1
|
Zeming KK, Llanora G, Quek K, Goh CR, Ng NZH, Han J, Yeo KT. Whole blood biophysical immune profiling of newborn infants correlates with immune responses. Pediatr Res 2025:10.1038/s41390-025-03952-y. [PMID: 40164874 DOI: 10.1038/s41390-025-03952-y] [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] [Received: 09/07/2024] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND There is a current, absence of reliable, blood-sparing, diagnostic tools to measure and trend real-time changes in the levels of inflammation and its effects on the immune cells in the infant. METHODS We deployed the BiophysicaL Immune Profiling for Infants (BLIPI) system in the neonatal intensive care unit to describe immune cell biophysical profiles using 50 microliters of blood per sample from term and preterm infants. RESULTS A total of 19 infants (8 term, 11 preterm) were recruited and 24 blood samples were collected in their first month. Based on the profiles of immune cells' size and deformation, there was a clear distinction between term and preterm infants, with 48/50 markers significantly different. A preterm infant with late-onset bacterial sepsis had notable size and deformability differences compared to the rest of the preterm cohort. There was a significant correlation between immune cell biophysical profiles and clinical markers such as C-reactive protein, white blood cell counts, and immature-to-total neutrophil (I:T) ratios, with Pearson correlation coefficients for linear regression models of 0.98, 0.97 and 0.94 respectively. CONCLUSION This study highlights the potential for the biophysical immune cell profiling system to provide an overview of the infant's current immune activation and response. IMPACT We present a novel, minimally invasive diagnostic system that leverages the physical properties of immune cells to provide a rapid and direct assessment of the immune status, requiring 20 times less blood volume than standard tests. This study demonstrates the potential of a compact, deployable system that is capable of performing biophysical profiling to assess immune cell activation in term and preterm infants, by revealing distinct differences in cell size and deformation between groups. The system's sensitive, quantitative measures were correlated with routine clinical biomarkers, highlighting its ability to provide a rapid, minimally invasive, real-time monitoring of neonatal immune status.
Collapse
Affiliation(s)
- Kerwin Kwek Zeming
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Genevieve Llanora
- Department of Neonatology, KK Women's & Children's Hospital, Singapore, Singapore
| | - Kaiyun Quek
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Chin Ren Goh
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Nicholas Zhi Heng Ng
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalized Medicine, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Antimicrobial Resistance, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.
| | - Kee Thai Yeo
- Department of Neonatology, KK Women's & Children's Hospital, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
2
|
Li Q, Li C, Rao Y, Han D, Wang X, Yang L, Chen Y, Yang C, Chen X. Dynamic association between plasma interleukin-1 family concentrations and bronchopulmonary dysplasia in extremely premature infants. J Perinatol 2025:10.1038/s41372-025-02275-4. [PMID: 40159578 DOI: 10.1038/s41372-025-02275-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/17/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
OBJECTIVE To evaluate the longitudinal changes of IL-1α, IL-1β, and IL-1Ra in extremely preterm infants and investigate the dynamic association with bronchopulmonary dysplasia (BPD). METHODS Plasma samples were collected from extremely preterm infants at postnatal day (PD) 7,28 and PMA 36 weeks. IL-1 cytokines concentrations were measured by Bio-Plex Pro (human cytokine panel). Univariate and multivariate logistic regression analysis were conducted to explore the association between the cytokines and BPD. RESULTS BPD infants exhibited significantly higher concentrations of IL-1α (10.75 vs. 8.18 pg/ml, p = 0.026), IL-1β (2.00 vs. 1.50 pg/ml, p = 0.046), and IL-1Ra (878.50 vs. 262.40 pg/ml, p = 0.011) compared to non-BPD infants at PD 28. Higher IL-1α concentration (≥8.09 pg/ml) at PD 28 was independently associated with BPD development (OR: 8.272, 95% CI: 1.127-60.705, p = 0.038). CONCLUSIONS Increased IL-1α concentrations at PD 28 were independently associated with an increased risk of BPD.
Collapse
Affiliation(s)
- Qingling Li
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Cuihui Li
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yunbei Rao
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Dongshan Han
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Xuan Wang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Lingling Yang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Yiran Chen
- Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Chuanzhong Yang
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China.
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China.
| | - Xueyu Chen
- Department of Neonatology, Shenzhen Maternity and Child Healthcare Hospital, the First School of Clinical Medicine, Southern Medical University, Shenzhen, China.
- Shenzhen Key Laboratory of Maternal and Child Health and Diseases, Shenzhen, China.
| |
Collapse
|
3
|
Rais H, Pronovost E, Guillot M, Boutin A, Simonyan D, Mohamed I, Lavoie PM, Piedboeuf B, Marc I. Can prenatal conditions impact the effect of omega-3 on bronchopulmonary dysplasia in very preterm infants? A secondary analysis of a randomized controlled trial. Eur J Pediatr 2025; 184:243. [PMID: 40072608 DOI: 10.1007/s00431-025-06053-4] [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: 08/08/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025]
Abstract
To explore whether prenatal conditions (i.e. chorioamnionitis, preeclampsia or small-for-gestational age (SGA)) affect the very preterm infant's response to docosahexaenoic acid (DHA) on bronchopulmonary dysplasia (BPD), according to mode of delivery, an independent factor shown to modulate this association. Secondary exploratory analysis of the MOBYDIck randomized controlled trial (NCT02371460) evaluating the effect of a neonatal high-dose DHA supplementation through maternal breastmilk compared to placebo. Population was preterm infants born before 29 weeks of gestation in sixteen Canadian neonatal intensive care units. Primary outcome was physiological BPD based on pulse oximetry assessment. Secondary outcomes included "death or BPD"; "moderate-or-severe" BPD; severe BPD; death from any causes. Heterogeneity in the effect of DHA on outcomes was assessed by prenatal conditions and mode of delivery using generalized estimating equation logistic regression models. The trial intended to enroll 800 mothers but was stopped early for safety, likely making subgroup analysis underpowered. 230 mothers (271 infants) were included in DHA group and 226 mothers (252 infants) in placebo group. The association between high-dose DHA and BPD differed by chorioamnionitis status (heterogeneity P=0.04). In infants exposed to chorioamnionitis and vaginal delivery, DHA supplementation was associated with a reduced risk of physiological BPD (adjusted odds ratio, 0.18 [95% CI, 0.05 to 0.62], P=0.007). No heterogeneity was found by maternal preeclampsia (heterogeneity P=0.44) nor SGA status (heterogeneity P=0.17). CONCLUSION This secondary analysis generated hypotheses for a potential differential effect of neonatal enteral high-dose DHA supplementation on BPD in very preterm infants according to chorioamnionitis exposure. WHAT IS KNOWN • The MOBYDIck trial reported a potential protective effect of docosahexaenoic acid (DHA) supplementation on bronchopulmonary dysplasia (BPD) in infants born vaginally, but not in those born via cesarean section. • Placenta pathologies are associated with inflammation in the infants and could affect the very preterm infant's response to a high-dose DHA supplementation on BPD according to the mode of delivery. WHAT IS NEW • This study suggests that, in infants born very preterm before 29 weeks of gestation, the association between enteral high-dose DHA supplementation in neonatal period and BPD at 36 weeks' postmenstrual age differ according to the maternal status for chorioamnionitis at delivery.
Collapse
Affiliation(s)
- Hymel Rais
- Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - Etienne Pronovost
- Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - Mireille Guillot
- Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - Amélie Boutin
- Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - David Simonyan
- Clinical and Evaluative Research Platform, Centre de Recherche du CHU de Québec-Université Laval, Québec, Québec, Canada
| | - Ibrahim Mohamed
- Department of Pediatrics, Université de Montréal, CHU Sainte-Justine, Montréal, Québec, Canada
| | - Pascal M Lavoie
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruno Piedboeuf
- Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - Isabelle Marc
- Department of Pediatrics, CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada.
| |
Collapse
|
4
|
Shams SE, Dastgheib SA, Mousavi-Beni SA, Hosein Lookzadeh M, Mirjalili SR, Golshan-Tafti M, Bahrami R, Yeganegi M, Shahbazi A, Masoudi A, Shiri A, Noorishadkam M, Neamatzadeh H. Association of TNF-α genetic variants with neonatal bronchopulmonary dysplasia: consolidated results. Front Pediatr 2024; 12:1511355. [PMID: 39748810 PMCID: PMC11693615 DOI: 10.3389/fped.2024.1511355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/11/2024] [Indexed: 01/04/2025] Open
Abstract
Objectives Inflammation is increasingly recognized as a key factor in the pathophysiology of bronchopulmonary dysplasia (BPD). While previous research has established significant links between TNF-α polymorphisms and BPD susceptibility, further validation of these associations is needed. This study aims to examine the relationship between TNF-α polymorphisms and the risk of BPD. Methods All relevant articles published before October 1, 2024, have been screened in the PubMed, Web of Science, CNKI, and Scopus databases. Results A total of 14 case-control studies were conducted: five studies with 1,252 cases and 1,377 controls on -308G/A, three studies with 1,180 cases and 1,194 controls on -238G/A, four studies with 149 cases and 192 controls on -857C/T, and two studies with 82 cases and 162 controls on 1,031 T/C. A significant association was found between the TNF-α -238G/A polymorphism and the risk of BPD. However, no significant relationships were observed for the TNF-α -308G/A, -857C/T, and 1,031 T/C polymorphisms regarding BPD susceptibility. Conclusions Our findings indicate a significant association between the TNF-α -238G/A polymorphism and the susceptibility to BPD in preterm neonates, suggesting potential biomarkers for its pathogenesis. However, this meta-analysis has limitations, including possible publication bias and heterogeneity due to the limited number of studies, which may affect the reliability of our conclusions. Moreover, population variability further complicates the interpretation of the link between TNF-α polymorphisms and BPD risk.
Collapse
Affiliation(s)
- Seyedeh Elham Shams
- Department of Pediatrics, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyede Arefe Mousavi-Beni
- Afshar Hospital Cardiovascular Research Center, Non-Communicable Disease Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohamad Hosein Lookzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Reza Mirjalili
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Yeganegi
- Department of Obstetrics and Gynecology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amirhossein Shahbazi
- Student Research Committee, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Ali Masoudi
- Student Research Committee, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Amirmasoud Shiri
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences,Shiraz, Iran
| | - Mahmood Noorishadkam
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
5
|
Ito M, Sasaki A, Haga M, Iwatani A, Nishimura E, Arai H, Nagano N, Suga S, Araki S, Konishi A, Onouchi Y, Namba F. Association of hyaluronan and proteoglycan link protein 1 gene with the need of home oxygen therapy in premature Japanese infants with bronchopulmonary dysplasia. J Matern Fetal Neonatal Med 2024; 37:2332914. [PMID: 38522947 DOI: 10.1080/14767058.2024.2332914] [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: 12/28/2023] [Accepted: 03/13/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) has a lasting effect on the respiratory function of infants, imposing chronic health burdens. BPD is influenced by various prenatal, postnatal, and genetic factors. This study explored the connection between BPD and home oxygen therapy (HOT), and then we examined the association between HOT and a specific single-nucleotide polymorphism (SNP) in the hyaluronan and proteoglycan link protein 1 (HAPLN1) gene among premature Japanese infants. MATERIALS AND METHODS Prenatal and postnatal data from 212 premature infants were collected and analyzed by four SNPs (rs975563, rs10942332, rs179851, and rs4703570) around HAPLN1 using the TaqMan polymerase chain reaction method. The clinical characteristics and genotype frequencies of HAPLN1 were assessed and compared between HOT and non-HOT groups. RESULTS Individuals with AA/AC genotypes in the rs4703570 SNP exhibited significantly higher HOT rates at discharge than those with CC homozygotes (odds ratio, 1.20, 95% confidence interval, 1.07-1.35, p = .038). A logistic regression analysis determined that CC homozygotes in the rs4703570 SNP did not show a statistically significant independent association with HOT at discharge. CONCLUSIONS Although our study did not reveal a correlation between HAPLN1 and the onset of BPD, we observed that individuals with CC homozygosity at the rs4703570 SNP exhibit a reduced risk of HOT.
Collapse
Affiliation(s)
- Masato Ito
- Department of Pediatrics, Akita University Graduate School of Medicine, Akita, Japan
| | - Ayumi Sasaki
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Mitsuhiro Haga
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Ayaka Iwatani
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Eri Nishimura
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Hirokazu Arai
- Department of Neonatology, Akita Red Cross Hospital, Akita, Japan
| | - Nobuhiko Nagano
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Itabashi, Japan
| | - Shutaro Suga
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shunsuke Araki
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Asami Konishi
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Yoshihiro Onouchi
- Department of Public Health, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Fumihiko Namba
- Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| |
Collapse
|
6
|
Zhou J, Lei X. Early postnatal corticosteroids in preventing mortality and bronchopulmonary dysplasia: the pivotal importance of selecting an appropriate treated population. Eur J Pediatr 2024; 184:10. [PMID: 39540933 DOI: 10.1007/s00431-024-05881-0] [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: 08/28/2024] [Revised: 10/28/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
This study explores the efficacy of early systematic postnatal corticosteroids (PCS) in reducing bronchopulmonary dysplasia (BPD) and mortality among preterm infants, focusing on identifying the populations most likely to benefit. Although PCS has been extensively studied for its anti-inflammatory effects in preventing BPD, the ideal target population remains unclear. This meta-analysis included 26 randomized controlled trials (RCTs) focusing exclusively on systemic intravenous PCS. Studies were stratified by baseline BPD or mortality rates in control groups (< 50%, 50-65%, and > 65%). Results indicated that PCS effectiveness in reducing BPD or mortality was significantly associated with baseline risk, with rate differences (RD) for BPD or mortality of - 0.03 (95% CI - 0.08, 0.01) in lower-risk groups (< 50%), - 0.07 (95% CI - 0.12, - 0.01) in moderate-risk groups (50-65%), and - 0.18 (95% CI - 0.32, - 0.04) in high-risk groups (> 65%). Linear logistic analysis demonstrated a significant trend, with higher baseline event rates in control groups associated with a more substantial RD (RD ~ rates in controls, R2 = 0.228, p = 0.014). Both dexamethasone and hydrocortisone showed similar trends. CONCLUSION These findings underscore that the baseline event rates in the control group are potentially correlated with the efficacy of PCS in preventing BPD or mortality, offering more precise guidance beyond the general "high-risk" category and supporting baseline risk stratification for more targeted PCS therapy in clinical practice. WHAT IS KNOWN • Early postnatal corticosteroids (PCS) have been widely studied for their anti-inflammatory effects in preventing bronchopulmonary dysplasia (BPD) in preterm infants. • Determining the optimal target population for PCS remains challenging due to varying baseline risks and associated neurodevelopmental concerns. WHAT IS NEW • Stratifying infants by baseline BPD or mortality rates reveals that PCS shows greater efficacy in high-risk groups, particularly those with baseline event rates above 50%. • Considering baseline risk stratification represents a key direction for future clinical decision-making.
