1
|
Sankar MN, Chock V, Myers F, Davis AS, McDonald S, Rysavy MA, Laughon M, Bhombal S, Van Meurs KP, Das A, Benitz WE, Watterberg K. Patent Ductus Arteriosus, Hydrocortisone, and Outcome among Infants Born Extremely Preterm: Secondary Analysis of the Hydrocortisone Trial. J Pediatr 2025; 281:114535. [PMID: 40090543 DOI: 10.1016/j.jpeds.2025.114535] [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: 10/25/2024] [Revised: 02/20/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE To examine whether hydrocortisone (HC) modified the relationship of patent ductus arteriosus (PDA) to outcomes among infants born extremely preterm and enrolled in the National Institute of Child Health and Human Development Neonatal Research Network (NRN) HC trial. STUDY DESIGN This was a posthoc secondary analysis of infants born <30 weeks' gestation and enrolled in the NRN HC Trial. The primary outcome was moderate to severe bronchopulmonary dysplasia (BPD) or death. Secondary outcomes included moderate to severe BPD, death, necrotizing enterocolitis, late-onset sepsis, days of mechanical ventilation, oxygen supplementation, Z-scores for growth, home oxygen, BPD severity, neurodevelopmental impairment, and moderate to severe cerebral palsy. Analyses for interaction between PDA (defined as treatment to achieve PDA closure) and HC were performed for the primary and secondary outcomes. RESULTS Of 800 infants enrolled in the NRN HC trial, PDA was treated in 198 HC treated and 197 placebo-treated infants. HC did not modify the relationship of PDA with BPD or death (P = .93). Regardless of HC treatment, PDA was associated with a significant increase in duration of ventilatory support, oxygen supplementation at 36 weeks postmenstrual age (PMA), BPD severity, decreased weight-for-age Z-score at 36 weeks PMA, moderate to severe BPD, or death at 36 weeks PMA and home oxygen support. CONCLUSIONS HC after the second postnatal week did not alter the relationship between PDA and BPD or death among infants born extremely preterm. PDA was associated with several adverse outcomes regardless of HC treatment.
Collapse
Affiliation(s)
- Meera N Sankar
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA.
| | - Valerie Chock
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | - Faith Myers
- Children's Health Care of Minnesota, Minneapolis, MN
| | - Alexis S Davis
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | | | - Matthew A Rysavy
- Department of Pediatrics, McGovern Medical School at UTHealth Houston, Houston, TX
| | - Matthew Laughon
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Shazia Bhombal
- Emory University/Children's Healthcare of Atlanta, Atlanta, GA
| | - Krisa P Van Meurs
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | - Abhik Das
- RTI International, Research Triangle Park, NC
| | - William E Benitz
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| | - Kristi Watterberg
- Division of Neonatology, Department of Pediatrics, University of New Mexico Health Sciences, Albuquerque, NM
| |
Collapse
|
2
|
Schaeffer SF, Omer B, Vachharajani A, Panchangam C. Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension: Screening and Management. Neoreviews 2025; 26:e316-e327. [PMID: 40306676 DOI: 10.1542/neo.26-5-013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/21/2025] [Indexed: 05/02/2025]
Abstract
Screening for pulmonary hypertension (PH) in infants with bronchopulmonary dysplasia (BPD) was recommended by the American Thoracic Society in 2015. However, the definition of BPD has since changed. This review summarizes the current definition of BPD, the recommendations and tools for screening for PH in infants with BPD, and the various treatment options and outcomes in infants with BPD-PH.
Collapse
Affiliation(s)
| | - Bwaar Omer
- School of Medicine, University of Missouri, Columbia, Missouri
| | | | | |
Collapse
|
3
|
Schmiedl A, Mühlfeld C. Morphological and molecular aspects of lung development. Histol Histopathol 2025; 40:411-430. [PMID: 39344418 DOI: 10.14670/hh-18-807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Healthy breathing relies on normal morphological and functional development of the lung. This includes different prenatal and postnatal developmental stages. Depending on species and postnatal behavior as nest escapers or nest squatters, the duration of individual developmental phases and the state of differentiation of the lungs at birth differ. However, the sequence and morphology of the lung developmental stages are similar in all mammals, so knowledge gained from animal models about development-specific genetic control and regulatory mechanisms can be translated in principle to the human lung. Functional lung development comprises the maturation of the surfactant system, which is closely linked to the morphological development of the pulmonary acini. Although a number of reviews are found in the literature, a presentation that integrates the morphological and molecular regulatory mechanisms is missing. Therefore, the aim of this article was to provide an up-to-date comprehensive review of the main morphological steps and regulatory mechanisms of lung development, including clinical aspects related to developmental disorders.
Collapse
Affiliation(s)
- Andreas Schmiedl
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research, Hannover, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research, Hannover, Germany
| |
Collapse
|
4
|
Takahashi Y, Fee EL, Takahashi T, Usuda H, Ikeda H, Carter SW, Saito Y, Sato S, Mochii N, Chemtob S, Olson DM, Keelan JA, Kumagai Y, Choolani MA, Illanes SE, Saito M, Kemp MW. Interleukin-1 Receptor Antagonists Partially Inhibited Histological Injury but Not Tissue Inflammation in a Sheep Model of Pregnancy. Reprod Sci 2025; 32:1213-1227. [PMID: 39953369 DOI: 10.1007/s43032-024-01781-8] [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: 08/26/2024] [Accepted: 12/27/2024] [Indexed: 02/17/2025]
Abstract
Intrauterine inflammation is a significant cause of early preterm birth and fetal injury. There is a lack of effective interventions for intrauterine inflammation. This study aimed to determine whether direct fetal treatment with IL-1 receptor antagonists (IL-1RA), specifically anakinra (competitive IL-1RA) or rytvela (allosteric IL-1RA), could reduce intrauterine inflammation caused by intraamniotic injection of E. coli lipopolysaccharides (LPS) in a sheep model of pregnancy. We hypothesized the fetal intramuscular administration of IL1-RA therapy would comprehensively resolve intrauterine inflammation caused by LPS in the pregnant sheep model. Date-mated Merino ewes carrying single fetuses were randomized into four groups: LPS Group (10 mg intraamniotic LPS injection followed by saline), RYTVELA Group (10 mg LPS injection followed by 5 mg rytvela), ANAKINRA Group (LPS injection followed by 100 mg anakinra), and SALINE Group (saline injection followed by saline). All LPS-exposed fetuses had elevated bilirubin levels, leukopenia, and increased inflammatory mediators IL-1β, IL-8, tumour necrosis factor alpha (TNFα), and monocyte chemoattractant protein 1 (MCP-1) in amniotic fluid and lung tissue. Both anakinra and rytvela treatments reduced immunocyte infiltration in chorioamniotic membranes and lungs, and microglial staining, and increased the oligodendrocyte staining, but did not significantly resolve overall inflammation compared to the SALINE Group. In conclusion, fetal intramuscular administration of anakinra and rytvela did not effectively resolve intrauterine inflammation but showed potential in reducing tissue invasion and brain injury markers. These findings suggest that modest inflammation reduction may protect against brain injury and preterm birth, though no additional benefit was observed compared to intraamniotic IL-1RA treatment.
Collapse
Affiliation(s)
- Yuki Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan.
- Department of Obstetrics, Tohoku University, 1-1 Seiryomachi Aobaku, Sendai, Miyagi, Japan.
| | - Erin L Fee
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Tsukasa Takahashi
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Haruo Usuda
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Hideyuki Ikeda
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sean W Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuya Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shinichi Sato
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Noriyoshi Mochii
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
| | - Sylvain Chemtob
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Canada
| | - David M Olson
- Departments of Obstetrics and Gynecology, Pediatrics, and Physiology, University of Alberta, Alberta, Canada
| | - Jeffrey A Keelan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Yusaku Kumagai
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics, Tohoku University, 1-1 Seiryomachi Aobaku, Sendai, Miyagi, Japan
| | - Mahesh A Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sebastian E Illanes
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, University of the Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Masatoshi Saito
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Department of Obstetrics, Tohoku University, 1-1 Seiryomachi Aobaku, Sendai, Miyagi, Japan
| | - Matthew W Kemp
- Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Japan
- Division of Obstetrics and Gynecology, The University of Western Australia, Perth, Western Australia, Australia
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Veterinary and Life Sciences, Murdoch University, Perth, Australia
- Women and Infants Research Foundation, Level 2, Carson House, King Edward Memorial Hospital, Perth, Australia
| |
Collapse
|
5
|
Lingappan K, Matthay MA. Unlocking the Therapeutic Code of Mesenchymal Stromal Cells. Am J Respir Crit Care Med 2025; 211:313-315. [PMID: 39847724 PMCID: PMC11936120 DOI: 10.1164/rccm.202412-2349ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/22/2025] [Indexed: 01/25/2025] Open
Affiliation(s)
- Krithika Lingappan
- Children's Hospital of Philadelphia University of Pennsylvania Philadelphia, Pennsylvania
| | - Michael A Matthay
- Cardiovascular Research Institute University of California San Francisco, California
| |
Collapse
|
6
|
Ako AA, Ismaiel A, Rastogi S. Electrical impedance tomography in neonates: a review. Pediatr Res 2025:10.1038/s41390-025-03929-x. [PMID: 39987341 DOI: 10.1038/s41390-025-03929-x] [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: 09/11/2024] [Revised: 01/10/2025] [Accepted: 01/26/2025] [Indexed: 02/24/2025]
Abstract
Appropriate interventions informed by real-time assessment of pulmonary function in mechanically ventilated critically ill neonates can reduce the incidence of bronchopulmonary dysplasia, pneumothorax, intraventricular hemorrhage and other complications of newborn life. The respiratory system in neonates is uniquely different from older children, and its physiological and anatomic attributes increase neonatal vulnerability to respiratory distress and eventual failure. While significant advancements have been made in developing respiratory support for neonates, such support is accompanied by inherent risks to their delicate lungs. Ventilator-associated lung injury poses a critical concern that can be potentially decreased with more precise, non-invasive, non-radiating, bedside methods for assessing neonatal pulmonary function in real time. Electrical impedance tomography (EIT) is one such tool, with immense potential for real-time pulmonary function monitoring in neonates. Still relatively new and in the earliest stages of clinical adoption, EIT use in neonatal critical care has been reported in several studies. This review discusses the basic features of EIT, its distinct advantages over traditional pulmonary function monitoring tools, the scope of its adoption in neonatal clinical practice, challenges associated with clinical adoption, and prospects for future applications. IMPACT: 1. Individualized care assisted by bedside pulmonary function monitoring can positively impact neonatal critical care and outcomes. 2. Electrical impedance tomography (EIT) has the potential to improve neonatal pulmonary function monitoring and treatment outcomes. 3. Electrical impedance tomography can be adopted as a part of routine neonatal respiratory critical care, especially in the population of patients most at risk for bronchopulmonary dysplasia and acute respiratory complications.
Collapse
Affiliation(s)
- Ako A Ako
- Division of Neonatology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, 10467, USA
| | - Ahmed Ismaiel
- Division of Neonatology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, 10467, USA
| | - Shantanu Rastogi
- Division of Neonatology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York, 10467, USA.
| |
Collapse
|
7
|
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
|
8
|
Callahan KP, Farrell K, Gibbs K, Kielt MJ, Morris H, Nilan K, Thomas S, DeMauro SB. Childhood outcomes following discharge from a referral bronchopulmonary dysplasia program. J Perinatol 2024; 44:1832-1838. [PMID: 38937610 PMCID: PMC11969747 DOI: 10.1038/s41372-024-02035-w] [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: 02/29/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
OBJECTIVES The primary objective of this study was to profile the childhood health, development, and health-related quality of life (HR QoL) for children with the most severe bronchopulmonary dysplasia (BPD), those discharged from a quaternary referral program. STUDY DESIGN We collected cross-sectional data through telephone interviews with 282 families of children ages 18 months to 11 years who had been discharged from a BPD referral program. RESULTS Respiratory morbidities were near universal, with 42% of children ever having required a tracheostomy and severity of these morbidities correlated with parent-reported health and QoL. Developmental morbidities were also marked: 97% required an individualized educational plan. While respiratory morbidities and overall health improved over time, developmental morbidities were increasingly prominent, resulting in lower quality of life. CONCLUSIONS Among children referred to a quaternary BPD program, respiratory and developmental morbidities are on numerous counts more severe than any reported in the literature.
Collapse
Affiliation(s)
- Katharine P Callahan
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Medical Ethics and Health Policy, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Kathryn Farrell
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen Gibbs
- Department of Medical Ethics and Health Policy, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Heidi Morris
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen Nilan
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Sara B DeMauro
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
9
|
Poggi C, Sarcina D, Miselli F, Ciarcià M, Dani C. Neonatal Sequential Organ Failure Assessment Score Predicts Respiratory Outcomes in Preterm Newborns with Late-Onset Sepsis: A Retrospective Study. Neonatology 2024; 122:56-65. [PMID: 39500295 DOI: 10.1159/000539526] [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: 01/31/2024] [Accepted: 05/24/2024] [Indexed: 02/11/2025]
Abstract
INTRODUCTION Neonatal sequential organ failure assessment (nSOFA) score predicts mortality in preterm newborns. The aim of the study was to assess whether nSOFA score could predict respiratory outcomes in preterm infants with late-onset sepsis (LOS). METHODS This retrospective, observational, single-center study enrolled infants with gestational age <32 weeks born between January 2016 and June 2023 who experienced an episode of LOS during NICU stay. The primary outcome was death or bronchopulmonary dysplasia (BPD); secondary outcomes were BPD, death or mechanical ventilation (MV) on day 5 after the onset of LOS, and MV on day 5 after the onset of LOS. The nSOFA score was assessed at the onset of LOS and after 6 ± 1, 12 ± 3, and 24 ± 3 h. RESULTS Neonatal SOFA score was significantly higher in patients who developed each outcome versus those who did not at all timings. Maximal nSOFA score during the first 24 h after onset of LOS was an independent predictive factor for death or BPD (p = 0.007), BPD (p = 0.009), and death or MV on day 5 (p = 0.009), areas under the curve (AUC) were 0.740 (95% CI: 0.656-0.828), 0.700 (95% CI: 0.602-0.800), and 0.800 (95% CI: 0.710-0.889), respectively. Maximal nSOFA score also predicted moderate to severe BPD (p = 0.019) and death or moderate to severe BPD (p < 0.001). Maximal nSOFA ≥4 was associated with odds ratio (OR) of 7.37 (95% CI: 2.42-22.44) for death or BPD, 4.86 (95% CI: 1.54-15.28) for BPD, and 7.99 (95% CI: 3.47-18.36) for death or MV on day 5. AUC of the predicting model was 0.895 (95% CI: 0.801-0.928) for BPD, 0.897 (95% CI: 0.830-0.939) for death or BPD, 0.904 (95% CI: 0.851-0.956) for MV on day 5, 0.923 (95% CI: 0.892-0.973) for death or MV on day 5. CONCLUSION Maximal nSOFA score during the first 24 h after the onset of LOS predicts respiratory outcomes and allows identification of patients who may crucially benefit from lung-protective measures.
