1
|
Groves AM, Bennett MM, Loyd J, Clark RH, Tolia VN. Trajectory of Postnatal Oxygen Requirement in Extremely Preterm Infants. J Pediatr 2025; 277:114414. [PMID: 39577761 DOI: 10.1016/j.jpeds.2024.114414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
OBJECTIVE To describe the trends in percentage oxygen requirement and mode of respiratory support delivered to extremely premature infants in the 12 weeks after birth. STUDY DESIGN This is a retrospective study of extremely premature infants (≤276/7 weeks) discharged from neonatal intensive care units managed by Pediatrix Medical Group between January 1, 2016, and December 31, 2021. Demographic and daily clinical data (mode of respiratory support and fraction of inspired oxygen [FiO2]) were extracted from the Pediatrix Clinical Data Warehouse. RESULTS A total of 16 386 infants with a median gestational age of 25 weeks and birthweight of 765 g were included. There were 3808 (23.2%) infants who died. Of the cohort, 6019 (43.1%) infants who survived to 36 weeks' gestation had bronchopulmonary dysplasia. Median FiO2 at all gestations followed a biphasic pattern with a peak on day of life 1, reduction to a nadir by day 4 to 5, and an increase to a second peak around day 14. Infants born at lower gestational ages had a higher median FiO2 at each time point. At lower gestations, there were higher proportions of infants receiving mechanical ventilation and a later introduction of noninvasive modes. CONCLUSIONS Extremely premature infants show a consistent biphasic pattern in percentage of supplemental oxygen required after birth.
Collapse
Affiliation(s)
- Alan M Groves
- Pediatrix Neonatology of Texas, Austin, TX; Department of Pediatrics, Dell Medical School, University of Texas at Austin, Austin, TX.
| | | | - John Loyd
- Pediatrix Neonatology of Texas, Austin, TX; Department of Pediatrics, Dell Medical School, University of Texas at Austin, Austin, TX
| | - Reese H Clark
- Pediatrix Center for Research Education, Quality, and Safety, Sunrise, FL
| | - Veeral N Tolia
- Pediatrix Center for Research Education, Quality, and Safety, Sunrise, FL; Division of Neonatology, Department of Pediatrics, Baylor University Medical Center, Dallas, TX; Pediatrix Medical Group, Dallas, TX
| |
Collapse
|
2
|
Pereira-Fantini PM, Byars SG, Kamlin COF, Manley BJ, Davis PG, Tingay DG. Plasma Proteome Profiles Associated with Early Development of Lung Injury in Extremely Preterm Infants. Am J Respir Cell Mol Biol 2024; 71:677-687. [PMID: 39051934 DOI: 10.1165/rcmb.2024-0034oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 07/27/2024] Open
Abstract
The biological mediators that initiate lung injury in extremely preterm infants during early postnatal life remain largely unidentified, limiting opportunities for early treatment and diagnosis. In this exploratory study, we used sequential window acquisition of all theoretical mass spectra mass spectrometry to identify bronchopulmonary dysplasia (BPD)-specific changes in protein abundance in plasma samples obtained in the first 72 hours of life from extremely preterm infants and bioinformatic analysis to identify BPD-related biological categories and pathways. Last, binary logistic regression analysis was used to test the BPD predictive potential of a base model alone (gestational age, birth weight, sex) and with the protein biomarker added, with bootstrap resampling used to internally validate protein predictors and adjust for overoptimism. We observed disturbance of key processes, including coagulation, complement activation, development, and extracellular matrix organization, in the first days of life in extremely preterm infants who later received diagnoses of BPD. In the BPD prediction analysis, 49 plasma proteins were identified; when each singularly was combined with birth characteristics the optimism-adjusted C index was 0.65-0.84, suggesting predictive potential for BPD outcomes. Taken together, the results of this study demonstrate that alterations in plasma proteins can be detected from 4 hours of age in extremely preterm infants who later develop BPD and that protein biomarkers, when combined with three birth characteristics, have the potential to predict BPD development within the first 72 hours of life.
Collapse
Affiliation(s)
- Prue M Pereira-Fantini
- Neonatal Research Group and
- Department of Paediatrics and
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Sean G Byars
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - C Omar F Kamlin
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Brett J Manley
- Victorian Infant Brain Studies Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Peter G Davis
- Neonatal Research Group and
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - David G Tingay
- Neonatal Research Group and
- Department of Paediatrics and
- Newborn Research, The Royal Women's Hospital, Parkville, Victoria, Australia; and
| |
Collapse
|
3
|
El-Atawi K, Abdul Wahab MG, Alallah J, Osman MF, Hassan M, Siwji Z, Saleh M. Beyond Bronchopulmonary Dysplasia: A Comprehensive Review of Chronic Lung Diseases in Neonates. Cureus 2024; 16:e64804. [PMID: 39156276 PMCID: PMC11329945 DOI: 10.7759/cureus.64804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
In neonates, pulmonary diseases such as bronchopulmonary dysplasia and other chronic lung diseases (CLDs) pose significant challenges due to their complexity and high degree of morbidity and mortality. This review discusses the etiology, pathophysiology, clinical presentation, and diagnostic criteria for these conditions, as well as current management strategies. The review also highlights recent advancements in understanding the pathophysiology of these diseases and evolving strategies for their management, including gene therapy and stem cell treatments. We emphasize how supportive care is useful in managing these diseases and underscore the importance of a multidisciplinary approach. Notably, we discuss the emerging role of personalized medicine, enabled by advances in genomics and precision therapeutics, in tailoring therapy according to an individual's genetic, biochemical, and lifestyle factors. We conclude with a discussion on future directions in research and treatment, emphasizing the importance of furthering our understanding of these conditions, improving diagnostic criteria, and exploring targeted treatment modalities. The review underscores the need for multicentric and longitudinal studies to improve preventative strategies and better understand long-term outcomes. Ultimately, a comprehensive, innovative, and patient-centered approach can enhance the quality of care and outcomes for neonates with CLDs.
Collapse
Affiliation(s)
| | | | - Jubara Alallah
- Neonatology, King Saud Bin Abdulaziz University for Health Sciences, Jeddah, SAU
- Neonatology, King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Jeddah, SAU
| | | | | | | | - Maysa Saleh
- Pediatrics and Child Health, Al Jalila Children's Specialty Hospital, Dubai, ARE
| |
Collapse
|
4
|
Chen Y, Song Y, Peng H, Li J, Zhao C, Liu D, Tan J, Liu Y. Changes in Thymic Size and Immunity Are Associated with Bronchopulmonary Dysplasia. Am J Perinatol 2024; 41:e1732-e1739. [PMID: 37192653 DOI: 10.1055/s-0043-1768704] [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] [Indexed: 05/18/2023]
Abstract
OBJECTIVE Preterm infants with bronchopulmonary dysplasia (BPD) are at increased risk for dysfunctional immune responses in the postnatal period. This study aimed to verify the hypothesis that thymic function is altered in infants with BPD and changes in the expression of thymic function-related genes affect thymic development. STUDY DESIGN Included in the study were infants who had a gestational age ≤32 weeks and survived to a postmenstrual age of ≥36 weeks. The clinical features and thymic size were comparatively studied between infants with and without BPD. Thymic function and the expression of thymic function-related genes were determined in BPD infants at birth, week 2, and 4 of life. The thymic size was ultrasonographically assessed in terms of the thymic index (TI) and thymic weight index (TWI). T-cell receptor excision circles (TRECs) and gene expression were quantitatively determined by real-time quantitative reverse transcription polymerase chain reaction. RESULTS Compared to non-BPD infants, their BPD counterparts had a shorter GA, lower birth weight, lower Apgar scores at birth, and were more likely to be of the male gender. BPD infants had an elevated incidence of respiratory distress syndrome and sepsis. TI was 1.73 ± 0.68 versus 2.87 ± 0.70 cm3 and TWI was 1.38 ± 0.45 versus 1.72 ± 0.28 cm3/kg in the BPD group versus the non-BPD group (p < 0.05). In BPD infants, no significant changes were observed in thymic size, lymphocyte counts, and TREC copy numbers at the first 2 weeks (p > 0.05), but they all exhibited a significant increase at week 4 (p < 0.05). BPD infants presented a trend toward increased expression of transforming growth factor-β1 and decreased expression of forkhead box protein 3 (Foxp3) from birth to week 4 (p < 0.05). Nonetheless, no significant difference was found in IL-2 or IL-7 expression at all time points (p > 0.05). CONCLUSION For preterm infants with BPD, reduced thymic size at birth might be associated with impaired thymic function. Thymic function was developmentally regulated in the BPD process. KEY POINTS · For preterm infants with BPD, reduced thymic size at birth might be associated with impaired thymic.. · BPD infants had an elevated incidence of respiratory distress syndrome and sepsis.. · Thymic function was developmentally regulated in the BPD process..
Collapse
Affiliation(s)
- Yan Chen
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Lab of Molecular Imaging, China
| | - Hua Peng
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Li
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Zhao
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Liu
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Tan
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yalan Liu
- Department of Pediatric, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
D’Amico F, Lugarà C, Luppino G, Giuffrida C, Giorgianni Y, Patanè EM, Manti S, Gambadauro A, La Rocca M, Abbate T. The Influence of Neurotrophins on the Brain-Lung Axis: Conception, Pregnancy, and Neonatal Period. Curr Issues Mol Biol 2024; 46:2528-2543. [PMID: 38534776 PMCID: PMC10968818 DOI: 10.3390/cimb46030160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Neurotrophins (NTs) are four small proteins produced by both neuronal and non-neuronal cells; they include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4). NTs can exert their action through both genomic and non-genomic mechanisms by interacting with specific receptors. Initial studies on NTs have identified them only as functional molecules of the nervous system. However, recent research have shown that some tissues and organs (such as the lungs, skin, and skeletal and smooth muscle) as well as some structural cells can secrete and respond to NTs. In addition, NTs perform several roles in normal and pathological conditions at different anatomical sites, in both fetal and postnatal life. During pregnancy, NTs are produced by the mother, placenta, and fetus. They play a pivotal role in the pre-implantation process and in placental and embryonic development; they are also involved in the development of the brain and respiratory system. In the postnatal period, it appears that NTs are associated with some diseases, such as sudden infant death syndrome (SIDS), asthma, congenital central hypoventilation syndrome (CCHS), and bronchopulmonary dysplasia (BPD).
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, AOUP G. Martino, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (F.D.); (C.L.); (G.L.); (C.G.); (Y.G.); (E.M.P.); (A.G.); (M.L.R.); (T.A.)
| | | | | | | |
Collapse
|
6
|
Wozniak PS, Makhoul L, Botros MM. Bronchopulmonary dysplasia in adults: Exploring pathogenesis and phenotype. Pediatr Pulmonol 2024; 59:540-551. [PMID: 38050796 DOI: 10.1002/ppul.26795] [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: 06/05/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023]
Abstract
This review highlights both the longstanding impact of bronchopulmonary dysplasia (BPD) on the health of adult survivors of prematurity and the pressing need for prospective, longitudinal studies of this population. Conservatively, there are an estimated 1,000,000 survivors of BPD in the United States alone. Unfortunately, most of the available literature regarding outcomes of lung disease due to prematurity naturally focuses on pediatric patients in early or middle childhood, and the relative amount of literature on adult survivors is scant. As the number of adult survivors of BPD continues to increase, it is essential that both adult and pediatric pulmonologists have a comprehensive understanding of the pathophysiology and underlying disease process, including the molecular signaling pathways and pro-inflammatory modulators that contribute to the pathogenesis of BPD. We summarize the most common presenting symptoms for adults with BPD and identify the critical challenges adult pulmonologists face in managing the care of survivors of prematurity. Specifically, these challenges include the wide variability of the clinical presentation of adult patients, comorbid cardiopulmonary complications, and the paucity of longitudinal data available on these patients. Adult survivors of BPD have even required lung transplantation, indicating the high burden of morbidity that can result from premature birth and subsequent lung injury. In addition, we analyze the disparate symptoms and management approach to adults with "old" BPD versus "new" BPD. The aim of this review is to assist pulmonologists in understanding the underlying pathophysiology of BPD and to improve clinical recognition of this increasingly common pulmonary disease.
Collapse
Affiliation(s)
- Phillip S Wozniak
- Department of Internal Medicine, Kansas City, Missouri, USA
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri, USA
- University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Lara Makhoul
- University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Mena M Botros
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
7
|
Zhang S, Li X, Yuan T, Guo X, Jin C, Jin Z, Li J. Glutamine inhibits inflammation, oxidative stress, and apoptosis and ameliorates hyperoxic lung injury. J Physiol Biochem 2023; 79:613-623. [PMID: 37145351 DOI: 10.1007/s13105-023-00961-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/05/2023] [Indexed: 05/06/2023]
Abstract
Glutamine (Gln) is the most widely acting and abundant amino acid in the body and has anti-inflammatory properties, regulates body metabolism, and improves immune function. However, the mechanism of Gln's effect on hyperoxic lung injury in neonatal rats is unclear. Therefore, this work focused on examining Gln's function in lung injury of newborn rats mediated by hyperoxia and the underlying mechanism. We examined body mass and ratio of wet-to-dry lung tissue weights of neonatal rats. Hematoxylin and eosin (HE) staining was performed to examine histopathological alterations of lung tissues. In addition, enzyme-linked immunoassay (ELISA) was conducted to measure pro-inflammatory cytokine levels within bronchoalveolar lavage fluid (BALF). Apoptosis of lung tissues was observed using TUNEL assay. Western blotting was performed for detecting endoplasmic reticulum stress (ERS)-associated protein levels. The results showed that Gln promoted body weight gain, significantly reduced pathological damage and oxidative stress in lung tissue, and improved lung function in neonatal rats. Gln reduced pro-inflammatory cytokine release as well as inflammatory cell production in BALF and inhibited apoptosis in lung tissue cells. Furthermore, we found that Gln could downregulate ERS-associated protein levels (GRP78, Caspase-12, CHOP) and inhibit c-Jun N-terminal kinase (JNK) and inositol-requiring enzyme 1 alpha (IRE1α) phosphorylation. These results in an animal model of bronchopulmonary dysplasia (BPD) suggest that Gln may have a therapeutic effect on BPD by reducing lung inflammation, oxidative stress, and apoptosis and improving lung function; its mechanism of action may be related to the inhibition of the IRE1α/JNK pathway.