Collapse
Affiliation(s)
- Jianguo Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoping Lei
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
7
|
Goates M, Shrestha A, Thapa S, Bettini M, Barrios R, Shivanna B. Blocking IL-17a Signaling Decreases Lung Inflammation and Improves Alveolarization in Experimental Bronchopulmonary Dysplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2023-2035. [PMID: 39117111 DOI: 10.1016/j.ajpath.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/13/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease of preterm infants that is associated with life-long morbidities. Inflammatory insults contribute to BPD pathogenesis. Although the proinflammatory cytokine, IL-17a, plays a role in various neonatal inflammatory disorders, its role in BPD pathogenesis is unclear. To test the hypothesis that blocking IL-17a signaling decreases lipopolysaccharide (LPS)-mediated experimental BPD in neonatal mice, wild-type mice were injected intraperitoneally with phosphate-buffered saline or LPS during the saccular lung developmental phase. Pulmonary IL-17a expression was determined by enzyme-linked immunosorbent assay and by flow cytometry. LPS-injected mice had higher pulmonary IL-17a protein levels and IL-17a+ and IL-22+ cells. γδ T cells, followed by non-T lymphoid cells, were the primary producers of IL-17a. Wild-type mice were then injected intraperitoneally with isotype antibody (Ab) or IL-17a Ab, while they were treated with phosphate-buffered saline or LPS, followed by quantification of lung inflammatory markers, alveolarization, vascularization, cell proliferation, and apoptosis. LPS-mediated alveolar simplification, apoptosis, and cell proliferation inhibition were significantly greater in mice treated with isotype Ab than in those treated with IL-17a Ab. Furthermore, STAT1 activation and IL-6 levels were significantly greater in LPS-exposed mice treated with isotype Ab than in those treated with IL-17a Ab. The study results indicate that blocking IL-17a signaling decreases LPS-mediated experimental BPD.
Collapse
Affiliation(s)
- Meagan Goates
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amrit Shrestha
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Matthew Bettini
- Department of Microbiology and Immunology, University of Utah, Salt Lake City, Utah
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
8
|
Chen C, Jin Y, Jin H, Chen S, Wang L, Ji L, Wang S, Zhang X, Sheng A, Sun Y. Adipose mesenchymal stem cells-derived exosomes attenuated hyperoxia-induced lung injury in neonatal rats via inhibiting the NF-κB signaling pathway. Pediatr Pulmonol 2024; 59:2523-2534. [PMID: 38771197 DOI: 10.1002/ppul.27057] [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: 09/16/2023] [Revised: 04/02/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVE Bronchopulmonary dysplasia (BPD) is the most common chronic morbidity in extremely preterm infants. Mesenchymal stem cells-derived exosomes (MSC-Exos) therapies have shown prospects in animal models of BPD. Our study aimed to evaluate the effect of adipose mesenchymal stem cells-derived exosomes (AMSC-Exos) on BPD and the role of the NF-κB signaling pathway in this process. METHODS The AMSCs were extracted and AMSC-Exos were isolated by ultracentrifugation method. Newborn rats were exposed to hyperoxia (90% O2) continuously for 7 days to establish a BPD model. The rats were treated with AMSC-Exos by intratracheal administration on postnatal day 4 (P4). Pulmonary morphology, pulmonary vasculature, inflammatory factors, and NF-κB were assessed. Hyperoxia-induced primary type II alveolar epithelial cells (AECIIs) and AMSC-Exos treatment with or without a pan-NF-κB inhibitor (PDTC) were established to explore the potential mechanism. RESULTS Hyperoxia-exposed rats showed alveolar simplification with decreased radial alveolar count and increased mean linear intercept, low CD31, and vascular endothelial growth factor expression, reduced microvessel density, increased the expression of TNF-α, IL-1β, and IL-6 and decreased the expression of IL-10, and induced NF-κB phosphorylation. AMSC-Exos protected the neonatal lung from the hyperoxia-induced arrest of alveolar and vascular development, alleviated inflammation, and inhibited NF-κB phosphorylation. Hyperoxia decreased viability, increased apoptosis, enhanced inflammation, and induced NF-κB phosphorylation of AECIIs but improved by AMSC-Exos, PDTC, or AMSC-Exos+PDTC. The effect of AMSC-Exos+PDTC in AECIIs was the same as AMSC-Exos, but more notable than PDTC alone. CONCLUSION AMSC-Exos attenuated the hyperoxia-induced lung injury in neonatal rats by inhibiting the NF-κB signaling pathway partly.
Collapse
Affiliation(s)
- Cuie Chen
- Department of Pediatrics, Yiwu Maternity and Children Hospital, Jinhua, Zhejiang, China
| | - Yuxia Jin
- Department of Prenatal Diagnostic Center, Yiwu Maternity and Children Hospital, Jinhua, Zhejiang, China
| | - Hongxing Jin
- Department of Pediatrics, Yiwu Maternity and Children Hospital, Jinhua, Zhejiang, China
| | - Shujun Chen
- Department of Pediatrics, Yiwu Maternity and Children Hospital, Jinhua, Zhejiang, China
| | - Lu Wang
- Department of Prenatal Diagnostic Center, Yiwu Maternity and Children Hospital, Jinhua, Zhejiang, China
| | - Liuqing Ji
- Department of Pediatrics, Yiwu Maternity and Children Hospital, Jinhua, Zhejiang, China
| | - Shi Wang
- Department of Anesthesiology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, China
| | - Xixi Zhang
- Department of Pediatrics, Yuhuan People's Hospital, Taizhou, Zhejiang, China
| | - Anqun Sheng
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanyuan Sun
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatrics, The Quzhou Affiliated Hospital of Wenzhou Medical University (Quzhou People's Hospital), Quzhou, Zhejiang, China
| |
Collapse
|
9
|
Teng M, Wu TJ, Jing X, Day BW, Pritchard KA, Naylor S, Teng RJ. Temporal Dynamics of Oxidative Stress and Inflammation in Bronchopulmonary Dysplasia. Int J Mol Sci 2024; 25:10145. [PMID: 39337630 PMCID: PMC11431892 DOI: 10.3390/ijms251810145] [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: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common lung complication of prematurity. Despite extensive research, our understanding of its pathophysiology remains limited, as reflected by the stable prevalence of BPD. Prematurity is the primary risk factor for BPD, with oxidative stress (OS) and inflammation playing significant roles and being closely linked to premature birth. Understanding the interplay and temporal relationship between OS and inflammation is crucial for developing new treatments for BPD. Animal studies suggest that OS and inflammation can exacerbate each other. Clinical trials focusing solely on antioxidants or anti-inflammatory therapies have been unsuccessful. In contrast, vitamin A and caffeine, with antioxidant and anti-inflammatory properties, have shown some efficacy, reducing BPD by about 10%. However, more than one-third of very preterm infants still suffer from BPD. New therapeutic agents are needed. A novel tripeptide, N-acetyl-lysyltyrosylcysteine amide (KYC), is a reversible myeloperoxidase inhibitor and a systems pharmacology agent. It reduces BPD severity by inhibiting MPO, enhancing antioxidative proteins, and alleviating endoplasmic reticulum stress and cellular senescence in a hyperoxia rat model. KYC represents a promising new approach to BPD treatment.
Collapse
Affiliation(s)
- Michelle Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Tzong-Jin Wu
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Xigang Jing
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| | - Billy W. Day
- ReNeuroGen LLC, 2160 San Fernando Dr., Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Kirkwood A. Pritchard
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
- ReNeuroGen LLC, 2160 San Fernando Dr., Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
- Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Stephen Naylor
- ReNeuroGen LLC, 2160 San Fernando Dr., Elm Grove, WI 53122, USA; (B.W.D.); (S.N.)
| | - Ru-Jeng Teng
- Department of Pediatrics, Medical College of Wisconsin, Suite C410, Children Corporate Center, 999N 92nd Street, Milwaukee, WI 53226, USA; (M.T.); (T.-J.W.); (X.J.)
- Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd., Wauwatosa, WI 53226, USA;
| |
Collapse
|
10
|
Chen X, Huang X, Zhou Q, Kang H, Qiu H, Shi L, Tang H, Zeng S. Association between Ureaplasma urealyticum colonization and bronchopulmonary dysplasia in preterm infants: a systematic review and meta-analysis. Front Pediatr 2024; 12:1436568. [PMID: 39175806 PMCID: PMC11338929 DOI: 10.3389/fped.2024.1436568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is the most prevalent chronic lung disease in preterm infants. Studies have shown that Ureaplasma urealyticum (UU) infection is linked to its pathogenesis. However, it remains controversial whether UU colonization in preterm infants increases the risk of developing BPD. Objective This study aimed to conduct a systematic review and meta-analysis to summarize the correlation between UU and BPD. Methods We searched PubMed, Embase, the Cochrane Library, Web of Science, Wanfang Database, Chinese National Knowledge Infrastructure Database, Chinese Science and Technique Journal Database, and the China Biology Medicine disc from their inception to March 15, 2024. We included cohort and case-control studies investigating the association between UU infections and BPD in preterm infants, adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The Newcastle-Ottawa Scale was used for quality assessment. The outcome was defined as the continued need for oxygen or respiratory support at 28 days after birth (BPD28) or at 36 weeks postmenstrual age (BPD36). Considering the potential publication bias in observational studies, we used a random-effects meta-analysis model, assessed heterogeneity (I2), performed subgroup analyses, evaluated publication bias, and graded the quality of evidence. Results The meta-analysis included 36 cohort studies encompassing 5,991 participants. Among these, 20 reported on BPD28, 13 on BPD36, and 3 on both. The results indicated a significant association between UU colonization and BPD28 (odds ratio (OR): 2.26, 95% confidence interval (CI): 1.78-2.85, P < 0.00001, 23 studies, very low certainty of evidence) and BPD36 (OR: 2.13, 95% CI: 1.47-3.07, P < 0.0001, 16 studies, very low certainty of evidence). Conclusion There is a correlation between UU colonization and the development of BPD in preterm infants. Future research should prioritize well-designed, large-scale, high-quality randomized controlled trials (RCTs) to comprehensively assess the risk of BPD in neonates following UU infection and to provide stronger evidence for clinical screening and prevention strategies to improve the prognosis of affected newborns. Systematic Review Registration https://www.crd.york.ac.uk/, identifier (CRD42024524846).
Collapse
Affiliation(s)
- Xianhong Chen
- Shenzhen Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xunbin Huang
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Qiujing Zhou
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Houxin Kang
- Neonatal·Child Critical Child Health Care Division, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi Tujia and Miao Autonomous Prefecture, Hubei, China
| | - Huixian Qiu
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Lindong Shi
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Hong Tang
- Division of Neonatology, Shenzhen Yantian District People’s Hospital, Shenzhen, Guangdong, China
| | - Shujuan Zeng
- Division of Neonatology, Longgang District Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Liu J, Bao T, Zhou Y, Ma M, Tian Z. Deficiency of Secreted Phosphoprotein 1 Alleviates Hyperoxia-induced Bronchopulmonary Dysplasia in Neonatal Mice. Inflammation 2024:10.1007/s10753-024-02088-1. [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] [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
|
12
|
Tamatam CM, Venkareddy LK, Ankireddy A, Machireddy N, Reddy SP. Myeloid Nrf2 Protects against Neonatal Oxidant-Stress-Induced Lung Inflammation and Alveolar Simplification in Mice. Antioxidants (Basel) 2024; 13:698. [PMID: 38929137 PMCID: PMC11200887 DOI: 10.3390/antiox13060698] [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: 04/16/2024] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic condition affecting preterm infants, characterized by lung alveolar simplification/hypoalveolarization and vascular remodeling. The nuclear factor erythroid 2 like 2 (Nfe2l2, or Nrf2) plays a critical role in the cytoprotective response to neonatal hyperoxia, and its global deficiency exacerbates hypoalveolarization in mice. The abnormal recruitment and activation of myeloid cells are associated with the pathogenesis of BPD. Therefore, we employed a genetic approach to investigate the role of myeloid Nrf2 in regulating hyperoxia-induced hypoalveolarization. Pups, both wild-type (Nrf2f/f) and those with a myeloid Nrf2 deletion (abbreviated as Nrf2∆/∆mye), were exposed to hyperoxia for 72 h at postnatal day 1 (Pnd1), and then sacrificed at either Pnd4 or Pnd18 following a two-week recovery period. We analyzed the hypoalveolarization, inflammation, and gene expression related to cytoprotective and inflammatory responses in the lungs of these pups. The hypoalveolarization induced by hyperoxia was significantly greater in Nrf2∆/∆mye pups compared to their Nrf2f/f counterparts (35.88% vs. 21.01%, respectively) and was accompanied by increased levels of inflammatory cells and IL-1β activation in the lungs. Antioxidant gene expression in response to neonatal hyperoxia was lower in Nrf2∆/∆mye pups compared to their Nrf2f/f counterparts. Furthermore, Nrf2-deficient macrophages exposed to hyperoxia exhibited markedly decreased cytoprotective gene expression and increased IL-1β levels compared to Nrf2-sufficient cells. Our findings demonstrate the crucial role of myeloid Nrf2 in mitigating hyperoxia-induced lung hypoalveolarization and inflammatory responses in neonatal mice.