Collapse
Affiliation(s)
- Chiara Poggi
- Neonatal Intensive Care Unit, Department of Mother and Child Care, Careggi University Hospital, Florence, Italy
| | - Davide Sarcina
- Department of Mother and Child Health, University of Florence, Florence, Italy
| | - Francesca Miselli
- Department of Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Martina Ciarcià
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Carlo Dani
- Neonatal Intensive Care Unit, Department of Mother and Child Care, Careggi University Hospital, Florence, Italy
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| |
Collapse
|
10
|
El Sherif DF, El Raggal NM, Nasef MW, Saleh GA, Youssef NH, Metwally MH. Oral ibuprofen versus placebo in closure of patent ductus arteriosus in preterm neonates, a randomized control trial. J Neonatal Perinatal Med 2024; 17:810-820. [PMID: 40016984 DOI: 10.1177/19345798241302264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
BACKGROUND A consensus on treatment of patent ductus arteriosus (PDA) in preterm neonates remains elusive. Conservative management has recently gained popularity due to medical treatment lacking the evidence of benefit in terms of mortality and morbidity. OBJECTIVE This study compares the efficacy of the standard dose of oral Ibuprofen versus placebo in the closure of PDA in preterm neonates ≤34 weeks gestation. METHODS Eighty enrolled preterm neonates with a hemodynamically significant PDA (hsPDA) were randomized to either receive an early 3-day course oral ibuprofen or placebo (1:1). The open-label option and an extended oral ibuprofen therapy were offered if there were concerns over patient's clinical condition being attributed to PDA and only if showing evidence of systemic and/or pulmonary hyperperfusion. RESULTS No significant difference in PDA closure between both groups (62.5% vs 65% in Ibuprofen & placebo group respectively, p = .816). No significant difference was observed between groups in mortality (p = 1), Bronchopulmonary dysplasia (BPD) (p = 1), or necrotizing enterocolitis (NEC) (0.5). Placebo group required a longer duration of inotropic support (median of 10 vs 7.5 days in Ibuprofen group, p = .013) and were prescribed more diuretic therapy (27 vs 18 in Ibuprofen group, p = .043). CONCLUSION The standard dose of oral ibuprofen was comparable to placebo in early treatment of hsPDA in preterm neonates less than 34 weeks in terms of effect on ductal closure after one course of treatment, incidence of mortality, and morbidities like NEC, and BPD. Failure of ductal closure itself was associated in both groups with increased mortality, failure to reach full feeds, and more use of vasopressors and inotropes.
Collapse
|
11
|
Leek C, Cantu A, Sonti S, Gutierrez MC, Eldredge L, Sajti E, Xu HN, Lingappan K. Role of sex as a biological variable in neonatal alveolar macrophages. Redox Biol 2024; 75:103296. [PMID: 39098263 PMCID: PMC11345582 DOI: 10.1016/j.redox.2024.103296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
The lung macrophages play a crucial role in health and disease. Sexual dimorphism significantly impacts the phenotype and function of tissue-resident macrophages. The primary mechanisms responsible for sexually dimorphic outcomes in bronchopulmonary dysplasia (BPD) remain unidentified. We tested the hypothesis that biological sex plays a crucial role in the transcriptional state of alveolar macrophages, using neonatal murine hyperoxia-induced lung injury as a relevant model for human BPD. The effects of neonatal hyperoxia exposure (95 % FiO2, PND1-5: saccular stage) on the lung myeloid cells acutely after injury and during normoxic recovery were measured. Alveolar macrophages (AM) from room air- and hyperoxia exposed from male and female neonatal murine lungs were subjected to bulk-RNA Sequencing. AMs are significantly depleted in the hyperoxia-exposed lung acutely after injury, with subsequent recovery in both sexes. The transcriptome of the alveolar macrophages is impacted by neonatal hyperoxia exposure and by sex as a biological variable. Pathways related to DNA damage and interferon-signaling were positively enriched in female AMs. Metabolic pathways related to glucose and carbohydrate metabolism were positively enriched in the male AMs, while oxidative phosphorylation was negatively enriched. These pathways were shared with monocytes and airway macrophages from intubated male and female human premature neonates.
Collapse
Affiliation(s)
- Connor Leek
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Abiud Cantu
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Shilpa Sonti
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Manuel Cantu Gutierrez
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Laurie Eldredge
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Washington School of Medicine, Seattle Children's Hospital, WA, USA
| | - Eniko Sajti
- Department of Pediatrics, Division of Neonatology, University of California San Diego, San Diego, CA, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Krithika Lingappan
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA.
| |
Collapse
|
12
|
De Luca D, Ferraioli S, Watterberg KL, Baud O, Gualano MR. Hydrocortisone in very preterm neonates for BPD prevention: meta-analysis and effect size modifiers. Arch Dis Child Fetal Neonatal Ed 2024; 109:481-487. [PMID: 38237961 DOI: 10.1136/archdischild-2023-326254] [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: 08/27/2023] [Accepted: 01/10/2024] [Indexed: 08/18/2024]
Abstract
OBJECTIVES To clarify if systemic hydrocortisone, in protocols allowing to start it before the 15th day of life, prevents bronchopulmonary dysplasia (BPD) or other adverse outcomes in very preterm neonates, and to identify any possible effect size modifiers. STUDY DESIGN Systematic review and meta-analysis following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Additional analyses included meta-regressions and review of biological plausibility. RESULTS Seven trials were included, they were of general good quality and accounted for a total of 2193 infants. Hydrocortisone treatment did not reduce BPD (risk ratio (RR) 0.84 (95% CI 0.64 to 1.04)), but heterogeneity was evident (I2=51.6%). The effect size for BPD is greatest for 10-12 days duration of treatment (β=0.032 (0.01), p=0.007) and tended to be greater in patients with chorioamnionitis (β=-1.5 (0.841), p=0.07). Hydrocortisone treatment may significantly reduce mortality (RR 0.75 (95% CI 0.59 to 0.91)), there is no heterogeneity (I2=0) and the reduction tended to be greater in males (β=-0.06 (0.03), p=0.07). Hydrocortisone may significantly reduce necrotising enterocolitis (NEC; RR 0.72 (95% CI 0.53 to 0.92)); there is neither heterogeneity (I2=0%) nor any effect size modifiers. Hydrocortisone did not affect other adverse outcomes of prematurity. CONCLUSIONS Systemic hydrocortisone may be considered, on a case-by-case evaluation, to reduce mortality and NEC, while it does not affect BPD. There are some potential effect size modifiers for mortality and BPD which should be addressed in future explanatory trials. PROSPERO REGISTRATION NUMBER CRD42023400520.
Collapse
Affiliation(s)
- Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, APHP, Paris Sac;ay University Hospitals, 'A. Beclere' Medical Centre, Paris, France
- Pathophysiology and Therapeutic Innovation Unit-U999, Paris-Saclay University, Gif-sur-Yvette, France
| | | | - Kristi L Watterberg
- Department of Paediatrics, University of New Mexico, Albuquerque, New Mexico, USA
| | - Olivier Baud
- DFEA, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | | |
Collapse
|
13
|
House M, Lagoski M, DiGeronimo R, Eldredge LC, Manimtim W, Baker CD, Coghill C, Fernandes CJ, Griffiths P, Ibrahim J, Kielt MJ, Lagatta J, Machry JS, Mikhael M, Vyas-Read S, Weems MF, Yallapragada SG, Murthy K, Nelin LD. Interdisciplinary clinical bronchopulmonary dysplasia programs: development, evolution, and maturation. J Perinatol 2024:10.1038/s41372-024-02049-4. [PMID: 39020027 DOI: 10.1038/s41372-024-02049-4] [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: 01/24/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024]
Abstract
Multidisciplinary bronchopulmonary dysplasia (BPD) programs provide improved and consistent medical management, care of the developing infant, family support, and smoother transitions in care resulting in improved survival, pulmonary, and extra-pulmonary outcomes. This review summarizes the benefits of interdisciplinary BPD management, as well as strategies for initial programmatic development, program growth, and maintenance at centers across the United States factoring in institutional, provider, and parent reported goals that were derived from a consensus conference on BPD management.
Collapse
Affiliation(s)
- Melissa House
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA.
| | - Megan Lagoski
- Ann & Robert H. Lurie Children's Hospitals of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert DiGeronimo
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - Laurie C Eldredge
- Division of Pulmonology and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle Children's Hospital, Seattle, WA, USA
| | - Winston Manimtim
- Divison of Neonatology, Children's Mercy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Christopher D Baker
- Section of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Carl Coghill
- Children's of Alabama, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caraciolo J Fernandes
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - John Ibrahim
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew J Kielt
- Comprehensive Center for BPD, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Joana Silva Machry
- Division of Neonatology, Maternal Fetal & Neonatal Institute at Johns Hopkins All Children's Hospital St. Petersburg, St. Petersburg, FL, USA
| | - Michel Mikhael
- Division of Neonatology, Children's Hospital of Orange County, Orange, CA, USA
| | - Shilpa Vyas-Read
- Division of Neonatology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mark F Weems
- Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Karna Murthy
- Ann & Robert H. Lurie Children's Hospitals of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Children's Hospitals Neonatal Consortium, Dover, DE, USA
| | - Leif D Nelin
- Comprehensive Center for BPD, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
14
|
Jeong J, Lee Y, Han J, Kang E, Kim D, Kim KS, Kim EAR, Lee BS, Jung E. Mitochondrial DNA mutations in extremely preterm infants with bronchopulmonary dysplasia. Gene 2024; 910:148337. [PMID: 38432533 DOI: 10.1016/j.gene.2024.148337] [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: 10/01/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a serious chronic lung disease affecting extremely preterm infants. While mitochondrial dysfunction has been investigated in various medical conditions, limited research has explored mitochondrial DNA (mtDNA) gene mutations, specifically in BPD. This study aimed to evaluate mitochondrial mtDNA gene mutations in extremely preterm infants with BPD. In this prospective observational study, we enrolled a cohort of extremely preterm infants diagnosed with BPD. Clinical data were collected to provide comprehensive patient profiles. Peripheral blood mononuclear cells were isolated from whole-blood samples obtained within a defined timeframe. Subsequently, mtDNA extraction and sequencing using next-generation sequencing technology were performed to identify mtDNA gene mutations. Among the cohort of ten extremely preterm infants with BPD, mtDNA sequencing revealed the presence of mutations in seven patients, resulting in a total of twenty-one point mutations. Notably, many of these mutations were identified in loci associated with critical components of the respiratory chain complexes, vital for proper mitochondrial function and cellular energy production. This pilot study provides evidence of mtDNA point mutations in a subset of extremely preterm infants with BPD. These findings suggest a potential association between mitochondrial dysfunction and the pathogenesis of BPD. Further extensive investigations are warranted to unravel the mechanisms underlying mtDNA mutations in BPD.
Collapse
Affiliation(s)
- Jiyoon Jeong
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Yeonmi Lee
- Department of Convergence Medicine and Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea.
| | - Jongsuk Han
- Department of Convergence Medicine and Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea.
| | - Eunju Kang
- Department of Convergence Medicine and Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea.
| | - Deokhoon Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Ki-Soo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Ellen Ai-Rhan Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Byong Sop Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Euiseok Jung
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Moreira A, Noronha M, Joy J, Bierwirth N, Tarriela A, Naqvi A, Zoretic S, Jones M, Marotta A, Valadie T, Brick J, Winter C, Porter M, Decker I, Bruschettini M, Ahuja SK. Rates of bronchopulmonary dysplasia in very low birth weight neonates: a systematic review and meta-analysis. Respir Res 2024; 25:219. [PMID: 38790002 PMCID: PMC11127341 DOI: 10.1186/s12931-024-02850-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
IMPORTANCE Large-scale estimates of bronchopulmonary dysplasia (BPD) are warranted for adequate prevention and treatment. However, systematic approaches to ascertain rates of BPD are lacking. OBJECTIVE To conduct a systematic review and meta-analysis to assess the prevalence of BPD in very low birth weight (≤ 1,500 g) or very low gestational age (< 32 weeks) neonates. DATA SOURCES A search of MEDLINE from January 1990 until September 2019 using search terms related to BPD and prevalence was performed. STUDY SELECTION Randomized controlled trials and observational studies evaluating rates of BPD in very low birth weight or very low gestational age infants were eligible. Included studies defined BPD as positive pressure ventilation or oxygen requirement at 28 days (BPD28) or at 36 weeks postmenstrual age (BPD36). DATA EXTRACTION AND SYNTHESIS Two reviewers independently conducted all stages of the review. Random-effects meta-analysis was used to calculate the pooled prevalence. Subgroup analyses included gestational age group, birth weight group, setting, study period, continent, and gross domestic product. Sensitivity analyses were performed to reduce study heterogeneity. MAIN OUTCOMES AND MEASURES Prevalence of BPD defined as BPD28, BPD36, and by subgroups. RESULTS A total of 105 articles or databases and 780,936 patients were included in this review. The pooled prevalence was 35% (95% CI, 28-42%) for BPD28 (n = 26 datasets, 132,247 neonates), and 21% (95% CI, 19-24%) for BPD36 (n = 70 studies, 672,769 neonates). In subgroup meta-analyses, birth weight category, gestational age category, and continent were strong drivers of the pooled prevalence of BPD. CONCLUSIONS AND RELEVANCE This study provides a global estimation of BPD prevalence in very low birth weight/low gestation neonates.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA.
