliated to Nanjing Medical University (Nanjing Children's Hospital), Nanjing, 210008, China.
| | - Ning Li
- Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan, 523325, China.
| | - Ya Pan
- Shenzhen Longhua Maternity and Child Health Care Hospital, Shenzhen, 518000, China.
| |
Collapse
|
35
|
Tanaka K, Hayashi R, Ariyama Y, Takahashi N, Namba F. Management of bronchopulmonary dysplasia in Japan: A nationwide survey. Early Hum Dev 2023; 186:105867. [PMID: 37788509 DOI: 10.1016/j.earlhumdev.2023.105867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/05/2023]
Abstract
BACKGROUND The incidence of bronchopulmonary dysplasia (BPD) and respiratory management practices for extremely low birth weight infants (ELBWIs) widely vary among institutions and countries. AIMS To clarify the variation and characteristics of the current practices of Japanese neonatologists managing patients with BPD. STUDY DESIGN Questionnaire-based survey. PARTICIPANTS Level II and III perinatal centers certified by the Japan Society of Perinatal and Neonatal Medicine. OUTCOME MEASURES Policies of the neonatal intensive care units (NICUs) regarding respiratory care and medications for BPD prevention and treatment. RESULTS A total of 76 % of facilities (207/274) responded to our survey. The response rates of level III and II facilities were 91 % (102/112) and 35 % (105/296), respectively. INtubation-SURfactant-Extubation and Less Invasive Surfactant Administration methods were performed in 23 % (47/206) and 1 % (3/206) of facilities, respectively. For the prophylactic purpose, systemic and inhaled steroids were administered "frequently" or "occasionally" in 14 % (28/205) and 42 % (86/204) of NICUs, respectively. For the therapeutic purpose, systemic and inhaled steroids were administered "frequently" or "occasionally" in 84 % (171/204) and 29 % (59/204) of NICUs, respectively. Approximately half of the NICUs (99/202) used volume-targeted ventilation (VTV) "frequently" or "occasionally" in progressing BPD. High-frequency oscillation ventilation (HFOV) was used for progressing BPD "frequently" and "occasionally" in 89 % (180/202) of the facilities. CONCLUSIONS Our study provided an overview and characteristics of BPD management in Japan in recent years. Noninvasive approaches with surfactant administration remain not widely used in Japan. HFOV is a widely accepted management for progressing BPD.
Collapse
Affiliation(s)
- Kosuke Tanaka
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan.
| | - Ryo Hayashi
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| | - Yuta Ariyama
- Department of Neonatology, Tokyo Metropolitan Children's Medical Center, Fuchu, Tokyo, Japan
| | - Naoto Takahashi
- Department of Pediatrics, The University of Tokyo Hospital, Bunkyo, Tokyo, Japan
| | - Fumihiko Namba
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| |
Collapse
|
36
|
Xu YP, Chen Z, Dorazio RM, Bai GN, Du LZ, Shi LP. Risk factors for bronchopulmonary dysplasia infants with respiratory score greater than four: a multi-center, prospective, longitudinal cohort study in China. Sci Rep 2023; 13:17868. [PMID: 37857836 PMCID: PMC10587148 DOI: 10.1038/s41598-023-45216-x] [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: 05/23/2023] [Accepted: 10/17/2023] [Indexed: 10/21/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common complication of prematurity involving both pre- and post-natal factors. A large, prospective, longitudinal cohort study was conducted to determine whether inflammation-related factors are associated with an increased risk of BPD in preterm infants who were born at a gestational age < 32 weeks, < 72 h after birth and respiratory score > 4. The study included infants from 25 participating hospitals in China between March 1, 2020 and March 31, 2022. The primary outcomes were BPD and severity of BPD at 36 weeks post-menstrual age. A total of 1362 preterm infants were enrolled in the study. After exclusion criteria, the remaining 1088 infants were included in this analysis, of whom, 588 (54.0%) infants were in the BPD group and 500 (46.0%) were in the non-BPD group. In the BPD III model, the following six factors were identified: birth weight (OR 0.175, 95% CI 0.060-0.512; p = 0.001), surfactant treatment (OR 8.052, 95% CI 2.658-24.399; p < 0.001), mean airway pressure (MAP) ≥ 12 cm H2O (OR 3.338, 95% CI 1.656-6.728; p = 0.001), late-onset sepsis (LOS) (OR 2.911, 95% CI 1.514-5.599; p = 0.001), ventilator-associated pneumonia (VAP) (OR 18.236, 95% CI 4.700-70.756; p < 0.001) and necrotizing enterocolitis (NEC) (OR 2.725, 95% CI 1.182-6.281; p = 0.019). Premature infants remained at high risk of BPD and with regional variation. We found that post-natal inflammation-related risk factors were associated with an increased risk of severe BPD, including LOS, VAP, NEC, MAP ≥ 12 cm H2O and use of surfactant.