Collapse
Affiliation(s)
- Chandra Mohan Tamatam
- Department of Pediatrics, University of Illinois, Chicago, IL 60612, USA; (L.K.V.); (A.A.); (N.M.)
| | - Lalith Kumar Venkareddy
- Department of Pediatrics, University of Illinois, Chicago, IL 60612, USA; (L.K.V.); (A.A.); (N.M.)
- Department of Cell Biology and Molecular Genetics, Sri Devaraj Urs Academy of Higher Education and Research, Tamaka, Kolar 563103, Karnataka, India
| | - Aparna Ankireddy
- Department of Pediatrics, University of Illinois, Chicago, IL 60612, USA; (L.K.V.); (A.A.); (N.M.)
| | - Narsa Machireddy
- Department of Pediatrics, University of Illinois, Chicago, IL 60612, USA; (L.K.V.); (A.A.); (N.M.)
| | - Sekhart P. Reddy
- Department of Pediatrics, University of Illinois, Chicago, IL 60612, USA; (L.K.V.); (A.A.); (N.M.)
- Department of Pathology, The University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Zhang L, Zhuo Z, Chen L, Liu J, Huang J, Deng J, Lu W, Jiang X. Nesfatin-1 alleviates hyperoxia-induced bronchopulmonary dysplasia (BPD) via the nuclear factor-κB (NF-κB) p65 signaling pathway. J Biochem Mol Toxicol 2024; 38:e23680. [PMID: 38511245 DOI: 10.1002/jbt.23680] [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: 07/24/2023] [Revised: 01/16/2024] [Accepted: 02/23/2024] [Indexed: 03/22/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic respiratory disease in newborns, which severely influences the health of infants and lacks effective clinical treatment strategies. The pathogenesis of BPD is correlated to enhanced inflammation and activated oxidative stress (OS). The application of antioxidants and anti-inflammatory treatment could be hot spots for BPD treatment. Nesfatin-1, a peptide with a suppressive property against inflammation, was tested herein for its potential therapeutic value in BPD. Neonatal SD rats were stimulated with hyperoxia, followed by being intraperitoneally administered with 20 μg/kg/day Nesfatin-1 for 2 weeks. Decreased RAC value in lung tissues, increased wet weight/dry weight (W/D) pulmonary ratio and bronchoalveolar lavage fluid (BALF) proteins, elevated cytokine release in BALF, increased malondialdehyde (MDA) content, and declined superoxide dismutase (SOD) activity were observed in BPD rats, all of which were sharply mitigated by Nesfatin-1. Rat epithelial type II cells (AECIIs) were handled with hyperoxia, and then cultured with 1 and 10 nM Nesfatin-1. Reduced cell viability, elevated lactate dehydrogenase production, elevated cytokine secretion, elevated MDA content, and decreased SOD activity were observed in hyperoxia-handled AECIIs, all of which were markedly alleviated by Nesfatin-1. Furthermore, activated nuclear factor-κB (NF-κB) signaling observed in both BPD rats and hyperoxia-handled AECIIs were notably repressed by Nesfatin-1. Collectively, Nesfatin-1 alleviated hyperoxia-triggered BPD by repressing inflammation and OS via the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Li Zhang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, China
| | - Zhang Zhuo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Lijuan Chen
- Department of Pharmacy, Stomatology Hospital Affiliated to Southwest Medical University, Luzhou, China
| | - Jiao Liu
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiang Huang
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Deng
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Wei Lu
- Department of Emergency, Luzhou People's Hospital, Luzhou, China
| | - Xian Jiang
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, China
| |
Collapse
|
14
|
Yang X, Bao Z, Lei X, Wang X, Zhao S, Du F, Liu X, Dong W. Omeprazole activates aryl hydrocarbon receptor to reduce hyperoxia-induced oxidative stress in the peripheral blood mononuclear cells from premature infants. J Matern Fetal Neonatal Med 2023; 36:2272577. [PMID: 37884440 DOI: 10.1080/14767058.2023.2272577] [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: 12/14/2021] [Accepted: 10/14/2023] [Indexed: 10/28/2023]
Abstract
OBJECTIVE To investigate the correlation between the aryl hydrocarbon receptor (AhR) and reactive oxygen species (ROS) in peripheral blood mononuclear cells (PBMCs) of premature infants, to demonstrate the protective role of AhR against hyperoxia-induced oxidative stress in premature infants and to provide a rational basis for the use of omeprazole (OM) as a new treatment for bronchopulmonary dysplasia (BPD). METHODS From January 2021 to June 2021, 1-3 ml of discarded peripheral blood was collected from premature infants of gestational age less than 32 weeks who were not taking inhaled oxygen and were admitted to the Department of Neonatology of the Affiliated Hospital of Southwest Medical University. Using a random number table, the PBMCs were randomly assigned to each of the following groups: the control group, air + OM group, hyperoxia group, and hyperoxia + OM group. After 48 h of in vitro modeling and culture, PBMCs and the culture medium of each group were collected. Immunofluorescence analysis was used to examine ROS levels in PBMCs. A full-spectrum spectrophotometer was used to examine malondialdehyde (MDA) levels in the culture medium. Enzyme-linked immunosorbent assay (ELISA) was used to examine monocyte chemotactic protein 1 (MCP-1) levels in culture medium. Immunofluorescence analysis was used to examine the intracellular localization of AhR. Western blotting was used to examine the expression level of AhR in PBMCs. RESULTS Compared with those in the control group, the levels of ROS, MDA, and MCP-1 and the cytoplasm-nuclear translocation rate of AhR in the air + OM group did not change significantly (p > 0.05), but the expression level of AhR increased significantly (p < 0.05). The levels of ROS, MDA, and MCP-1 and the cytoplasm-nuclear translocation rate of AhR significantly increased in the hyperoxia group (p < 0.05), and the expression level of AhR was significantly reduced (p < 0.05). Compared with those in the hyperoxia group, the levels of ROS, MDA, and MCP-1 in the hyperoxia + OM group were significantly reduced (p < 0.05), and the cytoplasm-nuclear translocation rate of AhR and the expression level of AhR were significantly increased (p < 0.05), but did not reach the level of the control group (p < 0.05). CONCLUSION OM can activate AhR to inhibit hyperoxia-induced oxidative stress in the PBMCs from 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 Centre for Birth Defects, Luzhou, China
| | - Zhengrong Bao
- 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 Centre for Birth Defects, Luzhou, China
| | - Xiaoping Lei
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Centre 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 Centre for Birth Defects, Luzhou, China
| | - Shuai Zhao
- 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 Centre for Birth Defects, Luzhou, China
| | - Fengling Du
- 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 Centre for Birth Defects, Luzhou, China
| | - Xingling Liu
- 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 Centre 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 Centre for Birth Defects, Luzhou, China
| |
Collapse
|
15
|
Aslan M, Gokce IK, Turgut H, Tekin S, Cetin Taslidere A, Deveci MF, Kaya H, Tanbek K, Gul CC, Ozdemir R. Molsidomine decreases hyperoxia-induced lung injury in neonatal rats. Pediatr Res 2023; 94:1341-1348. [PMID: 37179436 DOI: 10.1038/s41390-023-02643-w] [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: 09/21/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND The study's objective is to evaluate if Molsidomine (MOL), an anti-oxidant, anti-inflammatory, and anti-apoptotic drug, is effective in treating hyperoxic lung injury (HLI). METHODS The study consisted of four groups of neonatal rats characterized as the Control, Control+MOL, HLI, HLI + MOL groups. Near the end of the study, the lung tissue of the rats were evaluated with respect to apoptosis, histopathological damage, anti-oxidant and oxidant capacity as well as degree of inflammation. RESULTS Compared to the HLI group, malondialdehyde and total oxidant status levels in lung tissue were notably reduced in the HLI + MOL group. Furthermore, mean superoxide dismutase, glutathione peroxidase, and glutathione activities/levels in lung tissue were significantly higher in the HLI + MOL group as compared to the HLI group. Tumor necrosis factor-α and interleukin-1β elevations associated with hyperoxia were significantly reduced following MOL treatment. Median histopathological damage and mean alveolar macrophage numbers were found to be higher in the HLI and HLI + MOL groups when compared to the Control and Control+MOL groups. Both values were increased in the HLI group when compared to the HLI + MOL group. CONCLUSIONS Our research is the first to demonstrate that bronchopulmonary dysplasia may be prevented through the protective characteristics of MOL, an anti-inflammatory, anti-oxidant, and anti-apoptotic drug. IMPACT Molsidomine prophylaxis significantly decreased the level of oxidative stress markers. Molsidomine administration restored the activities of antioxidant enzymes. Molsidomine prophylaxis significantly reduced the levels of inflammatory cytokines. Molsidomine may provide a new and promising therapy for BPD in the future. Molsidomine prophylaxis decreased lung damage and macrophage infiltration in the tissue.
Collapse
Affiliation(s)
- Mehmet Aslan
- Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Ismail Kursat Gokce
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Hatice Turgut
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Suat Tekin
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Asli Cetin Taslidere
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Mehmet Fatih Deveci
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Huseyin Kaya
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Kevser Tanbek
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Cemile Ceren Gul
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Ramazan Ozdemir
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey.
| |
Collapse
|
16
|
Moore CM, D'Amore A, Fustolo-Gunnink S, Hudson C, Newton A, Santamaria BL, Deary A, Hodge R, Hopkins V, Mora A, Llewelyn C, Venkatesh V, Khan R, Willoughby K, Onland W, Fijnvandraat K, New HV, Clarke P, Lopriore E, Watts T, Stanworth S, Curley A. Two-year outcomes following a randomised platelet transfusion trial in preterm infants. Arch Dis Child Fetal Neonatal Ed 2023; 108:452-457. [PMID: 36810309 PMCID: PMC10447411 DOI: 10.1136/archdischild-2022-324915] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023]
Abstract
OBJECTIVE Assess mortality and neurodevelopmental outcomes at 2 years of corrected age in children who participated in the PlaNeT-2/MATISSE (Platelets for Neonatal Transfusion - 2/Management of Thrombocytopenia in Special Subgroup) study, which reported that a higher platelet transfusion threshold was associated with significantly increased mortality or major bleeding compared to a lower one. DESIGN Randomised clinical trial, enrolling from June 2011 to August 2017. Follow-up was complete by January 2020. Caregivers were not blinded; however, outcome assessors were blinded to treatment group. SETTING 43 level II/III/IV neonatal intensive care units (NICUs) across UK, Netherlands and Ireland. PATIENTS 660 infants born at less than 34 weeks' gestation with platelet counts less than 50×109/L. INTERVENTIONS Infants were randomised to undergo a platelet transfusion at platelet count thresholds of 50×109/L (higher threshold group) or 25×109/L (lower threshold group). MAIN OUTCOMES MEASURES Our prespecified long-term follow-up outcome was a composite of death or neurodevelopmental impairment (developmental delay, cerebral palsy, seizure disorder, profound hearing or vision loss) at 2 years of corrected age. RESULTS Follow-up data were available for 601 of 653 (92%) eligible participants. Of the 296 infants assigned to the higher threshold group, 147 (50%) died or survived with neurodevelopmental impairment, as compared with 120 (39%) of 305 infants assigned to the lower threshold group (OR 1.54, 95% CI 1.09 to 2.17, p=0.017). CONCLUSIONS Infants randomised to a higher platelet transfusion threshold of 50×109/L compared with 25×109/L had a higher rate of death or significant neurodevelopmental impairment at a corrected age of 2 years. This further supports evidence of harm caused by high prophylactic platelet transfusion thresholds in preterm infants. TRIAL REGISTRATION NUMBER ISRCTN87736839.
Collapse
Affiliation(s)
- Carmel Maria Moore
- School of Medicine, University College Dublin, Dublin, Ireland
- Neonatology, National Maternity Hospital, Dublin, Ireland
| | - Angela D'Amore
- Neonatology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Cara Hudson
- Clinical Trials Unit, NHS Blood and Transplant, Bristol, UK
| | - Alice Newton
- Clinical Trials Unit, NHS Blood and Transplant, Bristol, UK
| | | | - Alison Deary
- Clinical Trials Unit, NHS Blood and Transplant, Bristol, UK
| | - Renate Hodge
- Clinical Trials Unit, NHS Blood and Transplant, Bristol, UK
| | | | - Ana Mora
- Clinical Trials Unit, NHS Blood and Transplant, Bristol, UK
| | | | | | - Rizwan Khan
- NICU, University Maternity Hospital Limerick, Limerick, Ireland
| | - Karen Willoughby
- Neonatology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Wes Onland
- Neonatology, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Karin Fijnvandraat
- Pediatrics, Emma Children's Hospital, Pediatric Hematology, University of Amsterdam, Amsterdam, The Netherlands
| | - Helen V New
- Paediatric Transfusion Medicine, NHS Blood and Transplant, London, UK
| | - Paul Clarke
- Neonatal Intensive Care Unit, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Enrico Lopriore
- Neonatology, Leiden Universitair Medisch Centrum, Leiden, The Netherlands
| | - Timothy Watts
- Neonatal Intensive Care Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Simon Stanworth
- National Health Service Blood and Transplant, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Anna Curley
- School of Medicine, University College Dublin, Dublin, Ireland
- Neonatology, National Maternity Hospital, Dublin, Ireland
| |
Collapse
|
17
|
Burns J, Phung R, McNeill S, Hanlon-Dearman A, Ricci MF. Comorbidities Affecting Children with Autism Spectrum Disorder: A Retrospective Chart Review. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1414. [PMID: 37628413 PMCID: PMC10453739 DOI: 10.3390/children10081414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Autism spectrum disorder (ASD) is a developmental disorder characterized by deficits in social interaction/communication, restricted interests, and repetitive behaviors. Recent discussions have emerged worldwide regarding the heterogeneity around presentation/etiology and comorbidities. This study aimed to determine the frequency and characteristics of comorbidities among children diagnosed with ASD in Manitoba and to evaluate differences in presentation between those with and without medical comorbidities. We conducted a retrospective chart review of >1900 electronic charts at the only publicly funded referral site for children ≤6 years requiring evaluation for ASD in Manitoba. All children aged 0-6 years diagnosed with ASD at this site between May 2016 and September 2021 were identified. χ2 and t-tests were used to compare groups. Of the total of 1858 children identified, 1452 (78.1%) were boys, 251 (13.5%) were prematurely born, and 539 (29.0%) had ≥1 medical comorbidity. Global developmental delay (GDD) was diagnosed in 428 (23.0%). The age of referral and diagnosis did not differ between groups. Comorbidities were more common among premature children (16.0% vs. 12.5%, p: 0.005) and children with comorbid GDD (34.9% vs. 18.2%, p < 0.001). Neurological comorbidities were most common (37.1%). No sex difference in the overall presence of comorbidities was found (boys = 77.1% vs. 78.5%, p: 0.518); however, girls had a higher incidence of neurological comorbidities, e.g., cerebral palsy, seizures, hypotonia (14.8% vs. 9.64%, p: 0.009), as well as genetic comorbidities (4.92% vs. 2.75%, p: 0.04). The high rates of associated neurological conditions, GDD, and prematurity add heterogeneity to this group leading to potential difficulties with prognosis and service allocation. Primary vs. secondary ASD can be a way of separating individuals based on relevant medical comorbidities.