| | - Michelle Noronha
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Jooby Joy
- University of Texas Rio Grande Valley School of Medicine, Edinburg, TX, USA
| | - Noah Bierwirth
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Aina Tarriela
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Aliha Naqvi
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Sarah Zoretic
- Department of Pediatrics, University of Texas Southwestern, Dallas, TX, USA
| | - Maxwell Jones
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Ali Marotta
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Taylor Valadie
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Jonathan Brick
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Caitlyn Winter
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Melissa Porter
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Isabelle Decker
- Department of Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | | | - Sunil K Ahuja
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, USA
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, TX, USA
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
16
|
Al Mandhari H, Khan A, Al Saadi A, AboAnza M, Rizvi SG, Panchatcharam SM, Abdulatif M, Al Qassabi S, Quach S. Prevalence, Severity Patterns and Risk Factors of Bronchopulmonary Dysplasia in Preterm Infants Younger than 32 Weeks of Gestation in a Tertiary Centre in Oman. Sultan Qaboos Univ Med J 2024; 24:259-267. [PMID: 38828256 PMCID: PMC11139367 DOI: 10.18295/squmj.3.2024.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/31/2024] [Accepted: 02/18/2024] [Indexed: 06/05/2024] Open
Abstract
Objectives This study aimed to determine the rate and severity patterns of bronchopulmonary dysplasia (BPD) and identify antenatal and postnatal factors associated with BPD in preterm infants <32 weeks of gestational age (GA). Methods This retrospective observational study included preterm neonates <32 weeks of gestation admitted into the neonatal intensive care unit between January 2010 and December 2017 at Sultan Qaboos University Hospital, Muscat, Oman. A data set of antenatal and perinatal factors were collected. BPD was defined as the need for oxygen and/or respiratory support at 36 weeks post-menstrual age (PMA). Infants with and without BPD were compared in their antenatal and perinatal factors. Results A total of 589 preterm infants <32 weeks were admitted. Among them, 505 (85.7%) survived to 36 weeks' PMA and 90 (17.8%) had BPD. The combined BPD and mortality rate was 28.4%. Grades 1, 2 and 3 BPD constituted 77.8%, 7.8% and 14.4%, respectively. BPD was associated with lower GA, lower birth weight, need for intubation at resuscitation, lower Apgar scores, longer duration of ventilation, surfactant therapy and higher rates of neonatal morbidities. On binary logistic regression analysis, predictors of BPD were longer duration of ventilation, intraventricular haemorrhage (IVH) and necrotising enterocolitis (NEC). Conclusion In an Omani centre, 17.8% of preterm infants (<32 weeks GA) developed BPD. Various perinatal and neonatal factors were associated with BPD. However, longer duration of ventilation, IVH grades 1 and 2 and NEC stages II and III were significant predictors. Future multicentre research is necessary to provide the overall prevalence of BPD in Oman to help optimise the resources for BPD prevention and management in preterm infants.
Collapse
Affiliation(s)
- Hilal Al Mandhari
- Child Health Department, Sultan Qaboos University Hospital, Muscat, Oman
| | - Ashfaq Khan
- Child Health Department, Sultan Qaboos University Hospital, Muscat, Oman
| | | | - Mazen AboAnza
- Child Health Department, Sultan Qaboos University Hospital, Muscat, Oman
| | - Syed G.A. Rizvi
- Department of Family Medicine & Public Health, Sultan Qaboos University, Muscat, Oman
| | | | - Mohammed Abdulatif
- Child Health Department, Sultan Qaboos University Hospital, Muscat, Oman
| | | | - Shirley Quach
- Department of Respiratory Therapy, SickKids Research Institute, Toronto, Canada
| |
Collapse
|
17
|
Salsabili S, Chan ADC, Ukwatta E. Multiresolution semantic segmentation of biological structures in digital histopathology. J Med Imaging (Bellingham) 2024; 11:037501. [PMID: 38737492 PMCID: PMC11086667 DOI: 10.1117/1.jmi.11.3.037501] [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: 12/13/2022] [Revised: 10/30/2023] [Accepted: 11/17/2023] [Indexed: 05/14/2024] Open
Abstract
Purpose Semantic segmentation in high-resolution, histopathology whole slide images (WSIs) is an important fundamental task in various pathology applications. Convolutional neural networks (CNN) are the state-of-the-art approach for image segmentation. A patch-based CNN approach is often employed because of the large size of WSIs; however, segmentation performance is sensitive to the field-of-view and resolution of the input patches, and balancing the trade-offs is challenging when there are drastic size variations in the segmented structures. We propose a multiresolution semantic segmentation approach, which is capable of addressing the threefold trade-off between field-of-view, computational efficiency, and spatial resolution in histopathology WSIs. Approach We propose a two-stage multiresolution approach for semantic segmentation of histopathology WSIs of mouse lung tissue and human placenta. In the first stage, we use four different CNNs to extract the contextual information from input patches at four different resolutions. In the second stage, we use another CNN to aggregate the extracted information in the first stage and generate the final segmentation masks. Results The proposed method reported 95.6%, 92.5%, and 97.1% in our single-class placenta dataset and 97.1%, 87.3%, and 83.3% in our multiclass lung dataset for pixel-wise accuracy, mean Dice similarity coefficient, and mean positive predictive value, respectively. Conclusions The proposed multiresolution approach demonstrated high accuracy and consistency in the semantic segmentation of biological structures of different sizes in our single-class placenta and multiclass lung histopathology WSI datasets. Our study can potentially be used in automated analysis of biological structures, facilitating the clinical research in histopathology applications.
Collapse
Affiliation(s)
- Sina Salsabili
- Carleton University, Department of Systems and Computer Engineering, Ottawa, Ontario, Canada
| | - Adrian D. C. Chan
- Carleton University, Department of Systems and Computer Engineering, Ottawa, Ontario, Canada
- University of Ottawa, School of Human Kinetics, Ottawa, Ontario, Canada
- Bruyère Research Institute, Ottawa, Ontario, Canada
| | - Eranga Ukwatta
- Carleton University, Department of Systems and Computer Engineering, Ottawa, Ontario, Canada
- University of Guelph, School of Engineering, Guelph, Ontario, Canada
| |
Collapse
|
18
|
Reçica R, Kryeziu I, Thaçi Q, Avtanski D, Mladenov M, Basholli-Salihu M, Sopi RB. Protective Effects of Resveratrol Against Airway Hyperreactivity, Oxidative Stress, and Lung Inflammation in a Rat Pup Model of Bronchopulmonary Dysplasia. Physiol Res 2024; 73:239-251. [PMID: 38710061 PMCID: PMC11081184 DOI: 10.33549/physiolres.935239] [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: 09/25/2023] [Accepted: 01/10/2024] [Indexed: 12/20/2024] Open
Abstract
Oxygen therapy provides an important treatment for preterm and low-birth-weight neonates, however, it has been shown that prolonged exposure to high levels of oxygen (hyperoxia) is one of the factors contributing to the development of bronchopulmonary dysplasia (BPD) by inducing lung injury and airway hyperreactivity. There is no effective therapy against the adverse effects of hyperoxia. Therefore, this study was undertaken to test the hypothesis that natural phytoalexin resveratrol will overcome hyperoxia-induced airway hyperreactivity, oxidative stress, and lung inflammation. Newborn rats were exposed to hyperoxia (fraction of inspired oxygen - FiO2>95 % O2) or ambient air (AA) for seven days. Resveratrol was supplemented either in vivo (30 mg·kg-1·day-1) by intraperitoneal administration or in vitro to the tracheal preparations in an organ bath (100 mikroM). Contractile and relaxant responses were studied in tracheal smooth muscle (TSM) using the in vitro organ bath system. To explain the involvement of nitric oxide in the mechanisms of the protective effect of resveratrol against hyperoxia, a nitric oxide synthase inhibitor - Nomega-nitro-L-arginine methyl ester (L-NAME), was administered in some sets of experiments. The superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities and the tumor necrosis factor-alpha (TNF-alpha) and interleukin-1beta (IL-1beta) levels in the lungs were determined. Resveratrol significantly reduced contraction and restored the impaired relaxation of hyperoxia-exposed TSM (p<0.001). L-NAME reduced the inhibitory effect of resveratrol on TSM contractility, as well as its promotion relaxant effect (p<0.01). Resveratrol preserved the SOD and GPx activities and decreased the expression of TNF-alpha and IL-1beta in hyperoxic animals. The findings of this study demonstrate the protective effect of resveratrol against hyperoxia-induced airway hyperreactivity and lung damage and suggest that resveratrol might serve as a therapy to prevent the adverse effects of neonatal hyperoxia. Keywords: Bronchopulmonary dysplasia, Hyperoxia, Airway hyperreactivity, Resveratrol, Pro-inflammatory cytokines.
Collapse
Affiliation(s)
- R Reçica
- Faculty of Medicine, University of Prishtina, Prishtina, Kosovo
| | | | | | | | | | | | | |
Collapse
|
19
|
McGinn EA, Bye E, Gonzalez T, Sosa A, Bilodeaux J, Seedorf G, Smith BJ, Abman SH, Mandell EW. Antenatal Endotoxin Induces Dysanapsis in Experimental Bronchopulmonary Dysplasia. Am J Respir Cell Mol Biol 2024; 70:283-294. [PMID: 38207120 PMCID: PMC11478127 DOI: 10.1165/rcmb.2023-0157oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/10/2024] [Indexed: 01/13/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD), the chronic lung disease of prematurity, is characterized by impaired lung development with sustained functional abnormalities due to alterations of airways and the distal lung. Although clinical studies have shown striking associations between antenatal stress and BPD, little is known about the underlying pathogenetic mechanisms. Whether dysanapsis, the concept of discordant growth of the airways and parenchyma, contributes to late respiratory disease as a result of antenatal stress is unknown. We hypothesized that antenatal endotoxin (ETX) impairs juvenile lung function as a result of altered central airway and distal lung structure, suggesting the presence of dysanapsis in this preclinical BPD model. Fetal rats were exposed to intraamniotic ETX (10 μg) or saline solution (control) 2 days before term. We performed extensive structural and functional evaluation of the proximal airways and distal lung in 2-week-old rats. Distal lung structure was quantified by stereology. Conducting airway diameters were measured using micro-computed tomography. Lung function was assessed during invasive ventilation to quantify baseline mechanics, response to methacholine challenge, and spirometry. ETX-exposed pups exhibited distal lung simplification, decreased alveolar surface area, and decreased parenchyma-airway attachments. ETX-exposed pups exhibited decreased tracheal and second- and third-generation airway diameters. ETX increased respiratory system resistance and decreased lung compliance at baseline. Only Newtonian resistance, specific to large airways, exhibited increased methacholine reactivity in ETX-exposed pups compared with controls. ETX-exposed pups had a decreased ratio of FEV in 0.1 second to FVC and a normal FEV in 0.1 second, paralleling the clinical definition of dysanapsis. Antenatal ETX causes abnormalities of the central airways and distal lung growth, suggesting that dysanapsis contributes to abnormal lung function in juvenile rats.
Collapse
Affiliation(s)
- Elizabeth A. McGinn
- Pediatric Heart Lung Center, Department of Pediatrics
- Department of Pediatric Critical Care Medicine
| | - Elisa Bye
- Pediatric Heart Lung Center, Department of Pediatrics
| | | | - Alexander Sosa
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jill Bilodeaux
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Bradford J. Smith
- Pediatric Heart Lung Center, Department of Pediatrics
- Department of Pediatric Pulmonary and Sleep Medicine, and
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Steven H. Abman
- Pediatric Heart Lung Center, Department of Pediatrics
- Department of Pediatric Pulmonary and Sleep Medicine, and
| | - Erica W. Mandell
- Pediatric Heart Lung Center, Department of Pediatrics
- Department of Neonatology, University of Colorado School of Medicine, Aurora, Colorado; and
| |
Collapse
|
20
|
Matlock DN, Ratcliffe SJ, Courtney SE, Kirpalani H, Firestone K, Stein H, Dysart K, Warren K, Goldstein MR, Lund KC, Natarajan A, Demissie E, Foglia EE. The Diaphragmatic Initiated Ventilatory Assist (DIVA) trial: study protocol for a randomized controlled trial comparing rates of extubation failure in extremely premature infants undergoing extubation to non-invasive neurally adjusted ventilatory assist versus non-synchronized nasal intermittent positive pressure ventilation. Trials 2024; 25:201. [PMID: 38509583 PMCID: PMC10953115 DOI: 10.1186/s13063-024-08038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Invasive mechanical ventilation contributes to bronchopulmonary dysplasia (BPD), the most common complication of prematurity and the leading respiratory cause of childhood morbidity. Non-invasive ventilation (NIV) may limit invasive ventilation exposure and can be either synchronized or non-synchronized (NS). Pooled data suggest synchronized forms may be superior. Non-invasive neurally adjusted ventilatory assist (NIV-NAVA) delivers NIV synchronized to the neural signal for breathing, which is detected with a specialized catheter. The DIVA (Diaphragmatic Initiated Ventilatory Assist) trial aims to determine in infants born 240/7-276/7 weeks' gestation undergoing extubation whether NIV-NAVA compared to non-synchronized nasal intermittent positive pressure ventilation (NS-NIPPV) reduces the incidence of extubation failure within 5 days of extubation. METHODS This is a prospective, unblinded, pragmatic, multicenter phase III randomized clinical trial. Inclusion criteria are preterm infants 24-276/7 weeks gestational age who were intubated within the first 7 days of life for at least 12 h and are undergoing extubation in the first 28 postnatal days. All sites will enter an initial run-in phase, where all infants are allocated to NIV-NAVA, and an independent technical committee assesses site performance. Subsequently, all enrolled infants are randomized to NIV-NAVA or NS-NIPPV at extubation. The primary outcome is extubation failure within 5 days of extubation, defined as any of the following: (1) rise in FiO2 at least 20% from pre-extubation for > 2 h, (2) pH ≤ 7.20 or pCO2 ≥ 70 mmHg; (3) > 1 apnea requiring positive pressure ventilation (PPV) or ≥ 6 apneas requiring stimulation within 6 h; (4) emergent intubation for cardiovascular instability or surgery. Our sample size of 478 provides 90% power to detect a 15% absolute reduction in the primary outcome. Enrolled infants will be followed for safety and secondary outcomes through 36 weeks' postmenstrual age, discharge, death, or transfer. DISCUSSION The DIVA trial is the first large multicenter trial designed to assess the impact of NIV-NAVA on relevant clinical outcomes for preterm infants. The DIVA trial design incorporates input from clinical NAVA experts and includes innovative features, such as a run-in phase, to ensure consistent technical performance across sites. TRIAL REGISTRATION www. CLINICALTRIALS gov , trial identifier NCT05446272 , registered July 6, 2022.