Collapse
Affiliation(s)
- Shujian Zhang
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Xuewei Li
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Tiezheng Yuan
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, Jilin, China
| | - Xiangyu Guo
- Center of Morphological Experiment, Medical College of Yanbian University, Yanji, Jilin, China
| | - Can Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Zhengyong Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin, China.
- Department of Pediatrics, Second Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Jinliang Li
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
8
|
Kandasamy J, Li R, Vamesu BM, Olave N, Halloran B, Jilling T, Ballinger SW, Ambalavanan N. Mitochondrial DNA Variations Modulate Alveolar Epithelial Mitochondrial Function and Oxidative Stress in Newborn Mice Exposed to Hyperoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.17.541177. [PMID: 37292719 PMCID: PMC10245974 DOI: 10.1101/2023.05.17.541177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Oxidative stress is an important contributor to bronchopulmonary dysplasia (BPD), a form of chronic lung disease that is the most common morbidity in very preterm infants. Mitochondrial functional differences due to inherited and acquired mutations influence the pathogenesis of disorders in which oxidative stress plays a critical role. We previously showed using mitochondrial-nuclear exchange (MNX) mice that mitochondrial DNA (mtDNA) variations modulate hyperoxia-induced lung injury severity in a model of BPD. In this study, we studied the effects of mtDNA variations on mitochondrial function including mitophagy in alveolar epithelial cells (AT2) from MNX mice. We also investigated oxidant and inflammatory stress as well as transcriptomic profiles in lung tissue in mice and expression of proteins such as PINK1, Parkin and SIRT3 in infants with BPD. Our results indicate that AT2 from mice with C57 mtDNA had decreased mitochondrial bioenergetic function and inner membrane potential, increased mitochondrial membrane permeability and were exposed to higher levels of oxidant stress during hyperoxia compared to AT2 from mice with C3H mtDNA. Lungs from hyperoxia-exposed mice with C57 mtDNA also had higher levels of pro-inflammatory cytokines compared to lungs from mice with C3H mtDNA. We also noted changes in KEGG pathways related to inflammation, PPAR and glutamatergic signaling, and mitophagy in mice with certain mito-nuclear combinations but not others. Mitophagy was decreased by hyperoxia in all mice strains, but to a greater degree in AT2 and neonatal mice lung fibroblasts from hyperoxia-exposed mice with C57 mtDNA compared to C3H mtDNA. Finally, mtDNA haplogroups vary with ethnicity, and Black infants with BPD had lower levels of PINK1, Parkin and SIRT3 expression in HUVEC at birth and tracheal aspirates at 28 days of life when compared to White infants with BPD. These results indicate that predisposition to neonatal lung injury may be modulated by variations in mtDNA and mito-nuclear interactions need to be investigated to discover novel pathogenic mechanisms for BPD.
Collapse
|
9
|
Windhorst AC, Heydarian M, Schwarz M, Oak P, Förster K, Frankenberger M, Gonzalez Rodriguez E, Zhang X, Ehrhardt H, Hübener C, Flemmer AW, Hossain H, Stoeger T, Schulz C, Hilgendorff A. Monocyte signature as a predictor of chronic lung disease in the preterm infant. Front Immunol 2023; 14:1112608. [PMID: 37090732 PMCID: PMC10113536 DOI: 10.3389/fimmu.2023.1112608] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
IntroductionInflammation is a key driver of morbidity in the vulnerable preterm infant exposed to pre- and postnatal hazards and significantly contributes to chronic lung disease, i.e. bronchopulmonary dysplasia (BPD). However, the early changes in innate immunity associated with BPD development are incompletely understood.MethodsIn very immature preterm infants below 32 weeks gestational age (GA; n=30 infants), monocyte subtypes were identified by Flow Cytometry at birth and throughout the postnatal course including intracellular TNF expression upon LPS stimulation. Complementing these measurements, cytokine end growth factor expression profiles (Luminex® xMAP®; n=110 infants) as well as gene expression profiles (CodeLinkTM Human I Bioarray; n=22) were characterized at birth.ResultsThe abundance of monocyte subtypes differed between preterm and term neonates at birth. Specifically, CD14++CD16+ (intermediate) monocytes demonstrated a dependency on PMA and elevated levels of nonclassical (CD14+CD16++) monocytes characterized preterm infants with developing BPD. Postnatally, lung injury was associated with an increase in intermediate monocytes, while high levels of nonclassical monocytes persisted. Both subtypes were revealed as the main source of intracellular TNF-α expression in the preterm infant. We identified a cytokine and growth factor expression profile in cord blood specimen of preterm infants with developing BPD that corresponded to the disease-dependent regulation of monocyte abundances. Multivariate modeling of protein profiles revealed FGF2, sIL-2 Rα, MCP-1, MIP1a, and TNF-α as predictors of BPD when considering GA. Transcriptome analysis demonstrated genes predicting BPD to be overrepresented in inflammatory pathways with increased disease severity characterized by the regulation of immune and defense response pathways and upstream regulator analysis confirmed TNF-α, interleukin (IL) -6, and interferon α as the highest activated cytokines in more severe disease. Whereas all BPD cases showed downstream activation of chemotaxis and activation of inflammatory response pathways, more severe cases were characterized by an additional activation of reactive oxygen species (ROS) synthesis.DiscussionIn the present study, we identified the early postnatal presence of nonclassical (CD14+CD16++) and intermediate (CD14++CD16+) monocytes as a critical characteristic of BPD development including a specific response pattern of monocyte subtypes to lung injury. Pathophysiological insight was provided by the protein and transcriptome signature identified at birth, centered around monocyte and corresponding granulocyte activation and highlighting TNFα as a critical regulator in infants with developing BPD. The disease severity-dependent expression patterns could inform future diagnostic and treatment strategies targeting the monocytic cell and its progeny.
Collapse
Affiliation(s)
- Anita C. Windhorst
- Institute of Medical Informatics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Motaharehsadat Heydarian
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Maren Schwarz
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
- Department of Neonatology, Dr. von Hauner Childre's Hospital, University Hospital, Ludwig-Maximilian-University, Munich, Germany
| | - Prajakta Oak
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Kai Förster
- Department of Neonatology, Dr. von Hauner Childre's Hospital, University Hospital, Ludwig-Maximilian-University, Munich, Germany
- Center for Comprehensive Developmental Care (CDeCLMU) at the Social Pediatric Center, Dr. von Hauner Children`s Hospital, Ludwig Maximilian University (LMU) Hospital, Ludwig-Maximilian-University, Munich, Germany
| | - Marion Frankenberger
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Erika Gonzalez Rodriguez
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Xin Zhang
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Harald Ehrhardt
- Division of Neonatology and Pediatric Intensive Care Medicine, University Medical Center Ulm, Ulm, Germany
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Christoph Hübener
- Department of Gynecology and Obstetrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilian-University, Munich, Germany
| | - Andreas W. Flemmer
- Department of Neonatology, Dr. von Hauner Childre's Hospital, University Hospital, Ludwig-Maximilian-University, Munich, Germany
| | - Hamid Hossain
- Institute for Medical Microbiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Tobias Stoeger
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
| | - Christian Schulz
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Medicine I, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Anne Hilgendorff
- Institute for Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München, German Center for Lung Research (DZL), Munich, Germany
- Center for Comprehensive Developmental Care (CDeCLMU) at the Social Pediatric Center, Dr. von Hauner Children`s Hospital, Ludwig Maximilian University (LMU) Hospital, Ludwig-Maximilian-University, Munich, Germany
- *Correspondence: Anne Hilgendorff,
| |
Collapse
|
10
|
Yee AJ, Kandasamy J, Ambalavanan N, Ren C, Halloran B, Olave N, Nicola T, Jilling T. Platelet Activating Factor Activity Modulates Hyperoxic Neonatal Lung Injury Severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532697. [PMID: 36993203 PMCID: PMC10055044 DOI: 10.1101/2023.03.14.532697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Hyperoxia-induced inflammation contributes significantly to developmental lung injury and bronchopulmonary dysplasia (BPD) in preterm infants. Platelet activating factor (PAF) is known to be a major driver of inflammation in lung diseases such as asthma and pulmonary fibrosis, but its role in BPD has not been previously investigated. Therefore, to determine whether PAF signaling independently modulates neonatal hyperoxic lung injury and BPD pathogenesis, lung structure was assessed in 14 day-old C57BL/6 wild-type (WT) and PAF receptor knockout (PTAFR KO) mice that were exposed to 21% (normoxia) or 85% O 2 (hyperoxia) from postnatal day 4. Lung morphometry showed that PTAFR KO mice had attenuated hyperoxia-induced alveolar simplification when compared to WT mice. Functional analysis of gene expression data from hyperoxia-exposed vs. normoxia-exposed lungs of WT and PTAFR KO showed that the most upregulated pathways were the hypercytokinemia/hyperchemokinemia pathway in WT mice, NAD signaling pathway in PTAFR KO mice, and agranulocyte adhesion and diapedesis as well as other pro-fibrotic pathways such as tumor microenvironment and oncostatin-M signaling in both mice strains, indicating that PAF signaling may contribute to inflammation but may not be a significant mediator of fibrotic processes during hyperoxic neonatal lung injury. Gene expression analysis also indicated increased expression of pro-inflammatory genes such as CXCL1, CCL2 and IL-6 in the lungs of hyperoxia-exposed WT mice and metabolic regulators such as HMGCS2 and SIRT3 in the lungs of PTAFR KO mice, suggesting that PAF signaling may modulate BPD risk through changes in pulmonary inflammation and/or metabolic reprogramming in preterm infants.
Collapse
|
11
|
Moreira A, Tovar M, Smith AM, Lee GC, Meunier JA, Cheema Z, Moreira A, Winter C, Mustafa SB, Seidner S, Findley T, Garcia JGN, Thébaud B, Kwinta P, Ahuja SK. Development of a peripheral blood transcriptomic gene signature to predict bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L76-L87. [PMID: 36472344 PMCID: PMC9829478 DOI: 10.1152/ajplung.00250.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/27/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common lung disease of extreme prematurity, yet mechanisms that associate with or identify neonates with increased susceptibility for BPD are largely unknown. Combining artificial intelligence with gene expression data is a novel approach that may assist in better understanding mechanisms underpinning chronic lung disease and in stratifying patients at greater risk for BPD. The objective of this study is to develop an early peripheral blood transcriptomic signature that can predict preterm neonates at risk for developing BPD. Secondary analysis of whole blood microarray data from 97 very low birth weight neonates on day of life 5 was performed. BPD was defined as positive pressure ventilation or oxygen requirement at 28 days of age. Participants were randomly assigned to a training (70%) and testing cohort (30%). Four gene-centric machine learning models were built, and their discriminatory abilities were compared with gestational age or birth weight. This study adheres to the transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD) statement. Neonates with BPD (n = 62 subjects) exhibited a lower median gestational age (26.0 wk vs. 30.0 wk, P < 0.01) and birth weight (800 g vs. 1,280 g, P < 0.01) compared with non-BPD neonates. From an initial pool (33,252 genes/patient), 4,523 genes exhibited a false discovery rate (FDR) <1%. The area under the receiver operating characteristic curve (AUC) for predicting BPD utilizing gestational age or birth weight was 87.8% and 87.2%, respectively. The machine learning models, using a combination of five genes, revealed AUCs ranging between 85.8% and 96.1%. Pathways integral to T cell development and differentiation were associated with BPD. A derived five-gene whole blood signature can accurately predict BPD in the first week of life.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Miriam Tovar
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Alisha M Smith
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Grace C Lee
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Justin A Meunier
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Zoya Cheema
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Axel Moreira
- Division of Critical Care, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Caitlyn Winter
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Shamimunisa B Mustafa
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Steven Seidner
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Tina Findley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, Texas
| | - Joe G N Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Przemko Kwinta
- Neonatal Intensive Care Unit, Department of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Sunil K Ahuja
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
12
|
Lun Y, Hu J, Zuming Y. Circular RNAs expression profiles and bioinformatics analysis in bronchopulmonary dysplasia. J Clin Lab Anal 2022; 37:e24805. [PMID: 36514862 PMCID: PMC9833990 DOI: 10.1002/jcla.24805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/26/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) has long been considered the most challenging chronic lung disease for neonatologists and researchers due to its complex pathological mechanisms and difficulty in prediction. Growing evidence indicates that BPD is associated with the dysregulation of circular RNAs (circRNAs). Therefore, we aimed to explore the expression profiles of circRNAs and investigate the underlying molecular network associated with BPD. METHODS Peripheral blood was collected from very-low-birth-weight (VLBW) infants at 5-8 days of life to extract PBMCs. Microarray analysis and qRT-PCR tests were performed to determine the differentially expressed circRNAs (DEcircRNAs) between BPD and non-BPD VLBW infants. Simultaneous analysis of GSE32472 was conducted to obtain differentially expressed mRNAs (DEmRNA) from BPD infants. The miRNAs were predicted by DEcircRNAs and DEmRNAs of upregulated, respectively, and then screened for overlapping ones. GO and KEGG analysis was performed following construction of the competing endogenous RNA regulatory network (ceRNA) for further investigation. RESULTS A total of 65 circRNAs (52 upregulated and 13 downregulated) were identified as DEcircRNAs between the two groups (FC >2.0 and p.adj <0.05). As a result, the ceRNA network was constructed based on three upregulated DEcircRNAs validated by qRT-PCR (hsa_circ_0007054, hsa_circ_0057950, and hsa_circ_0120151). Bioinformatics analysis indicated these DEcircRNAs participated in response to stimulus, IL-1 receptor activation, neutrophil activation, and metabolic pathways. CONCLUSIONS In VLBW infants with a high risk for developing BPD, the circRNA expression profiles in PBMCs were significantly altered in the early post-birth period, suggesting immune dysregulation caused by infection and inflammatory response already existed.