Collapse
Affiliation(s)
- Jessy Burns
- SSCY Centre, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3G1, Canada; (S.M.); (A.H.-D.); (M.F.R.)
| | - Ryan Phung
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 0Z3, Canada;
| | - Shayna McNeill
- SSCY Centre, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3G1, Canada; (S.M.); (A.H.-D.); (M.F.R.)
| | - Ana Hanlon-Dearman
- SSCY Centre, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3G1, Canada; (S.M.); (A.H.-D.); (M.F.R.)
| | - M. Florencia Ricci
- SSCY Centre, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB R3E 3G1, Canada; (S.M.); (A.H.-D.); (M.F.R.)
| |
Collapse
|
18
|
Chen X, Huang X, Lin Y, Lin B, Yang C, Huang Z, Yang C. Association of Ureaplasma infection pattern and azithromycin treatment effect with bronchopulmonary dysplasia in Ureaplasma positive infants: a cohort study. BMC Pulm Med 2023; 23:229. [PMID: 37365524 DOI: 10.1186/s12890-023-02522-4] [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: 05/17/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND It is unclear whether Ureaplasma-associated pneumonia and azithromycin treatment affect the risk for bronchopulmonary dysplasia (BPD). METHODS A retrospective cohort study was performed in very low birth weight (VLBW) infants who tested positive for Ureaplasma within 72 h after birth in a tertiary unit. Chest X-ray (CXR) and laboratory test were performed before and after azithromycin treatment. Multivariate logistic regression analysis was used to identify the independent association between BPD and Ureaplasma-associated pneumonia, as well as BPD and effective azithromycin treatment. RESULTS A total of 118 infants were included in the current study, of whom 36 developed BPD (defined as supplemental oxygen needed at postmenstrual age 36 weeks or discharge). The rate of BPD was significantly higher in infants with Ureaplasma-associated pneumonia (44.6%) compared to infants with Ureaplasma colonization (17.7%, P = 0.002). After adjusting for confounders, an effective azithromycin treatment was significantly associated with reduced risk of BPD [odd ratio (OR) 0.011; 95% confidence interval (CI): 0.000-0.250), whereas Ureaplasma-associated pneumonia was not significantly associated with BPD (OR 1.835; 95% CI: 0.548-6.147). CONCLUSION Effective Azithromycin treatment in Ureaplasma positive VLBW infants was associated with a reduced risk of BPD.
Collapse
Affiliation(s)
- Xueyu Chen
- Department of Neonatology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xuemei Huang
- Department of Neonatology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
- Department of Neonatology, Liuzhou Maternity and Child Healthcare Hospital, Affiliated Maternity Hospital and Affiliated Children's Hospital of Guangxi University of Science and Technology, Guangxi, China
| | - Yanqing Lin
- Department of Neonatology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Bingchun Lin
- Department of Neonatology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Chunyu Yang
- Department of Radiology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Guangzhou, China
| | - Zhifeng Huang
- Department of Neonatology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Chuanzhong Yang
- Department of Neonatology, The First School of Clinical Medicine, Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
19
|
Ahmed M, Casanova NG, Zaghloul N, Gupta A, Rodriguez M, Robbins IR, Kempf CL, Sun X, Song JH, Hernon VR, Sammani S, Camp SM, Moreira A, Hsu CD, Garcia JGN. The eNAMPT/TLR4 inflammatory cascade drives the severity of intra-amniotic inflammation in pregnancy and predicts infant outcomes. Front Physiol 2023; 14:1129413. [PMID: 37415908 PMCID: PMC10319582 DOI: 10.3389/fphys.2023.1129413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction: Intra-amniotic inflammation (IAI) or chorioamnionitis is a common complication of pregnancy producing significant maternal morbidity/mortality, premature birth and neonatal risk of chronic lung diseases such as bronchopulmonary dysplasia (BPD). We examined eNAMPT (extracellular nicotinamide phosphoribosyltransferase), a critical inflammatory DAMP and TLR4 ligand, as a potential therapeutic target to reduce IAI severity and improve adverse fetal/neonatal outcomes. Methods: Blood/tissue samples were examined in: 1) women with histologically-proven chorioamnionitis, 2) very low birth weight (VLBW) neonates, and 3) a preclinical murine pregnancy model of IAI. Groups of pregnant IAI-exposed mice and pups were treated with an eNAMPT-neutralizing mAb. Results: Human placentas from women with histologically-proven chorioamnionitis exhibited dramatic NAMPT expression compared to placentas without chorioamnionitis. Increased NAMPT expression in whole blood from VLBW neonates (day 5) significantly predicted BPD development. Compared to untreated LPS-challenged murine dams (gestational day 15), pups born to eNAMPT mAb-treated dams (gestational days 15/16) exhibited a > 3-fold improved survival, reduced neonate lung eNAMPT/cytokine levels, and reduced development and severity of BPD and pulmonary hypertension (PH) following postnatal exposure to 100% hyperoxia days 1-14. Genome-wide gene expression studies of maternal uterine and neonatal cardiac tissues corroborated eNAMPT mAb-induced reductions in inflammatory pathway genes. Discussion: The eNAMPT/TLR4 inflammatory pathway is a highly druggable contributor to IAI pathobiology during pregnancy with the eNAMPT-neutralizing mAb a novel therapeutic strategy to decrease premature delivery and improve short- and long-term neonatal outcomes. eNAMPT blood expression is a potential biomarker for early prediction of chronic lung disease among premature neonates.
Collapse
Affiliation(s)
- Mohamed Ahmed
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nancy G. Casanova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Nahla Zaghloul
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Akash Gupta
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Marisela Rodriguez
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Ian R. Robbins
- Departments of Pediatrics, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Carrie L. Kempf
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Xiaoguang Sun
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Jin H. Song
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Vivian Reyes Hernon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Alvaro Moreira
- Department of Pediatrics, UT Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Chaur-Dong Hsu
- Department of Obstetrics and Gynecology, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| |
Collapse
|
20
|
Bulbul A, Bacak T, Avsar H. Role of Postnatal Corticosteroids in the Treatment or Prevention of Bronchopulmonary Dysplasia. SISLI ETFAL HASTANESI TIP BULTENI 2023; 57:171-181. [PMID: 37899802 PMCID: PMC10600625 DOI: 10.14744/semb.2023.80688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/09/2023]
Abstract
As the frequency of viable low birth weight preterm babies increases, bronchopulmonary dysplasia (BPD), one of the most important morbidities in these babies, also increases. Using postnatal steroids to reduce the development of BPD has not been fully enlightened. Besides all prevention strategies for reducing the development of BPD, it is known that steroid therapy used in the 1st week of life could induce negative neuromotor development according to current data. It may be recommended to administer low-dose dexamethasone between 8 and 49 days in infants dependent on mechanical ventilators in the postnatal period. It is seen that the use of hydrocortisone in the early period does not cause negative neuromotor development, but it cannot prevent the development of BPD as much as dexamethasone. All intensive care units must have their steroid protocol for BPD and use steroids in cases when the BPD development scale score is >60-65% and should have a goal of trying to keep the cumulative dose at the lowest level.
Collapse
Affiliation(s)
- Ali Bulbul
- Department of Pediatrics, Division of Neonatology, University of Health Sciences Türkiye, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Türkiye
| | - Tolga Bacak
- Department of Pediatrics, Division of Neonatology, University of Health Sciences Türkiye, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Türkiye
| | - Hasan Avsar
- Department of Pediatrics, Division of Neonatology, University of Health Sciences Türkiye, Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Türkiye
| |
Collapse
|
21
|
Shankar N, Thapa S, Shrestha AK, Sarkar P, Gaber MW, Barrios R, Shivanna B. Hyperoxia Disrupts Lung Lymphatic Homeostasis in Neonatal Mice. Antioxidants (Basel) 2023; 12:620. [PMID: 36978868 PMCID: PMC10045755 DOI: 10.3390/antiox12030620] [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: 12/27/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammation causes bronchopulmonary dysplasia (BPD), a common lung disease of preterm infants. One reason this disease lacks specific therapies is the paucity of information on the mechanisms regulating inflammation in developing lungs. We address this gap by characterizing the lymphatic phenotype in an experimental BPD model because lymphatics are major regulators of immune homeostasis. We hypothesized that hyperoxia (HO), a major risk factor for experimental and human BPD, disrupts lymphatic endothelial homeostasis using neonatal mice and human dermal lymphatic endothelial cells (HDLECs). Exposure to 70% O2 for 24-72 h decreased the expression of prospero homeobox 1 (Prox1) and vascular endothelial growth factor c (Vegf-c) and increased the expression of heme oxygenase 1 and NAD(P)H dehydrogenase [quinone]1 in HDLECs, and reduced their tubule formation ability. Next, we determined Prox1 and Vegf-c mRNA levels on postnatal days (P) 7 and 14 in neonatal murine lungs. The mRNA levels of these genes increased from P7 to P14, and 70% O2 exposure for 14 d (HO) attenuated this physiological increase in pro-lymphatic factors. Further, HO exposure decreased VEGFR3+ and podoplanin+ lymphatic vessel density and lymphatic function in neonatal murine lungs. Collectively, our results validate the hypothesis that HO disrupts lymphatic endothelial homeostasis.
Collapse
Affiliation(s)
- Nithyapriya Shankar
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Poonam Sarkar
- Division of Hematology-Oncology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - M. Waleed Gaber
- Division of Hematology-Oncology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| |
Collapse
|
22
|
Rumph JT, Stephens VR, Ameli S, Brown LK, Rayford KJ, Nde PN, Osteen KG, Bruner-Tran KL. A Paternal Fish Oil Diet Preconception Reduces Lung Inflammation in a Toxicant-Driven Murine Model of New Bronchopulmonary Dysplasia. Mar Drugs 2023; 21:161. [PMID: 36976210 PMCID: PMC10052688 DOI: 10.3390/md21030161] [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: 12/22/2022] [Revised: 02/25/2023] [Accepted: 02/25/2023] [Indexed: 03/08/2023] Open
Abstract
New bronchopulmonary dysplasia (BPD) is a neonatal disease that is theorized to begin in utero and manifests as reduced alveolarization due to inflammation of the lung. Risk factors for new BPD in human infants include intrauterine growth restriction (IUGR), premature birth (PTB) and formula feeding. Using a mouse model, our group recently reported that a paternal history of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure increased his offspring's risk of IUGR, PTB, and new BPD. Additionally, formula supplementation of these neonates worsened the severity of pulmonary disease. In a separate study, we reported that a paternal preconception fish oil diet prevented TCDD-driven IUGR and PTB. Not surprisingly, eliminating these two major risk factors for new BPD also significantly reduced development of neonatal lung disease. However, this prior study did not examine the potential mechanism for fish oil's protective effect. Herein, we sought to determine whether a paternal preconception fish oil diet attenuated toxicant-associated lung inflammation, which is an important contributor to the pathogenesis of new BPD. Compared to offspring of standard diet TCDD-exposed males, offspring of TCDD-exposed males provided a fish oil diet prior to conception exhibited a significant reduction in pulmonary expression of multiple pro-inflammatory mediators (Tlr4, Cxcr2, Il-1 alpha). Additionally, neonatal lungs of pups born to fish oil treated fathers exhibited minimal hemorrhaging or edema. Currently, prevention of BPD is largely focused on maternal strategies to improve health (e.g., smoking cessation) or reduce risk of PTB (e.g., progesterone supplementation). Our studies in mice support a role for also targeting paternal factors to improve pregnancy outcomes and child health.
Collapse
Affiliation(s)
- Jelonia T. Rumph
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Victoria R. Stephens
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Sharareh Ameli
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - LaKendria K. Brown
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kayla J. Rayford
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Pius N. Nde
- Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, TN 37208, USA
| | - Kevin G. Osteen
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System, Nashville, TN 37232, USA
| | - Kaylon L. Bruner-Tran
- Women’s Reproductive Health Research Center, Department of Obstetrics and Gynecology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
23
|
Li DB, Xu XX, Hu YQ, Cui Q, Xiao YY, Sun SJ, Chen LJ, Ye LC, Sun Q. Congenital heart disease-associated pulmonary dysplasia and its underlying mechanisms. Am J Physiol Lung Cell Mol Physiol 2023; 324:L89-L101. [PMID: 36472329 PMCID: PMC9925164 DOI: 10.1152/ajplung.00195.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical observation indicates that exercise capacity, an important determinant of survival in patients with congenital heart disease (CHD), is most decreased in children with reduced pulmonary blood flow (RPF). However, the underlying mechanism remains unclear. Here, we obtained human RPF lung samples from children with tetralogy of Fallot as well as piglet and rat RPF lung samples from animals with pulmonary artery banding surgery. We observed impaired alveolarization and vascularization, the main characteristics of pulmonary dysplasia, in the lungs of RPF infants, piglets, and rats. RPF caused smaller lungs, cyanosis, and body weight loss in neonatal rats and reduced the number of alveolar type 2 cells. RNA sequencing demonstrated that RPF induced the downregulation of metabolism and migration, a key biological process of late alveolar development, and the upregulation of immune response, which was confirmed by flow cytometry and cytokine detection. In addition, the immunosuppressant cyclosporine A rescued pulmonary dysplasia and increased the expression of the Wnt signaling pathway, which is the driver of postnatal lung development. We concluded that RPF results in pulmonary dysplasia, which may account for the reduced exercise capacity of patients with CHD with RPF. The underlying mechanism is associated with immune response activation, and immunosuppressants have a therapeutic effect in CHD-associated pulmonary dysplasia.