Collapse
Affiliation(s)
- David N Matlock
- University of Arkansas for Medical Sciences, 4301 W. Markham St., Slot 512-5B, Little Rock, AR, 72205, USA.
- University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | | | | | - Haresh Kirpalani
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- McMaster University, Hamilton, ON, Canada
| | | | | | - Kevin Dysart
- Nemours Children's Health Wilmington, Philadelphia, PA, USA
| | - Karen Warren
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Aruna Natarajan
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ejigayehu Demissie
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth E Foglia
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
21
|
Dubner SE, Rickerich L, Bruckert L, Poblaciones RV, Sproul D, Scala M, Feldman HM, Travis KE. Early, low-dose hydrocortisone and near-term brain connectivity in extremely preterm infants. Pediatr Res 2024; 95:1028-1034. [PMID: 38030826 DOI: 10.1038/s41390-023-02903-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Postnatal steroids are used to prevent bronchopulmonary dysplasia in extremely preterm infants but may have adverse effects on brain development. We assessed connectivity metrics of major cerebral and cerebellar white matter pathways at near-term gestational age among infants who did or did not receive a standardized regimen of hydrocortisone during the first 10 days of life. METHODS Retrospective cohort study. PARTICIPANTS Infants born <28 weeks: Protocol group (n = 33) received at least 50% and not more than 150% of an intended standard dose of 0.5 mg/kg hydrocortisone twice daily for 7 days, then 0.5 mg/kg per day for 3 days; Non-Protocol group (n = 22), did not receive protocol hydrocortisone or completed <50% of the protocol dose. We assessed group differences in near-term diffusion MRI mean fractional anisotropy (FA) and mean diffusivity (MD) across the corticospinal tract, inferior longitudinal fasciculus, corpus callosum and superior cerebellar peduncle. RESULTS Groups were comparable in gestational age, post-menstrual age at scan, medical complications, bronchopulmonary dysplasia, and necrotizing enterocolitis. No significant large effect group differences were identified in mean FA or MD in any cerebral or cerebellar tract. CONCLUSION(S) Low dose, early, postnatal hydrocortisone was not associated with significant differences in white matter tract microstructure at near-term gestational age. IMPACT This study compared brain microstructural connectivity as a primary outcome among extremely preterm infants who did or did not receive early postnatal hydrocortisone. Low dose hydrocortisone in the first 10 days of life was not associated with significant differences in white matter microstructure in major cerebral and cerebellar pathways. Hydrocortisone did not have a significant effect on early brain white matter circuits.
Collapse
Affiliation(s)
- Sarah E Dubner
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Lucy Rickerich
- Program in Human Biology, Stanford University, Stanford, CA, USA
| | - Lisa Bruckert
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Rocío Velasco Poblaciones
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Dawson Sproul
- Program in Human Biology, Stanford University, Stanford, CA, USA
| | - Melissa Scala
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Heidi M Feldman
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Katherine E Travis
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
22
|
Kim J, Villarreal M, Arya S, Hernandez A, Moreira A. Bridging the Gap: Exploring Bronchopulmonary Dysplasia through the Lens of Biomedical Informatics. J Clin Med 2024; 13:1077. [PMID: 38398389 PMCID: PMC10889493 DOI: 10.3390/jcm13041077] [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: 12/28/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD), a chronic lung disease predominantly affecting premature infants, poses substantial clinical challenges. This review delves into the promise of biomedical informatics (BMI) in reshaping BPD research and care. We commence by highlighting the escalating prevalence and healthcare impact of BPD, emphasizing the necessity for innovative strategies to comprehend its intricate nature. To this end, we introduce BMI as a potent toolset adept at managing and analyzing extensive, diverse biomedical data. The challenges intrinsic to BPD research are addressed, underscoring the inadequacies of conventional approaches and the compelling need for data-driven solutions. We subsequently explore how BMI can revolutionize BPD research, encompassing genomics and personalized medicine to reveal potential biomarkers and individualized treatment strategies. Predictive analytics emerges as a pivotal facet of BMI, enabling early diagnosis and risk assessment for timely interventions. Moreover, we examine how mobile health technologies facilitate real-time monitoring and enhance patient engagement, ultimately refining BPD management. Ethical and legal considerations surrounding BMI implementation in BPD research are discussed, accentuating issues of privacy, data security, and informed consent. In summation, this review highlights BMI's transformative potential in advancing BPD research, addressing challenges, and opening avenues for personalized medicine and predictive analytics.
Collapse
Affiliation(s)
- Jennifer Kim
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| | - Mariela Villarreal
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| | - Shreyas Arya
- Division of Neonatal-Perinatal Medicine, Dayton Children’s Hospital, Dayton, OH 45404, USA
| | - Antonio Hernandez
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| | - Alvaro Moreira
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| |
Collapse
|
23
|
Stading R, Swanson L, Xia G, Jiang W, Wang L, Couroucli X, Lingappan K, Moorthy B. Prenatal exposure to polycyclic aromatic hydrocarbons (PAHs) augments neonatal hyperoxic lung injury: Role of cytochrome P450 (CYP)1A1, 1A2, and 1B1. Free Radic Biol Med 2024; 211:35-46. [PMID: 38081439 DOI: 10.1016/j.freeradbiomed.2023.12.001] [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: 06/14/2023] [Revised: 11/29/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
Pregnant women exposed to polycyclic aromatic hydrocarbons (PAHs) are at increased risk for premature delivery. Premature infants often require supplemental oxygen, a known risk factor for bronchopulmonary dysplasia (BPD). Cytochrome P450 (CYP) enzymes have been implicated in hyperoxic lung injury. We hypothesize that prenatal PAH exposure exacerbates oxygen-mediated lung injury in neonatal mice, and that this effect is differentially altered in mice lacking the gene for (Cyp)1a1, 1a2, or 1b1. Timed pregnant wild type (WT) (C57BL/6J) mice were orally administered a PAH mixture of benzo[a]pyrene (BP) and benzo[b]fluoranthene (BbF) or the vehicle corn oil (CO) once daily on gestational days 16-19, and the dose response on postnatal lung injury was examined. In addition, timed pregnant mice with one of four genotypes, WT, Cyp1a1-null, Cyp1a2-null, and Cyp1b1-null, were treated orally with CO or PAH on gestational days 16-19 and exposed to hyperoxia or room air for 14 days. Lung injury was assessed on PND15 by radial alveolar count (RAC) and mean linear intercept (MLI) Gene expression of DNA repair genes in lung and liver were measured. Results showed that neonatal hyperoxic lung injury is augmented by prenatal PAH exposure in a dose-dependent manner. This effect was differentially altered in the Cyp-null mice, with Cyp1a2-null showing the greatest extent of lung injury. We concluded that newborn mice exposed to PAH in utero had more significant lung injury in response to hyperoxia than non-PAH exposed pups, and that CYP1A1 and CYP1A2 are protective against lung injury while CYP1B1 augments lung injury.
Collapse
Affiliation(s)
- Rachel Stading
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Lauren Swanson
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Guobin Xia
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Weiwu Jiang
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Lihua Wang
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Xanthi Couroucli
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Krithika Lingappan
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Bhagavatula Moorthy
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States.
| |
Collapse
|
24
|
Melan N, Pradat P, Godbert I, Pastor-Diez B, Basson E, Picaud JC. Neurodevelopment at 24 months corrected age in extremely preterm infants treated with dexamethasone alternatives during the late postnatal period: a cohort study. Eur J Pediatr 2024; 183:677-687. [PMID: 37955745 PMCID: PMC10912127 DOI: 10.1007/s00431-023-05319-z] [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: 09/20/2023] [Revised: 10/18/2023] [Accepted: 10/28/2023] [Indexed: 11/14/2023]
Abstract
The administration of dexamethasone has been associated with suboptimal neurodevelopment. We aimed to compare the development of extremely premature infants treated or not with alternatives to dexamethasone: betamethasone, hydrocortisone hemisuccinate. This retrospective cohort study included infants born before 29 weeks of gestational age, treated or not with late (day ≥ 7) postnatal steroids (betamethasone, hydrocortisone hemisuccinate). The neurodevelopment outcome was evaluated at 24 months corrected age, after adjustment on comorbidities of extreme prematurity. In order to analyse their overall development, data about growth and respiratory outcomes were collected. Among the 192 infants included, 59 (30.7%) received postnatal steroids. Suboptimal neurodevelopment concerned 37/59 (62.7%) postnatal steroid-treated and 43/133 (38.1%; p = 0.002) untreated infants. However, in multivariable analysis, only severe neonatal morbidity (p = 0.007) and male gender (p = 0.027) were associated with suboptimal neurodevelopment outcome at 24 months. Conclusions: Betamethasone or hydrocortisone hemisuccinate treatment was not an independent risk for suboptimal neurological development, growth and respiratory outcomes assessed at 24 months corrected age in extremely premature infants. Registration number: The study was registered on the ClinicalTrials.gov register: NCT05055193. What is Known: • Late postnatal steroids are used to treat bronchopulmonary dysplasia • Meta-analyses warned against the neurological risk of dexamethasone use during neonatal period. Early or late hydrocortisone hemisuccinate has been evaluated in multiple studies, none of which have reported an adverse effect on neurodevelopment at least to 2 years. Data about the use of betamethasone are scarce. What is New: • The risk of suboptimal neurodevelopment was higher among extremely premature infants who received postnatal steroids when compared to those who did not. • Betamethasone and hydrocortisone hemisuccinate treatment was not an independent risk factor for suboptimal neurodevelopment at 24 months corrected age.
Collapse
Affiliation(s)
- Nathalie Melan
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Pierre Pradat
- Centre for Clinical Research, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Isabelle Godbert
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Blandine Pastor-Diez
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Eliane Basson
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France
| | - Jean-Charles Picaud
- Department of Neonatology, Hôpital de La Croix-Rousse, Hospices Civils de Lyon, 69004, Lyon, France.
- CarMen Laboratory, INSERM, INRA, Université Claude Bernard Lyon 1, Pierre-Bénite, 69310, Lyon, France.
| |
Collapse
|
25
|
Amitai N, Stafler P, Blau H, Kaplan E, Mussaffi H, Levine H, Bar-On O, Steuer G, Bar-Yishay E, Klinger G, Mei-Zahav M, Prais D. Cardiopulmonary exercise testing in adolescence following extremely premature birth. Pediatr Pulmonol 2024. [PMID: 38240499 DOI: 10.1002/ppul.26867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 01/03/2024] [Accepted: 01/06/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Although extremely premature birth disrupts lung development, adolescent survivors of extreme prematurity show good clinical and physiologic outcomes. Cardiopulmonary limitations may not be clinically evident at rest. Data regarding exercise limitation in adolescents following preterm birth in the postsurfactant era are limited. RESEARCH QUESTION What are the long-term effects of bronchopulmonary dysplasia (BPD) and extreme prematurity (<29 weeks) on ventilatory response during exercise in adolescents in the postsurfactant era? STUDY DESIGN AND METHODS We followed a longitudinally recruited cohort of children aged 13-19 years who were born at a gestational age of <29 weeks (study group - SG). We compared the cardiopulmonary exercise testing (CPET) results of those with and without BPD, to their own CPET results from elementary school age (mean 9.09 ± 1.05 years). RESULTS Thirty-seven children aged 15.73 ± 1.31 years, mean gestational age 26 weeks ( ± 1.19), completed the study. CPET parameters in adolescence were within the normal range for age, including mean V̇O2 peak of 91% predicted. The BPD and non-BPD subgroups had similar results. In the longitudinal analysis of the SG, improvement was observed in adolescence, compared with elementary school age, in breathing reserve (36.37 ± 18.99 vs. 26.58 ± 17.92, p = 0.044), tidal volume as a fraction of vital capacity achieved at maximal load (0.51 ± 0.13 vs. 0.37 ± 0.08, p < 0.001), and respiratory exchange ratio at maximal load (1.18 ± 0.13 vs. 1.11 ± 0.10, p = 0.021). INTERPRETATION In the current cohort, adolescents born extremely premature have essentially normal ventilatory response during exercise, unrelated to BPD diagnosis. CPET results in this population improve over time.