Collapse
Affiliation(s)
- Yu Lun
- Department of Neonatal Intensive Care UnitSuzhou Municipal HospitalJiangsu ProvinceChina
| | - Junlong Hu
- Department of Neonatal Intensive Care UnitSuzhou Municipal HospitalJiangsu ProvinceChina
| | - Yang Zuming
- Department of Neonatal Intensive Care UnitSuzhou Municipal HospitalJiangsu ProvinceChina
| |
Collapse
|
13
|
Barrett JS, Cala Pane M, Knab T, Roddy W, Beusmans J, Jordie E, Singh K, Davis JM, Romero K, Padula M, Thebaud B, Turner M. Landscape analysis for a neonatal disease progression model of bronchopulmonary dysplasia: Leveraging clinical trial experience and real-world data. Front Pharmacol 2022; 13:988974. [PMID: 36313352 PMCID: PMC9597633 DOI: 10.3389/fphar.2022.988974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022] Open
Abstract
The 21st Century Cures Act requires FDA to expand its use of real-world evidence (RWE) to support approval of previously approved drugs for new disease indications and post-marketing study requirements. To address this need in neonates, the FDA and the Critical Path Institute (C-Path) established the International Neonatal Consortium (INC) to advance regulatory science and expedite neonatal drug development. FDA recently provided funding for INC to generate RWE to support regulatory decision making in neonatal drug development. One study is focused on developing a validated definition of bronchopulmonary dysplasia (BPD) in neonates. BPD is difficult to diagnose with diverse disease trajectories and few viable treatment options. Despite intense research efforts, limited understanding of the underlying disease pathobiology and disease projection continues in the context of a computable phenotype. It will be important to determine if: 1) a large, multisource aggregation of real-world data (RWD) will allow identification of validated risk factors and surrogate endpoints for BPD, and 2) the inclusion of these simulations will identify risk factors and surrogate endpoints for studies to prevent or treat BPD and its related long-term complications. The overall goal is to develop qualified, fit-for-purpose disease progression models which facilitate credible trial simulations while quantitatively capturing mechanistic relationships relevant for disease progression and the development of future treatments. The extent to which neonatal RWD can inform these models is unknown and its appropriateness cannot be guaranteed. A component of this approach is the critical evaluation of the various RWD sources for context-of use (COU)-driven models. The present manuscript defines a landscape of the data including targeted literature searches and solicitation of neonatal RWD sources from international stakeholders; analysis plans to develop a family of models of BPD in neonates, leveraging previous clinical trial experience and real-world patient data is also described.
Collapse
Affiliation(s)
- Jeffrey S. Barrett
- Critical Path Institute, Tucson, AZ, United States
- *Correspondence: Jeffrey S. Barrett,
| | | | - Timothy Knab
- Metrum Research Group, Tariffville, CT, United States
| | | | - Jack Beusmans
- Metrum Research Group, Tariffville, CT, United States
| | - Eric Jordie
- Metrum Research Group, Tariffville, CT, United States
| | | | - Jonathan Michael Davis
- Tufts Medical Center and the Tufts Clinical and Translational Science Institute, Boston, MA, United States
| | - Klaus Romero
- Critical Path Institute, Tucson, AZ, United States
| | - Michael Padula
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Bernard Thebaud
- Department of Pediatrics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Mark Turner
- Department of Women’s and Children’s Health Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
14
|
Dong Y, Rivetti S, Lingampally A, Tacke S, Kojonazarov B, Bellusci S, Ehrhardt H. Insights into the Black Box of Intra-Amniotic Infection and Its Impact on the Premature Lung: From Clinical and Preclinical Perspectives. Int J Mol Sci 2022; 23:9792. [PMID: 36077187 PMCID: PMC9456379 DOI: 10.3390/ijms23179792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Intra-amniotic infection (IAI) is one major driver for preterm birth and has been demonstrated by clinical studies to exert both beneficial and injurious effects on the premature lung, possibly due to heterogeneity in the microbial type, timing, and severity of IAI. Due to the inaccessibility of the intra-amniotic cavity during pregnancies, preclinical animal models investigating pulmonary consequences of IAI are indispensable to elucidate the pathogenesis of bronchopulmonary dysplasia (BPD). It is postulated that on one hand imbalanced inflammation, orchestrated by lung immune cells such as macrophages, may impact on airway epithelium, vascular endothelium, and interstitial mesenchyme, resulting in abnormal lung development. On the other hand, excessive suppression of inflammation may as well cause pulmonary injury and a certain degree of inflammation is beneficial. So far, effective strategies to prevent and treat BPD are scarce. Therapeutic options targeting single mediators in signaling cascades and mesenchymal stromal cells (MSCs)-based therapies with global regulatory capacities have demonstrated efficacy in preclinical animal models and warrant further validation in patient populations. Ante-, peri- and postnatal exposome analysis and therapeutic investigations using multiple omics will fundamentally dissect the black box of IAI and its effect on the premature lung, contributing to precisely tailored and individualized therapies.
Collapse
Affiliation(s)
- Ying Dong
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Feulgen Street 12, 35392 Giessen, Germany
| | - Stefano Rivetti
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany
| | - Arun Lingampally
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany
| | - Sabine Tacke
- Clinic for Small Animals (Surgery), Faculty of Veterinary Medicine, Justus-Liebig-University, Frankfurter Street 114, 35392 Giessen, Germany
| | - Baktybek Kojonazarov
- Institute for Lung Health (ILH), Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany
| | - Saverio Bellusci
- Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University, Aulweg 130, 35392 Giessen, Germany
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Justus-Liebig-University, Feulgen Street 12, 35392 Giessen, Germany
| |
Collapse
|
15
|
The Patent Ductus Arteriosus in Extremely Preterm Neonates Is More than a Hemodynamic Challenge: New Molecular Insights. Biomolecules 2022; 12:biom12091179. [PMID: 36139018 PMCID: PMC9496182 DOI: 10.3390/biom12091179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Complications to preterm birth are numerous, including the presence of a patent ductus arteriosus (PDA). The biological understanding of the PDA is sparse and treatment remains controversial. Herein, we speculate whether the PDA is more than a cardiovascular imbalance, and may be a marker in response to immature core molecular and physiological processes driven by biological systems, such as inflammation. To achieve a new biological understanding of the PDA, we performed echocardiography and collected plasma samples on day 3 of life in 53 consecutively born neonates with a gestational age at birth below 28 completed weeks. The proteome of these samples was analyzed by mass spectrometry (nanoLC-MS/MS) and immunoassay of 17 cytokines and chemokines. We found differences in 21 proteins and 8 cytokines between neonates with a large PDA (>1.5 mm) compared to neonates without a PDA. Amongst others, we found increased levels of angiotensinogen, periostin, pro-inflammatory associations, including interleukin (IL)-1β and IL-8, and anti-inflammatory associations, including IL-1RA and IL-10. Levels of complement factors C8 and carboxypeptidases were decreased. Our findings associate the PDA with the renin-angiotensin-aldosterone system and immune- and complement systems, indicating that PDA goes beyond the persistence of a fetal circulatory connection of the great vessels.
Collapse
|
16
|
Cui H, Zhang S, Wu Z, Xu C, Xu D, Jin Z. Insulin-like growth factor-1 reduces hyperoxia-induced lung inflammation and oxidative stress and inhibits cell apoptosis through PERK/eIF2α/ATF4/CHOP signaling. Exp Lung Res 2022; 48:187-197. [PMID: 35924334 DOI: 10.1080/01902148.2022.2106388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background: Insulin-like growth factor-1 (IGF-1), a member of the insulin family, has a high degree of homology with insulin and exhibits anti-inflammatory and anti-oxidative stress properties. However, the potential protective effect of IGF-1 on hyperoxia-induced lung injury remains unknown. In this study, we aimed to explore the effects and mechanism of action of IGF-1 in hyperoxia-induced lung injury in neonatal rats. Materials and Methods: Hematoxylin-eosin staining was used to observe pathological changes in lung tissue; transmission electron microscopy was used to examine the ultrastructure, and ELISA was used to detect the level of pro-inflammatory cytokines in bronchoalveolar lavage fluid. Further, malondialdehyde, glutathione, and superoxide dismutase activities in lung tissue were evaluated. TUNEL staining was used to detect cell apoptosis, and western blot analysis was used to detect the expression of Bax, Bcl-2, Caspase-3, p-PERK, p-eIF2α, ATF4, and CHOP in the lung tissue. Moreover, the wet/dry weight ratio of lung tissue was determined. Results: Intraperitoneal injection of IGF-1 effectively reduced lung tissue damage induced by hyperoxia; production of inflammatory cells and release of pro-inflammatory cytokines, oxidative stress, and cell apoptosis. Further, IGF-1 down-regulated the expression of ATF4, CHOP, and Bax/Bcl-2, and inhibited the phosphorylation of PERK and eIF2α. Conclusion: The results suggest that IGF-1 reduces hyperoxia-induced lung inflammation and oxidative stress in neonatal rats through the PERK/eIF2α/ATF4/CHOP signaling pathway and inhibits cell apoptosis.
Collapse
Affiliation(s)
- Haixia Cui
- Department of Clinical Laboratory, Affiliated Hospital of Yanbian University, Jilin, China
| | - Shujian Zhang
- Department of Emergency and Critical Care Medicine, Second Hospital of Jilin University, Jilin, China.,Department of Pediatrics, Affiliated Hospital of Yanbian University, Jilin, China
| | - Zhengxie Wu
- Department of Clinical Laboratory, Affiliated Hospital of Yanbian University, Jilin, China
| | - Chunhua Xu
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Jilin, China
| | - Dongyuan Xu
- Center of Morphological Experiment, Medical College of Yanbian University, Jilin, China
| | - Zhengyong Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Jilin, China
| |
Collapse
|
17
|
Dong N, Zhou PP, Li D, Zhu HS, Liu LH, Ma HX, Shi Q, Ju XL. Intratracheal administration of umbilical cord-derived mesenchymal stem cells attenuates hyperoxia-induced multi-organ injury via heme oxygenase-1 and JAK/STAT pathways. World J Stem Cells 2022; 14:556-576. [PMID: 36157523 PMCID: PMC9350625 DOI: 10.4252/wjsc.v14.i7.556] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/04/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is not merely a chronic lung disease, but a systemic condition with multiple organs implications predominantly associated with hyperoxia exposure. Despite advances in current management strategies, limited progress has been made in reducing the BPD-related systemic damage. Meanwhile, although the protective effects of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) or their exosomes on hyperoxia-induced lung injury have been explored by many researchers, the underlying mechanism has not been addressed in detail, and few studies have focused on the therapeutic effect on systemic multiple organ injury.
AIM To investigate whether hUC-MSC intratracheal administration could attenuate hyperoxia-induced lung, heart, and kidney injuries and the underlying regulatory mechanisms.
METHODS Neonatal rats were exposed to hyperoxia (80% O2), treated with hUC-MSCs intratracheal (iT) or intraperitoneal (iP) on postnatal day 7, and harvested on postnatal day 21. The tissue sections of the lung, heart, and kidney were analyzed morphometrically. Protein contents of the bronchoalveolar lavage fluid (BALF), myeloperoxidase (MPO) expression, and malondialdehyde (MDA) levels were examined. Pulmonary inflammatory cytokines were measured via enzyme-linked immunosorbent assay. A comparative transcriptomic analysis of differentially expressed genes (DEGs) in lung tissue was conducted via RNA-sequencing. Subsequently, we performed reverse transcription-quantitative polymerase chain reaction and western blot analysis to explore the expression of target mRNA and proteins related to inflammatory and oxidative responses.