Collapse
Affiliation(s)
- Yan-Ping Xu
- NICU, Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, China
| | - Zheng Chen
- NICU, Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, China
| | - Robert M Dorazio
- Clinical Research Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Guan-Nan Bai
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Li-Zhong Du
- NICU, Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, China
| | - Li-Ping Shi
- NICU, Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, 3333 Binsheng Road, Hangzhou, 310052, China.
| |
Collapse
|
37
|
Zhang Z, Chen K, Pan D, Liu T, Hang C, Ying Y, He J, Lv Y, Ma X, Chen Z, Liu L, Zhu J, Du L. A predictive model for preterm infants with bronchopulmonary dysplasia based on ferroptosis-related lncRNAs. BMC Pulm Med 2023; 23:367. [PMID: 37784105 PMCID: PMC10544375 DOI: 10.1186/s12890-023-02670-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/22/2023] [Indexed: 10/04/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the most challenging chronic lung disease for prematurity, with difficulties in early identification. Given lncRNA emerging as a novel biomarker and the regulator of ferroptosis, this study aims to develop a BPD predictive model based on ferroptosis-related lncRNAs (FRLs). METHODS Using a rat model, we firstly explored mRNA levels of ferroptosis-related genes and ferrous iron accumulation in BPD rat lungs. Subsequently, a microarray dataset of umbilical cord tissue from 20 preterm infants with BPD and 34 preterm infants without BPD were downloaded from the Gene Expression Omnibus databases. Random forest and LASSO regression were conducted to identify diagnostic FRLs. Nomogram was used to construct a predictive BPD model based on the FRLs. Finally, umbilical cord blood lymphocytes of preterm infants born before 32 weeks gestational age and term infants were collected and determined the expression level of diagnostic FRLs by RT-qPCR. RESULTS Increased iron accumulation and several dysregulated ferroptosis-associated genes were found in BPD rat lung tissues, indicating that ferroptosis was participating in the development of BPD. By exploring the microarray dataset of preterm infants with BPD, 6 FRLs, namely LINC00348, POT1-AS1, LINC01103, TTTY8, PACRG-AS1, LINC00691, were determined as diagnostic FRLs for modeling. The area under the receiver operator characteristic curve of the model was 0.932, showing good discrimination of BPD. In accordance with our analysis of microarray dataset, the mRNA levels of FRLs were significantly upregulated in umbilical cord blood lymphocytes from preterm infants who had high risk of BPD. CONCLUSION The incorporation of FRLs into a predictive model offers a non-invasive approach to show promise in improving early detection and management of this challenging chronic lung disease in premature infant, enabling timely intervention and personalized treatment strategies.
Collapse
Affiliation(s)
- Ziming Zhang
- Neonatal Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Kewei Chen
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Dandan Pan
- Department of Neonatology, Guiyang Maternal and Child Health Care Hospital, Guiyang, China
| | - Tieshuai Liu
- Department of Anesthesiology, Sir Run Run Shaw Hospital School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengcheng Hang
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuhan Ying
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jia He
- Teaching Experimental Center of Public Health, Zhejiang University, Hangzhou, China
| | - Ying Lv
- Department and Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaolu Ma
- Neonatal Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zheng Chen
- Neonatal Intensive Care Unit, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ling Liu
- Department of Neonatology, Guiyang Maternal and Child Health Care Hospital, Guiyang, China
| | - Jiajun Zhu
- Department of Neonatology, Women's Hospital School of Medicine, Zhejiang University, Key Laboratory& Women's Hospital, Hangzhou, China.