Collapse
Affiliation(s)
- De-Bao Li
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xiu-Xia Xu
- 4Department of Radiology, Huangpu Branch, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yu-Qing Hu
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qing Cui
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ying-Ying Xiao
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Si-Juan Sun
- 5Department of Pediatric Intensive Care Unit, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Li-Jun Chen
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Lin-Cai Ye
- 2Department of Thoracic and Cardiovascular Surgery, Shanghai Institute for Pediatric Congenital Heart Disease, Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, Shanghai School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qi Sun
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
24
|
Rossholt ME, Wendel K, Bratlie M, Aas MF, Gunnarsdottir G, Fugelseth D, Pripp AH, Domellöf M, Størdal K, Stiris T, Moltu SJ. Vitamin A Status in Preterm Infants Is Associated with Inflammation and Dexamethasone Exposure. Nutrients 2023; 15:441. [PMID: 36678312 PMCID: PMC9861363 DOI: 10.3390/nu15020441] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023] Open
Abstract
Vitamin A has a key role in lung development and its deficiency is associated with an increased risk of bronchopulmonary dysplasia. This secondary cohort analysis of the ImNuT trial (Immature, Nutrition Therapy NCT03555019) aimed to (1) explore vitamin A status in preterm infants <29 weeks gestation and (2) assess the influence of inflammation and postnatal dexamethasone exposure on vitamin A concentrations in blood. We report detailed information on vitamin A biochemistry, vitamin A intake, markers of inflammation and dexamethasone exposure. After four weeks of age, infants exposed to dexamethasone (n = 39) showed higher vitamin A concentrations compared to unexposed infants (n = 41); median (IQR) retinol was 1.0 (0.74, 1.5) vs. 0.56 (0.41, 0.74) µmol/L, p < 0.001. Pretreatment retinol concentrations were lower in the dexamethasone group compared to non-exposed infants (p < 0.001); 88% vs. 60% of the infants were considered deficient in vitamin A (retinol < 0.7 µmol/L) at one week of age. Small size for gestational age, mechanical ventilation and elevated levels of interleukin-6 were factors negatively associated with first-week retinol concentrations. In conclusion, preterm infants <29 weeks gestation are at risk of vitamin A deficiency despite intakes that accommodate current recommendations. The presence of inflammation and dexamethasone exposure should be considered when interpreting vitamin A status.
Collapse
Affiliation(s)
- Madelaine Eloranta Rossholt
- Department of Neonatal Intensive Care, Oslo University Hospital, 0450 Oslo, Norway
- Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Kristina Wendel
- Department of Neonatal Intensive Care, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Marianne Bratlie
- Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, 0450 Oslo, Norway
| | - Marlen Fossan Aas
- Department of Neonatal Intensive Care, Oslo University Hospital, 0450 Oslo, Norway
- Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Gunnthorunn Gunnarsdottir
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
- Department of Pediatric Neurology, Oslo University Hospital, 0450 Oslo, Norway
| | - Drude Fugelseth
- Department of Neonatal Intensive Care, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Are Hugo Pripp
- Oslo Centre of Biostatistics and Epidemiology, Oslo University Hospital, 0450 Oslo, Norway
| | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics, Umeå University, 901 87 Umea, Sweden
| | - Ketil Størdal
- Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Tom Stiris
- Department of Neonatal Intensive Care, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| | - Sissel Jennifer Moltu
- Department of Neonatal Intensive Care, Oslo University Hospital, 0450 Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, 0450 Oslo, Norway
| |
Collapse
|
25
|
Halbmeijer NM, Onland W, Cools F, Swarte RM, van der Heide-Jalving M, Dijk P, Mulder-de Tollenaer S, Tan RNGB, Mohns T, Bruneel E, van Heijst AFJ, Kramer B, Debeer A, van Weissenbruch MM, Marechal Y, Blom H, Plaskie K, Offringa M, van Wassenaer-Leemhuis AG, van Kaam AH, Aarnoudse-Moens CSH. Effect of systemic hydrocortisone in ventilated preterm infants on parent-reported behavioural outcomes at 2 years' corrected age: follow-up of a randomised clinical trial. Arch Dis Child Fetal Neonatal Ed 2023:archdischild-2022-324179. [PMID: 36593110 DOI: 10.1136/archdischild-2022-324179] [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: 03/24/2022] [Accepted: 12/16/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To report the parent-reported behavioural outcomes of infants included in the Systemic Hydrocortisone To Prevent Bronchopulmonary Dysplasia in preterm infants study at 2 years' corrected age (CA). DESIGN Randomised placebo-controlled trial. SETTING Dutch and Belgian neonatal intensive care units. PATIENTS Infants born <30 weeks' gestation and/or birth weight <1250 g, and ventilator dependent in the second week of life. INTERVENTION Infants were randomly assigned to a 22-day course of systemic hydrocortisone (cumulative dose 72.5 mg/kg; n=182) or placebo (n=190). MAIN OUTCOME MEASURES Parent-reported behavioural outcomes at 2 years' CA assessed with the Child Behavior Checklist (CBCL 1½-5). RESULTS Parents completed the CBCL of 183 (70% (183/262)) infants (hydrocortisone group, n=96; placebo group, n=87). Multiple imputation was used to account for missing data. Infants with critically elevated T-scores (>55) were found in 22.9%, 19.1% and 29.4% of infants for total, internalising and externalising problems, respectively; these scores were not significantly different between groups (mean difference -1.52 (95% CI -4.00 to 0.96), -2.40 (95% CI -4.99 to 0.20) and -0.81 (95% CI -3.40 to 1.77), respectively). In the subscales, we found a significantly lower T-score for anxiety problems in the hydrocortisone group (mean difference -1.26, 95% CI -2.41 to -0.12). CONCLUSION This study found high rates of behaviour problems at 2 years' CA following very preterm birth, but these problems were not associated with hydrocortisone treatment initiated between 7 and 14 days after birth in ventilated preterm infants. TRIAL REGISTRATION NUMBER NTR2768; EudraCT 2010-023777-19.
Collapse
Affiliation(s)
- Nienke Marjolein Halbmeijer
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Wes Onland
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Filip Cools
- Neonatology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Renate M Swarte
- Section of Neonatology, Pediatrics Department, Erasmus MC Sophia Children's Hospital, Rotterdam, Netherlands
| | | | - Peter Dijk
- Neonatology, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, Netherlands
| | | | - Ratna N G B Tan
- Neonatology, Leiden University Medical Center, Leiden, Netherlands
| | - Thilo Mohns
- NICU, Woman-Mother-Child Center, Màxima Medical Centre, Veldhoven, Netherlands
| | - Els Bruneel
- Department of Neonatology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Arno F J van Heijst
- Department of Neonatology, Amalia Children's Hospital, Radboud Universiteit Nijmegen, Nijmegen, Netherlands
| | - Boris Kramer
- Pediatrics, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Anne Debeer
- Neonatology, KU Leuven University Hospitals, Leuven, Belgium
| | - Mirjam M van Weissenbruch
- Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands.,Neonatology, Amsterdam UMC, location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Yoann Marechal
- Department of Neonatology, Centre Hospitalier Universitaire de Charleroi, Charleroi, Belgium
| | - Henry Blom
- Neonatology, Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | - Katleen Plaskie
- Department of Neonatology, GZA Ziekenhuizen Campus Sint-Augustinus, Wilrijk, Belgium
| | - Martin Offringa
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Child Health Evaluative Sciences, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Aleid G van Wassenaer-Leemhuis
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Anton H van Kaam
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands
| | - Cornelieke S H Aarnoudse-Moens
- Neonatology, Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands .,Amsterdam Reproduction and Development Research Institute, Amsterdam, Netherlands.,Psychosocial Department, Universiteit van Amsterdam, Amsterdam, Netherlands
| | | |
Collapse
|
26
|
Wendel K, Aas MF, Gunnarsdottir G, Rossholt ME, Bratlie M, Nordvik T, Landsend ECS, Fugelseth D, Domellöf M, Pripp AH, Stiris T, Moltu SJ. Effect of arachidonic and docosahexaenoic acid supplementation on respiratory outcomes and neonatal morbidities in preterm infants. Clin Nutr 2023; 42:22-28. [PMID: 36473425 DOI: 10.1016/j.clnu.2022.11.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS Studies have suggested that supplementation with docosahexaenoic acid (DHA) to preterm infants might be associated with an increased risk of bronchopulmonary dysplasia (BPD). Our aim was to investigate the effect of enteral supplementation with arachidonic acid (ARA) and DHA on short-term respiratory outcomes and neonatal morbidities in very preterm infants. METHODS This is a secondary analysis of data from the ImNuT (Immature, Nutrition Therapy) study, a randomized double blind clinical trial. Infants with gestational age less than 29 weeks were randomized to receive a daily enteral supplement with ARA 100 mg/kg and DHA 50 mg/kg (intervention) or medium chain triglycerides (MCT) oil (control), from second day of life to 36 weeks postmenstrual age. Study outcomes included duration of respiratory support, incidence of BPD and other major morbidities associated with preterm birth. RESULTS 120 infants with mean (SD) gestational age 26.4 (1.7) weeks were randomized and allocated to either the intervention or control group. Supplementation with ARA and DHA led to a significant reduction in number of days with respiratory support (mean (95% CI) 63.4 (56.6-71.3) vs 80.6 (72.4-88.8); p = 0.03) and a lower oxygen demand (FiO2) (mean (95% CI) 0.26 (0.25-0.28) vs 0.29 (0.27-0.30); p = 0.03) compared to control treatment. There were no clinically important differences in incidence of BPD and other major morbidities between the treatment groups. CONCLUSIONS Supplementation with ARA and DHA to preterm infants was safe and might have a beneficial effect on respiratory outcomes. CLINICAL TRIAL REGISTRATION The trial has been registered in www. CLINICALTRIALS gov, ID: NCT03555019.
Collapse
Affiliation(s)
- Kristina Wendel
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway.
| | - Marlen Fossan Aas
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | - Gunnthorunn Gunnarsdottir
- Department of Pediatric Neurology, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | - Madelaine Eloranta Rossholt
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, Norway
| | - Marianne Bratlie
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Department of Pediatrics and Adolescence Medicine, Oslo University Hospital, Norway
| | - Tone Nordvik
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | | | - Drude Fugelseth
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | - Magnus Domellöf
- Department of Clinical Sciences, Pediatrics, Umea University, Sweden
| | - Are Hugo Pripp
- Oslo Centre of Biostatistics and Epidemiology, Oslo University Hospital, Norway
| | - Tom Stiris
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| | - Sissel Jennifer Moltu
- Department of Neonatal Intensive Care, Oslo University Hospital, Norway; Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
27
|
Dettman RW, Dizon MLV. How lung injury and therapeutic oxygen could alter white matter development. J Neurosci Res 2022; 100:2127-2137. [PMID: 33687103 PMCID: PMC8426430 DOI: 10.1002/jnr.24816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 01/07/2023]
Abstract
Developmental brain injury describes a spectrum of neurological pathologies resulting from either antenatal or perinatal injury. This includes both cognitive and motor defects that affect patients for their entire lives. Developmental brain injury can be caused by a spectrum of conditions including stroke, perinatal hypoxia-ischemia, and intracranial hemorrhage. Additional risk factors have been identified including very low birth weight, mechanical ventilation, and oxygen (O2 ) supplementation. In fact, infants with bronchopulmonary dysplasia, an inflammatory disease associated with disrupted lung development, have been shown to have decreased cerebral white matter and decreased intracranial volumes. Thus, there appears to be a developmental link between the lung, O2 , and the brain that leads to proper myelination. Here, we will discuss what is currently known about the link between O2 and myelination and how scientists are exploring mechanisms through which supplemental O2 and/or lung injury can affect brain development. Consideration of a link between the diseased lung and developing brain will allow clinicians to fine tune their approaches in managing preterm lung disease in order to optimize brain health.
Collapse
Affiliation(s)
- Robert W. Dettman
- Perinatal Origins of Disease, Stanley Manne Children’s Research Institute, Chicago, IL 60611
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago IL, 60611
| | - Maria L. V. Dizon
- Perinatal Origins of Disease, Stanley Manne Children’s Research Institute, Chicago, IL 60611
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago IL, 60611
| |
Collapse
|
28
|
Hu J, Wu Z, Wang H, Geng H, Huo J, Zhu X, Zhu X. Vitamin D Ameliorates Apoptosis and Inflammation by Targeting the Mitochondrial and MEK1/2-ERK1/2 Pathways in Hyperoxia-Induced Bronchopulmonary Dysplasia. J Inflamm Res 2022; 15:4891-4906. [PMID: 36046664 PMCID: PMC9423049 DOI: 10.2147/jir.s371906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/16/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose Bronchopulmonary dysplasia (BPD) is a common and severe complication in preterm infants. Vitamin D (VitD) has been reported to protect against BPD; however, its role in the mitochondria-mediated and MEK1/2-ERK1/2 pathways has not yet been reported. Methods We first performed in vivo studies using neonatal C57BL/6 mice in which we induced BPD by exposing them to a hyperoxic environment (85% O2). The mice were divided into room air (RA; 21% O2), RA+VitD, BPD, and BPD+VitD groups. Hematoxylin and eosin and Masson’s trichrome staining were used to evaluate lung injury. Inflammation and apoptosis were measured using ELISA, RT-qPCR, and TUNEL assays. We then analyzed BEAS-2B cells divided into the same groups along with an additional BPD+VitD+inhibitor group. Mitochondrial apoptosis was evaluated by transmission electron microscopy, mitochondrial membrane potential, and Western blotting. We then used VDR-shRNA to silence the Vitamin D Receptor (VDR) in the BEAS-2B cells. The inflammation, apoptotic rate, and the phosphorylated forms of MEK1/2 and ERK1/2 in cells were detected by RT-qPCR, flow cytometry, and Western blotting. Results The mean linear intercept, septal thickness, and abnormal fibrosis increased, while radial alveolar count decreased in BPD lungs compared to RA lungs. VitD administration was able to ameliorate the phenotype in BPD lungs. IL-6, IFN-γ, and TNF-α expression and the apoptotic rate decreased in the BPD+VitD lung group. VitD pretreatment restored abnormal mitochondrial morphology, reduced mitochondrial membrane loss, and reduced the expression of cleaved caspase-3, Bax, and Bcl-2 in BEAS-2B cells. VitD administration also reduced IL-6, IFN-γ, and TNF-α mRNA, as well as pMEK1/2 and pERK1/2 expression and apoptosis rate in cells exposed to hyperoxia. Conclusion We concluded that VitD treatment ameliorated apoptosis and inflammation by targeting the mitochondrial pathway and via the MEK1/2-ERK1/2 signaling pathway in BPD, thus supporting its potential therapeutic use in this condition.
