1
|
Shemesh Gilboa N, Aviram M, Goldbart A, Hazan G, Arwas N, Hazan I, Yafit D, Tsaregorodtsev S, Golan-Tripto I. Flexible bronchoscopy in preterm infants with bronchopulmonary dysplasia: findings and complications in a matched control study. Eur J Pediatr 2024; 183:4837-4845. [PMID: 39245659 DOI: 10.1007/s00431-024-05750-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Bronchopulmonary dysplasia (BPD) poses a significant challenge as the most common late morbidity of preterm infants. This study aimed to evaluate airway abnormalities in infants with BPD who underwent flexible bronchoscopy (FB) to gain insights into the prevalence of upper airway obstruction and associated complications. A retrospective case-control study was conducted on BPD patients who underwent FB at a tertiary center between 2013 and 2023. BPD patients were matched (1:3) with a reference group based on age, gender, and ethnicity, who also had undergone FB. Demographic data, comorbidities, indications for FB, findings, and complications during and after FB were collected. The study included 50 BPD patients (mean age 1.26 ± 0.9 years, 58% males), and 150 controls. As expected, BPD patients had a lower gestational age, lower birth weight, and longer hospitalizations and were treated with more medications. Abnormal bronchoscopy findings were significantly more common in the BPD group compared to the reference group, with an increased rate of turbinate hypertrophy (OR [95% CI]: 3.44 [1.27-9.37], P = 0.014), adenoid hypertrophy (OR: 2.7 [1.38-5.29], P = 0.004), lingual tonsils (OR: 5.44 [1.29-27.4], P = 0.0024), subglottic stenosis (OR: 6.95 [2.08-27.1], P = 0.002), and tracheomalacia (OR: 2.98 [1.06-8.19], P = 0.034). Complications including desaturation (OR: 3.89 [1.32-11.7], P = 0.013) and PICU admission (OR: 16.6 [2.58-322], P = 0.011) were more frequent in the BPD than in the reference group. CONCLUSION The study revealed a high prevalence of structural anomalies leading to upper airway obstruction and complications in infants with BPD undergoing FB. These findings emphasize the importance of careful consideration and preparation for bronchoscopic procedures in this vulnerable population. WHAT IS KNOWN • Bronchopulmonary dysplasia (BPD) represents the most prevalent late morbidity among preterm infants. • Preterm infants diagnosed with BPD frequently undergo diagnostic procedures, including flexible and rigid bronchoscopies, to identify structural pathologies within the respiratory tract. WHAT IS NEW • A significantly higher prevalence of structural anomalies leading to upper airway obstruction was observed in the BPD group compared to controls. • The incidence of complications during flexible bronchoscopy was higher in the BPD group than in controls.
Collapse
Affiliation(s)
- Noa Shemesh Gilboa
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Micha Aviram
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Aviv Goldbart
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Pediatrics Department, Soroka University Medical Center, Beer Sheva, Israel
- Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Guy Hazan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Pediatrics Department, Soroka University Medical Center, Beer Sheva, Israel
- Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Noga Arwas
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Pediatrics Department, Soroka University Medical Center, Beer Sheva, Israel
| | - Itai Hazan
- Pediatrics Department, Soroka University Medical Center, Beer Sheva, Israel
- Clinical Research Center, Soroka University Medical Center, Beer Sheva, Israel
| | - Daniel Yafit
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Department of Otorhinolaryngology and Head and Neck Surgery, Soroka University Medical Center, Beer Sheva, Israel
| | - Sergey Tsaregorodtsev
- Department of Anesthesiology and Intensive Care, Soroka University Medical Center, Beer-Sheva, Israel
| | - Inbal Golan-Tripto
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- Pediatrics Department, Soroka University Medical Center, Beer Sheva, Israel.