| | - Lizhong Du
- Department of Neonatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
38
|
Li H, Ma K, Dou H, Liu L, Qian Y, Li S, Chen J, Han S, Gu X, Yin J. CircABPD1 alleviates oxidative lung injury of bronchopulmonary dysplasia through regulating miR-330-3p/HIF1α axis. Int J Biochem Cell Biol 2023; 163:106464. [PMID: 37660980 DOI: 10.1016/j.biocel.2023.106464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
In the NICU, bronchopulmonary dysplasia (BPD) is a concerning common respiratory complication in preterm and low birth-weight infants. Clinical studies have confirmed that human milk has an important nutritional role for children with BPD, therefore, dentification of beneficial components in human milk that prevent BPD is urgently needed. Our previous work showed that human milk exosomes (HM-Exos) could inhibit apoptosis of alveolar type II epithelial cells (AT II), and the circular RNA (circRNA)-circABPD1 were highly expressed in preterm colostrum milk exosomes. Exosomes transport circRNAs that are stable and may exert anti-inflammatory and immune effects attracted the attention of researchers, but the role and mechanism of human milk exosome-derived circABPD1 in BPD remains unclear. Here, we constructed BPD in vivo and in vitro models through exposure to hyperoxia, verified the effect of circABPD1 and revealed its mechanism through rescue experiments. We found that circABPD1 had circRNA properties, and overexpression of circABPD1 could improve reduced alveolar number, enlarged the alveolar linear intercept in vivo models of BPD, promote cell proliferation, reduce oxidative stress levels and alleviate lung epithelial cell damage in vivo and in vitro models. Mechanistically, circABPD1 targets miR-330-3p and regulates the expression of HIF1α. These results suggest that circABPD1 can improve the pathologoical changes of bronchopulmonary dysplasia, promote cell proliferation, inhibit oxidative stress level, and alleviate lung injury by targeting the miR-330-3p/HIF1α axis, which provides a new idea for the prevention and treatment of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Huimin Li
- School of Pediatrics, Nanjing Medical University, Nanjing, Jiangsu, China; Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Ke Ma
- School of Pediatrics, Nanjing Medical University, Nanjing, Jiangsu, China; Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Heng Dou
- School of Pediatrics, Nanjing Medical University, Nanjing, Jiangsu, China; Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Linjie Liu
- School of Pediatrics, Nanjing Medical University, Nanjing, Jiangsu, China; Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Yun Qian
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Shushu Li
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Jingjing Chen
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Shuping Han
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China.
| | - Xiaoqi Gu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China.
| | - Jing Yin
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China.
| |
Collapse
|
39
|
Hu Z, Liu C, Mao Y, Shi J, Xu J, Zhou G, Jiang F. Integration of transcriptomics reveals ferroptosis-related signatures and immune cell infiltration in bronchopulmonary dysplasia. Heliyon 2023; 9:e21093. [PMID: 37928394 PMCID: PMC10622619 DOI: 10.1016/j.heliyon.2023.e21093] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 09/12/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
Ferroptosis has emerged as a significant factor in the development of bronchopulmonary dysplasia (BPD). Nevertheless, our understanding of the potential involvement of ferroptosis-related genes (FRGs) in BPD remains incomplete. In this study, we leveraged the Gene Expression Omnibus (GEO) database to investigate this aspect. We identified 20 differentially expressed FRGs that are associated with BPD, shedding light on their potential role in the condition.LASSO along with SVM-RFE algorithms found that 12 genes: MEG3, ACSL1, DPP4, GALNT14, MAPK14, CD82, SMPD1, NR1D1, PARP3, ACVR1B, H19, and SLC7A11 were closely related to ferroptosis modulation and immunological response. These genes were used to create a nomogram with good predictive power and were found to be involved in BPD-linked pathways. In addition, the marker genes-based prediction model performed well in external validation data sets. The study also showed a significance between BPD and control samples in terms of immune cell infiltration. These findings may help improve our understanding of FRGs in BPD and lead to the development of more effective immunotherapies.
Collapse
Affiliation(s)
- Zhengyun Hu
- Department of Pediatrics, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai, China
| | - Chong Liu
- Department of Pediatrics, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (Preparatory Stage), Shanghai, China
| | - Yan Mao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianwei Shi
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jinwen Xu
- Department of Pediatric Nephrology, Wuxi Children's Hospital, Wuxi, China
| | - Guoping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
40
|
刘 选, 谷 名, 孙 萌, 江 倩. [Clinical characteristics and pathogens of infancy lower respiratory tract infections in infants with bronchopulmonary dysplasia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:953-958. [PMID: 37718402 PMCID: PMC10511232 DOI: 10.7499/j.issn.1008-8830.2304076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVES To study the clinical characteristics and pathogen features of infants with bronchopulmonary dysplasia (BPD) who were readmitted during infancy due to lower respiratory tract infections. METHODS A retrospective analysis was conducted on 128 preterm infants with BPD who were admitted for lower respiratory tract infections in Qingdao Women and Children's Hospital from January 2020 to December 2022. An equal number of non-BPD preterm infants admitted during the same period were selected as controls. General information, clinical characteristics, lung function parameters, and respiratory pathogen results were compared between the two groups. RESULTS Compared with the non-BPD group, the BPD group had a lower gestational age and birth weight, were more likely to experience shortness of breath, wheezing, and cyanosis, and had a longer duration of wheezing relief (P<0.05). Compared with the non-BPD group, the BPD group had lower lung function parameters, including tidal volume per kilogram of body weight, ratio of time to peak tidal expiratory flow to total expiratory time, ratio of volume at peak tidal expiratory flow to expiratory tidal volume, tidal expiratory flow at 25%, 50%, and 75% of tidal volume, and increased respiratory rate (P<0.05). The detection rates of gram-negative bacteria, such as Klebsiella pneumoniae and Acinetobacter baumannii, were higher in the BPD group than in the non-BPD group (P<0.05). CONCLUSIONS Infants with BPD who develop infancy lower respiratory tract infections require closer attention to the clinical characteristics such as shortness of breath, wheezing, and cyanosis. Lung function is characterized by obstructive changes and small airway dysfunction. Gram-negative bacteria, including Klebsiella pneumoniae and Acinetobacter baumannii, are more likely to be detected as respiratory pathogens.