Collapse
Affiliation(s)
- Jinhui Hu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, People's Republic of China.,Neonatal Medical Center, Huai'an Maternity and Child Health Care Hospital, Xuzhou Medical University, Huai'an, People's Republic of China
| | - Zhixin Wu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Huawei Wang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Haifeng Geng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jie Huo
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, People's Republic of China.,Department of Neonatology, Yangzhou Maternity and Child Health Care Hospital, Yangzhou, People's Republic of China
| | - Xueping Zhu
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Xiaoli Zhu
- Department of Intervention, The First Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
29
|
Heydarian M, Schulz C, Stoeger T, Hilgendorff A. Association of immune cell recruitment and BPD development. Mol Cell Pediatr 2022; 9:16. [PMID: 35917002 PMCID: PMC9346035 DOI: 10.1186/s40348-022-00148-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
In the neonatal lung, exposure to both prenatal and early postnatal risk factors converge into the development of injury and ultimately chronic disease, also known as bronchopulmonary dysplasia (BPD). The focus of many studies has been the characteristic inflammatory responses provoked by these exposures. Here, we review the relationship between immaturity and prenatal conditions, as well as postnatal exposure to mechanical ventilation and oxygen toxicity, with the imbalance of pro- and anti-inflammatory regulatory networks. In these conditions, cytokine release, protease activity, and sustained presence of innate immune cells in the lung result in pathologic processes contributing to lung injury. We highlight the recruitment and function of myeloid innate immune cells, in particular, neutrophils and monocyte/macrophages in the BPD lung in human patients and animal models. We also discuss dissimilarities between the infant and adult immune system as a basis for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christian Schulz
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Tobias Stoeger
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany. .,Center for Comprehensive Developmental Care (CDeCLMU) at the interdisciplinary Social Pediatric Center, (iSPZ), University Hospital Ludwig-Maximilian University, Munich, Germany.
| |
Collapse
|
30
|
Ren Z, Mo W, Yang L, Wang J, Zhang Q, Zhong Z, Wei W, Liu Z, Wu Z, Yao Y, Yang J. Cord blood antimicrobial peptide LL37 levels in preterm neonates and association with preterm complications. Ital J Pediatr 2022; 48:111. [PMID: 35804392 PMCID: PMC9270758 DOI: 10.1186/s13052-022-01295-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/06/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cathelicidin/LL-37 plays a significant role in the human immune defense reaction. Preterm human immature organs being exposed to inflammation-induced injury was the critical denominator leading to the common preterm associated complications. Previous study showed LL37 concentration in preterm neonates was lower in tracheal aspirates and breast milk as compared to term infants. An adults study showed decreased LL-37 levels was a risk factor for patients in developing severe chronic obstructive pulmonary disease (COPD). However, little is known about the regulation of human cord blood LL37 in preterm neonates and the association with preterm complications. This study was designed to investigate the concentration of LL37 in cord blood of preterm infants and correlation with preterm complications. METHODS Singleton infants born in June 2017 to August 2021 in the study hospital were enrolled. Maternal and neonatal clinical characteristics were collected. LL37 levels, pro-inflammatory factor interleukin-6 (IL-6) and tumor necrosis factor-a (TNF-a) in cord blood and LL37 levels in serum 48-72 hours after birth were measured by enzyme-linked immunosorbent assay. The serum level of LL37 in preterm and term neonates were compared, the perinatal factors possibly affecting the LL37 levels were investigated and the relationship between LL37 level and preterm outcomes were analyzed. RESULTS Cord blood LL37 levels in preterm infants were lower than that in term neonates. Cord blood LL37 level was positively correlated with gestational age in preterm. Prenatal steroid administration in preterm neonates decreased cord blood LL37 level. LL37 level was obviously lower in patients with bronchopulmonary dysplasia (BPD). Multiple line regression analysis showed higher LL37 level in cord blood was an independent protective factor for BPD. The concentration of pro-inflammatory factor IL-6 was negatively correlated with LL37. CONCLUSION Cord blood LL37 levels increased during gestation and decreased after perinatal steroid usage. Very preterm infants who displayed higher cord blood LL37 level had reduced risk of developing BPD. Regulation of pro-inflammatory cytokine IL-6 may be associated with the protective effect of LL37 on BPD.
Collapse
Affiliation(s)
- Zhuxiao Ren
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wenhui Mo
- Department of Neonatology, Foshan fosun chancheng hospital, Foshan, China.
| | - Liling Yang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Jianlan Wang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China
| | - Qi Zhang
- Clinical Genetic Center, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhicheng Zhong
- Clinical Genetic Center, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wei Wei
- Guangdong Cord Blood Bank, Guangzhou, China
| | | | - Zhiping Wu
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Regeneration and Biological Therapies, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yao Yao
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Center for Cell Regeneration and Biological Therapies, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jie Yang
- Department of Neonatology, Guangdong Women and Children Hospital, Guangzhou, China. .,Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Wang X, Liu Y, Han D, Zhong J, Yang C, Chen X. Dose-dependent immunomodulatory effects of metformin on human neonatal monocyte-derived macrophages. Cell Immunol 2022; 377:104557. [PMID: 35679651 DOI: 10.1016/j.cellimm.2022.104557] [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: 01/11/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022]
Abstract
While the association of inflammation with bronchopulmonary dysplasia (BPD) has long been appreciated, M1 proinflammatory macrophage population is emerging as the key element in driving the BPD inflammatory environment. Previous study suggests that low-dose metformin elicits an anti-inflammatory response, possibly through modulating macrophages, to improve disease outcome in a rat BPD model. To investigate this concept further, we examined the dose-dependent immunomodulatory function of metformin directly on human macrophages derived from cord blood (CB) monocytes. We demonstrate that low-dose metformin promotes expansion of M2 anti-inflammatory macrophages, contrasted with high-dose treatment, which exacerbates inflammation by favoring M1 polarization and restricting M2 phenotype. These findings highlight that metformin hold immunomodulatory ability by regulating macrophage polarization in a dose-dependent manner, and only when applied at low dose, exhibiting potential for beneficial anti-inflammatory adjuvant in BPD setting.
Collapse
Affiliation(s)
- Xuan Wang
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Yijun Liu
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Dongshan Han
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Junyan Zhong
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Chuanzhong Yang
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xueyu Chen
- Laboratory of Neonatology, Department of Neonatology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
32
|
Wang X, Huo R, Liang Z, Xu C, Chen T, Lin J, Li L, Lin W, Pan B, Fu X, Chen S. Simvastatin Inhibits NLRP3 Inflammasome Activation and Ameliorates Lung Injury in Hyperoxia-Induced Bronchopulmonary Dysplasia via the KLF2-Mediated Mechanism. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8336070. [PMID: 35509841 PMCID: PMC9060986 DOI: 10.1155/2022/8336070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly found in premature infants. Excessive inflammation and oxidative stress contribute to BPD occurrence and development. Simvastatin, as an inhibitor of HMG-CoA reductase, has been reported to have antioxidative and anti-inflammatory effects. However, its effect and possible mechanisms in hyperoxia-induced lung injury are rarely reported. In this study, in vivo and in vitro experiments were conducted to investigate whether simvastatin could ameliorate hyperoxia-induced lung injury and explore its potential mechanism. For the in vivo study, simvastatin could improve alveolar development after hyperoxic lung injury and reduce hyperoxic stress and inflammation. The in vitro study revealed that simvastatin can reduce inflammation in A549 cells after high-oxygen exposure. Simvastatin suppressed NLRP3 inflammasome activation and played anti-inflammatory and antioxidant roles by increasing KLF2 (Krüppel-like factor 2) expression. In vitro experiments also revealed that these effects of simvastatin were partially reversed by KLF2 shRNA, indicating that KLF2 was involved in simvastatin effects. In summary, our findings indicate that simvastatin could downregulate NLRP3 inflammasome activation and attenuate lung injury in hyperoxia-induced bronchopulmonary dysplasia via KLF2-mediated mechanism.
Collapse
Affiliation(s)
- Xinye Wang
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
- Department of Pediatric, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ran Huo
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Zhongjie Liang
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Congcong Xu
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Tingting Chen
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Jingjing Lin
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Luyao Li
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Wei Lin
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Bingting Pan
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoqin Fu
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | - Shangqin Chen
- Department of Neonatology, The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
33
|
HLA-A2 Promotes the Therapeutic Effect of Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Hyperoxic Lung Injury. Bioengineering (Basel) 2022; 9:bioengineering9040177. [PMID: 35447737 PMCID: PMC9029550 DOI: 10.3390/bioengineering9040177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 12/03/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the most extensively studied stem cell types owing to their capacity for differentiation into multiple lineages as well as their ability to secrete regenerative factors and modulate immune functions. However, issues remain regarding their further application for cell therapy. Here, to demonstrate the superiority of the improvement of MSCs, we divided umbilical cord blood-derived MSCs (UCB-MSCs) from 15 donors into two groups based on efficacy and revealed donor-dependent variations in the anti-inflammatory effect of MSCs on macrophages as well as their immunoregulatory effect on T cells. Through surface marker analyses (242 antibodies), we found that HLA-A2 was positively related to the anti-inflammatory and immunoregulatory function of MSCs. Additionally, HLA-A2 mRNA silencing in MSCs attenuated their therapeutic effects in vitro; namely, the suppression of LPS-stimulated macrophages and phytohemagglutinin-stimulated T cells. Moreover, HLA-A2 silencing in MSCs significantly decreased their therapeutic effects in a rat model of hyperoxic lung damage. The present study provides novel insights into the quality control of donor-derived MSCs for the treatment of inflammatory conditions and diseases.
Collapse
|
34
|
Zhang Z, Jiang J, Li Z, Wan W. The Change of Cytokines and Gut Microbiome in Preterm Infants for Bronchopulmonary Dysplasia. Front Microbiol 2022; 13:804887. [PMID: 35387067 PMCID: PMC8978829 DOI: 10.3389/fmicb.2022.804887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Background Bronchopulmonary dysplasia (BPD) is a devastating form of chronic lung disease that develops in preterm infants. BPD is speculated to arise from abnormal inflammatory responses, which is related to the composition of commensal microbiota, leading us to hypothesize that BPD susceptibility could be influenced by gut microbiota through inflammatory responses. This study is aimed to detect cytokines and the differences in fecal gut microbial composition in the BPD patients. Methods Between June 2018 and June 2020, preterm infants born at gestational age ≤30 weeks were recruited. The clinical data of infant characteristics were collected. On days 3–7 and 14–28 after birth, fresh stool samples and serum were collected. The gut microbiota composition between the BPD group and controls was detected by 16S rRNA sequencing. On days 3–7 and days 14–28, ten cytokines including IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, IFN-γ, and TNF-α were detected in the serum. Results This study enrolled 38 preterm infants; the number of preterm infants in the BPD group and control group was, respectively, 18 and 20. The gestational age (27.4 ± 1.5 weeks vs. 29.5 ± 0.9 weeks, p = 0.000) and birth weight (971 ± 240 g vs. 1262 ± 335 g, p = 0.000) of the BPD group were lower than those of the control group. The present study found that the BPD group had high levels of IL-1β, IL-4, IL-6, IL-8, and TNF-α, whereas IL-10 was decreased. The Shannon diversity index of the BPD group was lower. The relative abundances of Proteobacteria in BPD group increased significantly from days 3–7 to days 14–28, while the Firmicutes was decreased. On days 14–28, the relative abundances of Proteobacteria in BPD group were significantly higher than those in the control group, while the Firmicutes was lower. Conclusion Bronchopulmonary dysplasia could be influenced by gut microbiota through inflammatory responses. More studies are needed to explore the imbalance of cytokines and microbiome in BPD infants and whether it could be reversed by probiotics. This study provided a novel perspective for treating BPD.
Collapse
Affiliation(s)
- Zhenjie Zhang
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingjing Jiang
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenghong Li
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weilin Wan
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
35
|
Zhang S, Luan X, Li H, Jin Z. Insulin-like growth factor-1: A potential target for bronchopulmonary dysplasia treatment (Review). Exp Ther Med 2022; 23:191. [PMID: 35126694 PMCID: PMC8794548 DOI: 10.3892/etm.2022.11114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/08/2021] [Indexed: 11/05/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common respiratory disorder among preterm infants, particularly low-birth-weight infants (LBWIs) and very-low-birth-weight infants (VLBWIs). Although BPD was first reported 50 years ago, no specific drugs or efficient measures are yet available for prevention or treatment. Insulin-like growth factor-1 (IGF-1) belongs to the insulin family. It promotes mitosis and stimulates cell proliferation and DNA synthesis, the primary factors involved in pulmonary development during the fetal and postnatal periods. Several studies have reported that IGF-1 exerts certain effects on BPD genesis and progression by regulating BPD-related biological processes. In addition, exogenous addition of IGF-1 can alleviate lung inflammation, cell apoptosis and eliminate alveolar development disorders in children with BPD. These findings suggest that IGF-1 could be a new target for treating BPD. Here, we summarize and analyze the definition, pathogenesis, and research status of BPD, as well as the pathogenesis of IGF-1 in BPD and the latest findings in related biological processes.