RESULTS iT hUC-MSCs administration improved pulmonary alveolarization and angiogenesis (P < 0.01, P < 0.01, P < 0.001, and P < 0.05 for mean linear intercept, septal counts, vascular medial thickness index, and microvessel density respectively). Meanwhile, treatment with hUC-MSCs iT ameliorated right ventricular hypertrophy (for Fulton’s index, P < 0.01), and relieved reduced nephrogenic zone width (P < 0.01) and glomerular diameter (P < 0.001) in kidneys. Among the beneficial effects, a reduction of BALF protein, MPO, and MDA was observed in hUC-MSCs groups (P < 0.01, P < 0.001, and P < 0.05 respectively). Increased pro-inflammatory cytokines tumor necrosis factor-alpha, interleukin (IL)-1β, and IL-6 expression observed in the hyperoxia group were significantly attenuated by hUC-MSCs administration (P < 0.01, P < 0.001, and P < 0.05 respectively). In addition, we observed an increase in anti-inflammatory cytokine IL-10 expression in rats that received hUC-MSCs iT compared with rats reared in hyperoxia (P < 0.05). Transcriptomic analysis showed that the DEGs in lung tissues induced by hyperoxia were enriched in pathways related to inflammatory responses, epithelial cell proliferation, and vasculature development. hUC-MSCs administration blunted these hyperoxia-induced dysregulated genes and resulted in a shift in the gene expression pattern toward the normoxia group. hUC-MSCs increased heme oxygenase-1 (HO-1), JAK2, and STAT3 expression, and their phosphorylation in the lung, heart, and kidney (P < 0.05). Remarkably, no significant difference was observed between the iT and iP administration.
CONCLUSION iT hUC-MSCs administration ameliorates hyperoxia-induced lung, heart, and kidney injuries by activating HO-1 expression and JAK/STAT signaling. The therapeutic benefits of local iT and iP administration are equivalent.
Collapse
Affiliation(s)
- Na Dong
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Pan-Pan Zhou
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Dong Li
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Hua-Su Zhu
- Department of Pediatrics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Ling-Hong Liu
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Hui-Xian Ma
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Qing Shi
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| | - Xiu-Li Ju
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
- Stem Cell and Regenerative Medicine Research Center, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
18
|
Collaco JM, McGrath-Morrow SA, Griffiths M, Chavez-Valdez R, Parkinson C, Zhu J, Northington FJ, Graham EM, Everett AD. Perinatal Inflammatory Biomarkers and Respiratory Disease in Preterm Infants. J Pediatr 2022; 246:34-39.e3. [PMID: 35460699 PMCID: PMC9264338 DOI: 10.1016/j.jpeds.2022.04.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/01/2022] [Accepted: 04/15/2022] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To measure plasma levels of vascular endothelial growth factor (VEGF) and several cytokines (Interleukin [IL]-6 IL-8, IL-10) during the first week of life to examine the relationship between protein expression and likelihood of developing respiratory distress syndrome (RDS) and bronchopulmonary dysplasia (BPD). STUDY DESIGN Levels of IL-6, IL-8, IL-10, and VEGF were measured from plasma obtained from preterm patients during the first week of life. Newborns were recruited from a single center between April 2009 and April 2019. Criteria for the study included being inborn, birth weight of less than 1500 grams, and a gestational age of less than 32 weeks at birth. RESULTS The development of RDS in preterm newborns was associated with lower levels of VEGF during the first week of life. Higher plasma levels of IL-6 and IL-8 plasma were associated with an increased likelihood and increased severity of BPD at 36 weeks postmenstrual age. In contrast, plasma levels of VEGF, IL-6, IL-8, and IL-10 obtained during the first week of life were not associated with respiratory symptoms and acute care use in young children with BPD in the outpatient setting. CONCLUSIONS During the first week of life, lower plasma levels of VEGF was associated with the diagnosis of RDS in preterm infants. Preterm infants with higher levels of IL-6 and IL-8 during the first week of life were also more likely to be diagnosed with BPD. These biomarkers may help to predict respiratory morbidities in preterm newborns during their initial hospitalization.
Collapse
Affiliation(s)
- Joseph M Collaco
- Department of Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD.
| | | | - Megan Griffiths
- Department of Pediatrics, Columbia University Medical Center, New York, NY
| | - Raul Chavez-Valdez
- Department of Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD
| | | | - Jie Zhu
- Department of Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD
| | | | - Ernest M Graham
- Department of Gynecology and Obstetrics, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Allen D Everett
- Department of Pediatrics, Johns Hopkins Medical Institutions, Baltimore, MD
| |
Collapse
|
19
|
Lao JC, Bui CB, Pang MA, Cho SX, Rudloff I, Elgass K, Schröder J, Maksimenko A, Mangan NE, Starkey MR, Skuza EM, Sun YBY, Beker F, Collins CL, Kamlin OF, König K, Malhotra A, Tan K, Theda C, Young MJ, McLean CA, Wilson NJ, Sehgal A, Hansbro PM, Pearson JT, Polo JM, Veldman A, Berger PJ, Nold-Petry CA, Nold MF. Type 2 immune polarization is associated with cardiopulmonary disease in preterm infants. Sci Transl Med 2022; 14:eaaz8454. [PMID: 35385341 DOI: 10.1126/scitranslmed.aaz8454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Postnatal maturation of the immune system is poorly understood, as is its impact on illnesses afflicting term or preterm infants, such as bronchopulmonary dysplasia (BPD) and BPD-associated pulmonary hypertension. These are both cardiopulmonary inflammatory diseases that cause substantial mortality and morbidity with high treatment costs. Here, we characterized blood samples collected from 51 preterm infants longitudinally at five time points, 20 healthy term infants at birth and age 3 to 16 weeks, and 5 healthy adults. We observed strong associations between type 2 immune polarization in circulating CD3+CD4+ T cells and cardiopulmonary illness, with odds ratios up to 24. Maternal magnesium sulfate therapy, delayed hepatitis B vaccination, and increasing fetal, but not maternal, chorioamnionitis severity were associated with attenuated type 2 polarization. Blocking type 2 mediators such as interleukin-4 (IL-4), IL-5, IL-13, or signal transducer and activator of transcription 6 (STAT6) in murine neonatal cardiopulmonary disease in vivo prevented changes in cell type composition, increases in IL-1β and IL-13, and losses of pulmonary capillaries, but not gains in larger vessels. Thereby, type 2 blockade ameliorated lung inflammation, protected alveolar and vascular integrity, and confirmed the pathological impact of type 2 cytokines and STAT6. In-depth flow cytometry and single-cell transcriptomics of mouse lungs further revealed complex associations between immune polarization and cardiopulmonary disease. Thus, this work advances knowledge on developmental immunology and its impact on early life disease and identifies multiple therapeutic approaches that may relieve inflammation-driven suffering in the youngest patients.
Collapse
Affiliation(s)
- Jason C Lao
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Christine B Bui
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Merrin A Pang
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Steven X Cho
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Ina Rudloff
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Kirstin Elgass
- Monash Micro Imaging, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Jan Schröder
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Victoria 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Anton Maksimenko
- Imaging and Medical Beamline, Australian Synchrotron, Melbourne, Victoria 3168, Australia
| | - Niamh E Mangan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne, Victoria 3168, Australia
| | - Malcolm R Starkey
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia.,Department of Immunology and Pathology, Monash University, Melbourne, Victoria 3004, Australia
| | - Elisabeth M Skuza
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Yu B Y Sun
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Victoria 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia
| | - Friederike Beker
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4101, Australia.,Neonatal Services, Mercy Hospital for Women, Melbourne, Victoria 3084, Australia
| | - Clare L Collins
- Neonatal Services, Mercy Hospital for Women, Melbourne, Victoria 3084, Australia
| | - Omar F Kamlin
- Department of Newborn Research, Royal Women's Hospital, Melbourne, Victoria 3052, Australia.,University of Melbourne, Melbourne, Victoria 3010, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria 3052, Australia
| | - Kai König
- Department of Paediatrics, Medicum Wesemlin, Lucerne 6006, Switzerland
| | - Atul Malhotra
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| | - Kenneth Tan
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| | - Christiane Theda
- Department of Newborn Research, Royal Women's Hospital, Melbourne, Victoria 3052, Australia.,University of Melbourne, Melbourne, Victoria 3010, Australia.,Murdoch Children's Research Institute, Melbourne, Victoria 3052, Australia
| | - Morag J Young
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Health, Melbourne, Victoria 3004, Australia.,Department of Medicine, Central Clinical School, Monash University, Melbourne, Victoria 3800, Australia
| | - Nicholas J Wilson
- CSL Limited, Bio21 Institute, Parkville, Melbourne, Victoria 3052, Australia
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| | - Philip M Hansbro
- Priority Research Centres for Healthy Lungs and GrowUpWell, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales 2308, Australia.,Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Ultimo, Sydney, New South Wales 2007, Australia
| | - James T Pearson
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Victoria 3800, Australia.,Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.,Victorian Heart Institute, Melbourne, Victoria 3168, Australia
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia.,Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Victoria 3800, Australia.,Australian Regenerative Medicine Institute, Monash University, Melbourne, Victoria 3800, Australia.,Adelaide Centre for Epigenetics, University of Adelaide, Adelaide, South Australia 5005, Australia.,South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Alex Veldman
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Department of Pediatrics, Helios HSK, Wiesbaden 65199, Germany.,Department of Pediatric Cardiology, J. Liebig University, Gießen 35392, Germany
| | - Philip J Berger
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Claudia A Nold-Petry
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia
| | - Marcel F Nold
- Department of Paediatrics, Monash University, Melbourne, Victoria 3168, Australia.,Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria 3168, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Victoria 3168, Australia
| |
Collapse
|
20
|
Zhang S, Luan X, Li H, Jin Z. Insulin-like growth factor-1: A potential target for bronchopulmonary dysplasia treatment (Review). Exp Ther Med 2022; 23:191. [PMID: 35126694 PMCID: PMC8794548 DOI: 10.3892/etm.2022.11114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/08/2021] [Indexed: 11/05/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common respiratory disorder among preterm infants, particularly low-birth-weight infants (LBWIs) and very-low-birth-weight infants (VLBWIs). Although BPD was first reported 50 years ago, no specific drugs or efficient measures are yet available for prevention or treatment. Insulin-like growth factor-1 (IGF-1) belongs to the insulin family. It promotes mitosis and stimulates cell proliferation and DNA synthesis, the primary factors involved in pulmonary development during the fetal and postnatal periods. Several studies have reported that IGF-1 exerts certain effects on BPD genesis and progression by regulating BPD-related biological processes. In addition, exogenous addition of IGF-1 can alleviate lung inflammation, cell apoptosis and eliminate alveolar development disorders in children with BPD. These findings suggest that IGF-1 could be a new target for treating BPD. Here, we summarize and analyze the definition, pathogenesis, and research status of BPD, as well as the pathogenesis of IGF-1 in BPD and the latest findings in related biological processes.
Collapse
Affiliation(s)
- Shujian Zhang
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Xue Luan
- Department of Pediatrics, First Hospital, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Huiwen Li
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| | - Zhengyong Jin
- Department of Pediatrics, Affiliated Hospital of Yanbian University, Yanji, Jilin 133000, P.R. China
| |
Collapse
|
21
|
Adiponectin ameliorates hyperoxia-induced lung endothelial dysfunction and promotes angiogenesis in neonatal mice. Pediatr Res 2022; 91:545-555. [PMID: 33767374 DOI: 10.1038/s41390-021-01442-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/31/2021] [Accepted: 02/08/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a common respiratory disease of preterm infants. Lower circulatory/intrapulmonary levels of the adipokine, adiponectin (APN), occur in premature and small-for-gestational-age infants and at saccular/alveolar stages of lung development in the newborn rat. However, the role of low intrapulmonary APN during hyperoxia exposure in developing lungs is unknown. METHODS We test the hypothesis that treatment of hyperoxia-exposed newborn mice with recombinant APN protein attenuates the BPD phenotype characterized by inflammation, impaired alveolarization, and dysregulated vascularization. We used developmentally appropriate in vitro and in vivo BPD modeling systems as well as human lung tissue. RESULTS We observed reduced levels of intrapulmonary APN in experimental BPD mice and human BPD lungs. APN-deficient (APN-/-) newborn mice exposed to moderate (60% O2) hyperoxia showed a worse BPD pulmonary phenotype (inflammation, enhanced endothelial dysfunction, impaired pulmonary vasculature, and alveolar simplification) as compared to wild-type (WT) mice. Treatment of hyperoxia-exposed newborn WT mice with recombinant APN protein attenuated the BPD phenotype (diminished inflammation, decreased pulmonary vascular injury, and improved pulmonary alveolarization) and improved pulmonary function tests. CONCLUSIONS Low intrapulmonary APN is associated with disruption of lung development during hyperoxia exposure, while recombinant APN protein attenuates the BPD pulmonary phenotype. IMPACT Intrapulmonary APN levels were significantly decreased in lungs of experimental BPD mice and human BPD lung tissue at various stages of BPD development. Correlative data from human lung samples with decreased APN levels were associated with increased lung adhesion markers (intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin). Decreased APN levels were associated with endothelial dysfunction and moderate BPD phenotype in APN-deficient, as compared to WT, experimental BPD mice. WT experimental BPD mice treated with recombinant APN protein had an improved pulmonary structural and functional phenotype. Exogenous APN may be considered as a potential therapeutic agent to prevent BPD.