Collapse
|
41
|
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
|
42
|
Lu X, Gao Y, Liu C, Pan M, Chen X. Effect of Breast Milk on the Frequency of Bronchopulmonary Dysplasia in Very Low Birth Weight Premature Infants: A Meta-analysis. Breastfeed Med 2023; 18:636-644. [PMID: 37729032 DOI: 10.1089/bfm.2023.0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Purpose: To analyze the effect of different feeding types on bronchopulmonary dysplasia (BPD) in very low birth weight preterm infants. Methods: The Cochrane Library, PubMed, Embase, China National Knowledge Infrastructure (CNKI), Wanfang Data Knowledge Service Platform, China Biomedical Literature Database (CBM) were searched for literature related to breastfeeding and BPD, with a search period from their inception to January 2023. Two researchers independently screened the literature, extracted data, and assessed the quality of included studies before analyzing the data using Stata16 and RevMan5.4.1 software. Results: A total of 17 studies were included. The results showed that there was no significant difference in the frequency of BPD between human milk (HM) and donor human milk (DHM) (OR = 0.54, 95% CI: 0.29-1.03, p = 0.07). However, DHM had a significant effect in reducing the frequency of BPD compared to preterm formula (PF) (OR = 0.62, 95% CI: 0.41-0.94, p = 0.02). Exclusive HM also had a significant effect in reducing the frequency of BPD compared to exclusive PF (OR = 0.51, 95% CI: 0.34-0.78, p = 0.002), as well as compared to any PF (OR = 0.57, 95% CI: 0.37-0.88, p = 0.01). Furthermore, mainly (>50%) HM had a significant effect in reducing the frequency of BPD compared to mainly PF (OR = 0.72, 95% CI: 0.55-0.93, p = 0.01). However, there was no statistically significant difference between any HM and exclusive PF (OR = 0.88, 95% CI: 0.62-1.23, p = 0.46). Conclusions: Our study findings suggest that both HM and DHM have a significant protective effect in reducing the frequency of BPD occurrence compared to PF. Furthermore, even when the amount of HM is insufficient, feeding more than 50% of the HM volume still provides a protective effect against the frequency of BPD. Therefore, we recommend feeding infants with more than 50% of HM to harness the protective effect of HM against BPD occurrence.
Collapse
Affiliation(s)
- Xiaoyu Lu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Gao
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chuntian Liu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengqing Pan
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaochun Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Leopoldino RWD, Marques DP, Rocha LC, Fernandes FEM, Oliveira AG, Martins RR. Temporal profile of adverse drug reactions and associated clinical factors: a prospective observational study in a neonatal intensive care unit. BMJ Open 2023; 13:e073304. [PMID: 37553191 PMCID: PMC10414078 DOI: 10.1136/bmjopen-2023-073304] [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: 03/02/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023] Open
Abstract
OBJECTIVE Although adverse drug reactions (ADRs) are quite common in hospitalised neonates, pharmacovigilance activities in this public are still incipient. This study aims to characterise ADRs in neonates in a neonatal intensive care unit (NICU), identifying causative drugs, temporal profile and associated factors. DESIGN Prospective observational study. SETTING NICU of a public maternity hospital in Natal/Brazil. PARTICIPANTS All neonates admitted to the NICU for more than 24 hours and using at least one medication were followed up during the time of hospitalisation. PRIMARY OUTCOME MEASURES Incidence rate and risk factors for ADRs. The ADRs were detected by an active search in electronic medical records and analysis of spontaneous reports in the hospital pharmacovigilance system. RESULTS Six hundred neonates were included in the study, where 118 neonates had a total of 186 ADRs. The prevalence of ADRs at the NICU was 19.7% (95% CI 16.7% to 23.0%). The most common ADRs were tachycardia (30.6%), polyuria (9.1%) and hypokalaemia (8.6%). Tachycardia (peak incidence rate: 57.1 ADR/1000 neonates) and hyperthermia (19.1 ADR/1000 neonates) predominated during the first 5 days of hospitalisation. The incidence rate of polyuria and hypokalaemia increased markedly after the 20th day, with both reaching a peak of 120.0 ADR/1000 neonates. Longer hospitalisation time (OR 0.018, 95% CI 0.007 to 0.029; p<0.01) and number of prescribed drugs (OR 0.127, 95% CI 0.075 to 0.178; p<0.01) were factors associated with ADRs. CONCLUSION ADRs are very common in NICU, with tachycardia and hyperthermia predominant in the first week of hospitalisation and polyuria and hypokalaemia from the third week onwards.