Collapse
Affiliation(s)
- Shujian Zhang
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Xue Luan
- Department of Pediatrics, First Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Huiwen Li
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Zhengyong Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
36
|
Slagle C, Gist KM, Starr MC, Hemmelgarn TS, Goldstein SL, Kent AL. Fluid Homeostasis and Diuretic Therapy in the Neonate. Neoreviews 2022; 23:e189-e204. [PMID: 35229135 DOI: 10.1542/neo.23-3-e189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Understanding physiologic water balance and homeostasis mechanisms in the neonate is critical for clinicians in the NICU as pathologic fluid accumulation increases the risk for morbidity and mortality. In addition, once this process occurs, treatment is limited. In this review, we will cover fluid homeostasis in the neonate, explain the implications of prematurity on this process, discuss the complexity of fluid accumulation and the development of fluid overload, identify mitigation strategies, and review treatment options.
Collapse
Affiliation(s)
- Cara Slagle
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Katja M Gist
- Division of Cardiology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Michelle C Starr
- Division of Pediatric Nephrology, Department of Pediatrics, Indiana University School of Medicine and Riley Hospital for Children, Indianapolis, IN
| | - Trina S Hemmelgarn
- Division of Pharmacology, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, College of Pharmacy, Cincinnati, OH
| | - Stuart L Goldstein
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center and the University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Alison L Kent
- Department of Pediatrics, University of Rochester, NY, and Australian National University Medical School, Canberra, ACT, Australia
| |
Collapse
|
37
|
Qing C, Ziyun L, Xuefei Y, Xinyi Z, Xindong X, Jianhua F. Protective Effects of 18β-Glycyrrhetinic Acid on Neonatal Rats with Hyperoxia Exposure. Inflammation 2022; 45:1224-1238. [PMID: 34989920 DOI: 10.1007/s10753-021-01616-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 11/05/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common devastating pulmonary complication in preterm infants. Supplemental oxygen is a lifesaving therapeutic measure used for premature infants with pulmonary insufficiency. However, oxygen toxicity is a significant trigger for BPD. Oxidative stress disrupts lung development, accompanied by increased pro-inflammatory cytokines and chemokines expression and immune cells infiltration in lung tissue. Licorice, a typical traditional herbal medicine, is commonly used in the medicine and food industries. 18β-Glycyrrhetinic acid (18β-GA), a primary active ingredient of licorice, has powerful anti-oxidative and anti-inflammatory effects. This study aimed to determine whether 18β-GA has a protective effect on neonatal rats with hyperoxia exposure. Newborn Sprague-Dawley rats were kept in either 21% (normoxia) or 80% O2 (hyperoxia) continuously from postnatal day (PN) 1 to 14. 18β-GA was injected intragastrically at 50 or 100 mg/kg body weight once a day from PN 1 to 14. We examined the body weight and alveolar development and measured ROS level and the markers of pulmonary inflammation. Mature-IL-1β and NF-κB pathway proteins, and the NLRP3 inflammasome, were assessed; concurrently, caspase-1 activity was measured. Our results indicated that hyperoxia resulted in alveolar simplification and decreased bodyweight of neonatal rats. Hyperoxia increased ROS level and pulmonary inflammation and activated NF-κB and the NLRP3 inflammasome. 18β-GA treatment inhibited the activation of NF-κB and the NLRP3 inflammasome, decreased ROS level and pulmonary inflammation, improved alveolar development, and increased the bodyweight of neonatal rats with hyperoxia exposure. Our study demonstrates that 18β-GA has a protective effect on neonatal rats with hyperoxia exposure.
Collapse
Affiliation(s)
- Cai Qing
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Liu Ziyun
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Yu Xuefei
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Zhao Xinyi
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Xue Xindong
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China
| | - Fu Jianhua
- Department of Pediatrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, China.
| |
Collapse
|
38
|
Ozdemir R, Gokce IK, Tekin S, Cetin Taslidere A, Turgut H, Tanbek K, Gul CC, Deveci MF, Aslan M. The protective effects of apocynin in hyperoxic lung injury in neonatal rats. Pediatr Pulmonol 2022; 57:109-121. [PMID: 34581514 DOI: 10.1002/ppul.25707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 11/08/2022]
Abstract
AIM Inflammation and oxidate stress are significant factors in the pathogenesis of bronchopulmonary dysplasia (BPD). The aim of this study is to investigate the efficacy of apocynin (APO), an anti-inflammatory, antioxidant, and antiapoptotic drug, in the prophylaxis of neonatal hyperoxic lung injury. METHOD This experimental study included 40 neonatal rats divided into the control, APO, BPD, and BPD + APO groups. The control and APO groups were kept in a normal room environment, while the BPD and BPD + APO groups were kept in a hyperoxic environment. The rats in the APO and BPD + APO groups were administered intraperitoneal APO, while the control and BPD rats were administered ordinary saline. At the end of the trial, lung tissue was evaluated with respect to the degree of histopathological injury, apoptosis, oxidant and antioxidant capacity, and severity of inflammation. RESULT The BPD and BPD + APO groups exhibited higher mean histopathological injury and alveolar macrophage scores compared to the control and APO groups. Both scores were lower in the BPD + APO group in comparison to the BPD group. The BPD + APO group had a significantly lower average of TUNEL positive cells than the BPD group. The lung tissue examination indicated significantly higher levels of mean malondialdehyde (MDA), total oxidant status (TOS), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) in the BPD group compared to the control and APO groups. While the TNF-α and IL-1β levels of the BPD + APO group were similar to that of the control group, the MDA and TOS levels were higher compared to the controls and lower compared to the BPD group. The BPD group demonstrated significantly lower levels/activities of mean total antioxidant status, glutathione reductase, superoxide dismutase, glutathione peroxidase in comparison to the control and APO groups. While the mean antioxidant enzyme activity of the BPD + APO group was lower than the control group, it was significantly higher compared to the BPD group. CONCLUSION This is the first study in the literature to reveal through an experimental neonatal hyperoxic lung injury that APO, an anti-inflammatory, antioxidant, and antiapoptotic drug, exhibits protective properties against the development of BPD.
Collapse
Affiliation(s)
- Ramazan Ozdemir
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Ismail Kursat Gokce
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Suat Tekin
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Asli Cetin Taslidere
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Hatice Turgut
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Kevser Tanbek
- Department of Physiology, Inonu University School of Medicine, Malatya, Turkey
| | - Cemile Ceren Gul
- Department of Histology and Embryology, Inonu University School of Medicine, Malatya, Turkey
| | - Mehmet Fatih Deveci
- Division of Neonatology, Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| | - Mehmet Aslan
- Department of Pediatrics, Inonu University School of Medicine, Malatya, Turkey
| |
Collapse
|
39
|
Belfort MB, Ramel SE, Martin CR, Fichorova R, Kuban KCK, Heeren T, Fry RC, O'Shea TM. Systemic Inflammation in the First 2 Weeks after Birth as a Determinant of Physical Growth Outcomes in Hospitalized Infants with Extremely Low Gestational Age. J Pediatr 2022; 240:37-43.e1. [PMID: 34508750 PMCID: PMC8712377 DOI: 10.1016/j.jpeds.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To examine associations of systemic inflammation with growth outcomes at neonatal intensive care unit discharge or transfer among infants with extremely low gestational ages. STUDY DESIGN We studied 850 infants at born at 23-27 weeks of gestation. We defined inflammatory protein elevation as the highest quartile of C-reactive protein (CRP), Interleukin (IL)-6, tumor necrosis factor-∝, or IL-8 on postnatal days 1, 7, and 14. We compared z-scores of weight, length, and head circumference at neonatal intensive care unit discharge or transfer between infants with vs without inflammatory protein elevation, adjusting in linear regression for birth size z-score, sex, gestational age, diet, comorbidities, medications, and length of hospitalization. RESULTS The mean gestational age was 25 weeks (range, 23-27 weeks) and birth weight z-score 0.14 (range, -2.73 to 3.28). Infants with a high CRP on day 7 had lower weights at discharge or transfer (-0.17 z-score; 95% CI, -0.27 to -0.06) than infants without CRP elevation, with similar results on day 14. Infants with CRP elevation on day 14 were also shorter (-0.21 length z-scores; 95% CI, -0.38 to -0.04), and had smaller head circumferences (-0.18 z-scores; 95% CI, -0.33 to -0.04) at discharge or transfer. IL-6 elevation on day 14 was associated with lower weight (-0.12; 95% CI, -0.22 to -0.02); IL-6 elevation on day 7 was associated with shorter length (-0.27; 95% CI, -0.43 to -0.12). Tumor necrosis factor-∝ and IL-8 elevation on day 14 were associated with a lower weight at discharge or transfer. CONCLUSIONS Postnatal systemic inflammation may contribute to impaired nutrient accretion during a critical period in development in infants with extremely low gestational ages.
Collapse
Affiliation(s)
- Mandy B Belfort
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| | - Sara E Ramel
- University of Minnesota School of Medicine, Minneapolis, MN
| | - Camilia R Martin
- Harvard Medical School, Boston, MA; Beth Israel Deaconess Medical Center, Boston, MA
| | - Raina Fichorova
- Harvard Medical School, Boston, MA; Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA
| | | | | | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, University of North Carolina School of Medicine, Chapel Hill, NC
| | - T Michael O'Shea
- Division of Neonatal-Perinatal Medicine, University of North Carolina School of Medicine, Chapel Hill, NC
| |
Collapse
|
40
|
Tolg C, Messam BJA, McCarthy JB, Nelson AC, Turley EA. Hyaluronan Functions in Wound Repair That Are Captured to Fuel Breast Cancer Progression. Biomolecules 2021; 11:1551. [PMID: 34827550 PMCID: PMC8615562 DOI: 10.3390/biom11111551] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/14/2022] Open
Abstract
Signaling from an actively remodeling extracellular matrix (ECM) has emerged as a critical factor in regulating both the repair of tissue injuries and the progression of diseases such as metastatic cancer. Hyaluronan (HA) is a major component of the ECM that normally functions in tissue injury to sequentially promote then suppress inflammation and fibrosis, a duality in which is featured, and regulated in, wound repair. These essential response-to-injury functions of HA in the microenvironment are hijacked by tumor cells for invasion and avoidance of immune detection. In this review, we first discuss the numerous size-dependent functions of HA and emphasize the multifunctional nature of two of its receptors (CD44 and RHAMM) in regulating the signaling duality of HA in excisional wound healing. This is followed by a discussion of how HA metabolism is de-regulated in malignant progression and how targeting HA might be used to better manage breast cancer progression.
Collapse
Affiliation(s)
- Cornelia Tolg
- London Regional Cancer Program, Lawson Health Research Institute, London, ON N6A 5W9, Canada;
| | - Britney Jodi-Ann Messam
- Department Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada;
| | - James Benjamin McCarthy
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Andrew Cook Nelson
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Eva Ann Turley
- London Regional Cancer Program, Lawson Health Research Institute, Department Oncology, Biochemistry and Surgery, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| |
Collapse
|
41
|
RHAMM Is a Multifunctional Protein That Regulates Cancer Progression. Int J Mol Sci 2021; 22:ijms221910313. [PMID: 34638654 PMCID: PMC8508827 DOI: 10.3390/ijms221910313] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 01/09/2023] Open
Abstract
The functional complexity of higher organisms is not easily accounted for by the size of their genomes. Rather, complexity appears to be generated by transcriptional, translational, and post-translational mechanisms and tissue organization that produces a context-dependent response of cells to specific stimuli. One property of gene products that likely increases the ability of cells to respond to stimuli with complexity is the multifunctionality of expressed proteins. Receptor for hyaluronan-mediated motility (RHAMM) is an example of a multifunctional protein that controls differential responses of cells in response-to-injury contexts. Here, we trace its evolution into a sensor-transducer of tissue injury signals in higher organisms through the detection of hyaluronan (HA) that accumulates in injured microenvironments. Our goal is to highlight the domain and isoform structures that generate RHAMM's function complexity and model approaches for targeting its key functions to control cancer progression.
Collapse
|
42
|
Lv Y, Li Y, Wang J, Li M, Zhang W, Zhang H, Shen Y, Li C, Du Y, Jiang L. MiR-382-5p suppresses M1 macrophage polarization and inflammatory response in response to bronchopulmonary dysplasia through targeting CDK8: Involving inhibition of STAT1 pathway. Genes Cells 2021; 26:772-781. [PMID: 34228857 DOI: 10.1111/gtc.12883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/11/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is an inflammation-related respiratory disorder in infants. MiR-382-5p has displayed low expression in developing lungs with BPD, while the effect of miR-382-5p on BPD remains elusive. Here, a hyperoxia (85% oxygen)-induced BPD model in neonatal mice was established. On postnatal days 10 and 15, hyperoxia reduced miR-382-5p expression in lungs of mice. Besides, CDK8, CD68 and CD86 levels were elevated on day 15 after birth, implying the involvement of CDK8 in M1 macrophage polarization. In addition, in vitro injury in RAW264.7 macrophages was induced by IFN-γ and LPS stimulation. Lentivirus-encoding miR-382-5p decreased CDK8 expression, alleviated the production of inflammatory cytokines TNF-α, IL-1β and IL-6, and restricted the levels of CD40 and CD86 in response to IFN-γ and LPS. Moreover, miR-382-5p inhibited the phosphorylation of STAT1. Luciferase reporter assay verified that miR-382-5p might target the 3'UTR of CDK8. Rescue assays revealed that CDK8 reversed the mitigating roles of miR-382-5p in inflammatory response and M1 macrophage polarization, as reflected by increased IL-6 and CD40 levels. Taken together, these findings indicate that miR-382-5p may suppress M1 macrophage activation and inflammatory response via inhibiting CDK8, thereby regulating the development of BPD, which is possibly mediated by STAT1 signaling.