Collapse
|
22
|
Tamura K, Nagaoka M, Inomata S, Kawasaki Y, Makimoto M, Yoshida T. Effects of postnatal hydrocortisone on cytokine profile in extremely preterm infants. Pediatr Int 2022; 64:e15205. [PMID: 35831259 DOI: 10.1111/ped.15205] [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: 12/15/2021] [Revised: 02/25/2022] [Accepted: 03/25/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Systemic hydrocortisone administration has been widely used in preterm infants who are at risk of bronchopulmonary dysplasia (BPD). However, the effects of hydrocortisone on cytokine profiles have not been examined. We aimed to investigate the effects of postnatal hydrocortisone treatment on serum cytokine levels in extremely preterm infants. METHODS This is a retrospective study of 29 extremely preterm infants born at <28 weeks of gestational age. We obtained serum from blood samples collected during an early phase (5-20 days) and a late phase (28-60 days) after birth. We measured the levels of proinflammatory cytokines (tumor necrosis factors α and β, interleukin (IL)-1β, and IL-6), T-helper (Th) 1 cytokines (interferon-γ, IL-2, and IL-12p70), Th2 cytokines (IL-4, IL-5, and IL-10), Th17 cytokine IL-17A, and chemokine IL-8. The cytokine levels between the early and late phases were compared between infants who received postnatal hydrocortisone and those who did not. RESULTS Thirteen infants (45%) received systemic hydrocortisone treatment at a median age of 15 days (IQR: 10.0-21.5) after birth due to respiratory deterioration. The percentage of BPD was higher in the steroid group than in the non-steroid group (P = 0.008). The ratio of IL-6 for the late-to-early phase was significantly lower in the steroid group than in the non-steroid group (P = 0.04). The concentration of the other cytokines remained unchanged between the phases. CONCLUSIONS Although the postnatal hydrocortisone treatment provided for respiratory deterioration did not prevent the BPD development, hydrocortisone treatment might suppress IL-6 overproduction in extremely preterm infants.
Collapse
Affiliation(s)
- Kentaro Tamura
- Division of Neonatology, Maternal and Perinatal Center, Toyama University Hospital, Toyama, Japan
| | - Mitsuhide Nagaoka
- Division of Neonatology, Maternal and Perinatal Center, Toyama University Hospital, Toyama, Japan
| | - Satomi Inomata
- Division of Neonatology, Maternal and Perinatal Center, Toyama University Hospital, Toyama, Japan
| | - Yukako Kawasaki
- Division of Neonatology, Maternal and Perinatal Center, Toyama University Hospital, Toyama, Japan
| | - Masami Makimoto
- Division of Neonatology, Maternal and Perinatal Center, Toyama University Hospital, Toyama, Japan
| | - Taketoshi Yoshida
- Division of Neonatology, Maternal and Perinatal Center, Toyama University Hospital, Toyama, Japan
| |
Collapse
|
23
|
Yang Y, Li J, Mao J. Early diagnostic value of C-reactive protein as an inflammatory marker for moderate-to-severe bronchopulmonary dysplasia in premature infants with birth weight less than 1500 g. Int Immunopharmacol 2021; 103:108462. [PMID: 34952464 DOI: 10.1016/j.intimp.2021.108462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/04/2021] [Accepted: 12/11/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a serious respiratory complication in premature infants and moderate-to-severe BPD may affect the long-term quality of life and lack of specific treatment once it happened. Therefore, it is necessary to identify early diagnostic biomarkers for moderate-to-severe BPD. METHODS This retrospective cohort study included all premature infants with birth weight <1500 g from March 1, 2015 to June 30, 2017. Patients were categorized into mild BPD, moderate-to-severe BPD and non BPD groups. Data collected included patient characteristics, C-reactive protein (CRP) tested at six time points, including 1d (2 h after birth and before the first feeding), 3d, 7d, 2w, 3w, and 4w after birth, and maternal factors. Ordinal regression analysis was used to identify independent predictors of moderate-to-severe BPD and receiver operating characteristic (ROC) curve was used to evaluate the value of CRP as an early diagnostic marker for moderate-to-severe BPD. RESULTS A total of 831 patients were recruited. BPD occurred in 156/831 premature infants with birth weight less than 1500 g. Lower birth weight (OR = 0.998, 95% CI 0.997-0.999, P = 0.004), higher CRP level 3 days after birth (OR = 1.287, 95% CI 1.195-1.384, P = 0.000), and hemodynamically significant patent ductus arteriosus (HsPDA) (OR = 12.256, 95% CI 3.766-39.845, P = 0.000) were independent risk factors for moderate-to-severe BPD. The area under curve of the CRP level 3 days after birth for diagnosing moderate-to-severe BPD was 0.867 (95% CI, 0.823-0.912, P = 0.000). The sensitivity was 83.0% and the specificity was 78.3% when the cut-off value was set at 4.105 mg/L. CONCLUSION The CRP level 3 days after birth may be used as an early diagnostic marker for moderate-to-severe BPD in preterm infants who have the risk factors for BPD with birth weight less than 1500 g.
Collapse
Affiliation(s)
- Yuchen Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jian Mao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
24
|
Inflammatory biomarkers in very preterm infants during early intravenous paracetamol administration. Early Hum Dev 2021; 161:105464. [PMID: 34536795 DOI: 10.1016/j.earlhumdev.2021.105464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Paracetamol promotes early closure of patent ductus arteriosus (PDA), and it may affect inflammation after preterm birth. OBJECTIVE The aim of this study was to evaluate the association between paracetamol treatment and serum inflammatory biomarkers in very preterm infants with respiratory distress. STUDY DESIGN The infants were randomly assigned to intravenous paracetamol or placebo during the first 4 days of life, and others received a lower dose of paracetamol unblinded. Serum samples were used for the analysis of 10 cytokines, C-reactive protein (CRP) and malondialdehyde (MDA). The impact of paracetamol on the biomarkers was evaluated, based on the levels during the early (<60 h) and the later (60-120 h) postnatal age. RESULTS Altogether, 296 serum samples from 31 paracetamol and 25 placebo group infants were analysed. Paracetamol had no effect on cytokine levels during the first 60 h when most induced PDA contractions took place. Later paracetamol treatment was associated with lower serum levels of several cytokines, including interleukin (IL-) 10, interferon gamma-induced protein (IP-) 10, and monocyte chemoattractant protein-1. CRP levels were lower in the paracetamol group during the early treatment. Amongst the infants who had severe morbidities, MDA was higher (p = .045), regardless of paracetamol treatment. CONCLUSION No significant differences in the cytokine levels were evident between the treatment and placebo groups. However, during early treatment, CRP levels were lower in the paracetamol group. To clarify whether this was due to a decrease in cardiopulmonary distress, or a distinct anti-inflammatory effect, requires further studies.
Collapse
|
25
|
Nascimento CP, Maia LP, Alves PT, Paula ATD, Cunha Junior JP, Abdallah VOS, Ferreira DMDLM, Goulart LR, Azevedo VMGDO. Invasive mechanical ventilation and biomarkers as predictors of bronchopulmonary dysplasia in preterm infants. J Pediatr (Rio J) 2021; 97:280-286. [PMID: 32407675 PMCID: PMC9432278 DOI: 10.1016/j.jped.2020.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 11/02/2022] Open
Abstract
OBJECTIVES To evaluate the impact of invasive mechanical ventilation associated with two serum inflammatory cytokines and clinical indicators, on the second day of life, as predictors of bronchopulmonary dysplasia in very low birth weight preterm infants. It was hypothesized that the use of invasive mechanical ventilation in the first hours of life is associated with biomarkers that may predict the chances of preterm infants to develop bronchopulmonary dysplasia. METHODS Prospective cohort of 40 preterm infants with gestational age <34 weeks and birth weight <1500 g. The following were analyzed: clinical variables; types of ventilator support used (there is a higher occurrence of bronchopulmonary dysplasia when oxygen supplementation is performed by long periods of invasive mechanical ventilation); hospitalization time; quantification of two cytokines (granulocyte and macrophage colony stimulating factor [GM-CSF] and eotaxin) in blood between 36 and 48 h of life. The preterm infants were divided in two groups: with and without bronchopulmonary dysplasia. RESULTS The GM-CSF levels presented a significantly higher value in the bronchopulmonary dysplasia group (p = 0.002), while eotaxin presented higher levels in the group without bronchopulmonary dysplasia (p = 0.02). The use of continuous invasive mechanical ventilation was associated with increased ratios between GM-CSF and eotaxin (100% sensitivity and 80% specificity; receiver operating characteristic area = 0.9013, CI = 0.7791-1.024, p < 0.0001). CONCLUSIONS The duration of invasive mechanical ventilation performed in the first 48 h of life in the very low birth weight infants is a significant clinical predictor of bronchopulmonary dysplasia. The use of continuous invasive mechanical ventilation was associated with increased ratios between GM-CSF and eotaxin, suggesting increased lung injury and consequent progression of the disease.
Collapse
Affiliation(s)
- Camila Piqui Nascimento
- Universidade Federal de Uberlândia, Programa de Pós-Graduação em Ciências da Saúde, Uberlândia, MG, Brazil
| | - Larissa Prado Maia
- Universidade Federal de Uberlândia, Instituto Nacional de Ciência e Tecnologia - Teranóstica e Nanobiotecnologia (INCT-TeraNano), Laboratório de Nanobiotecnologia, Uberlândia, MG, Brazil
| | - Patrícia Terra Alves
- Universidade Federal de Uberlândia, Instituto Nacional de Ciência e Tecnologia - Teranóstica e Nanobiotecnologia (INCT-TeraNano), Laboratório de Nanobiotecnologia, Uberlândia, MG, Brazil
| | - Aline Teodoro de Paula
- Universidade Federal de Uberlândia, Instituto Nacional de Ciência e Tecnologia - Teranóstica e Nanobiotecnologia (INCT-TeraNano), Laboratório de Nanobiotecnologia, Uberlândia, MG, Brazil
| | - Jair Pereira Cunha Junior
- Universidade Federal de Uberlândia, Departamento de Imunologia, Laboratório de Imunoquímica e Imunotecnologia, Uberlândia, MG, Brazil
| | | | | | - Luiz Ricardo Goulart
- Universidade Federal de Uberlândia, Programa de Pós-Graduação em Ciências da Saúde, Uberlândia, MG, Brazil; Universidade Federal de Uberlândia, Instituto Nacional de Ciência e Tecnologia - Teranóstica e Nanobiotecnologia (INCT-TeraNano), Laboratório de Nanobiotecnologia, Uberlândia, MG, Brazil
| | - Vivian Mara Gonçalves de Oliveira Azevedo
- Universidade Federal de Uberlândia, Programa de Pós-Graduação em Ciências da Saúde, Uberlândia, MG, Brazil; Universidade Federal de Uberlândia, Faculdade de Educação Física e Fisioterapia, Uberlândia, MG, Brazil.
| |
Collapse
|
26
|
Ali A, Zambrano R, Duncan MR, Chen S, Luo S, Yuan H, Chen P, Benny M, Schmidt A, Young K, Kerr N, de Rivero Vaccari JP, Keane RW, Dietrich WD, Wu S. Hyperoxia-activated circulating extracellular vesicles induce lung and brain injury in neonatal rats. Sci Rep 2021; 11:8791. [PMID: 33888735 PMCID: PMC8062626 DOI: 10.1038/s41598-021-87706-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/16/2021] [Indexed: 01/02/2023] Open
Abstract
Hyperoxia-induced lung injury plays a key role in the development of bronchopulmonary dysplasia (BPD), characterized by inflammatory injury and impaired lung development in preterm infants. Although BPD is a predictor of poor neurodevelopmental outcomes, currently it is uncertain how lung injury contributes to brain injury in preterm infants. Extracellular vesicles (EVs) are a heterogeneous group of cell-derived membranous structures that regulate intercellular and inter-organ communications. Gasdermin D (GSDMD) has emerged as a key executor of inflammasome-mediated cell death and inflammation. In this study, we utilized a neonatal rat model of BPD to assess if hyperoxia stimulates lung release of circulating EVs and if these EVs induce lung and brain injury. We found that hyperoxia-exposed rats had elevated numbers of plasma-derived EVs compared to rats maintained in room air. These EVs also had increased cargos of surfactant protein C, a marker of type II alveolar epithelial cells (AEC), and the active (p30) form of GSDMD. When these EVs were adoptively transferred into normal newborn rats via intravenous injection, they were taken up both by lung and brain tissues. Moreover, EVs from hyperoxic animals induced not only the pathological hallmarks of BPD, but also brain inflammatory injury in recipient rats, as well as inducing cell death in cultured pulmonary vascular endothelial cells and neural stem cells (NSC). Similarly, hyperoxia-exposed cultured AEC-like cells released EVs that also contained increased GSDMD-p30 and these EVs induced pyroptotic cell death in NSC. Overall, these data indicate that hyperoxia-activated circulating EVs mediate a lung to brain crosstalk resulting in brain injury and suggest a mechanism that links lung injury and neurodevelopmental impairment in BPD infants.