Collapse
Affiliation(s)
- Ramon Weyler Duarte Leopoldino
- Graduate Program of Pharmaceutical Science, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Daniel Paiva Marques
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Luan Carvalho Rocha
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Flavia Evelyn Medeiros Fernandes
- School Maternity Januário Cicco (MEJC/EBSERH), Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Antonio Gouveia Oliveira
- Graduate Program of Pharmaceutical Science, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| | - Rand Randall Martins
- Graduate Program of Pharmaceutical Science, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
- Department of Pharmacy, Federal University of Rio Grande do Norte, Center for Health Sciences, Natal, Brazil
| |
Collapse
|
44
|
Verd S, Porta R, Ginovart G, Avila-Alvarez A, García-Muñoz Rodrigo F, Izquierdo Renau M, Ventura PS. Human Milk Feeding Is Associated with Decreased Incidence of Moderate-Severe Bronchopulmonary Dysplasia in Extremely Preterm Infants. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1267. [PMID: 37508764 PMCID: PMC10378323 DOI: 10.3390/children10071267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND An increased rate of bronchopulmonary dysplasia (BPD) is reported in extremely preterm infants. A potential role of human milk feeding in protecting against this condition has been suggested. METHODS A retrospective descriptive study was conducted based on data about morbidity in the population of infants born between 22+0 and 26+6 weeks of gestation, included in the Spanish network SEN1500 during the period 2004-2019 and discharged alive. The primary outcome was moderate-severe BPD. Associated conditions were studied, including human milk feeding at discharge. The temporal trends of BPD and human milk feeding rates at discharge were also studied. RESULTS In the study population of 4341 infants, the rate of moderate-severe BPD was 43.7% and it increased to >50% in the last three years. The factors significantly associated with a higher risk of moderate-severe BPD were birth weight, male sex, high-frequency oscillatory ventilation, duration of invasive mechanical ventilation, inhaled nitric oxide, patent ductus arteriosus, and late-onset sepsis. Exclusive human milk feeding and any amount of human milk at discharge were associated with a lower incidence of moderate-severe BPD (OR 0.752, 95% CI 0.629-0.901 and OR 0.714, 95% CI 0.602-0.847, respectively). During the study period, the proportion of infants with moderate-severe BPD fed any amount of human milk at discharge increased more than twofold. And the proportion of infants with moderate-severe BPD who were exclusively fed human milk at discharge increased at the same rate. CONCLUSIONS Our work shows an inverse relationship between human milk feeding at discharge from the neonatal unit and the occurrence of BPD.
Collapse
Affiliation(s)
- Sergio Verd
- Department of Pediatric Care Primary Unit, La Vileta Surgery, Health Sciences Research Institute (IUNICS), Balearic University, 07122 Palma, Spain
| | - Roser Porta
- Department of Pediatrics, Division of Neonatology, Hospital Universitari Germans Trias i Pujol, Badalona, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Gemma Ginovart
- Department of Pediatrics, Division of Neonatology, Hospital Universitari Germans Trias i Pujol, Badalona, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Alejandro Avila-Alvarez
- Division of Neonatology, Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, 15006 A Coruña, Spain
| | - Fermín García-Muñoz Rodrigo
- Division of Neonatology, Complejo Hospitalario Universitario Insular Materno-Infantil, 35016 Las Palmas, Spain
| | | | - Paula Sol Ventura
- Department of Pediatrics, Division of Neonatology, Hospital Universitari Germans Trias i Pujol, Badalona, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
45
|
Rosenboom I, Pust MM, Pirr S, Bakker A, Willers M, Davenport CF, Wiehlmann L, Viemann D, Tümmler B. Longitudinal development of the airway metagenome of preterm very low birth weight infants during the first two years of life. ISME COMMUNICATIONS 2023; 3:75. [PMID: 37474785 PMCID: PMC10359316 DOI: 10.1038/s43705-023-00285-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
Preterm birth is accompanied with many complications and requires severe therapeutic regimens at the neonatal intensive care unit. The influence of the above-mentioned factors on the premature-born infants' respiratory metagenome or more generally its maturation is unknown. We therefore applied shotgun metagenome sequencing of oropharyngeal swabs to analyze the airway metagenome development of 24 preterm infants from one week postpartum to 15 months of age. Beta diversity analysis revealed a distinct clustering of airway microbial communities from hospitalized preterms and samples after hospital discharge. At nine and 15 months of age, the preterm infants lost their hospital-acquired individual metagenome signatures towards a common taxonomic structure. However, ecological network analysis and Random Forest classification of cross-sectional data revealed that by this age the preterm infants did not succeed in establishing the uniform and stable bacterial community structures that are characteristic for healthy full-term infants.