Collapse
Affiliation(s)
- Yuanyuan Lv
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Pediatrics, Baoding First Central Hospital, Baoding, China
| | - Yang Li
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiangya Wang
- Department of Pediatrics, Hebei General Hospital, Shijiazhuang, China
| | - Mei Li
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenhao Zhang
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huifen Zhang
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ying Shen
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chao Li
- Department of Pediatrics, Baoding First Central Hospital, Baoding, China
| | - Yuan Du
- Department of Laboratory Medicine, Baoding No. 1 Hospital of TCM, Baoding, China
| | - Lian Jiang
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
43
|
Qing C, Xinyi Z, Xuefei Y, Xindong X, Jianhua F. The Specific Connexin 43-Inhibiting Peptide Gap26 Improved Alveolar Development of Neonatal Rats With Hyperoxia Exposure. Front Pharmacol 2021; 12:587267. [PMID: 34290603 PMCID: PMC8287833 DOI: 10.3389/fphar.2021.587267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 04/20/2021] [Indexed: 11/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common devastating pulmonary complication in preterm infants. Alveolar maldevelopment is the crucial pathological change of BPD highly associated with oxidative stress–mediated excessive apoptosis. Cellular injury can be propagated and amplified by gap junction (GJ)–mediated intercellular communication. Connexin 43 (Cx43) is the most ubiquitous and critical GJ protein. Gap26 is a specific Cx43 mimic peptide, playing as a Cx43-GJ inhibitor. We hypothesized that Cx43-GJ was involved in alveolar maldevelopment of BPD via amplifying oxidative stress signaling and inducing excessive apoptosis. Neonatal Sprague Dawley rats were kept in either normoxia (21% O2) or hyperoxia (85% O2) continuously from postnatal day (PN) 1 to 14 in the presence or absence of Gap26. Moreover, RLE-6TN cells (type II alveolar epithelial cells of rats) were cultured in vitro under normoxia (21% O2) or hyperoxia (85% O2). RLE-6TN cells were treated by N-acetyl cysteine (NAC) (a kind of reactive oxygen species (ROS) scavenger) or Gap26. Morphological properties of lung tissue are detected. Markers associated with Cx43 expression, ROS production, the activity of the ASK1-JNK/p38 signaling pathway, and apoptotic level are detected in vivo and in vitro, respectively. In vitro, the ability of GJ-mediated intercellular communication was examined by dye-coupling assay. In vitro, our results demonstrated ROS increased Cx43 expression and GJ-mediated intercellular communication and Gap26 treatment decreased ROS production, inhibited ASK1-JNK/p38 signaling, and decreased apoptosis. In vivo, we found that hyperoxia exposure resulted in increased ROS production and Cx43 expression, activated ASK1-JNK/p38 signaling, and induced excessive apoptosis. However, Gap26 treatment reversed these changes, thus improving alveolar development in neonatal rats with hyperoxia exposure. In summary, oxidative stress increased Cx43 expression and Cx43-GJ–mediated intercellular communication. And Cx43-GJ–mediated intercellular communication amplified oxidative stress signaling, inducing excessive apoptosis via the ASK1-JNK/p38 signaling pathway. The specific connexin 43–inhibiting peptide Gap26 was a novel therapeutic strategy to improve the alveolar development of BPD.
Collapse
Affiliation(s)
- Cai Qing
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhao Xinyi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Xuefei
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xue Xindong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fu Jianhua
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
44
|
Extracellular Vesicles in Organ Fibrosis: Mechanisms, Therapies, and Diagnostics. Cells 2021; 10:cells10071596. [PMID: 34202136 PMCID: PMC8305303 DOI: 10.3390/cells10071596] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is the unrelenting deposition of excessively large amounts of insoluble interstitial collagen due to profound matrigenic activities of wound-associated myofibroblasts during chronic injury in diverse tissues and organs. It is a highly debilitating pathology that affects millions of people globally and leads to decreased function of vital organs and increased risk of cancer and end-stage organ disease. Extracellular vesicles (EVs) produced within the chronic wound environment have emerged as important vehicles for conveying pro-fibrotic signals between many of the cell types involved in driving the fibrotic response. On the other hand, EVs from sources such as stem cells, uninjured parenchymal cells, and circulation have in vitro and in vivo anti-fibrotic activities that have provided novel and much-needed therapeutic options. Finally, EVs in body fluids of fibrotic individuals contain cargo components that may have utility as fibrosis biomarkers, which could circumvent current obstacles to fibrosis measurement in the clinic, allowing fibrosis stage, progression, or regression to be determined in a manner that is accurate, safe, minimally-invasive, and conducive to repetitive testing. This review highlights the rapid and recent progress in our understanding of EV-mediated fibrotic pathogenesis, anti-fibrotic therapy, and fibrosis staging in the lung, kidney, heart, liver, pancreas, and skin.
Collapse
|
45
|
Schiliro M, Bartman CM, Pabelick C. Understanding hydrogen sulfide signaling in neonatal airway disease. Expert Rev Respir Med 2021; 15:351-372. [PMID: 33086886 PMCID: PMC10599633 DOI: 10.1080/17476348.2021.1840981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Airway dysfunction leading to chronic lung disease is a common consequence of premature birth and mechanisms responsible for early and progressive airway remodeling are not completely understood. Current therapeutic options are only partially effective in reducing the burden of neonatal airway disease and premature decline of lung function. Gasotransmitter hydrogen sulfide (H2S) has been recently recognized for its therapeutic potential in lung diseases. AREAS COVERED Contradictory to its well-known toxicity at high concentrations, H2S has been characterized to have anti-inflammatory, antioxidant, and antiapoptotic properties at physiological concentrations. In the respiratory system, endogenous H2S production participates in late lung development and exogenous H2S administration has a protective role in a variety of diseases such as acute lung injury and chronic pulmonary hypertension and fibrosis. Literature searches performed using NCBI PubMed without publication date limitations were used to construct this review, which highlights the dichotomous role of H2S in the lung, and explores its promising beneficial effects in lung diseases. EXPERT OPINION The emerging role of H2S in pathways involved in chronic lung disease of prematurity along with its recent use in animal models of BPD highlight H2S as a potential novel candidate in protecting lung function following preterm birth.
Collapse
Affiliation(s)
- Marta Schiliro
- Departments of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | | | - Christina Pabelick
- Departments of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
46
|
Zhang L, Bai X, Yan W. LncRNA-MALAT1, as a biomarker of neonatal BPD, exacerbates the pathogenesis of BPD by targeting miR-206. Am J Transl Res 2021; 13:462-479. [PMID: 33594304 PMCID: PMC7868848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/18/2020] [Indexed: 06/12/2023]
Abstract
Neonatal bronchopulmonary dysplasia (BPD) is one of the common causes of premature birth complications, which is caused by lung dysplasia. Long non-coding RNA (LncRNA) has been proved to be related to BPD and other disease processes, but the molecular mechanism of metastasis-related lung adenocarcinoma transcript 1 (MALAT1) in BPD has not been fully understood. This study focused on exploring the clinical and molecular mechanism of MALAT1 in neonatal BPD, aiming to provide new insights for the management of neonatal BPD. In our study, we first found that serum MALAT1 was up-regulated in neonatal BPD and severe BPD. Further, through receiver operating characteristic curve (ROC) analysis, it was found that the area under the curve of MALAT1 for differentiating neonatal BPD from severe BPD was 0.943 and 0.866, respectively. Then, we established BPD models in vivo and in vitro with C57BL/6J mice and BEAS-2B cells, and found that MALAT1 was also highly expressed in them and increased with the induction time of the models. Pathological evaluation confirmed that down-regulating MALAT1 or up-regulating miR-206 might improve the pathological condition of BPD. Obvious inflammatory response, oxidative stress and up-regulated apoptosis were observed in BPD models in vivo and in vitro. However, after MALAT1 knockdown treatment, the above abnormal phenomena were alleviated to varying degrees. Furthermore, we also found that MALAT1 has a targeted relationship with miR-206, and miR-206 is down-regulated in BPD in vivo and in vitro. Down-regulating miR-206 could also eliminate the anti-BPD effect after knocking down MALAT1. The above results indicated that MALAT1 has the potential as a blood biomarker of neonatal BPD, and MALAT1-miR-206 axis mediates BPD process, which may be a new target for neonatal BPD treatment.
Collapse
Affiliation(s)
- Limin Zhang
- Neonatal Intensive Care Unit, Zhoukou Central Hospital Zhoukou 466000, Henan, China
| | - Xueyan Bai
- Neonatal Intensive Care Unit, Zhoukou Central Hospital Zhoukou 466000, Henan, China
| | - Wenpeng Yan
- Neonatal Intensive Care Unit, Zhoukou Central Hospital Zhoukou 466000, Henan, China
| |
Collapse
|
47
|
Gentle SJ, Ahmed KA, Yi N, Morrow CD, Ambalavanan N, Lal CV, Patel RP. Bronchopulmonary dysplasia is associated with reduced oral nitrate reductase activity in extremely preterm infants. Redox Biol 2021; 38:101782. [PMID: 33166868 PMCID: PMC7658701 DOI: 10.1016/j.redox.2020.101782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 01/08/2023] Open
Abstract
Oral microbiome mediated nitrate reductase (NR) activity regulates nitric oxide (NO) bioavailability and signaling. While deficits in NO-bioavailability impact several morbidities of extreme prematurity including bronchopulmonary dysplasia (BPD), whether oral NR activity is associated with morbidities of prematurity is not known. We characterized NR activity in extremely preterm infants from birth until 34 weeks' post menstrual age (PMA), determined whether changes in the oral microbiome contribute to changes in NR activity, and determined whether changes in NR activity correlated with disease. In this single center prospective cohort study (n = 28), we observed two surprising findings: (1) NR activity unexpectedly peaked at 29 weeks' PMA (p < 0.05) and (2) when infants were stratified for BPD status, infants who developed BPD had significantly less NR activity at 29 weeks' PMA compared to infants who did not develop BPD. Oral microbiota and NR activity may play a role in BPD development in extremely preterm infants, indicating potential for disease prediction and therapeutic targeting.
Collapse
Affiliation(s)
- Samuel J Gentle
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Khandaker A Ahmed
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nengjun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Casey D Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Charitharth V Lal
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
48
|
Abstract
Purpose of Review Detail normal adaptive immune maturation during fetal and neonatal life and review the clinical implications of arrested immune development. Recent Findings Advancements in the field of immunology have enabled investigations of the adaptive immunity starting during fetal life. New insights have drawn important distinctions between the neonatal and adult immune systems. The presence of diverse immunologic responses in the perinatal period suggests the importance of in utero immune development. Disruption of immune maturation due to premature birth may have significant implications for clinical pathology. Summary Establishing protective adaptive immunity during the perinatal period is critical for effective immune responses later in life. Preterm infants are susceptible to aberrant immune system maturation and inflammatory immune responses have been associated with the development of necrotizing enterocolitis (NEC) and bronchopulmonary dysplasia (BPD). Improving our understanding of how immune responses contribute to the pathogenesis of NEC and BPD may offer new opportunities for future treatment and prevention of these diseases.
Collapse
|
49
|
Rocha G. Pulmonary pneumatoceles in neonates. Pediatr Pulmonol 2020; 55:2532-2541. [PMID: 32691976 DOI: 10.1002/ppul.24969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/12/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022]
Abstract
Pulmonary pneumatoceles were relatively common in neonates in the pre-surfactant era. In the current era of surfactant, noninvasive and gentle invasive ventilation there is a paucity of data on clinical characteristics and outcomes of pneumatoceles in neonates. The lesion generally resolves spontaneously, but a few cases follow a complicated course with formation of extensive and expanding lesions. To better understand the pathophysiology, clinical significance, natural history, complications, treatment options and prognosis of pulmonary pneumatoceles in neonates, an extensive research was performed on the databases of medical literature. The information collected in this review is important for the clinicians in decision-making, especially in the most difficult cases.
Collapse
Affiliation(s)
- Gustavo Rocha
- Department of Neonatology, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
50
|
Shrestha AK, Menon RT, El-Saie A, Barrios R, Reynolds C, Shivanna B. Interactive and independent effects of early lipopolysaccharide and hyperoxia exposure on developing murine lungs. Am J Physiol Lung Cell Mol Physiol 2020; 319:L981-L996. [PMID: 32901520 DOI: 10.1152/ajplung.00013.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH) is a chronic infantile lung disease that lacks curative therapies. Infants with BPD-associated PH are often exposed to hyperoxia and additional insults such as sepsis that contribute to disease pathogenesis. Animal models that simulate these scenarios are necessary to develop effective therapies; therefore, we investigated whether lipopolysaccharide (LPS) and hyperoxia exposure during saccular lung development cooperatively induce experimental BPD-PH in mice. C57BL/6J mice were exposed to normoxia or 70% O2 (hyperoxia) during postnatal days (PNDs) 1-5 and intraperitoneally injected with varying LPS doses or a vehicle on PNDs 3-5. On PND 14, we performed morphometry, echocardiography, and gene and protein expression studies to determine the effects of hyperoxia and LPS on lung development, vascular remodeling and function, inflammation, oxidative stress, cell proliferation, and apoptosis. LPS and hyperoxia independently and cooperatively affected lung development, inflammation, and apoptosis. Growth rate and antioxidant enzyme expression were predominantly affected by LPS and hyperoxia, respectively, while cell proliferation and vascular remodeling and function were mainly affected by combined exposure to LPS and hyperoxia. Mice treated with lower LPS doses developed adaptive responses and hyperoxia exposure did not worsen their BPD phenotype, whereas those mice treated with higher LPS doses displayed the most severe BPD phenotype when exposed to hyperoxia and were the only group that developed PH. Collectively, our data suggest that an additional insult such as LPS may be necessary for models utilizing short-term exposure to moderate hyperoxia to recapitulate human BPD-PH.
Collapse
Affiliation(s)
- Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Ahmed El-Saie
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Corey Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, Texas
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|