- Pediatric Pulmonary Unit, Soroka University Medical Center, Beer Sheva, Israel.
| |
Collapse
|
2
|
Chioma R, Healy DB, Finn D, Walsh BH, Reynolds C, O'Sullivan D, Livingstone V, Bussmann N, Dempsey EM. The Bronchopulmonary Dysplasia score: A predictive model for bronchopulmonary dysplasia or death in high-risk preterm infants. Acta Paediatr 2024; 113:1781-1790. [PMID: 38798138 DOI: 10.1111/apa.17304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
AIM Progressive respiratory deterioration in infants at high risk of bronchopulmonary dysplasia (BPD) is associated with patent ductus arteriosus (PDA) exposure. This study aimed to design an early predictive model for BPD or death in preterm infants using early echocardiographic markers and clinical data. METHODS Infants born with gestational age (GA) ≤ 29 weeks and/or birth weight (BW) < 1500 g at Cork University Maternity Hospital, Ireland were retrospectively evaluated. Those with echocardiography performed between 36 h and 7 days of life were eligible for inclusion. Exclusion criteria were pulmonary hypertension and major congenital anomalies. The primary outcome was a composite of BPD and death before discharge. RESULTS The study included 99 infants. A predictive model for the primary outcome was developed, which included three variables (BW, Respiratory Severity Score and flow pattern across the PDA), and yielding an area under the curve of 0.98 (95% CI 0.96-1.00, p < 0.001). Higher scores were predictive of the primary outcome. A cut-off of -1.0 had positive and negative predictive values of 89% and 98%, and sensitivity and specificity of 98% and 88%, respectively. CONCLUSION Our prediction model is an accessible bedside tool that predicts BPD or death in premature infants.
Collapse
Affiliation(s)
- Roberto Chioma
- INFANT Research Centre, University College Cork, Cork, Ireland
- Catholic University of Sacred Heart, Rome, Italy
| | - David B Healy
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Daragh Finn
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Brian H Walsh
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Cole Reynolds
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Deirdre O'Sullivan
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Vicki Livingstone
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Neidin Bussmann
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - Eugene M Dempsey
- INFANT Research Centre, University College Cork, Cork, Ireland
- Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Cortesi V, Cavallaro G, Raffaeli G, Ghirardello S, Mosca F, Klei TR, Fustolo-Gunnink S, Stanworth S, New HV, Deschmann E, Lopriore E. Why might cord blood be a better source of platelets for transfusion to neonates? BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2024; 22:292-302. [PMID: 38557319 PMCID: PMC11251836 DOI: 10.2450/bloodtransfus.566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/04/2023] [Indexed: 04/04/2024]
Abstract
Thrombocytopenia (defined as a platelet count <150×109/L) is a common condition in preterm neonates and may occur in 18-35% of all infants admitted to the Neonatal Intensive Care Unit (NICU). Neonatal platelet functionality in terms of reactivity is often described as reduced compared to adults, even in healthy, term neonates. However, this platelet "hyporeactivity" does not correspond to a global functional impairment of the normal delicately balanced neonatal hemostatic system. The extent to which neonatal thrombocytopenia and platelet hyporeactivity contribute to the bleeding risk in preterm neonates remains unknown. Prophylactic platelet transfusions are often administered to them to reduce the risk of bleeding. However, recent literature indicates that adopting a higher platelet transfusion threshold than a lower one results in significantly higher death rates or major bleeding and can be harmful. Although the mechanism by which this occurs is not entirely clear, a mismatch between adult transfused platelets and the neonatal hemostatic system, as well as volume overload, are speculated to be potentially involved. Therefore, future research should consider novel transfusion products that may be more suitable for premature neonates. Blood products derived from umbilical cord blood (UCB) are promising, as they might perfectly match neonatal blood features. Here, we discuss the current knowledge about UCB-derived products, focusing on UCB-derived platelet concentrates and their potential for future clinical application. We will discuss how they may overcome the potential risks of transfusing adult-derived platelets to premature infants while maintaining efficacy.