Collapse
Affiliation(s)
- Nofar Amitai
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Patrick Stafler
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hannah Blau
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Kaplan
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Intensive Care Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Huda Mussaffi
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hagit Levine
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ophir Bar-On
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Steuer
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Ephraim Bar-Yishay
- Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheba, Israel
| | - Gil Klinger
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neonatal Intensive Care Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Meir Mei-Zahav
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dario Prais
- Pulmonary Institute, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- School of Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Dini G, Santini MG, Celi F. Less Invasive Surfactant Administration (LISA) Versus INSURE Method in Preterm Infants: a Retrospective Study. Med Arch 2024; 78:112-116. [PMID: 38566872 PMCID: PMC10983101 DOI: 10.5455/medarh.2024.78.112-116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Background Respiratory distress syndrome (RDS) is a major cause of morbidity and mortality in preterm infants. Early nasal CPAP and selective administration of surfactant via the endotracheal tube are widely used in the treatment of RDS in preterm infants. Objective The aim of this study was to compare the need for intubation and mechanical ventilation after surfactant delivery between LISA-treated and INSURE-treated premature infants with respiratory distress syndrome (RDS). Methods Retrospective registry-based cohort study enrolled 36 newborns admitted to the neonatal intensive care unit of the "Santa Maria" Hospital of Terni between 2016 and 2023. As a primary outcome, we followed the need for intubation and mechanical ventilation within 72 hours of life, while the secondary outcomes were major neonatal morbidities and death before discharge. Results The LISA group and the INSURE group included 13 and 23 newborns respectively. Demographic features showed no significant differences between the two groups. The need for mechanical ventilation in the first 72 hours of life was similar in both groups (p >0.99). There were no significant differences in morbidities. Conclusion LISA and INSURE are equally effective modalities for surfactant administration for the treatment of RDS in preterm infants.
Collapse
Affiliation(s)
- Gianluca Dini
- Neonatal Intensive Care Unit, “Santa Maria” Hospital, Terni, Italy
| | | | - Federica Celi
- Neonatal Intensive Care Unit, “Santa Maria” Hospital, Terni, Italy
| |
Collapse
|
27
|
Mani S, Rawat M. Less Invasive Surfactant Administration: A Viewpoint. Am J Perinatol 2024; 41:211-227. [PMID: 36539205 PMCID: PMC10791155 DOI: 10.1055/a-2001-9139] [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: 09/05/2022] [Accepted: 12/12/2022] [Indexed: 02/17/2023]
Abstract
The standard of care in treating respiratory distress syndrome in preterm infants is respiratory support with nasal continuous positive airway pressure or a combination of continuous positive airway pressure and exogenous surfactant replacement. Endotracheal intubation, the conventional method for surfactant administration, is an invasive procedure associated with procedural and mechanical ventilation complications. The INSURE (intubation, surfactant administration, and extubation soon after) technique is an accepted method aimed at reducing the short-term complications and long-term morbidities related to mechanical ventilation but does not eliminate risks associated with endotracheal intubation and mechanical ventilation. Alternative methods of surfactant delivery that can overcome the problems associated with the INSURE technique are surfactant through a laryngeal mask, surfactant through a thin intratracheal catheter, and aerosolized surfactant delivered using nebulizers. The three alternative methods of surfactant delivery studied in the last two decades have advantages and limitations. More than a dozen randomized controlled trials have aimed to study the benefits of the three alternative techniques of surfactant delivery compared with INSURE as the control arm, with promising results in terms of reduction in mortality, need for mechanical ventilation, and bronchopulmonary dysplasia. The need to find a less invasive surfactant administration technique is a clinically relevant problem. Before broader adoption in routine clinical practice, the most beneficial technique among the three alternative strategies should be identified. This review aims to summarize the current evidence for using the three alternative techniques of surfactant administration in neonates, compare the three techniques, highlight the knowledge gaps, and suggest future directions. KEY POINTS: · The need to find a less invasive alternative method of surfactant delivery is a clinically relevant problem.. · Clinical trials that have studied alternative surfactant delivery methods have shown promising results but are inconclusive for broader adoption into clinical practice.. · Future studies should explore novel clinical trial methodologies and select clinically significant long term outcomes for comparison..
Collapse
Affiliation(s)
- Srinivasan Mani
- Department of Pediatrics, University of Toledo, Toledo, Ohio
| | - Munmun Rawat
- Department of Pediatrics, University at Buffalo, Buffalo, New York
| |
Collapse
|
28
|
Riccetti MR, Green J, Taylor TJ, Perl AKT. Prenatal FGFR2 Signaling via PI3K/AKT Specifies the PDGFRA + Myofibroblast. Am J Respir Cell Mol Biol 2024; 70:63-77. [PMID: 37734036 PMCID: PMC10768833 DOI: 10.1165/rcmb.2023-0245oc] [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: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 09/23/2023] Open
Abstract
It is well known that FGFR2 (fibroblast growth factor receptor 2) signaling is critical for proper lung development. Recent studies demonstrate that epithelial FGFR2 signaling during the saccular phase of lung development (sacculation) regulates alveolar type 1 (AT1) and AT2 cell differentiation. During sacculation, PDGFRA (platelet-derived growth factor receptor-α)-positive lung fibroblasts exist as three functional subtypes: contractile myofibroblasts, extracellular matrix-producing matrix fibroblasts, and lipofibroblasts. All three subtypes are required during alveolarization to establish a niche that supports AT2 epithelial cell self-renewal and AT1 epithelial cell differentiation. FGFR2 signaling directs myofibroblast differentiation in PDGFRA+ fibroblasts during alveolar reseptation after pneumonectomy. However, it remains unknown if FGFR2 signaling regulates PDGFRA+ myo-, matrix, or lipofibroblast differentiation during sacculation. In this study, FGFR2 signaling was inhibited by temporal expression of a secreted dominant-negative FGFR2b (dnFGFR2) by AT2 cells from embryonic day (E) 16.5 to E18.5. Fibroblast and epithelial differentiation were analyzed at E18.5 and postnatal days 7 and 21. At all time points, the number of myofibroblasts was reduced and the number of lipo-/matrix fibroblasts was increased. AT2 cells are increased and AT1 cells are reduced postnatally, but not at E18.5. Similarly, in organoids made with PDGFRA+ fibroblasts from dnFGFR2 lungs, increased AT2 cells and reduced AT1 cells were observed. In vitro treatment of primary wild-type E16.5 adherent saccular lung fibroblasts with recombinant dnFGFR2b/c resulted in reduced myofibroblast contraction. Treatment with the PI3K/AKT activator 740 Y-P rescued the lack of myofibroblast differentiation caused by dnFGFR2b/2c. Moreover, treatment with the PI3K/AKT activator 740 Y-P rescued myofibroblast differentiation in E18.5 fibroblasts isolated from dnFGFR2 lungs.
Collapse
Affiliation(s)
- Matthew R. Riccetti
- Division of Neonatology and Pulmonary Biology and
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Jenna Green
- Division of Neonatology and Pulmonary Biology and
| | - Thomas J. Taylor
- Division of Neonatology and Pulmonary Biology and
- College of Arts and Sciences, University of Cincinnati, Cincinnati, Ohio; and
| | - Anne-Karina T. Perl
- Division of Neonatology and Pulmonary Biology and
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
29
|
Kuwana S, Sato S, Sakurai Y, Koshinami S, Watanabe S, Watanabe T, Hanita T. Reduced number of alveoli after birth in rats exposed to iron in utero. Pediatr Int 2024; 66:e15820. [PMID: 39692218 DOI: 10.1111/ped.15820] [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: 07/02/2023] [Revised: 04/16/2024] [Accepted: 06/13/2024] [Indexed: 12/19/2024]
Abstract
BACKGROUND Clinical studies have shown that diffuse chorioamniotic hemosiderosis (DCH) is a risk factor for bronchopulmonary dysplasia (BPD). However, the details of the underlying mechanism are unknown. We focused on iron, one of the blood components that diffuses within the amniotic cavity in DCH. Specifically, we conducted a histological evaluation of its effects on fetal lung development. METHODS Iron dextran 10 mg (Fe group) or physiological saline (control group) was injected into the individual amniotic cavities of pregnant Sprague Dawley rats on gestational day 19. The pups were born naturally, and the placentas and lungs collected on postnatal days 1, 14, and 60 were subjected to histological analysis. RESULTS The placenta and lungs of the Fe group on day 14 contained significantly larger numbers of iron-positive granules. Morphological analysis of the lungs showed that on day 60, the total number of alveoli was significantly lower in the Fe group (p = 0.015). Immunohistochemical staining of the lung tissue for 8-hydroxy-2'-deoxyguanosine showed that, on day 1, there were significantly more positive cells in the Fe group (p = 0.003). The number of ectodermal dysplasia 1 positive cells on day 14 was significantly increased in the Fe group (p = 0.001). CONCLUSIONS These results suggest that diffuse iron deposition in immature fetal lungs increases oxidative stress and decreases the number of postnatal alveoli associated with impaired lung development.
Collapse
Affiliation(s)
- Shouta Kuwana
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shinichi Sato
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Yoshie Sakurai
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shouta Koshinami
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Shimpei Watanabe
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Tatsuya Watanabe
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Takushi Hanita
- Center for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, Miyagi, Japan
| |
Collapse
|
30
|
Sehgal A, Gauli B. Changes in respiratory mechanics in response to crystalloid infusions in extremely premature infants. Am J Physiol Lung Cell Mol Physiol 2023; 325:L819-L825. [PMID: 37933458 DOI: 10.1152/ajplung.00179.2023] [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: 06/08/2023] [Revised: 09/08/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023] Open
Abstract
Extremely premature infants are at a higher risk of developing respiratory distress syndrome and circulatory impairments in the first few weeks of life. Administration of normal saline boluses to manage hypotension is a common practice in preterm infants. As a crystalloid, a substantial proportion might leak into the interstitium; most consequently the lungs in the preterm cohorts, putatively affecting ventilation. We downloaded and analyzed ventilator mechanics data in infants managed by conventional mechanical ventilation and administered normal saline bolus for clinical reasons. Data were downloaded for 30 min prebolus, 60 min during the bolus followed by 30 min postbolus. Sixteen infants (mean gestational age 25.2 ± 1 wk and birth weight 620 ± 60 g) were administered 10 mL/kg normal saline over 60 min. The most common clinical indication for saline was hypotension. No significant increase was noted in mean blood pressure after the saline bolus. A significant reduction in pulmonary compliance (mL/cmH2O/kg) was noted (0.43 ± 0.07 vs. 0.38 ± 0.07 vs. 0.33 ± 0.07, P = 0.003, ANOVA). This was accompanied by an elevation in the required peak inspiratory pressure to deliver set volume-guarantee (19 ± 2 vs. 22 ± 2 vs. 22 ± 3 mmHg, P < 0.0001, ANOVA), resulting in a higher respiratory severity score. Normal saline infusion therapy was associated with adverse pulmonary mechanics. Relevant pathophysiologic mechanisms might include translocation of fluid across pulmonary capillaries affected by low vascular tone and heightened permeability in extremes of prematurity, back-pressure effects from raised left atrial volume due to immature left-ventricular myocardium; complemented by the effect of cytokine release from positive pressure ventilation.NEW & NOTEWORTHY Administration of saline boluses is common in premature infants although hypovolemia is an uncommon underlying cause of hypotension. This crystalloid can redistribute into pulmonary interstitial space. In the presence of an immature myocardium and diastolic dysfunction, excess fluid can also be "edemagenic." This study on extremely premature infants (25 wk gestation) noted adverse influence on respiratory physiology after saline infusion. Clinicians need to choose judiciously and reconsider routine use of saline boluses in premature infants.
Collapse
Affiliation(s)
- Arvind Sehgal
- Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| | - Bishal Gauli
- Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
31
|
Lavizzari A, Esposito B, Pesenti N, Shaykhova A, Vizzari G, Ophorst M, Gangi S, Morniroli D, Colnaghi M, Mosca F, Giannì ML. Dose-dependent impact of human milk feeding on tidal breathing flow-volume loop parameters across the first 2 years of life in extremely low-birth-weight infants: a cohort study. Eur J Pediatr 2023; 182:4969-4976. [PMID: 37610435 DOI: 10.1007/s00431-023-05163-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023]
Abstract
The purpose of this study is to test the hypothesis that higher consumption of human milk (HM) in preterm infants with birth weight (BW) <1000 g is associated with improved lung function in a dose-dependent manner over the first 2 years of corrected age (CA). This retrospective study at an academic medical center included infants with BW <1000g. They had lung function assessment by the tidal breathing flow-volume loop (TBFVL) follow-up visits at 0-3-, 3-6-, 6-12-, 12-18-, and 18-24-month CA. One hundred eighty infants were included in the study with a mean (SD) gestational age 26.5 (1.90) weeks and BW 772.4 (147.0) g, 50% were female, and 60% developed BPD. 62.8% of infants received HM during the NICU stay. According to a general linear model (including GA, being small for GA (SGA), sex, human milk percentage, sepsis, and BPD), on average, each week of GA resulted in a higher tPTEF/tE of 1.24 (p = 0.039) and being SGA in a lower tPTEF/tE of 5.75 (p = 0.013) at 0-3-month CA. A higher percentage of human milk out of the total enteral intake was associated with better tPTEF/tE z-scores at 0-3 months (p = 0.004) and 18-24 months of CA (p = 0.041). BPD diagnosis was associated with a relevantly worse tPTEF/tE z-score at 6-12 months of CA (p = 0.003). CONCLUSION Preterm infants with higher consumption of HM had significantly less airway obstruction across the first 2 years, suggesting that human milk may contribute in a dose-dependent manner to improve lung function in early childhood in former preterm infants born ELBW. WHAT IS KNOWN • Human milk feeding reduces the risk of prematurity-related morbidities, including necrotizing enterocolitis, sepsis, lower respiratory tract infections, and BPD. Both exclusive and partial human milk feeding appear to be associated with a lower risk of BPD in preterm infants. WHAT IS NEW • This cohort study of 180 preterm infants with birth weight < 1000 g found that exposure to human milk during hospitalization improves airway obstruction markers tPTEF/tE z-score over the first 2 years of corrected age in a dose-dependent manner.