Collapse
Affiliation(s)
- Ilona Rosenboom
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Marie-Madlen Pust
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Sabine Pirr
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Alina Bakker
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Maike Willers
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Colin F Davenport
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Lutz Wiehlmann
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Dorothee Viemann
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Translational Pediatrics, Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Center for Infection Research, University Würzburg, Würzburg, Germany
| | - Burkhard Tümmler
- Department for Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
46
|
Xu L, Shi Z, Pan Z, Wu R. METTL3 promotes hyperoxia-induced pyroptosis in neonatal bronchopulmonary dysplasia by inhibiting ATG8-mediated autophagy. Clinics (Sao Paulo) 2023; 78:100253. [PMID: 37478627 PMCID: PMC10387564 DOI: 10.1016/j.clinsp.2023.100253] [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: 02/08/2023] [Revised: 06/07/2023] [Accepted: 07/03/2023] [Indexed: 07/23/2023] Open
Abstract
OBJECTIVES N6-Methyladenosine (m6A) modification plays a vital role in lung disorders. However, the potential of m6A in neonatal Bronchopulmonary Dysplasia (BPD) has not been reported. This study aimed to investigate the roles of METTL3 in BPD. METHODS BPD models were established by hyperoxia in vivo and in vitro. Histological analysis was determined using HE staining. Gene expression was determined using Western blotting, qRT-PCR, and immunofluorescence. The release of IL-1β and IL-18 was detected using ELISA. The m6A sites of ATG8 were predicted by SCRAPM and verified by MeRIP assay. The location of GSDMD and ATG8 was determined by FISH assay. The interaction between ATG8 and GSDMD was detected using Coimmunoprecipitation (Co-IP). Cell pyroptosis was determined using flow cytometry and TUNEL assays. RESULTS METTL3 was overexpressed in BPD, which was accompanied by an increase in m6A levels. Interestingly, METTL3 suppressed hyperoxia-mediated damage and pyroptosis in BEAS-2B cells and promoted cell autophagy. METTL3-mediated m6A modification of ATG8 suppressed its expression and disrupted the interaction between ATG8 and GSDMD. However, autophagy inhibition induced pyroptosis in BEAS-2B cells. In vivo assays showed that METTL3-mediated autophagy inhibition induced a decrease in the radial alveolar count and an increase in the mean linear intercept and promoted cell pyroptosis. CONCLUSION In conclusion, METTL3-mediated cell pyroptosis promotes BPD by regulating the m6A modification of ATG8. This may provide new insight into the development of BPD.
Collapse
Affiliation(s)
- Lili Xu
- Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical College, Neonatal Medical Center, Huai'an, China
| | - Zhan Shi
- Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical College, Neonatal Medical Center, Huai'an, China
| | - Zhaojun Pan
- Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical College, Neonatal Medical Center, Huai'an, China
| | - Rong Wu
- Huai'an Maternity and Child Healthcare Hospital, Yangzhou University Medical College, Neonatal Medical Center, Huai'an, China.
| |
Collapse
|
47
|
He W, Zhang L, Feng R, Fang WH, Cao Y, Sun SQ, Shi P, Zhou JG, Tang LF, Zhang XB, Qi YY. Risk factors and machine learning prediction models for bronchopulmonary dysplasia severity in the Chinese population. World J Pediatr 2023; 19:568-576. [PMID: 36357648 PMCID: PMC10198877 DOI: 10.1007/s12519-022-00635-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 10/07/2022] [Indexed: 11/12/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in extremely preterm neonates. The outcome and clinical burden vary dramatically according to severity. Although some prediction tools for BPD exist, they seldom pay attention to disease severity and are based on populations in developed countries. This study aimed to develop machine learning prediction models for BPD severity based on selected clinical factors in a Chinese population. METHODS In this retrospective, single-center study, we included patients with a gestational age < 32 weeks who were diagnosed with BPD in our neonatal intensive care unit from 2016 to 2020. We collected their clinical information during the maternal, birth and early postnatal periods. Risk factors were selected through univariable and ordinal logistic regression analyses. Prediction models based on logistic regression (LR), gradient boosting decision tree, XGBoost (XGB) and random forest (RF) models were implemented and assessed by the area under the receiver operating characteristic curve (AUC). RESULTS We ultimately included 471 patients (279 mild, 147 moderate, and 45 severe cases). On ordinal logistic regression, gestational diabetes mellitus, initial fraction of inspiration O2 value, invasive ventilation, acidosis, hypochloremia, C-reactive protein level, patent ductus arteriosus and Gram-negative respiratory culture were independent risk factors for BPD severity. All the XGB, LR and RF models (AUC = 0.85, 0.86 and 0.84, respectively) all had good performance. CONCLUSIONS We found risk factors for BPD severity in our population and developed machine learning models based on them. The models have good performance and can be used to aid in predicting BPD severity in the Chinese population.