Collapse
Affiliation(s)
- Valeria Cortesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Stefano Ghirardello
- Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Thomas R.L. Klei
- Department of Product and Process Development, Sanquin Blood Supply, Amsterdam, the Netherlands
| | - Suzanne Fustolo-Gunnink
- Sanquin Blood Supply Foundation, Amsterdam, the Netherlands
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pediatric Hematology, Emma Children’s Hospital, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Simon Stanworth
- Department of Hematology, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| | - Helen V. New
- Clinical Directorate, NHS Blood and Transplant, London, United Kingdom
| | - Emöke Deschmann
- Department of Neonatology, Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | - Enrico Lopriore
- Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
4
|
Zheng S, Ye L. Hemodynamic Melody of Postnatal Cardiac and Pulmonary Development in Children with Congenital Heart Diseases. BIOLOGY 2024; 13:234. [PMID: 38666846 PMCID: PMC11048247 DOI: 10.3390/biology13040234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Hemodynamics is the eternal theme of the circulatory system. Abnormal hemodynamics and cardiac and pulmonary development intertwine to form the most important features of children with congenital heart diseases (CHDs), thus determining these children's long-term quality of life. Here, we review the varieties of hemodynamic abnormalities that exist in children with CHDs, the recently developed neonatal rodent models of CHDs, and the inspirations these models have brought us in the areas of cardiomyocyte proliferation and maturation, as well as in alveolar development. Furthermore, current limitations, future directions, and clinical decision making based on these inspirations are highlighted. Understanding how CHD-associated hemodynamic scenarios shape postnatal heart and lung development may provide a novel path to improving the long-term quality of life of children with CHDs, transplantation of stem cell-derived cardiomyocytes, and cardiac regeneration.
Collapse
Affiliation(s)
- Sixie Zheng
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| | - Lincai Ye
- Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China;
- Shanghai Institute for Pediatric Congenital Heart Disease, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, National Children’s Medical Center, Shanghai 200127, China
| |
Collapse
|
5
|
Roeder F, Knudsen L, Schmiedl A. The expression of the surfactant proteins SP-A and SP-B during postnatal alveolarization of the rat lung. PLoS One 2024; 19:e0297889. [PMID: 38483982 PMCID: PMC10939297 DOI: 10.1371/journal.pone.0297889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/03/2024] [Indexed: 03/17/2024] Open
Abstract
OBJECTIVE Surfactant-specific proteins (SP) are responsible for the functional and structural integrity as well as for the stabilization of the intra-alveolar surfactant. Morphological lung maturation starts in rat lungs after birth. The aim of this study was to investigate whether the expression of the hydrophilic SP-A and the hydrophobic SP-B is associated with characteristic postnatal changes characterizing morphological lung maturation. METHODS Stereological methods were performed on the light microscope. Using immunohistochemical and molecular biological methods (Western Blot, RT-qPCR), the SP-A and SP-B of adult rat lungs and of those with different postnatal developmental stages (3, 7, 14 and 21 days after birth) were characterized. RESULTS As signs of alveolarization the total septal surface and volume increased and the septal thickness decreased. The significantly highest relative surface fraction of SP-A labeled alveolar epithelial cells type II (AEII) was found together with the highest relative SP-A gene expression before the alveolarization (3th postnatal day). With the downregulation of SP-A gene expression during and after alveolarization (between postnatal days 7 and 14), the surface fraction of the SP-A labeled AEII also decreased, so they are lowest in adult animals. The surface fraction of SP-B labeled AEII and the SP-B gene expression showed the significantly highest levels in adults, the protein expression increased also significantly at the end of morphological lung maturation. There were no alterations in the SP-B expression before and during alveolarization until postnatal day 14. The protein expression as well as the gene expression of SP-A and SP-B correlated very well with the total surface of alveolar septa independent of the postnatal age. CONCLUSION The expression of SP-A and SP-B is differentially associated with morphological lung maturation and correlates with increased septation of alveoli as indirect clue for alveolarization.