Collapse
Affiliation(s)
- Anum Ali
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Ronald Zambrano
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Matthew R Duncan
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Shaoyi Chen
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Shihua Luo
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Huijun Yuan
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Pingping Chen
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Merline Benny
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Augusto Schmidt
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Karen Young
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA
| | - Nadine Kerr
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Shu Wu
- Division of Neonatology and Batchelor Children's Research Institute, Department of Pediatrics, University of Miami Miller School of Medicine, P. O. Box 016960, Miami, FL, 33101, USA.
| |
Collapse
|
27
|
Go H, Ohto H, Nollet KE, Sato K, Ichikawa H, Kume Y, Kanai Y, Maeda H, Kashiwabara N, Ogasawara K, Sato M, Hashimoto K, Hosoya M. Red cell distribution width as a predictor for bronchopulmonary dysplasia in premature infants. Sci Rep 2021; 11:7221. [PMID: 33790386 PMCID: PMC8012706 DOI: 10.1038/s41598-021-86752-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/19/2021] [Indexed: 01/21/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common morbidity complicating preterm birth. Red blood cell distribution width (RDW), a measure of the variation of red blood cell size, could reflect oxidative stress and chronic inflammation in many diseases such as cardiovascular, pulmonary, and other diseases. The objectives of the present study were to evaluate perinatal factors affecting RDW and to validate whether RDW could be a potential biomarker for BPD. A total of 176 preterm infants born at < 30 weeks were included in this study. They were categorized into BPD (n = 85) and non-BPD (n = 91) infants. RDW at birth and 14 days and 28 days of life (DOL 14, DOL 28) were measured. Clinical data were obtained from all subjects at Fukushima Medical University (Fukushima, Japan). The mean RDW at birth, DOL 14 and DOL 28 were 16.1%, 18.6%, 20.1%, respectively. Small for gestational age (SGA), chorioamnionitis (CAM), hypertensive disorders of pregnancy (HDP), gestational age and birth weight were significantly associated with RDW at birth. SGA, BPD and red blood cell (RBC) transfusion before DOL 14 were associated with RDW at DOL 14. BPD and RBC transfusion before DOL 14 were associated with RDW at DOL 28. Compared with non-BPD infants, mean RDW at DOL 14 (21.1% vs. 17.6%, P < 0.001) and DOL 28 (22.2% vs. 18.2%, P < 0.001) were significantly higher in BPD infants. Multivariate analysis revealed that RDW at DOL 28 was significantly higher in BPD infants (P = 0.001, odds ratio 1.63; 95% CI 1.22–2.19). Receiver operating characteristic analysis for RDW at DOL 28 in infants with and without BPD yielded an area under the curve of 0.87 (95% CI 0.78–0.91, P < 0.001). RDW at DOL 28 with mild BPD (18.1% vs. 21.3%, P < 0.001), moderate BPD (18.1% vs. 21.2%, P < 0.001), and severe BPD (18.1% vs. 24.0%, P < 0.001) were significantly higher than those with non-BPD, respectively. Furthermore, there are significant differences of RDW at DOL 28 among mild, moderate, and severe BPD. In summary, we conclude that RDW at DOL 28 could serve as a biomarker for predicting BPD and its severity. The mechanism by which RDW at DOL 28 is associated with the pathogenesis of BPD needs further elucidation.
Collapse
Affiliation(s)
- Hayato Go
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan.
| | | | - Kenneth E Nollet
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenichi Sato
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Hirotaka Ichikawa
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Yohei Kume
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Yuji Kanai
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Hajime Maeda
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Nozomi Kashiwabara
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Kei Ogasawara
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Maki Sato
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Koichi Hashimoto
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| | - Mitsuaki Hosoya
- Department of Pediatrics, Fukushima Medical University School of Medicine, Hikarigaoka 1, Fukushima, Japan
| |
Collapse
|
28
|
Bhattacharya S, Mereness JA, Baran AM, Misra RS, Peterson DR, Ryan RM, Reynolds AM, Pryhuber GS, Mariani TJ. Lymphocyte-Specific Biomarkers Associated With Preterm Birth and Bronchopulmonary Dysplasia. Front Immunol 2021; 11:563473. [PMID: 33552042 PMCID: PMC7859626 DOI: 10.3389/fimmu.2020.563473] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 12/07/2020] [Indexed: 01/11/2023] Open
Abstract
Many premature babies who are born with neonatal respiratory distress syndrome (RDS) go on to develop Bronchopulmonary Dysplasia (BPD) and later Post-Prematurity Respiratory Disease (PRD) at one year corrected age, characterized by persistent or recurrent lower respiratory tract symptoms frequently related to inflammation and viral infection. Transcriptomic profiles were generated from sorted peripheral blood CD8+ T cells of preterm and full-term infants enrolled with consent in the NHLBI Prematurity and Respiratory Outcomes Program (PROP) at the University of Rochester and the University at Buffalo. We identified outcome-related gene expression patterns following standard methods to identify markers for oxygen utilization and BPD as outcomes in extremely premature infants. We further identified predictor gene sets for BPD based on transcriptomic data adjusted for gestational age at birth (GAB). RNA-Seq analysis was completed for CD8+ T cells from 145 subjects. Among the subjects with highest risk for BPD (born at <29 weeks gestational age (GA); n=72), 501 genes were associated with oxygen utilization. In the same set of subjects, 571 genes were differentially expressed in subjects with a diagnosis of BPD and 105 genes were different in BPD subjects as defined by physiologic challenge. A set of 92 genes could predict BPD with a moderately high degree of accuracy. We consistently observed dysregulation of TGFB, NRF2, HIPPO, and CD40-associated pathways in BPD. Using gene expression data from both premature and full-term subjects (n=116), we identified a 28 gene set that predicted the PRD status with a moderately high level of accuracy, which also were involved in TGFB signaling. Transcriptomic data from sort-purified peripheral blood CD8+ T cells from 145 preterm and full-term infants identified sets of molecular markers of inflammation associated with independent development of BPD in extremely premature infants at high risk for the disease and of PRD among the preterm and full-term subjects.
Collapse
Affiliation(s)
- Soumyaroop Bhattacharya
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Jared A. Mereness
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Andrea M. Baran
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Ravi S. Misra
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Derick R. Peterson
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY, United States
| | - Rita M. Ryan
- Department of Pediatrics, University at Buffalo, Buffalo, NY, United States
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| | | | - Gloria S. Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Thomas J. Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester, Rochester, NY, United States
| |
Collapse
|
29
|
Sahoo D, Zaramela LS, Hernandez GE, Mai U, Taheri S, Dang D, Stouch AN, Medal RM, McCoy AM, Aschner JL, Blackwell TS, Zengler K, Prince LS. Transcriptional profiling of lung macrophages identifies a predictive signature for inflammatory lung disease in preterm infants. Commun Biol 2020; 3:259. [PMID: 32444859 PMCID: PMC7244484 DOI: 10.1038/s42003-020-0985-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 05/01/2020] [Indexed: 02/06/2023] Open
Abstract
Lung macrophages mature after birth, placing newborn infants, particularly those born preterm, within a unique window of susceptibility to disease. We hypothesized that in preterm infants, lung macrophage immaturity contributes to the development of bronchopulmonary dysplasia (BPD), the most common serious complication of prematurity. By measuring changes in lung macrophage gene expression in preterm patients at risk of BPD, we show here that patients eventually developing BPD had higher inflammatory mediator expression even on the first day of life. Surprisingly, the ex vivo response to LPS was similar across all samples. Our analysis did however uncover macrophage signature genes whose expression increased in the first week of life specifically in patients resilient to disease. We propose that these changes describe the dynamics of human lung macrophage differentiation. Our study therefore provides new mechanistic insight into both neonatal lung disease and human developmental immunology.
Collapse
Affiliation(s)
- Debashis Sahoo
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92037, USA
| | - Livia S Zaramela
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Gilberto E Hernandez
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Uyen Mai
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Sahar Taheri
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Dharanidhar Dang
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ashley N Stouch
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rachel M Medal
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Alyssa M McCoy
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Judy L Aschner
- Department of Pediatrics, Joseph M Sanzari Children's Hospital, Hackensack University Medical Center, Hackensack Meridian School of Medicine at Seton Hall, Hackensack, NJ, 07110, USA
| | - Timothy S Blackwell
- Departments of Medicine, Cancer Biology, and Developmental Cell Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Karsten Zengler
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Lawrence S Prince
- Department of Pediatrics, Rady Children's Hospital, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
30
|
Gilfillan M, Das P, Shah D, Alam MA, Bhandari V. Inhibition of microRNA-451 is associated with increased expression of Macrophage Migration Inhibitory Factor and mitgation of the cardio-pulmonary phenotype in a murine model of Bronchopulmonary Dysplasia. Respir Res 2020; 21:92. [PMID: 32321512 PMCID: PMC7178994 DOI: 10.1186/s12931-020-01353-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Macrophage migration inhibitory factor (MIF) has been implicated as a protective factor in the development of bronchopulmonary dysplasia (BPD) and is known to be regulated by MicroRNA-451 (miR-451). The aim of this study was to evaluate the role of miR-451 and the MIF signaling pathway in in vitro and in vivo models of BPD. Methods Studies were conducted in mouse lung endothelial cells (MLECs) exposed to hyperoxia and in a newborn mouse model of hyperoxia-induced BPD. Lung and cardiac morphometry as well as vascular markers were evaluated. Results Increased expression of miR-451 was noted in MLECs exposed to hyperoxia and in lungs of BPD mice. Administration of a miR-451 inhibitor to MLECs exposed to hyperoxia was associated with increased expression of MIF and decreased expression of angiopoietin (Ang) 2. Treatment with the miR-451 inhibitor was associated with improved lung morphometry indices, significant reduction in right ventricular hypertrophy, decreased mean arterial wall thickness and improvement in vascular density in BPD mice. Western blot analysis demonstrated preservation of MIF expression in BPD animals treated with a miR-451 inhibitor and increased expression of vascular endothelial growth factor-A (VEGF-A), Ang1, Ang2 and the Ang receptor, Tie2. Conclusion We demonstrated that inhibition of miR-451 is associated with mitigation of the cardio-pulmonary phenotype, preservation of MIF expression and increased expression of several vascular growth factors.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,St Christopher's Hospital for Children, Philadelphia, PA, 19134, USA
| | - Pragnya Das
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA
| | - Dilip Shah
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA
| | - Mohammad Afaque Alam
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Temple University, Philadelphia, PA, 19140, USA
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA. .,St Christopher's Hospital for Children, Philadelphia, PA, 19134, USA. .,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA. .,Temple University, Philadelphia, PA, 19140, USA. .,Pediatrics, Obstetrics and Gynecology and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, USA. .,Neonatology, The Children's Regional Hospital at Cooper, One Cooper Plaza, Camden, NJ, 08103, USA.
| |
Collapse
|
31
|
McEvoy CT, Ballard PL, Ward RM, Rower JE, Wadhawan R, Hudak ML, Weitkamp JH, Harris J, Asselin J, Chapin C, Ballard RA. Dose-escalation trial of budesonide in surfactant for prevention of bronchopulmonary dysplasia in extremely low gestational age high-risk newborns (SASSIE). Pediatr Res 2020; 88:629-636. [PMID: 32006953 PMCID: PMC7223897 DOI: 10.1038/s41390-020-0792-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Initial trials of lung-targeted budesonide (0.25 mg/kg) in surfactant to prevent bronchopulmonary dysplasia (BPD) in premature infants have shown benefit; however, the optimal safe dose is unknown. METHODS Dose-escalation study of budesonide (0.025, 0.05, 0.10 mg/kg) in calfactatant in extremely low gestational age neonates (ELGANs) requiring intubation at 3-14 days. Tracheal aspirate (TA) cytokines, blood budesonide concentrations, and untargeted blood metabolomics were measured. Outcomes were compared with matched infants receiving surfactant in the Trial Of Late SURFactant (TOLSURF). RESULTS Twenty-four infants with mean gestational age 25.0 weeks and 743 g birth weight requiring mechanical ventilation were enrolled at mean age 6 days. Budesonide was detected in the blood of all infants with a half-life of 3.4 h. Of 11 infants with elevated TA cytokine levels at baseline, treatment was associated with sustained decrease (mean 65%) at all three dosing levels. There were time- and dose-dependent decreases in blood cortisol concentrations and changes in total blood metabolites. Respiratory outcomes did not differ from the historic controls. CONCLUSIONS Budesonide/surfactant had no clinical respiratory benefit at any dosing levels for intubated ELGANs. One-tenth the dose used in previous trials had minimal systemic metabolic effects and appeared effective for lung-targeted anti-inflammatory action.
Collapse
Affiliation(s)
- Cindy T. McEvoy
- grid.5288.70000 0000 9758 5690Department of Pediatrics, Oregon Health & Science University, Portland, OR USA
| | - Philip L. Ballard
- grid.266102.10000 0001 2297 6811Department of Pediatrics, University of California San Francisco, San Francisco, CA USA
| | - Robert M. Ward
- grid.223827.e0000 0001 2193 0096Department of Pediatrics, University of Utah, Salt Lake City, UT USA
| | - Joseph E. Rower
- grid.223827.e0000 0001 2193 0096Department of Pediatrics, University of Utah, Salt Lake City, UT USA ,grid.223827.e0000 0001 2193 0096Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT USA
| | - Rajan Wadhawan
- grid.468438.50000 0004 0441 8332Department of Pediatrics, AdventHealth for Children, Orlando, FL USA
| | - Mark L. Hudak
- grid.413116.00000 0004 0625 1409Department of Pediatrics, University of Florida College of Medicine-Jacksonville, Jacksonville, FL USA
| | - Joern-Hendrik Weitkamp
- grid.412807.80000 0004 1936 9916Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN USA
| | - Julia Harris
- grid.5288.70000 0000 9758 5690Department of Pediatrics, Oregon Health & Science University, Portland, OR USA
| | - Jeanette Asselin
- grid.414016.60000 0004 0433 7727Department of Pediatrics, Oakland Children’s Hospital, Oakland, CA USA
| | - Cheryl Chapin
- grid.266102.10000 0001 2297 6811Department of Pediatrics, University of California San Francisco, San Francisco, CA USA
| | - Roberta A. Ballard
- grid.266102.10000 0001 2297 6811Department of Pediatrics, University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
32
|
Exploration of potential biochemical markers for persistence of patent ductus arteriosus in preterm infants at 22-27 weeks' gestation. Pediatr Res 2019; 86:333-338. [PMID: 30287890 DOI: 10.1038/s41390-018-0182-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/03/2018] [Accepted: 07/09/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND Early identification of infants at risk for complications from patent ductus arteriosus (PDA) may improve treatment outcomes. The aim of this study was to identify biochemical markers associated with persistence of PDA, and with failure of pharmacological treatment for PDA, in extremely preterm infants. METHODS Infants born at 22-27 weeks' gestation were included in this prospective study. Blood samples were collected on the second day of life. Fourteen biochemical markers associated with factors that may affect PDA closure were analyzed and related to persistent PDA and to the response of pharmacological treatment with ibuprofen. RESULTS High levels of B-type natriuretic peptide, interleukin-6, -8, -10, and -12, growth differentiation factor 15 and monocyte chemotactic protein 1 were associated with persistent PDA, as were low levels of platelet-derived growth factor. High levels of erythropoietin were associated with both persistent PDA and failure to close PDA within 24 h of the last dose of ibuprofen. CONCLUSIONS High levels of inflammatory markers were associated with the persistence of PDA. High levels of erythropoietin were associated with both the persistence of PDA and failure to respond to pharmacological treatment.