Collapse
Affiliation(s)
- Anna Lavizzari
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy.
| | - Benedetta Esposito
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Nicola Pesenti
- Revelo Datalabs Srl, Via Spezia 1, 20142, Milan, Italy
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, University of Milan-Bicocca, Milan, Italy
| | - Alina Shaykhova
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giulia Vizzari
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Marijke Ophorst
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Silvana Gangi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Daniela Morniroli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Mariarosa Colnaghi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Maria L Giannì
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 28, 20122, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
32
|
Romijn M, Onland W, van Keulen BJ, Heijboer AC, Rotteveel J, van Kaam AH, Finken MJJ. Glucocorticoid signature of preterm infants developing bronchopulmonary dysplasia. Pediatr Res 2023; 94:1804-1809. [PMID: 37355738 DOI: 10.1038/s41390-023-02690-3] [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: 12/29/2022] [Revised: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Systemic inflammation plays a key role in the development of bronchopulmonary dysplasia (BPD). Cortisol is known to dampen inflammation. However, adrenal function following preterm birth is characterized by insufficient cortisol levels for the degree of inflammation, and a relative abundancy of cortisol precursors. We investigated whether this pattern could contribute to the development of BPD in preterm infants born <30 weeks of gestation. METHODS Cortisol, cortisone, 17-OH progesterone (17-OHP) and 11-deoxycortisol were measured in serum obtained at postnatal days 1, 3, 7, 14 and 28, using liquid-chromatography-tandem-mass-spectrometry. The presence of BPD was ascertained at 36 weeks postmenstrual age. RESULTS Sixty-five infants were included for analysis, of whom 32 (49%) developed BPD. Preterm infants developing BPD, as compared to those without BPD, had higher levels of 17-OHP, 11-deoxycortisol and cortisone relative to cortisol in their first week of life, but not at birth or beyond day 7. CONCLUSION Preterm infants developing BPD had higher levels of cortisol precursors and cortisone relative to cortisol in their first week of life than infants without BPD. These findings suggest that BPD is preceded by an activated hypothalamus-pituitary-adrenal axis that could not meet the high cortisol demands, which may predispose to inflammation and BPD. IMPACT Relative adrenal insufficiency is common in the first weeks after preterm birth, resulting in insufficient cortisol production for the degree of inflammation and a relative abundance of cortisol precursors; Whether this pattern contributes to the development of bronchopulmonary dysplasia (BPD) is not fully elucidated, since most studies focused on cortisol levels; Preterm infants developing BPD had higher levels of cortisol precursors and cortisone relative to cortisol in the first week of life, suggestive of a hypothalamus-pituitary-adrenal-axis activation during BPD development which cannot meet the high cortisol demands in tissues; This glucocorticoid pattern is likely to dispose to inflammation and BPD.
Collapse
Affiliation(s)
- Michelle Romijn
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric Endocrinology, Boelelaan, 1117, Amsterdam, The Netherlands.
- Amsterdam UMC location University of Amsterdam, Department of Neonatology, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands.
| | - Wes Onland
- Amsterdam UMC location University of Amsterdam, Department of Neonatology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Britt J van Keulen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric Endocrinology, Boelelaan, 1117, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Annemieke C Heijboer
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
- Amsterdam UMC location University of Amsterdam and location Vrije Universiteit Amsterdam, Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam, The Netherlands
- Amsterdam Gastroenterology, Endocrinology & Metabolism, Amsterdam, The Netherlands
| | - Joost Rotteveel
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric Endocrinology, Boelelaan, 1117, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Anton H van Kaam
- Amsterdam UMC location University of Amsterdam, Department of Neonatology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - Martijn J J Finken
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric Endocrinology, Boelelaan, 1117, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Hysinger EB, Critser P. How is preterm birth working out: cardiopulmonary response to exercise in extreme prematurity. Eur Respir J 2023; 62:2301787. [PMID: 37973173 DOI: 10.1183/13993003.01787-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Affiliation(s)
- Erik B Hysinger
- Division of Pulmonary Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul Critser
- The Heart Institute, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
34
|
Zhang S, Mulder C, Riddle S, Song R, Yue D. Mesenchymal stromal/stem cells and bronchopulmonary dysplasia. Front Cell Dev Biol 2023; 11:1247339. [PMID: 37965579 PMCID: PMC10642488 DOI: 10.3389/fcell.2023.1247339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication in preterm infants, leading to chronic respiratory disease. There has been an improvement in perinatal care, but many infants still suffer from impaired branching morphogenesis, alveolarization, and pulmonary capillary formation, causing lung function impairments and BPD. There is an increased risk of respiratory infections, pulmonary hypertension, and neurodevelopmental delays in infants with BPD, all of which can lead to long-term morbidity and mortality. Unfortunately, treatment options for Bronchopulmonary dysplasia are limited. A growing body of evidence indicates that mesenchymal stromal/stem cells (MSCs) can treat various lung diseases in regenerative medicine. MSCs are multipotent cells that can differentiate into multiple cell types, including lung cells, and possess immunomodulatory, anti-inflammatory, antioxidative stress, and regenerative properties. MSCs are regulated by mitochondrial function, as well as oxidant stress responses. Maintaining mitochondrial homeostasis will likely be key for MSCs to stimulate proper lung development and regeneration in Bronchopulmonary dysplasia. In recent years, MSCs have demonstrated promising results in treating and preventing bronchopulmonary dysplasia. Studies have shown that MSC therapy can reduce inflammation, mitochondrial impairment, lung injury, and fibrosis. In light of this, MSCs have emerged as a potential therapeutic option for treating Bronchopulmonary dysplasia. The article explores the role of MSCs in lung development and disease, summarizes MSC therapy's effectiveness in treating Bronchopulmonary dysplasia, and delves into the mechanisms behind this treatment.
Collapse
Affiliation(s)
- Shuqing Zhang
- School of Pharmacy, China Medical University, Shenyang, China
| | - Cassidy Mulder
- Liberty University College of Osteopathic Medicine, Lynchburg, VA, United States
| | - Suzette Riddle
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Dongmei Yue
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
35
|
Al-Mudares F, Cantu Gutierrez M, Cantu A, Jiang W, Wang L, Dong X, Moorthy B, Sajti E, Lingappan K. Loss of growth differentiation factor 15 exacerbates lung injury in neonatal mice. Am J Physiol Lung Cell Mol Physiol 2023; 325:L314-L326. [PMID: 37368978 PMCID: PMC10625832 DOI: 10.1152/ajplung.00086.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/11/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023] Open
Abstract
Growth differentiation factor 15 (GDF15) is a divergent member of the transforming growth factor-β (TGF-β) superfamily, and its expression increases under various stress conditions, including inflammation, hyperoxia, and senescence. GDF15 expression is increased in neonatal murine bronchopulmonary dysplasia (BPD) models, and GDF15 loss exacerbates oxidative stress and decreases cellular viability in vitro. Our overall hypothesis is that the loss of GDF15 will exacerbate hyperoxic lung injury in the neonatal lung in vivo. We exposed neonatal Gdf15-/- mice and wild-type (WT) controls on a similar background to room air or hyperoxia (95% [Formula: see text]) for 5 days after birth. The mice were euthanized on postnatal day 21 (PND 21). Gdf15-/- mice had higher mortality and lower body weight than WT mice after exposure to hyperoxia. Hyperoxia exposure adversely impacted alveolarization and lung vascular development, with a greater impact in Gdf15-/- mice. Interestingly, Gdf15-/- mice showed lower macrophage count in the lungs compared with WT mice both under room air and after exposure to hyperoxia. Analysis of the lung transcriptome revealed marked divergence in gene expression and enriched biological pathways in WT and Gdf15-/- mice and differed markedly by biological sex. Notably, pathways related to macrophage activation and myeloid cell homeostasis were negatively enriched in Gdf15-/- mice. Loss of Gdf15 exacerbates mortality, lung injury, and the phenotype of the arrest of alveolarization in the developing lung with loss of female-sex advantage in Gdf15-/- mice.NEW & NOTEWORTHY We show for the first time that loss of Gdf15 exacerbates mortality, lung injury, and the phenotype of the arrest of alveolarization in the developing lung with loss of female-sex advantage in Gdf15-/- mice. We also highlight the distinct pulmonary transcriptomic response in the Gdf15-/- lung including pathways related to macrophage recruitment and activation.
Collapse
Affiliation(s)
- Faeq Al-Mudares
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Manuel Cantu Gutierrez
- Divsion of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Abiud Cantu
- Divsion of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Weiwu Jiang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Lihua Wang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Xiaoyu Dong
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Eniko Sajti
- Division of Neonatology, Department of Pediatrics, University of California, San Diego, California, United States
| | - Krithika Lingappan
- Divsion of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
36
|
Carr H, Gunnerbeck A, Eisenlauer P, Johansson S, Cnattingius S, Ludvigsson JF, Edstedt Bonamy AK. Severity of preterm birth and the risk of pulmonary hypertension in childhood: A population-based cohort study in Sweden. Paediatr Perinat Epidemiol 2023; 37:630-640. [PMID: 37414733 DOI: 10.1111/ppe.12997] [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: 12/04/2022] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Preterm birth (<37 completed gestational weeks) has been linked to pulmonary hypertension (PH), but the relationship to severity of preterm birth has not been studied. OBJECTIVES We investigated associations between extremely (<28 weeks), very (28-31 weeks), moderately (32-36 weeks) preterm birth, early-term birth (37-38 weeks) and later PH. Additionally, we explored associations between birthweight for gestational age and PH. METHODS This registry-based cohort study followed 3.1 million individuals born in Sweden (1987-2016) from 1 up to a maximum of 30 years of age. The outcome was diagnosis or death from PH in national health registers. Adjusted hazard ratios (HR) were estimated using Cox regression analysis. Unadjusted and confounder-adjusted incidence rate differences were also calculated. RESULTS Of 3,142,812 individuals, there were 543 cases of PH (1.2 per 100,000 person-years), 153 of which in individuals without malformations. Compared with individuals born at 39 weeks, adjusted HRs with 95% confidence interval (CI) for PH for extremely, moderately, and very preterm birth were 68.78 (95% CI 49.49, 95.57), 13.86 (95% CI 9.27, 20.72) and 3.42 (95% CI 2.46, 4.74), respectively, and for early-term birth 1.74 (1.31, 2.32). HRs were higher in subjects without malformations. There were 90 additional cases of PH per 100,000 person-years in the extremely preterm group (50 after excluding malformations). Very small for gestational age (below 2 SD from estimated birthweight for gestational age and sex) was also associated with increased risk of PH (adjusted HR 2.02, 95% CI 1.14, 3.57). CONCLUSIONS We found an inverse association between gestational age and later PH, but the incidence and absolute risks are low. The severity of preterm birth adds clinically relevant information to the assessment of cardiovascular risks in childhood.
Collapse
Affiliation(s)
- Hanna Carr
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Anna Gunnerbeck
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Peter Eisenlauer
- Department of Neonatology, Karolinska University Hospital, Solna, Sweden
| | - Stefan Johansson
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Stockholm, Sweden
| | - Sven Cnattingius
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatrics, Örebro University Hospital, Örebro, Sweden
| | - Anna-Karin Edstedt Bonamy
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Kryeziu I, Reçica S, Thaçi Q, Kurshumliu F, Hadzi-Petrushev N, Basholli-Salihu M, Mladenov M, Sopi RB. Quercetin supplementation attenuates airway hyperreactivity and restores airway relaxation in rat pups exposed to hyperoxia. Exp Biol Med (Maywood) 2023; 248:1492-1499. [PMID: 37837396 PMCID: PMC10666724 DOI: 10.1177/15353702231199468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/20/2023] [Indexed: 10/16/2023] Open
Abstract
Hyperoxia exposure of immature lungs contributes to lung injury and airway hyperreactivity. Up to now, treatments of airway hyperreactivity induced by hyperoxia exposure have been ineffective. The aim of this study was to investigate the effects of quercetin on hyperoxia-induced airway hyperreactivity, impaired relaxation, and lung inflammation. Newborn rats were exposed to hyperoxia (FiO2 > 95%) or ambient air (AA) for seven days. Subgroups were injected with quercetin (10 mg·kg-1·day-1). After exposures, tracheal cylinders were prepared for in vitro wire myography. Contraction to methacholine was measured in the presence or absence of organ bath quercetin and/or Nω-nitro-L-arginine methyl ester (L-NAME). Relaxation responses were evoked in preconstricted tissues using electrical field stimulation (EFS). Lung tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) levels were measured by enzyme-linked immunosorbent assay (ELISA). A P < 0.05 was considered statistically significant. Contractile responses of tracheal smooth muscle (TSM) of hyperoxic animals were significantly increased compared with AA animals (P < 0.001). Treatment with quercetin significantly reduced contraction in hyperoxic groups compared with hyperoxic control (P < 0.01), but did not have any effect in AA groups. In hyperoxic animals, relaxation of TSM was significantly reduced compared with AA animals (P < 0.001), while supplementation of quercetin restored the lost relaxation in hyperoxic groups. Incubation of preparations in L-NAME significantly reduced the quercetin effects on both contraction and relaxation (P < 0.01). Treatment of hyperoxic animals with quercetin significantly decreased the expression of TNF-α and IL-1β compared with hyperoxic controls (P < 0.001 and P < 0.01, respectively).The findings of this study demonstrate the protective effect of quercetin on airway hyperreactivity and suggest that quercetin might serve as a novel therapy to prevent and treat neonatal hyperoxia-induced airway hyperreactivity and inflammation.
Collapse
Affiliation(s)
- Islam Kryeziu
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Shkëlzen Reçica
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Qëndrim Thaçi
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Fisnik Kurshumliu
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| | | | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University in Skopje, 1000 Skopje, North Macedonia
| | - Ramadan B Sopi
- Faculty of Medicine, University of Prishtina, 10 000 Prishtina, Kosovo
| |
Collapse
|
38
|
Shi ZS, Wang XB, Wang MC, Zeng YY. Effects of WeChat follow-up management on the psychological distress, care burden, and quality of life of parents of infants with bronchopulmonary dysplasia: a retrospective cohort study. Front Pediatr 2023; 11:1239527. [PMID: 37635790 PMCID: PMC10449024 DOI: 10.3389/fped.2023.1239527] [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: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Objective The objective was to explore the impact of WeChat follow-up management on the psychological distress, care burden, and quality of life of parents of infants with bronchopulmonary dysplasia (BPD) receiving in-home care. Methods This was a retrospective cohort study. A total of 101 parents of infants with BPD who were followed up from January 2016 to January 2022 were included in this study. According to different follow-up methods, these patients were classified into the WeChat group and the routine group. The Depression, Anxiety, and Stress Scale-21 (DASS-21), Zarit Caregiver Burden Interview (ZBI), and WHOQOL-BREF were used. The data on the psychological distress, care burden, and quality of life of the parents in the two groups were analyzed and compared at discharge and at the 3-month follow-up. Results There was no significant difference in the DASS-21 and ZBI scores at discharge between the parents in the two groups. During the 3-month follow-up, the scores of the DASS-21 anxiety and stress subscale and the ZBI of parents in the WeChat group were significantly lower than those of parents in the routine group (P < 0.05); however, there was no significant difference in the depression subscale score between the two groups (P > 0.05). A comparison of the WHOQOL-BREF score between the two groups showed that the total quality of life score in the WeChat group was significantly higher than that in the routine group (P < 0.05). The scores of the psychological and social relationship fields in the WeChat group were significantly higher than those in the routine group (P < 0.05). The incidence of adverse events during follow-up was significantly lower in the WeChat group than in the routine group (P < 0.05). Conclusion WeChat follow-up management is helpful to decrease the anxiety and stress, reduce the care burden, and improve the quality of life of parents of infants with BPD receiving in-home care.