Collapse
Affiliation(s)
- Franziska Roeder
- Institute of Functional and Applied Anatomy, Medical Hannover School, Hannover, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Medical Hannover School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Andreas Schmiedl
- Institute of Functional and Applied Anatomy, Medical Hannover School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
6
|
Mehra K, Kresch M. Trends in the Incidence of Bronchopulmonary Dysplasia after the Introduction of Neurally Adjusted Ventilatory Assist (NAVA). CHILDREN (BASEL, SWITZERLAND) 2024; 11:113. [PMID: 38255426 PMCID: PMC10814022 DOI: 10.3390/children11010113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/04/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
OBJECTIVE This study investigates the difference in the rates of bronchopulmonary dysplasia in very low birth weight infants before and after the introduction of neurally adjusted ventilatory assist (NAVA). STUDY DESIGN A retrospective cohort study comparing rates of Bronchopulmonary dysplasia (BPD) before and after implementation of NAVA. Eligibility criteria included all very low birth weight VLBW neonates needing ventilation. For analysis, each cohort was divided into three subgroups based on gestational age. Changes in the rate of BPD, length of stay, tracheostomy rates, invasive ventilator days, and home oxygen therapy were compared. RESULTS There were no differences in the incidence of BPD in neonates at 23-25 6/7 weeks' and 29-32 weeks' gestation between the two cohorts. A higher incidence of BPD was seen in the 26-28 5/7 weeks' gestation NAVA subgroup compared to controls (86% vs. 68%, p = 0.05). No significant difference was found for ventilator days, but infants in the 26-28 6/7 subgroup in the NAVA cohort had a longer length of stay (98 ± 34 days vs. 82 ± 24 days, p = 0.02), a higher percentage discharged on home oxygen therapy (45% vs. 18%, respectively, p = 0.006), and higher tracheostomy rates (3/36 vs. 0/60, p = 0.02), compared to the control group. CONCLUSIONS The NAVA mode was not associated with a reduction in BPD when compared to other modes of ventilation. Unexpected increases were seen in BPD rates, home oxygen therapy rates, tracheostomy rates, and the length of stay in the NAVA subgroup born at 26-28 6/7 weeks' gestation.
Collapse
Affiliation(s)
- Kashish Mehra
- Division of Neonatal-Perinatal Medicine, Penn State Health Children’s Hospital, Hershey, PA 17033, USA;
| | | |
Collapse
|
7
|
Perveen S, Chen CM, Sobajima H, Zhou X, Chen JY. Editorial: Bronchopulmonary dysplasia: latest advances. Front Pediatr 2023; 11:1303761. [PMID: 38027276 PMCID: PMC10679731 DOI: 10.3389/fped.2023.1303761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Affiliation(s)
- Shahana Perveen
- Department of Pediatrics, Cohen Children Medical Center, Manhasset, NY, United States
- Department of Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Chung-Ming Chen
- Department of Pediatrics, Taipei Medical University, Taipei, Taiwan
| | - Hisanori Sobajima
- Division of Neonatology, Department of Pediatrics, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Xiaoguang Zhou
- Department of Neonatology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Kangdon, China
| | - Jia-Yuh Chen
- Division of Neonatology, Changhua Christian Children’s Hospital, Changhua, Taiwan
| |
Collapse
|
8
|
Ou W, Lei K, Wang H, Ma H, Deng X, He P, Zhao L, Lv Y, Tang G, Zhang B, Li J. Development of a blood proteins-based model for bronchopulmonary dysplasia prediction in premature infants. BMC Pediatr 2023; 23:304. [PMID: 37330491 PMCID: PMC10276448 DOI: 10.1186/s12887-023-04065-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 05/10/2023] [Indexed: 06/19/2023] Open
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is the most common chronic pulmonary disease in premature infants. Blood proteins may be early predictors of the development of this disease. METHODS In this study, protein expression profiles (blood samples during their first week of life) and clinical data of the GSE121097 was downloaded from the Gene Expression Omnibus. Weighted gene co-expression network analysis (WGCNA) and differential protein analysis were carried out for variable dimensionality reduction and feature selection. Least absolute shrinkage and selection operator (LASSO) were conducted for BPD prediction model development. The performance of the model was evaluated by the receiver operating characteristic (ROC) curve, calibration curve, and decision curve. RESULTS The results showed that black module, magenta module and turquoise module, which included 270 proteins, were significantly correlated with the occurrence of BPD. 59 proteins overlapped between differential analysis results and above three modules. These proteins were significantly enriched in 253 GO terms and 11 KEGG signaling pathways. Then, 59 proteins were reduced to 8 proteins by LASSO analysis in the training cohort. The proteins model showed good BPD predictive performance, with an AUC of 1.00 (95% CI 0.99-1.00) and 0.96 (95% CI 0.90-1.00) in training cohort and test cohort, respectively. CONCLUSION Our study established a reliable blood-protein based model for early prediction of BPD in premature infants. This may help elucidate pathways to target in lessening the burden or severity of BPD.