Collapse
|
33
|
Bhandari V. Designing a better definition of bronchopulmonary dysplasia. Pediatr Pulmonol 2019; 54:678-679. [PMID: 30864236 DOI: 10.1002/ppul.24305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 01/28/2019] [Accepted: 02/12/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Vineet Bhandari
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Baker EK, Malhotra A, Lim R, Jacobs SE, Hooper SB, Davis PG, Wallace EM. Human amnion cells for the prevention of bronchopulmonary dysplasia: a protocol for a phase I dose escalation study. BMJ Open 2019; 9:e026265. [PMID: 30826799 PMCID: PMC6398764 DOI: 10.1136/bmjopen-2018-026265] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Bronchopulmonary dysplasia (BPD), an important sequela of preterm birth, is associated with long-term abnormalities of lung function and adverse neurodevelopmental outcomes. Inflammation, inhibition of secondary septation and vascular maldevelopment play key roles in the pathogenesis of BPD. Human amnion epithelial cells (hAECs), stem-like cells, derived from placental tissues are able to modulate the inflammatory milieu and, in preclinical studies of BPD-like injury, restore lung architecture and function. Allogeneic hAECs may present a new preventative and reparative therapy for BPD. METHODS AND ANALYSIS In this two centre, phase I cell dose escalation study we will evaluate the safety of intravenous hAEC infusions in preterm infants at high risk of severe BPD. Twenty-four infants born at less than 29 weeks' gestation will each receive intravenous hAECs beginning day 14 of life. We will escalate the dose of cells contained in a single intravenous hAEC infusion in increments from 2 million cells/kg to 10 million cells/kg. Further dose escalation will be achieved with repeat infusions given at 5 day intervals to a maximum total dose of 30 million cells/kg (three infusions). Safety is the primary outcome. Infants will be followed-up until 2 years corrected age. Additional outcome measures include a description of infants' cytokine profile following hAEC infusion, respiratory outcomes including BPD and pulmonary hypertension and other neonatal morbidities including neurodevelopmental assessment at 2 years. ETHICS AND DISSEMINATION This study was approved on the June12th, 2018 by the Human Research Ethics Committee of Monash Health and Monash University. Recruitment commenced in August 2018 and is expected to take 18 months. Accordingly, follow-up will be completed mid-2022. The findings of this study will be disseminated via peer-reviewed journals and at conferences. PROTOCOL VERSION 5, 21 May 2018. TRIAL REGISTRATION NUMBER ACTRN12618000920291; Pre-results.
Collapse
Affiliation(s)
- Elizabeth Kate Baker
- Newborn Research Centre, The Royal Women's Hospital, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Atul Malhotra
- Monash Newborn, Monash Children's Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Susan E Jacobs
- Newborn Research Centre, The Royal Women's Hospital, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Stuart B Hooper
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Peter G Davis
- Newborn Research Centre, The Royal Women's Hospital, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Euan M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Abstract
Bronchopulmonary dysplasia (BPD) continues to be one of the most common complications of preterm birth and is characterized histopathologically by impaired lung alveolarization. Extremely preterm born infants remain at high risk for the development of BPD, highlighting a pressing need for continued efforts to understand the pathomechanisms at play in affected infants. This brief review summarizes recent progress in our understanding of the how the development of the newborn lung is stunted, highlighting recent reports on roles for growth factor signaling, oxidative stress, inflammation, the extracellular matrix and proteolysis, non-coding RNA, and fibroblast and epithelial cell plasticity. Additionally, some concerns about modeling BPD in experimental animals are reviewed, as are new developments in the in vitro modeling of pathophysiological processes relevant to impaired lung alveolarization in BPD.
Collapse
Affiliation(s)
- Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
36
|
The definition of bronchopulmonary dysplasia: an evolving dilemma. Pediatr Res 2018; 84:586-588. [PMID: 30188504 DOI: 10.1038/s41390-018-0167-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/19/2018] [Indexed: 12/11/2022]
Abstract
The definition of bronchopulmonary dysplasia (BPD) continues to be a challenging one. The evolution of the disease as well as its definition are discussed. Limitations of the current as well as newer suggested definitions of BPD are summarized. Recognition of the variability of the pulmonary phenotype is a critical aspect of the disease that needs to identified, utilizing biomarkers that will potentially better inform the next iteration of the definition of BPD.
Collapse
|
37
|
Hagman C, Björklund LJ, Hellgren G, Tufvesson E, Hansen-Pupp I. Club cell secretory protein (CC16) in gastric fluid at birth and subsequent lung disease in preterm infants. Pediatr Pulmonol 2018; 53:1399-1406. [PMID: 29992757 DOI: 10.1002/ppul.24128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 05/31/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Club cell secretory protein (CC16) probably has a role in protecting the lung from inflammation. AIM To evaluate if low levels of CC16 in gastric fluid at birth, reflecting low levels of CC16 in the lung, would be associated with lung inflammation and respiratory morbidity. METHODS A study of 64 infants with mean gestational age 26.1 weeks. CC16 was analyzed in gastric fluid at birth. CC16, pro-inflammatory cytokines, and MMP-9 were analyzed in tracheal aspirate within 24 h from birth. RESULTS CC16 in gastric fluid increased with gestational age (P = 0.033). Lower concentrations of CC16 in gastric fluid at birth were associated with higher concentrations of IL-1β (P = 0.028), TNF-α (P = 0.034), and MMP-9 (P = 0.015) in tracheal aspirate. Infants who needed mechanical ventilation at 24 and 72 h of age had lower CC16 in gastric fluid than those not ventilated at these ages (P = 0.011 and P = 0.024, respectively). Lower CC16 in gastric fluid was associated with higher FiO2 at 6 h (P = 0.009), higher PaCO2 at 24 h (P = 0.03), more ventilator days (P = 0.012) and more days with supplemental oxygen (P = 0.03). Infants who had either died or were still treated with supplemental oxygen at 36 weeks postmenstrual age had lower CC16 in gastric fluid than infants with none of these outcomes (P = 0.049). CONCLUSION A low CC16 concentration in gastric fluid at birth was associated with increased inflammation in the trachea within the first 24 h of life and with more need for respiratory support in the neonatal period.
Collapse
Affiliation(s)
- Cecilia Hagman
- Lund University, Skåne University Hospital, Department of Clinical Sciences Lund, Pediatrics, Lund, Sweden
| | - Lars J Björklund
- Lund University, Skåne University Hospital, Department of Clinical Sciences Lund, Pediatrics, Lund, Sweden
| | - Gunnel Hellgren
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ellen Tufvesson
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Respiratory Medicine and Allergology, Lund, Sweden
| | - Ingrid Hansen-Pupp
- Lund University, Skåne University Hospital, Department of Clinical Sciences Lund, Pediatrics, Lund, Sweden
| |
Collapse
|
38
|
Liu Y, Dong WB. [Preventive effect of caffeine on bronchopulmonary dysplasia in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:598-602. [PMID: 30022766 PMCID: PMC7389204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/05/2018] [Indexed: 11/12/2023]
Abstract
With the increase in the rescue success rate of critically ill preterm infants and extremely preterm infants, the incidence rate of bronchopulmonary dysplasia (BPD) is increasing year by year. BPD has a high mortality rate and high possibility of sequelae, which greatly affects the quality of life of preterm infants and brings a heavy burden to their families, and so the treatment of BPD is of vital importance. At present, no consensus has been reached on the treatment measures for BPD. However, recent studies have shown that early application of caffeine can prevent BPD. With reference to the latest studies on the effect of caffeine in the prevention of BPD, this article reviews the mechanism of action of caffeine in reducing pulmonary inflammation, improving morphological abnormalities of lung injury, reducing oxidative stress injury, and improving pulmonary function.
Collapse
MESH Headings
- Animals
- Bronchopulmonary Dysplasia/genetics
- Bronchopulmonary Dysplasia/metabolism
- Bronchopulmonary Dysplasia/physiopathology
- Bronchopulmonary Dysplasia/prevention & control
- Caffeine/administration & dosage
- Humans
- Infant, Premature/growth & development
- Infant, Premature/metabolism
- Infant, Premature, Diseases/genetics
- Infant, Premature, Diseases/metabolism
- Infant, Premature, Diseases/physiopathology
- Infant, Premature, Diseases/prevention & control
- Oxidative Stress/drug effects
Collapse
Affiliation(s)
- Yang Liu
- Department of Neonatology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | | |
Collapse
|
39
|
Liu Y, Dong WB. [Preventive effect of caffeine on bronchopulmonary dysplasia in preterm infants]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:598-602. [PMID: 30022766 PMCID: PMC7389204 DOI: 10.7499/j.issn.1008-8830.2018.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
With the increase in the rescue success rate of critically ill preterm infants and extremely preterm infants, the incidence rate of bronchopulmonary dysplasia (BPD) is increasing year by year. BPD has a high mortality rate and high possibility of sequelae, which greatly affects the quality of life of preterm infants and brings a heavy burden to their families, and so the treatment of BPD is of vital importance. At present, no consensus has been reached on the treatment measures for BPD. However, recent studies have shown that early application of caffeine can prevent BPD. With reference to the latest studies on the effect of caffeine in the prevention of BPD, this article reviews the mechanism of action of caffeine in reducing pulmonary inflammation, improving morphological abnormalities of lung injury, reducing oxidative stress injury, and improving pulmonary function.
Collapse
MESH Headings
- Animals
- Bronchopulmonary Dysplasia/genetics
- Bronchopulmonary Dysplasia/metabolism
- Bronchopulmonary Dysplasia/physiopathology
- Bronchopulmonary Dysplasia/prevention & control
- Caffeine/administration & dosage
- Humans
- Infant, Premature/growth & development
- Infant, Premature/metabolism
- Infant, Premature, Diseases/genetics
- Infant, Premature, Diseases/metabolism
- Infant, Premature, Diseases/physiopathology
- Infant, Premature, Diseases/prevention & control
- Oxidative Stress/drug effects
Collapse
Affiliation(s)
- Yang Liu
- Department of Neonatology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | | |
Collapse
|
40
|
Mao X, Qiu J, Zhao L, Xu J, Yin J, Yang Y, Zhang M, Cheng R. Vitamin D and IL-10 Deficiency in Preterm Neonates With Bronchopulmonary Dysplasia. Front Pediatr 2018; 6:246. [PMID: 30246004 PMCID: PMC6137192 DOI: 10.3389/fped.2018.00246] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/17/2018] [Indexed: 11/25/2022] Open
Abstract
Introduction: Vitamin D deficiency and inflammation are involved with bronchopulmonary dysplasia (BPD) in preterm neonates; however, the clinical evidence still remains scarce. We hypothesized that vitamin D and inflammatory cytokines may be risk factors for BPD in infants. Methods: Preterm infants born between 28 and 31 weeks' gestation were recruited between January 2016 and 2017. Blood samples were all collected at corresponding time points. Vitamin D was measured using an automatic biochemical analyzer, and inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-10) were measured using ELISA. Results: The baseline characteristics for preterm infants without BPD (non-BPD control, n = 20) or with BPD (n = 19) were similar. In the blood samples collected 24-h post birth, vitamin D was significantly reduced in the BPD neonates (non-BPD vs. BPD, 28.96 ± 3.404 vs. 17.99 ± 2.233 nmol/l, p = 0.0134). Inflammatory cytokines TNF-α, IL-1β, and IL-6 were comparable in both groups. The anti-inflammatory cytokine IL-10, however, was significantly decreased in 24-h blood samples from BPD preterm infants (non-BPD vs. BPD, 44.61 ± 10.48 vs. 11.64 ± 2.351 pg/ml, p = 0.0054). In the BPD infants with mild or moderate disease, vitamin D deficiency was quite similar. IL-10 deficiency, however, was more aggravated in the BPD infants with moderate disease. No changes in Vitamin D or cytokines (TNF-α, IL-1β, IL-6, and IL-10) were observed for blood samples collected 2 or 4 weeks after birth. Conclusion: In our pilot study, Vitamin D and IL-10 levels at 24-h of life were risk factors for the development of BPD in very preterm infants.