Collapse
Affiliation(s)
- Zhong-Shan Shi
- Department of Pediatrics, Shishi General Hospital, Quanzhou, China
| | | | | | | |
Collapse
|
39
|
Cantu A, Cantu Gutierrez M, Zhang Y, Dong X, Lingappan K. Endothelial to mesenchymal transition in neonatal hyperoxic lung injury: role of sex as a biological variable. Physiol Genomics 2023; 55:345-354. [PMID: 37395632 PMCID: PMC10625841 DOI: 10.1152/physiolgenomics.00037.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/12/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by an arrest in alveolarization, abnormal vascular development, and variable interstitial fibroproliferation in the premature lung. Endothelial to mesenchymal transition (EndoMT) may be a source of pathological fibrosis in many organ systems. Whether EndoMT contributes to the pathogenesis of BPD is not known. We tested the hypothesis that pulmonary endothelial cells will show increased expression of EndoMT markers upon exposure to hyperoxia and that sex as a biological variable will modulate differences in expression. Wild-type (WT) and Cdh5-PAC CreERT2 (endothelial reporter) neonatal male and female mice (C57BL6) were exposed to hyperoxia (0.95 [Formula: see text]) either during the saccular stage of lung development (95% [Formula: see text]; postnatal day 1-5 [PND1-5]) or through the saccular and early alveolar stages of lung development (75% [Formula: see text]; PND1-14). Expression of EndoMT markers was measured in whole lung and endothelial cell mRNA. Sorted lung endothelial cells (from room air- and hyperoxia-exposed lungs) were subjected to bulk RNA-Seq. We show that exposure of the neonatal lung to hyperoxia leads to upregulation of key markers of EndoMT. Furthermore, using lung sc-RNA-Seq data from neonatal lung we were able to show that all endothelial cell subpopulations including the lung capillary endothelial cells show upregulation of EndoMT-related genes. Markers related to EndoMT are upregulated in the neonatal lung upon exposure to hyperoxia and show sex-specific differences. Mechanisms mediating EndoMT in the injured neonatal lung can modulate the response of the neonatal lung to hyperoxic injury and need further investigation.NEW & NOTEWORTHY We show that neonatal hyperoxia exposure increased EndoMT markers in the lung endothelial cells and this biological process exhibits sex-specific differences.
Collapse
Affiliation(s)
- Abiud Cantu
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Manuel Cantu Gutierrez
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Yuhao Zhang
- Division of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States
| | - Xiaoyu Dong
- Division of Neonatology, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States
| | - Krithika Lingappan
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| |
Collapse
|
40
|
Lemyre B, Deguise MO, Benson P, Kirpalani H, Ekhaguere OA, Davis PG. Early nasal intermittent positive pressure ventilation (NIPPV) versus early nasal continuous positive airway pressure (NCPAP) for preterm infants. Cochrane Database Syst Rev 2023; 7:CD005384. [PMID: 37466143 PMCID: PMC10355255 DOI: 10.1002/14651858.cd005384.pub3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
BACKGROUND Nasal continuous positive airway pressure (NCPAP) is a strategy to maintain positive airway pressure throughout the respiratory cycle through the application of a bias flow of respiratory gas to an apparatus attached to the nose. Early treatment with NCPAP is associated with decreased risk of mechanical ventilation exposure and might reduce chronic lung disease. Nasal intermittent positive pressure ventilation (NIPPV) is a form of noninvasive ventilation delivered through the same nasal interface during which patients are exposed to short inflations, along with background end-expiratory pressure. OBJECTIVES To examine the risks and benefits of early (within the first six hours after birth) NIPPV versus early NCPAP for preterm infants at risk of or with respiratory distress syndrome (RDS). Primary endpoints are respiratory failure and the need for intubated ventilatory support during the first week of life. Secondary endpoints include the incidence of mortality, chronic lung disease (CLD) (oxygen therapy at 36 weeks' postmenstrual age), pneumothorax, duration of respiratory support, duration of oxygen therapy, and intraventricular hemorrhage (IVH). SEARCH METHODS Searches were conducted in January 2023 in CENTRAL, MEDLINE, Embase, Web of Science, and Dissertation Abstracts. The reference lists of related systematic reviews and of studies selected for inclusion were also searched. SELECTION CRITERIA We considered all randomized and quasi-randomized controlled trials. Eligible studies compared NIPPV versus NCPAP treatment, starting within six hours after birth in preterm infants (< 37 weeks' gestational age (GA)). DATA COLLECTION AND ANALYSIS We collected and analyzed data using the recommendations of the Cochrane Neonatal Review Group. MAIN RESULTS We included 17 trials, enrolling 1958 infants in this review. NIPPV likely reduces the rate of respiratory failure (risk ratio (RR) 0.65, 95% confidence interval (CI) 0.54 to 0.78; risk difference (RD) -0.08, 95% CI -0.12 to -0.05; 17 RCTs, 1958 infants; moderate-certainty evidence) and needing endotracheal tube ventilation (RR 0.67, 95% CI 0.56 to 0.81; RD -0.07, 95% CI -0.11 to -0.04; 16 RCTs; 1848 infants; moderate-certainty evidence) amongst infants treated with early NIPPV compared with early NCPAP. The meta-analysis demonstrated that NIPPV may reduce the risk of developing CLD compared to CPAP (RR 0.70, 95% CI 0.52 to 0.92; 12 RCTs, 1284 infants; low-certainty evidence) slightly. NIPPV may result in little to no difference in mortality (RR 0.82, 95% CI 0.62 to 1.10; 17 RCTs; 1958 infants; I2 of 0%; low-certainty evidence), the incidence of pneumothorax (RR 0.92, 95% CI 0.60 to 1.41; 16 RCTs; 1674 infants; I2 of 0%; low-certainty evidence), and rates of severe IVH (RR 0.98, 95% CI 0.53 to 1.79; 8 RCTs; 977 infants; I2 of 0%; low-certainty evidence). AUTHORS' CONCLUSIONS When applied within six hours after birth, NIPPV likely reduces the risk of respiratory failure and the need for intubation and endotracheal tube ventilation in very preterm infants (GA 28 weeks and above) with respiratory distress syndrome or at risk for RDS. It may also decrease the rate of CLD slightly. However, most trials enrolled infants with a gestational age of approximately 28 to 32 weeks with an overall mean gestational age of around 30 weeks. As such, the results of this review may not apply to extremely preterm infants that are most at risk of needing mechanical ventilation or developing CLD. Additional studies are needed to confirm these results and to assess the safety of NIPPV compared with NCPAP alone in a larger patient population.
Collapse
Affiliation(s)
- Brigitte Lemyre
- Division of Neonatology, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Marc-Olivier Deguise
- Dept. of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Paige Benson
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | | | - Osayame A Ekhaguere
- Division of Neonatal-Perinatal Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Peter G Davis
- Newborn Research Centre, The Royal Women's Hospital, Parkville, Australia
| |
Collapse
|
41
|
Aleem S, Greenberg RG. Accurate Prediction of Bronchopulmonary Dysplasia: Are We There Yet? J Pediatr 2023; 258:113389. [PMID: 36933768 DOI: 10.1016/j.jpeds.2023.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/20/2023]
Affiliation(s)
- Samia Aleem
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | | |
Collapse
|
42
|
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
|
43
|
Zhang EY, Bartman CM, Prakash YS, Pabelick CM, Vogel ER. Oxygen and mechanical stretch in the developing lung: risk factors for neonatal and pediatric lung disease. Front Med (Lausanne) 2023; 10:1214108. [PMID: 37404808 PMCID: PMC10315587 DOI: 10.3389/fmed.2023.1214108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023] Open
Abstract
Chronic airway diseases, such as wheezing and asthma, remain significant sources of morbidity and mortality in the pediatric population. This is especially true for preterm infants who are impacted both by immature pulmonary development as well as disproportionate exposure to perinatal insults that may increase the risk of developing airway disease. Chronic pediatric airway disease is characterized by alterations in airway structure (remodeling) and function (increased airway hyperresponsiveness), similar to adult asthma. One of the most common perinatal risk factors for development of airway disease is respiratory support in the form of supplemental oxygen, mechanical ventilation, and/or CPAP. While clinical practice currently seeks to minimize oxygen exposure to decrease the risk of bronchopulmonary dysplasia (BPD), there is mounting evidence that lower levels of oxygen may carry risk for development of chronic airway, rather than alveolar disease. In addition, stretch exposure due to mechanical ventilation or CPAP may also play a role in development of chronic airway disease. Here, we summarize the current knowledge of the impact of perinatal oxygen and mechanical respiratory support on the development of chronic pediatric lung disease, with particular focus on pediatric airway disease. We further highlight mechanisms that could be explored as potential targets for novel therapies in the pediatric population.
Collapse
Affiliation(s)
- Emily Y. Zhang
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Colleen M. Bartman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Elizabeth R. Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
44
|
Valsecchi C, Croce S, Lenta E, Acquafredda G, Comoli P, Avanzini MA. TITLE: New therapeutic approaches in pediatric diseases: Mesenchymal stromal cell and mesenchymal stromal cell-derived extracellular vesicles as new drugs. Pharmacol Res 2023; 192:106796. [PMID: 37207738 DOI: 10.1016/j.phrs.2023.106796] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Mesenchymal Stromal Cell (MSC) clinical applications have been widely reported and their therapeutic potential has been documented in several diseases. MSCs can be isolated from several human tissues and easily expanded in vitro, they are able to differentiate in a variety of cell lineages, and they are known to interact with most immunological cells, showing immunosuppressive and tissue repair properties. Their therapeutic efficacy is closely associated with the release of bioactive molecules, namely Extracellular Vesicles (EVs), effective as their parental cells. EVs isolated from MSCs act by fusing with target cell membrane and releasing their content, showing a great potential for the treatment of injured tissues and organs, and for the modulation of the host immune system. EV-based therapies provide, as major advantages, the possibility to cross the epithelium and blood barrier and their activity is not influenced by the surrounding environment. In the present review, we deal with pre-clinical reports and clinical trials to provide data in support of MSC and EV clinical efficacy with particular focus on neonatal and pediatric diseases. Considering pre-clinical and clinical data so far available, it is likely that cell-based and cell-free therapies could become an important therapeutic approach for the treatment of several pediatric diseases.
Collapse
Affiliation(s)
- Chiara Valsecchi
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Stefania Croce
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Elisa Lenta
- Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Gloria Acquafredda
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Patrizia Comoli
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | - Maria Antonietta Avanzini
- Pediatric Hematology Oncology Unit and Cell Factory, Fondazione IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| |
Collapse
|
45
|
Dani A, Hayes D, Guzman-Gomez A, Hossain MM, Woods JC, Morales DLS, Hirsch R, Zafar F, Hysinger EB. Lung Transplantation for Bronchopulmonary Dysplasia. Chest 2023; 163:1166-1175. [PMID: 36610665 PMCID: PMC10206512 DOI: 10.1016/j.chest.2022.12.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Patients with bronchopulmonary dysplasia (BPD) have poor respiratory trajectories and are at increased risk of lung function decline with age. Lung transplant (LTx) is a possible treatment option for this growing patient population, but little has been published on LTx in this patient group. RESEARCH QUESTION What are the characteristics of patients with BPD who are listed for LTx? How do waitlist and post-LTx outcomes for BPD compare with LTx for other diagnoses? STUDY DESIGN AND METHODS The United Network for Organ Sharing (UNOS) registry was queried for patients of all ages listed for or who underwent LTx (2000-2020). Descriptive analysis, waitlist outcomes, and post-LTx survival at 1, 5, and 10 years were assessed comparing patients with BPD vs LTx patients with other diagnoses. Post-LTx survival for patients with BPD born in the pre-surfactant era (pre-SE, before 1990) and those born in the post-surfactant era (post-SE) was compared. Propensity score matching was performed to control for the risk factors and match patients with BPD with other LTx patients on a 1:1 ratio. RESULTS BPD was reported in 65 patients, of whom 32 (49.2%) underwent LTx. Patients with BPD at listing were younger than those with other diagnoses (median age, 21 [interquartile range, 5-31] years vs 57 [45-63] years; P < .001), and more were likely to receive mechanical ventilation at listing (23% vs 3.7%; P < .001). Patients with BPD had an FEV1 of 17% compared with 34% predicted in other patients (P = .002). Patients with BPD had an overall similar post-LTx survival compared with patients with other diagnoses (P = .106), even following propensity score matching (P = .41). INTERPRETATION LTx for BPD has increased over the last 20 years. Patients with BPD have similar post-LTx outcomes compared with those of other patient populations in the modern era. Thus, LTx could be considered for patients with BPD experiencing progressive respiratory deterioration.