Collapse
Affiliation(s)
- Wanting Ou
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - KeJing Lei
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Huanhuan Wang
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Hongmei Ma
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Xiaojuan Deng
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Pengcheng He
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Liping Zhao
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Youdao Lv
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Guohong Tang
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Benjin Zhang
- Department of Pediatrics, Dazhou Central Hospital, Dazhou, Sichuan, China.
| | - Jie Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China.
| |
Collapse
|
9
|
Chioma R, Ghirardello S, Włodarczyk K, Ulan-Drozdowska J, Spagarino A, Szumska M, Krasuska K, Seliga-Siwecka J, Philip RK, Al Assaf N, Pierro M. Association between the development of bronchopulmonary dysplasia and platelet transfusion: a protocol for a systematic review and meta-analysis. Front Pediatr 2023; 11:1049014. [PMID: 37360369 PMCID: PMC10289060 DOI: 10.3389/fped.2023.1049014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/10/2023] [Indexed: 06/28/2023] Open
Abstract
Background There is a lack of consensus on the management of thrombocytopenia in preterm infants, and the threshold for prophylactic platelet transfusion varies widely among clinicians and institutions. Reports in animal models suggested that platelets may play a relevant role in lung alveolarization and regeneration. Bronchopulmonary dysplasia (BPD) is a severe respiratory condition with a multifactorial origin that affects infants born at the early stages of lung development. Recent randomized controlled trials on the platelets count threshold for prophylactic transfusions in preterm infants with thrombocytopenia suggest that a higher exposition to platelet transfusion may increase the risk of BPD. Here, we report a protocol for a systematic review, which aims to assist evidence-based clinical practice and clarify if the administration of platelet products may be associated with the incidence of BPD and/or death in preterm infants. Methods MEDLINE, Embase, Cochrane databases, and sources of gray literature for conference abstracts and trial registrations will be searched with no time or language restrictions. Case-control studies, cohort studies, and nonrandomized or randomized trials that evaluated the risk for BPD and/or death in preterm infants exposed to platelet transfusion will be included. Data from studies that are sufficiently similar will be pooled as appropriate. Data extraction forms will be developed a priori. Observational studies and nonrandomized and randomized clinical trials will be analyzed separately. Odds ratio with 95% confidence interval (CI) for dichotomous outcomes and the mean difference (95% CI) for continuous outcomes will be combined. The expected heterogeneity will be accounted for using a random-effects model. Subgroup analysis will be performed based on a priori-determined covariate of interest. In case of sufficient homogeneity of interventions and outcomes evaluated, results from subgroups of studies will be pooled together in a meta-analysis. Discussion This systematic review will investigate the association of BPD/death with platelet components administration in preterm infants, and, consequently, it will provide reliable indications for the evidence-based management of premature patients with thrombocytopenia.