Collapse
Affiliation(s)
- Wen He
- Department of Respirology, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Lan Zhang
- Department of Neonatology, Children's Hospital, Fudan University, Shanghai, China
| | - Rui Feng
- Shanghai Key Laboratory of Intelligent Information Processing, School of Management and Statistics, Fudan University, Shanghai, China
| | - Wei-Han Fang
- Shanghai Pinghe Bilingual School, Shanghai, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital, Fudan University, Shanghai, China
| | - Si-Qi Sun
- Shanghai Key Laboratory of Intelligent Information Processing, School of Management and Statistics, Fudan University, Shanghai, China
| | - Peng Shi
- Department of Data Management and Statistics, Children's Hospital of Fudan University, Shanghai, China
| | - Jian-Guo Zhou
- Department of Neonatology, Children's Hospital, Fudan University, Shanghai, China
| | - Liang-Feng Tang
- Department of Urology, Children's Hospital, Fudan University, Shanghai, China
| | - Xiao-Bo Zhang
- Department of Respirology, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| | - Yuan-Yuan Qi
- Department of Respirology, Children's Hospital, Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| |
Collapse
|
48
|
Xu RZ, Sun B, Zhao NC. [Association between early parenteral nutrition and the development of bronchopulmonary dysplasia in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:362-367. [PMID: 37073840 PMCID: PMC10120333 DOI: 10.7499/j.issn.1008-8830.2210128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
OBJECTIVES To study the relationship between early parenteral nutrient intake and the development of bronchopulmonary dysplasia (BPD) in preterm infants with gestational age less than 32 weeks who could not receive enteral nutrition within one week after birth. METHODS A retrospective study was conducted on preterm infants born between October 2017 and August 2022 with gestational age less than 32 weeks who were admitted to the Neonatal Intensive Care Unit in Children's Hospital of Soochow University within 24 hours after birth and relied solely on parenteral nutrition within the first week of life. The study population included 79 infants with BPD and 73 infants without BPD. Clinical data during hospitalization were compared between the two groups. RESULTS The proportions of infants with weight loss of more than 10% after birth, extrauterine growth retardation, and parenteral nutrition-associated cholestasis in the BPD group were higher than in the non-BPD group (P<0.05). The time to regain birth weight, time to achieve full enteral feeding, and corrected gestational age at discharge were longer in the BPD group than in the non-BPD group. The Z-scores of physical growth at corrected gestational age of 36 weeks were lower in the BPD group than in the non-BPD group (P<0.05). The BPD group had a higher fluid intake and a lower calories intake in the first week than the non-BPD group (P<0.05). The starting dose and total amount of amino acids, glucose, and lipids in the first week were lower in the BPD group than in the non-BPD group (P<0.05). The BPD group had a higher glucose-to-lipid ratio on the third day and higher energy-to-nitrogen and glucose-to-lipid ratios on the seventh day after birth than the non-BPD group (P<0.05). CONCLUSIONS Preterm infants with BPD had lower intake of amino acids and lipids and a lower proportion of calories provided by amino acids and lipids in the first week of life, which suggests an association between early parenteral nutrition intake and the development of BPD.