Collapse
Affiliation(s)
- Xiaonan Mao
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Qiu
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Li Zhao
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Junjie Xu
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jiao Yin
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Yang
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mingshun Zhang
- Department of Immunology, Nanjing Medical University, Nanjing, China
| | - Rui Cheng
- Department of neonates, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
Leroy S, Caumette E, Waddington C, Hébert A, Brant R, Lavoie PM. A Time-Based Analysis of Inflammation in Infants at Risk of Bronchopulmonary Dysplasia. J Pediatr 2018; 192:60-65.e1. [PMID: 29092751 DOI: 10.1016/j.jpeds.2017.09.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/28/2017] [Accepted: 09/07/2017] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To precisely delineate the timing and contribution of inflammation to bronchopulmonary dysplasia (BPD) in preterm infants during the neonatal period. STUDY DESIGN Longitudinal study of blood inflammatory biomarkers (interleukin [IL]-6, IL-8, and granulocyte colony-stimulating factor) measured between birth and 42 days of age, at high temporal (daily) resolution, in infants born at or below 30 weeks of gestation. Cytokine predictors of BPD at 36 weeks postmenstrual age were adjusted for infant-specific and time-dependent factors, using hierarchical mixed effects regressions models. RESULTS A total of 1518 data points were obtained in 62 infants (mean gestational age of 27 weeks). Infants who developed BPD later on presented increased inflammation after birth compared with infants without BPD. Inflammation was sustained, with gradual attenuation over 2 weeks (IL-8: OR: 6.5 [95% CI: 1.8-24]; granulocyte colony-stimulating factor: 3.3 [1.5-7.6]) and was higher in boys and in infants of lower birth weight. This inflammation preceded the clinical increased requirement in supplemental oxygen characteristic of BPD, and preceded the peak occurrence of neonatal sepsis or necrotizing enterocolitis. CONCLUSIONS Systemic inflammation occurs early in the neonatal period and precedes clinical symptoms in infants with BPD. These data provide a discrete vulnerability window period, supporting a role for targeted intensive care interventions during the early phase of BPD.
Collapse
Affiliation(s)
- Sandrine Leroy
- EA2415, Montpellier University, Montpellier, France; Mobile Pediatrics Intensive Care Unit, Avicenne hospital, AP-HP, Paris, France
| | | | - Chandra Waddington
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Audrey Hébert
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rollin Brant
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; Department of Statistics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pascal M Lavoie
- Neonatal Program, Children's and Women's Hospitals of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
42
|
Nelin LD, Logan JW. The use of inhaled corticosteroids in chronically ventilated preterm infants. Semin Fetal Neonatal Med 2017; 22:296-301. [PMID: 28768578 DOI: 10.1016/j.siny.2017.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is the most usual reason for preterm infants to require chronic mechanical ventilation. Inflammation is a key factor underlying the lung injury leading to the development of BPD, and the rationale for use of corticosteroids in the management of ventilator-dependent preterm infants is based on their anti-inflammatory effects. Because systemic corticosteroids are associated with significant adverse effects in preterm infants, attention has turned to the use of inhaled corticosteroids (ICS) as a potentially safer therapy for BPD. The aim of this review is to discuss what is known about the efficacy and safety of ICS in chronically ventilated preterm infants. However, this has been a challenge since there is a paucity of high-grade evidence for the use of ICS in these patients. Thus, there is a real need for well-powered randomized controlled trials examining short- and long-term outcomes of ICS use in this population.
Collapse
Affiliation(s)
- Leif D Nelin
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital, Columbus, OH, USA.
| | - J Wells Logan
- Comprehensive Center for Bronchopulmonary Dysplasia, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
43
|
Surate Solaligue DE, Rodríguez-Castillo JA, Ahlbrecht K, Morty RE. Recent advances in our understanding of the mechanisms of late lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1101-L1153. [PMID: 28971976 DOI: 10.1152/ajplung.00343.2017] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of lung development is to generate an organ of gas exchange that provides both a thin gas diffusion barrier and a large gas diffusion surface area, which concomitantly generates a steep gas diffusion concentration gradient. As such, the lung is perfectly structured to undertake the function of gas exchange: a large number of small alveoli provide extensive surface area within the limited volume of the lung, and a delicate alveolo-capillary barrier brings circulating blood into close proximity to the inspired air. Efficient movement of inspired air and circulating blood through the conducting airways and conducting vessels, respectively, generates steep oxygen and carbon dioxide concentration gradients across the alveolo-capillary barrier, providing ideal conditions for effective diffusion of both gases during breathing. The development of the gas exchange apparatus of the lung occurs during the second phase of lung development-namely, late lung development-which includes the canalicular, saccular, and alveolar stages of lung development. It is during these stages of lung development that preterm-born infants are delivered, when the lung is not yet competent for effective gas exchange. These infants may develop bronchopulmonary dysplasia (BPD), a syndrome complicated by disturbances to the development of the alveoli and the pulmonary vasculature. It is the objective of this review to update the reader about recent developments that further our understanding of the mechanisms of lung alveolarization and vascularization and the pathogenesis of BPD and other neonatal lung diseases that feature lung hypoplasia.
Collapse
Affiliation(s)
- David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
44
|
Mahlman M, Karjalainen MK, Huusko JM, Andersson S, Kari MA, Tammela OKT, Sankilampi U, Lehtonen L, Marttila RH, Bassler D, Poets CF, Lacaze-Masmonteil T, Danan C, Delacourt C, Palotie A, Muglia LJ, Lavoie PM, Hadchouel A, Rämet M, Hallman M. Genome-wide association study of bronchopulmonary dysplasia: a potential role for variants near the CRP gene. Sci Rep 2017; 7:9271. [PMID: 28839172 PMCID: PMC5571168 DOI: 10.1038/s41598-017-08977-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/20/2017] [Indexed: 11/28/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD), the main consequence of prematurity, has a significant heritability, but little is known about predisposing genes. The aim of this study was to identify gene loci predisposing infants to BPD. The initial genome-wide association study (GWAS) included 174 Finnish preterm infants of gestational age 24–30 weeks. Thereafter, the most promising single-nucleotide polymorphisms (SNPs) associated with BPD were genotyped in both Finnish (n = 555) and non-Finnish (n = 388) replication cohorts. Finally, plasma CRP levels from the first week of life and the risk of BPD were assessed. SNP rs11265269, flanking the CRP gene, showed the strongest signal in GWAS (odds ratio [OR] 3.2, p = 3.4 × 10−6). This association was nominally replicated in Finnish and French African populations. A number of other SNPs in the CRP region, including rs3093059, had nominal associations with BPD. During the first week of life the elevated plasma levels of CRP predicted the risk of BPD (OR 3.4, p = 2.9 × 10–4) and the SNP rs3093059 associated nominally with plasma CRP levels. Finally, SNP rs11265269 was identified as a risk factor of BPD (OR 1.8, p = 5.3 × 10−5), independently of the robust antenatal risk factors. As such, in BPD, a potential role for variants near CRP gene is proposed.
Collapse
Affiliation(s)
- Mari Mahlman
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland. .,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.
| | - Minna K Karjalainen
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Johanna M Huusko
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.,Perinatal Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sture Andersson
- Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - M Anneli Kari
- Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Outi K T Tammela
- Tampere University Hospital, Tampere University, and Center of Pediatric Child Health, Tampere, Finland
| | - Ulla Sankilampi
- Department of Pediatrics, Kuopio University Hospital, Kuopio, Finland
| | - Liisa Lehtonen
- Turku University Hospital, and the University of Turku, Turku, Finland
| | - Riitta H Marttila
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Dirk Bassler
- Department of Neonatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Hospital, Tuebingen, Germany
| | | | - Claude Danan
- Inserm, U955, Créteil, France.,CRB, CHI-Creteil, France.,Department of neonatology, CHI-Creteil, Creteil, France
| | - Christophe Delacourt
- Inserm, U955, Créteil, France.,AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Paris, France.,Université Paris-Descartes, Paris, France
| | - Aarno Palotie
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.,The Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Louis J Muglia
- Perinatal Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Vancouver Canada, Vancouver, Canada
| | - Alice Hadchouel
- Inserm, U955, Créteil, France.,AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Paris, France.,Université Paris-Descartes, Paris, France
| | - Mika Rämet
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.,BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere, Finland
| | - Mikko Hallman
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
45
|
Abstract
Despite recent advances in neonatal medicine, neonatal disorders, such as bronchopulmonary dysplasia and intraventricular hemorrhage in preterm neonates and hypoxic ischemic encephalopathy in term neonates, remain major causes of mortality and morbidities. Promising preclinical research results suggest that stem cell therapies represent the next breakthrough in the treatment of currently intractable and devastating neonatal disorders with complex multifactorial etiologies. This review focuses primarily on the potential role of stem cell therapy in the above mentioned neonatal disorders, highlighting the results of human clinical trials and the challenges that remain to be addressed for their safe and successful translation into clinical care of newborn infants.
Collapse
Affiliation(s)
- Yun Sil Chang
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - So Yoon Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sein Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Soon Park
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
46
|
Cantey JB, Huffman LW, Subramanian A, Marshall AS, Ballard AR, Lefevre C, Sagar M, Pruszynski JE, Mallett LH. Antibiotic Exposure and Risk for Death or Bronchopulmonary Dysplasia in Very Low Birth Weight Infants. J Pediatr 2017; 181:289-293.e1. [PMID: 27908652 DOI: 10.1016/j.jpeds.2016.11.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/03/2016] [Accepted: 11/01/2016] [Indexed: 12/23/2022]
Abstract
We assessed the association between antibiotic exposure in the first 2 weeks of life and development of bronchopulmonary dysplasia in a cohort of very low birth weight infants. After controlling for the severity of illness, each additional day of antibiotic therapy was associated with both an increased risk for and severity of bronchopulmonary dysplasia.
Collapse
Affiliation(s)
- Joseph B Cantey
- Department of Pediatrics, Texas A&M Health Science Center, Baylor Scott & White Health, Temple, TX.
| | - Landon W Huffman
- Department of Pediatrics, Texas A&M Health Science Center, Baylor Scott & White Health, Temple, TX
| | | | | | - A Rebecca Ballard
- Department of Pediatrics, Texas A&M Health Science Center, Baylor Scott & White Health, Temple, TX
| | - Cassandra Lefevre
- Department of Pediatrics, Texas A&M Health Science Center, Baylor Scott & White Health, Temple, TX
| | - Malvika Sagar
- Department of Pediatrics, Texas A&M Health Science Center, Baylor Scott & White Health, Temple, TX
| | | | - Lea H Mallett
- Department of Pediatrics, Texas A&M Health Science Center, Baylor Scott & White Health, Temple, TX
| |
Collapse
|
47
|
Abstract
The pathogenesis of Bronchopulmonary Dysplasia (BPD) is multifactorial and the clinical phenotype of BPD is extremely variable. Predicting BPD is difficult, as it is a disease with a clinical operational definition but many clinical phenotypes and endotypes. Most biomarkers studied over the years have low predictive accuracy, and none are currently used in routine clinical care or shown to be useful for predicting longer-term respiratory outcome. Targeted cellular and humoral biomarkers and novel systems biology 'omic' based approaches including genomic and microbiomic analyses are described in this review.
Collapse
|
48
|
Davidson LM, Berkelhamer SK. Bronchopulmonary Dysplasia: Chronic Lung Disease of Infancy and Long-Term Pulmonary Outcomes. J Clin Med 2017; 6:E4. [PMID: 28067830 PMCID: PMC5294957 DOI: 10.3390/jcm6010004] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/28/2016] [Accepted: 12/28/2016] [Indexed: 12/16/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease most commonly seen in premature infants who required mechanical ventilation and oxygen therapy for acute respiratory distress. While advances in neonatal care have resulted in improved survival rates of premature infants, limited progress has been made in reducing rates of BPD. Lack of progress may in part be attributed to the limited therapeutic options available for prevention and treatment of BPD. Several lung-protective strategies have been shown to reduce risks, including use of non-invasive support, as well as early extubation and volume ventilation when intubation is required. These approaches, along with optimal nutrition and medical therapy, decrease risk of BPD; however, impacts on long-term outcomes are poorly defined. Characterization of late outcomes remain a challenge as rapid advances in medical management result in current adult BPD survivors representing outdated neonatal care. While pulmonary disease improves with growth, long-term follow-up studies raise concerns for persistent pulmonary dysfunction; asthma-like symptoms and exercise intolerance in young adults after BPD. Abnormal ventilatory responses and pulmonary hypertension can further complicate disease. These pulmonary morbidities, combined with environmental and infectious exposures, may result in significant long-term pulmonary sequalae and represent a growing burden on health systems. Additional longitudinal studies are needed to determine outcomes beyond the second decade, and define risk factors and optimal treatment for late sequalae of disease.
Collapse
Affiliation(s)
- Lauren M Davidson
- Department of Pediatrics, University at Buffalo SUNY, Buffalo, NY 14228, USA.
| | - Sara K Berkelhamer
- Department of Pediatrics, University at Buffalo SUNY, Buffalo, NY 14228, USA.
| |
Collapse
|
49
|
Hibbs AM, Muhlebach MS. Infection and Inflammation: Catalysts of Pulmonary Morbidity in Bronchopulmonary Dysplasia. RESPIRATORY OUTCOMES IN PRETERM INFANTS 2017. [PMCID: PMC7121702 DOI: 10.1007/978-3-319-48835-6_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Anna Maria Hibbs
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio USA
| | - Marianne S. Muhlebach
- Department of Pediatrics, University of North Carolina Chapel Hill, Chapel Hill, North Carolina USA
| |
Collapse
|