Collapse
Affiliation(s)
- Roberto Chioma
- Department of Woman and Child Health and Public Health, Neonatology Unit, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Ghirardello
- Department of Woman and Child Health and Public Health, Neonatology Unit, Ospedale San Matteo, Pavia, Italy
| | - Krzysztof Włodarczyk
- Main Library, Medical University of Warsaw, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Marta Szumska
- Neonatal and Intensive Care Department, Medical University of Warsaw, Warsaw, Poland
| | - Klaudia Krasuska
- Neonatal and Intensive Care Department, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Seliga-Siwecka
- Neonatal and Intensive Care Department, Medical University of Warsaw, Warsaw, Poland
| | - Roy K. Philip
- Division of Neonatology, Department of Pediatrics, University Maternity Hospital Limerick and Medical School University of Limerick, Limerick, Ireland
| | - Niazy Al Assaf
- Division of Neonatology, Department of Pediatrics, University Maternity Hospital Limerick and Medical School University of Limerick, Limerick, Ireland
| | - Maria Pierro
- M. Bufalini Hospital, AUSL Romagna, Cesena, Italy
| |
Collapse
|
10
|
Zhong XQ, Hao TF, Zhu QJ, Zheng J, Zheng MF, Li XH, Luo LH, Xia CS, Fan YW, Gu J, Liu T, Chen DJ. Umbilical cord blood exosomes from very preterm infants with bronchopulmonary dysplasia aggravate lung injury in mice. Sci Rep 2023; 13:8648. [PMID: 37244977 DOI: 10.1038/s41598-023-35620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/21/2023] [Indexed: 05/29/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by abnormal development of the blood vessels and alveoli in lungs, which largely occurs in premature infants. Exosomes (EXO) from very preterm infants (VPI) with BPD (BPD-EXO) impair angiogenic activities of human umbilical vein endothelial cells (HUVECs) via EXO-miRNAs cargo. This study aimed to determine whether and how BPD-EXO affect the development of BPD in a mouse model. We showed that treating BPD mice with BPD-EXO chronically and irreversibly aggravated lung injury. BPD-EXO up-regulated 139 and down-regulated 735 genes in the mouse lung tissue. These differentially expressed genes were enriched to the MAPK pathway (e.g., Fgf9 and Cacna2d3), which is critical to angiogenesis and vascular remodeling. BPD-EXO suppressed expression of Fgf9 and Cacna2d3 in HUVECs and inhibited migration, tube formation, and increased cell apoptosis in HUVECs. These data demonstrate that BPD-EXO aggravate lung injury in BPD mice and impair lung angiogenesis, plausibly leading to adverse outcomes of VPI with BPD. These data also suggest that BPD-EXO could serve as promising targets for predicting and treating BPD.
Collapse
Affiliation(s)
- Xin-Qi Zhong
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China.
- Key Laboratory for Major Obstetric Disease of Guangdong Province, Guangzhou, China.
| | - Tao-Fang Hao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qi-Jiong Zhu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, 510632, China
- China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of WI-Madison, Madison, WI, USA
| | - Mao-Fei Zheng
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Xiu-Hong Li
- Department of Maternal and Child Health, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Li-Hua Luo
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Chang-Shun Xia
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Yu-Wei Fan
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Jian Gu
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Tao Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, 510632, China.
- China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China.
| | - Dun-Jin Chen
- Key Laboratory for Major Obstetric Disease of Guangdong Province, Guangzhou, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
11
|
Shankar N, Thapa S, Shrestha AK, Sarkar P, Gaber MW, Barrios R, Shivanna B. Hyperoxia Disrupts Lung Lymphatic Homeostasis in Neonatal Mice. Antioxidants (Basel) 2023; 12:620. [PMID: 36978868 PMCID: PMC10045755 DOI: 10.3390/antiox12030620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammation causes bronchopulmonary dysplasia (BPD), a common lung disease of preterm infants. One reason this disease lacks specific therapies is the paucity of information on the mechanisms regulating inflammation in developing lungs. We address this gap by characterizing the lymphatic phenotype in an experimental BPD model because lymphatics are major regulators of immune homeostasis. We hypothesized that hyperoxia (HO), a major risk factor for experimental and human BPD, disrupts lymphatic endothelial homeostasis using neonatal mice and human dermal lymphatic endothelial cells (HDLECs). Exposure to 70% O2 for 24-72 h decreased the expression of prospero homeobox 1 (Prox1) and vascular endothelial growth factor c (Vegf-c) and increased the expression of heme oxygenase 1 and NAD(P)H dehydrogenase [quinone]1 in HDLECs, and reduced their tubule formation ability. Next, we determined Prox1 and Vegf-c mRNA levels on postnatal days (P) 7 and 14 in neonatal murine lungs. The mRNA levels of these genes increased from P7 to P14, and 70% O2 exposure for 14 d (HO) attenuated this physiological increase in pro-lymphatic factors. Further, HO exposure decreased VEGFR3+ and podoplanin+ lymphatic vessel density and lymphatic function in neonatal murine lungs. Collectively, our results validate the hypothesis that HO disrupts lymphatic endothelial homeostasis.