Collapse
Affiliation(s)
- Ru-Zheng Xu
- Department of Cardiology, Children's Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Bin Sun
- Department of Cardiology, Children's Hospital of Nanjing Medical University, Nanjing 210000, China
| | | |
Collapse
|
49
|
Han W, Zhang F, Mo D, Zhang X, Chen B, Ding X, Guo H, Li F, Guo C. Involvement of HIF1 stabilization and VEGF signaling modulated by Grx-1 in murine model of bronchopulmonary dysplasia. Cell Biol Int 2023; 47:796-807. [PMID: 36640422 DOI: 10.1002/cbin.11985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/23/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023]
Abstract
Hypoxia inducible factor (HIF)-1α could be stabilized by Grx1 deletion, which is implicated critical in the pathogenesis of bronchopulmonary dysplasia (BPD). Until now, the stabilization of HIF-1α by glutathionylation to regulate the pulmonary microcirculation in BPD is not well addressed. In this study, we investigated whether the HIF-1α stabilization modulated by Grx1 ablation could ameliorate the pathological changes in the mouse model of BPD, including angiogenesis and alveolar formation. We found that depletion of Grx1 increased levels of GSH-protein adducts, which was associated with the improvement in the numbers of alveoli, the capillary density in the pulmonary microcirculation and the survival rate in the littermates with hyperoxic exposure. Grx1 ablation could promote HIF-1α glutathionylation by increasing GSH adducts to stabilize HIF-1α and to induce VEGF-A production in the lung tissue. The above phenotype of capillary density and VEGF-A production was removed by the pharmacological administration of YC-1, the HIF-1α inhibitor, suggesting the HIF-1α dependent manner for pulmonary microcirculatory perfusion. These data indicate that HIF-1α stabilization plays an critical role in modification pulmonary microcirculatory perfusion, which is associated with the pathological damage under hyperoxic conditions, suggesting that targeting with HIF-1α stabilization should be a potential clinical and therapeutic strategy for BPD treatment.
Collapse
Affiliation(s)
- Wenli Han
- School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China.,Department of Animal Center, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Fengmei Zhang
- School of Pharmacy, Chongqing Medical University, Chongqing, People's Republic of China.,Department of Animal Center, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Dandan Mo
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China.,Department of Animal Center, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Zhang
- Department of Animal Center, Children's Hospital of Chongqing Medical University, Chongqing, China.,Department of Burn, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Bailin Chen
- Department of General Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xionghui Ding
- Department of Burn, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongjie Guo
- Department of Animal Center, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Li
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China.,Department of Pediatrics, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China
| | - Chunbao Guo
- Department of Pediatrics, Chongqing Health Center for Women and Children, Chongqing, China.,Department of Pediatrics, Women and Chidren's Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
He F, Wang QF, Li L, Yu C, Liu CZ, Wei WC, Chen LP, Li HY. Melatonin Protects Against Hyperoxia-Induced Apoptosis in Alveolar Epithelial type II Cells by Activating the MT2/PI3K/AKT/ETS1 Signaling Pathway. Lung 2023; 201:225-234. [PMID: 36928143 DOI: 10.1007/s00408-023-00610-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE Hyperoxia-induced apoptosis in alveolar epithelial type II cells (AECIIs) plays a critical role in the development of bronchopulmonary dysplasia (BPD). Melatonin has been shown to improve BPD. However, the protective effect of melatonin on hyperoxia-induced apoptosis in AECIIs and the precise mechanisms involved remain unclear. METHODS Human alveolar epithelial type II A549 cells were treated with hyperoxia as an in vitro model to investigate the antiapoptotic mechanism of melatonin. CCK-8 assays were performed to investigate the viability of A549 cells. Hoechst 33,258 staining was carried out to quantify apoptosis in A549 cells. The protein expression levels of E26 oncogene homolog 1 (ETS1), Bcl-2, Bax, Bim, Wnt, β-catenin, AKT and phosphorylated AKT were measured by western blotting. LY294002, SC79 and the downregulation of ETS1, melatonin receptor 1 (MT1) and MT2 with specific siRNAs were used to investigate the role of the PI3K/AKT pathway, ETS1, MT1 and MT2 in hyperoxia-induced apoptosis in A549 cells. RESULTS Melatonin prevented hyperoxia-induced apoptosis in A549 cells, and the upregulation of E26 oncogene homolog 1 (ETS1) contributed to the antiapoptotic effect of melatonin. Melatonin activated the PI3K/AKT axis, which led to ETS1 upregulation and inhibited apoptosis in hyperoxia-exposed A549 cells. Furthermore, melatonin-induced activation of the PI3K/AKT axis, upregulation of ETS1 and inhibition of apoptosis were reversed by melatonin receptor 2 (MT2) siRNA in hyperoxia-exposed A549 cells. CONCLUSION Melatonin prevents hyperoxia-induced apoptosis by activating the MT2/PI3K/AKT/ETS1 axis in alveolar epithelial cells.
Collapse
Affiliation(s)
- Fan He
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Qiao-Fang Wang
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Lin Li
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Cong Yu
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Chun-Zi Liu
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Wen-Chao Wei
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China
| | - Li-Ping Chen
- Department of Neonatology, Jiangxi Provincial Children's Hospital, No. 1666, DieziHu Road, Honggutan District, Nanchang, 330038, Jiangxi, China.
| | - Huan-Yu Li
- Second Department of Respiratory Disease, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, No. 152, Aiguo Road, DongHu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|