Collapse
Affiliation(s)
- Alia Dani
- Department of Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Don Hayes
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Amalia Guzman-Gomez
- Department of Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Md Monir Hossain
- Deparment of Pediatrics, Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jason C Woods
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - David L S Morales
- Department of Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Russel Hirsch
- Department of Pediatrics, Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Farhan Zafar
- Department of Cardiothoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Erik B Hysinger
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
46
|
Zanini F, Che X, Knutsen C, Liu M, Suresh NE, Domingo-Gonzalez R, Dou SH, Zhang D, Pryhuber GS, Jones RC, Quake SR, Cornfield DN, Alvira CM. Developmental diversity and unique sensitivity to injury of lung endothelial subtypes during postnatal growth. iScience 2023; 26:106097. [PMID: 36879800 PMCID: PMC9984561 DOI: 10.1016/j.isci.2023.106097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/20/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
At birth, the lung is still immature, heightening susceptibility to injury but enhancing regenerative capacity. Angiogenesis drives postnatal lung development. Therefore, we profiled the transcriptional ontogeny and sensitivity to injury of pulmonary endothelial cells (EC) during early postnatal life. Although subtype speciation was evident at birth, immature lung EC exhibited transcriptomes distinct from mature counterparts, which progressed dynamically over time. Gradual, temporal changes in aerocyte capillary EC (CAP2) contrasted with more marked alterations in general capillary EC (CAP1) phenotype, including distinct CAP1 present only in the early alveolar lung expressing Peg3, a paternally imprinted transcription factor. Hyperoxia, an injury that impairs angiogenesis induced both common and unique endothelial gene signatures, dysregulated capillary EC crosstalk, and suppressed CAP1 proliferation while stimulating venous EC proliferation. These data highlight the diversity, transcriptomic evolution, and pleiotropic responses to injury of immature lung EC, possessing broad implications for lung development and injury across the lifespan.
Collapse
Affiliation(s)
- Fabio Zanini
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South Wales, Sydney, Kensington, NSW 2052, Australia
| | - Xibing Che
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carsten Knutsen
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Min Liu
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nina E. Suresh
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Racquel Domingo-Gonzalez
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steve H. Dou
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daoqin Zhang
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gloria S. Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Robert C. Jones
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Stephen R. Quake
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - David N. Cornfield
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Cristina M. Alvira
- Center for Excellence in Pulmonary Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
47
|
Marc I, Boutin A, Pronovost E, Perez Herrera NM, Guillot M, Bergeron F, Moore L, Sullivan TR, Lavoie PM, Makrides M. Association Between Enteral Supplementation With High-Dose Docosahexaenoic Acid and Risk of Bronchopulmonary Dysplasia in Preterm Infants: A Systematic Review and Meta-analysis. JAMA Netw Open 2023; 6:e233934. [PMID: 36943265 PMCID: PMC10031388 DOI: 10.1001/jamanetworkopen.2023.3934] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/03/2023] [Indexed: 03/23/2023] Open
Abstract
Importance High-dose docosahexaenoic acid (DHA), a long-chain polyunsaturated fatty acid, may affect the risk of bronchopulmonary dysplasia (BPD). However, high-level summative evidence supporting such clinical association in very preterm infants is lacking. Objective To examine the association between enteral supplementation with high-dose DHA during the neonatal period and the risk of BPD in preterm infants born at less than 29 weeks' gestation. Data Sources PubMed, Embase, Web of Science, Cochrane Central Register of Controlled Trials, medRxiv, and ClinicalTrials.gov were searched from inception to August 1, 2022, for eligible articles with no language restrictions. Study Selection Randomized clinical trials (RCTs) were eligible for inclusion (1) if their interventions involved direct administration of a minimum DHA supplementation of 40 mg/kg/d or breast milk or formula feeding of at least 0.4% of total fatty acids, and (2) if they reported data on either BPD, death, BPD severity, or a combined outcome of BPD and death. Data Extraction and Synthesis Two investigators completed independent review of titles and abstracts, full text screening, data extraction, and quality assessment using the Cochrane Risk of Bias 2.0. Risk ratios (RRs) with 95% CIs were pooled using random-effect meta-analyses. Main Outcomes and Measures Primary outcome was BPD using trial-specific definitions, which was further stratified for RCTs that used a more stringent BPD definition based on systematic pulse oximetry assessment at 36 weeks' postmenstrual age. Other outcomes were BPD, death, BPD severity, or combined BPD and death. Results Among the 2760 studies screened, 4 RCTs were included, which involved 2304 infants (1223 boys [53.1%]; mean [SD] gestational age, 26.5 [1.6] weeks). Enteral supplementation with high-dose DHA was associated with neither BPD (4 studies [n = 2186 infants]; RR, 1.07 [95% CI, 0.86-1.34]; P = .53; I2 = 72%) nor BPD or death (4 studies [n = 2299 infants]; RR, 1.04 [95% CI, 0.91-1.18]; P = .59; I2 = 61%). However, an inverse association with BPD was found in RCTs that used a more stringent BPD definition (2 studies [n = 1686 infants]; RR, 1.20 [95% CI, 1.01-1.42]; P = .04; I2 = 48%). Additionally, DHA was inversely associated with moderate-to-severe BPD (3 studies [n = 1892 infants]; RR, 1.16 [95% CI, 1.04-1.29]; P = .008; I2 = 0%). Conclusions and Relevance Results of this study showed that enteral supplementation with high-dose DHA in the neonatal period was not associated overall with BPD, but an inverse association was found in the included RCTs that used a more stringent BPD definition. These findings suggest that high-dose DHA supplementation should not be recommended to prevent BPD in very preterm infants.
Collapse
Affiliation(s)
- Isabelle Marc
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Amélie Boutin
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Etienne Pronovost
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Norma Maria Perez Herrera
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Mireille Guillot
- Department of Pediatrics, Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | | | - Lynne Moore
- Department of Social and Preventive Medicine, Université Laval, Québec, Québec, Canada
| | - Thomas R. Sullivan
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Pascal M. Lavoie
- Department of Pediatrics, Division of Neonatology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Maria Makrides
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
48
|
Romijn M, van Kaam AH, Fenn D, Bos LD, van den Akker CHP, Finken MJJ, Rotteveel J, Cerullo J, Brinkman P, Onland W. Exhaled Volatile Organic Compounds for Early Prediction of Bronchopulmonary Dysplasia in Infants Born Preterm. J Pediatr 2023:113368. [PMID: 36868304 DOI: 10.1016/j.jpeds.2023.02.014] [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: 12/21/2022] [Revised: 02/10/2023] [Accepted: 02/12/2023] [Indexed: 03/05/2023]
Abstract
OBJECTIVE(S) To investigate the predictive performances of exhaled breath volatile organic compounds (VOCs) for development of bronchopulmonary dysplasia (BPD) in infants born preterm. METHODS Exhaled breath was collected from infants born <30 weeks' gestation at days 3 and 7 of life. Ion-fragments detected by gas-Chromatography-mass-spectrometry analysis were used to derive and internally validate a VOC prediction model for moderate or severe BPD at 36 weeks postmenstrual age. We tested the predictive performance of the National Institute of Child Health and Human Development (NICHD) clinical BPD prediction model with and without VOCs. RESULTS Breath samples were collected from 117 infants (mean gestation 26.8 [±1.5] weeks). Thirty-three percent of the infants developed moderate or severe BPD. The VOC model showed a c-statistic of 0.89 (95% confidence interval [CI] 0.80-0.97) and 0.92 (95% CI 0.84-0.99)) for the prediction of BPD at days 3 and 7, respectively. Adding the VOCs to the clinical prediction model in non-invasive supported infants resulted in significant improvement in discriminative power on both days (day 3: c-statistic 0.83 versus 0.92, p-value 0.04; day 7: c-statistic 0.82 versus 0.94, p-value 0.03). CONCLUSIONS This study showed that VOC profiles in exhaled breath of preterm infants on non-invasive support in the first week of life differ between those developing and not developing BPD. Adding VOCs to a clinical prediction model significantly improved its discriminative performance.
Collapse
Affiliation(s)
- Michelle Romijn
- Amsterdam UMC location University of Amsterdam, Department of Pediatrics-Neonatology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric-Endocrinology, Boelelaan 1117, Amsterdam, the Netherlands
| | - Anton H van Kaam
- Amsterdam UMC location University of Amsterdam, Department of Pediatrics-Neonatology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Dominic Fenn
- Amsterdam UMC location University of Amsterdam, Department of Respiratory Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of laboratory of Experimental Intensive Care and Anaesthesiology, Meibergdreef 9, Amsterdam, the Netherlands
| | - Lieuwe D Bos
- Amsterdam UMC location University of Amsterdam, Department of Respiratory Medicine, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of laboratory of Experimental Intensive Care and Anaesthesiology, Meibergdreef 9, Amsterdam, the Netherlands
| | - Chris H P van den Akker
- Amsterdam UMC location University of Amsterdam, Department of Pediatrics-Neonatology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Martijn J J Finken
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric-Endocrinology, Boelelaan 1117, Amsterdam, the Netherlands
| | - Joost Rotteveel
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pediatric-Endocrinology, Boelelaan 1117, Amsterdam, the Netherlands
| | - Julia Cerullo
- Division of Neonatolgy "Villa dei Fiori" Hospital, Acerra, Naples, Italy
| | - Paul Brinkman
- Amsterdam UMC location University of Amsterdam, Department of Respiratory Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Wes Onland
- Amsterdam UMC location University of Amsterdam, Department of Pediatrics-Neonatology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands.
| |
Collapse
|
49
|
Park SJ, Bae MH, Jeong MH, Jeong SH, Lee N, Byun SY, Park KH. Risk factors and clinical outcomes of extubation failure in very early preterm infants: a single-center cohort study. BMC Pediatr 2023; 23:36. [PMID: 36681822 PMCID: PMC9863082 DOI: 10.1186/s12887-023-03833-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 01/02/2023] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Early extubation success (ES) in preterm infants may reduce various mechanical ventilation-associated complications; however, extubation failure (EF) can cause adverse short- and long-term outcomes. Therefore, the present study aimed to identify differences in risk factors and clinical outcomes between ES and EF in very early preterm infants. METHODS This retrospective study was conducted between January 2017 and December 2021. Premature infants born at 32 weeks' gestational age in whom extubation had failed at least once were assigned to the EF group. Successfully extubated patients with a similar gestational age and birth weight as those in the EF group were assigned to the ES group. EF was defined as the need for re-intubation within 120 h of extubation. Various variables were compared between groups. RESULTS The EF rate in this study was 18.6% (24/129), and approximately 80% of patients with EF required re-intubation within 90.17 h. In the ES group, there was less use of inotropes within 7 days of life (12 [63.2%] vs. 22 [91.7%], p = 0.022), a lower respiratory severity score (RSS) at 1 and 4 weeks (1.72 vs. 2.5, p = 0.026; 1.73 vs. 2.92, p = 0.010), and a faster time to reach full feeding (18.7 vs. 29.7, p = 0.020). There was a higher severity of bronchopulmonary dysplasia BPD (3 [15.8%] vs. 14 [58.3%], p = 0.018), longer duration of oxygen supply (66.5 vs. 92.9, p = 0.042), and higher corrected age at discharge (39.6 vs. 42.5, p = 0.043) in the EF group. The cutoff value, sensitivity, and specificity of the respiratory severity score (RSS) at 1 week were 1.98, 0.71, and 0.42, respectively, and the cutoff value, sensitivity, and specificity of RSS at 4 weeks were 2.22, 0.67, and 0.47, respectively. CONCLUSIONS EF caused adverse short-term outcomes such as a higher BPD severity and longer hospital stay. Therefore, extubation in very early preterm infants should be carefully evaluated. Using inotropes, feeding, and RSS at 1 week of age can help predict extubation success.
Collapse
Affiliation(s)
- Su Jeong Park
- Department of Pediatrics, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Mi Hye Bae
- Department of Pediatrics, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Mun Hui Jeong
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Seong Hee Jeong
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - NaRae Lee
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Shin Yun Byun
- Department of Pediatrics, Pusan National University Children's Hospital, Pusan National University School of Medicine, Yangsan, Korea
| | - Kyung Hee Park
- Department of Pediatrics, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea.
| |
Collapse
|
50
|
Yao H, Wallace J, Peterson AL, Scaffa A, Rizal S, Hegarty K, Maeda H, Chang JL, Oulhen N, Kreiling JA, Huntington KE, De Paepe ME, Barbosa G, Dennery PA. Timing and cell specificity of senescence drives postnatal lung development and injury. Nat Commun 2023; 14:273. [PMID: 36650158 PMCID: PMC9845377 DOI: 10.1038/s41467-023-35985-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
Senescence causes age-related diseases and stress-related injury. Paradoxically, it is also essential for organismal development. Whether senescence contributes to lung development or injury in early life remains unclear. Here, we show that lung senescence occurred at birth and decreased throughout the saccular stage in mice. Reducing senescent cells at this stage disrupted lung development. In mice (<12 h old) exposed to hyperoxia during the saccular stage followed by air recovery until adulthood, lung senescence increased particularly in type II cells and secondary crest myofibroblasts. This peaked during the alveolar stage and was mediated by the p53/p21 pathway. Decreasing senescent cells during the alveolar stage attenuated hyperoxia-induced alveolar and vascular simplification. Conclusively, early programmed senescence orchestrates postnatal lung development whereas later hyperoxia-induced senescence causes lung injury through different mechanisms. This defines the ontogeny of lung senescence and provides an optimal therapeutic window for mitigating neonatal hyperoxic lung injury by inhibiting senescence.
Collapse
Affiliation(s)
- Hongwei Yao
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA.
| | - Joselynn Wallace
- Center for Computational Biology of Human Disease and Center for Computation and Visualization, Brown University, Providence, RI, 02912, USA
| | - Abigail L Peterson
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Alejandro Scaffa
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Salu Rizal
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Katy Hegarty
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Hajime Maeda
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Jason L Chang
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Nathalie Oulhen
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Jill A Kreiling
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Kelsey E Huntington
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI, 02903, USA
| | - Monique E De Paepe
- Department of Pathology, Women and Infants Hospital, Providence, RI, 02905, USA
| | - Guilherme Barbosa
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA
| | - Phyllis A Dennery
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, RI, 02912, USA.
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI, 02903, USA.
| |
Collapse
|