Collapse
Affiliation(s)
- Nithyapriya Shankar
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Poonam Sarkar
- Division of Hematology-Oncology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - M. Waleed Gaber
- Division of Hematology-Oncology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine (BCM), Houston, TX 77030, USA
| |
Collapse
|
12
|
Li DB, Xu XX, Hu YQ, Cui Q, Xiao YY, Sun SJ, Chen LJ, Ye LC, Sun Q. Congenital heart disease-associated pulmonary dysplasia and its underlying mechanisms. Am J Physiol Lung Cell Mol Physiol 2023; 324:L89-L101. [PMID: 36472329 PMCID: PMC9925164 DOI: 10.1152/ajplung.00195.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical observation indicates that exercise capacity, an important determinant of survival in patients with congenital heart disease (CHD), is most decreased in children with reduced pulmonary blood flow (RPF). However, the underlying mechanism remains unclear. Here, we obtained human RPF lung samples from children with tetralogy of Fallot as well as piglet and rat RPF lung samples from animals with pulmonary artery banding surgery. We observed impaired alveolarization and vascularization, the main characteristics of pulmonary dysplasia, in the lungs of RPF infants, piglets, and rats. RPF caused smaller lungs, cyanosis, and body weight loss in neonatal rats and reduced the number of alveolar type 2 cells. RNA sequencing demonstrated that RPF induced the downregulation of metabolism and migration, a key biological process of late alveolar development, and the upregulation of immune response, which was confirmed by flow cytometry and cytokine detection. In addition, the immunosuppressant cyclosporine A rescued pulmonary dysplasia and increased the expression of the Wnt signaling pathway, which is the driver of postnatal lung development. We concluded that RPF results in pulmonary dysplasia, which may account for the reduced exercise capacity of patients with CHD with RPF. The underlying mechanism is associated with immune response activation, and immunosuppressants have a therapeutic effect in CHD-associated pulmonary dysplasia.
Collapse
Affiliation(s)
- De-Bao Li
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xiu-Xia Xu
- 4Department of Radiology, Huangpu Branch, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yu-Qing Hu
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qing Cui
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ying-Ying Xiao
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Si-Juan Sun
- 5Department of Pediatric Intensive Care Unit, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Li-Jun Chen
- 3Department of Cardiology, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Lin-Cai Ye
- 2Department of Thoracic and Cardiovascular Surgery, Shanghai Institute for Pediatric Congenital Heart Disease, Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, Shanghai School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qi Sun
- 1Department of Thoracic and Cardiovascular Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
13
|
Yang L, Bao Z, Zhang L, Lei X, Zhang L. Position management on pulmonary function and bronchopulmonary dysplasia in premature infants: study protocol for a randomised controlled trial. BMJ Open 2022; 12:e062291. [PMID: 36521889 PMCID: PMC9756205 DOI: 10.1136/bmjopen-2022-062291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Bronchopulmonary dysplasia (BPD) is a common disease caused by various factors and mechanisms in premature infants. Owing to lung hypoplasia and the lack of alveolar surfactants in premature infants, oxygen therapy is often needed to maintain adequate breathing. Nevertheless, prolonged oxygen therapy can easily induce BPD, and there is currently no effective treatment. Therefore, the prevention of BPD in premature infants during hospitalisation is essential. Studies have revealed that the prone position can effectively improve the oxygenation of premature infants. However, a few studies have reported whether prone positioning can improve lung function and reduce BPD incidence. This trial will determine whether the prone position, compared with the supine position, can reduce BPD incidence and improve lung function in preterm infants. METHODS AND ANALYSIS This study protocol is for a single-centre, single-blind, randomised controlled trial of the prone position in premature infants. Following daily feeding, premature infants will be placed in the lateral position for 30 min; then they will be turned to the supine position (control group) or prone position (intervention group) for 2 hours each in the morning and afternoon. Moreover, infants in both groups will be placed in the supine or lateral position alternately according to their medical needs for the remaining time. The study begins when the premature infants are stable within 5 days after admission and ends when they are discharged from the hospital or at 36 weeks postmenstrual age. The primary outcome is the survival rate without BPD. The secondary outcomes include lung function parameters and lung oxygen saturation. ETHICS AND DISSEMINATION This trial is approved by the ethics committee of the Affiliated Hospital of Southwest Medical University, (ref approval no.KY2021186). The results will be published in a peer-reviewed journal. TRIAL REGISTRATION NUMBER ChiCTR2100049847.
Collapse
Affiliation(s)
- Liu Yang
- School of Nursing, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhengrong Bao
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Lianyu Zhang
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Clinical Nursing Research Institute, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaoping Lei
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Perinatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Lingping Zhang
- Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Department of Neonatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Clinical Nursing Research Institute, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
14
|
Heydarian M, Oak P, Zhang X, Kamgari N, Kindt A, Koschlig M, Pritzke T, Gonzalez-Rodriguez E, Förster K, Morty RE, Häfner F, Hübener C, Flemmer AW, Yildirim AO, Sudheendra D, Tian X, Petrera A, Kirsten H, Ahnert P, Morrell N, Desai TJ, Sucre J, Spiekerkoetter E, Hilgendorff A. Relationship between impaired BMP signalling and clinical risk factors at early-stage vascular injury in the preterm infant. Thorax 2022; 77:1176-1186. [PMID: 35580897 PMCID: PMC9685723 DOI: 10.1136/thoraxjnl-2021-218083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 04/11/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chronic lung disease, that is, bronchopulmonary dysplasia (BPD) is the most common complication in preterm infants and develops as a consequence of the misguided formation of the gas-exchange area undergoing prenatal and postnatal injury. Subsequent vascular disease and its progression into pulmonary arterial hypertension critically determines long-term outcome in the BPD infant but lacks identification of early, disease-defining changes. METHODS We link impaired bone morphogenetic protein (BMP) signalling to the earliest onset of vascular pathology in the human preterm lung and delineate the specific effects of the most prevalent prenatal and postnatal clinical risk factors for lung injury mimicking clinically relevant conditions in a multilayered animal model using wild-type and transgenic neonatal mice. RESULTS We demonstrate (1) the significant reduction in BMP receptor 2 (BMPR2) expression at the onset of vascular pathology in the lung of preterm infants, later mirrored by reduced plasma BMP protein levels in infants with developing BPD, (2) the rapid impairment (and persistent change) of BMPR2 signalling on postnatal exposure to hyperoxia and mechanical ventilation, aggravated by prenatal cigarette smoke in a preclinical mouse model and (3) a link to defective alveolar septation and matrix remodelling through platelet derived growth factor-receptor alpha deficiency. In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. CONCLUSION We identified impaired BMP signalling as a hallmark of early vascular disease in the injured neonatal lung while outlining its promising potential as a future biomarker or therapeutic target in this growing, high-risk patient population.
Collapse
Affiliation(s)
- Motaharehsadat Heydarian
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Prajakta Oak
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xin Zhang
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Nona Kamgari
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Alida Kindt
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Markus Koschlig
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Tina Pritzke
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Erika Gonzalez-Rodriguez
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kai Förster
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Rory E Morty
- Department of Translational Pulmonology, University Hospital Heidelberg, Translational Lung Research Center campus of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Friederike Häfner
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Christoph Hübener
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Andreas W Flemmer
- Department of Neonatology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| | - Ali Oender Yildirim
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Deepti Sudheendra
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Xuefei Tian
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Agnese Petrera
- Research Unit Protein Science and Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Peter Ahnert
- Institute for Medical Informatics, Statistics, and Epidemiology (IMISE), associated partner of the German Center for Lung Research (DZL), University of Leipzig, Leipzig, Germany
| | - Nick Morrell
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Tushar J Desai
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Jennifer Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee, USA
| | - Edda Spiekerkoetter
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Anne Hilgendorff
- Institute for Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
- Center for Comprehensive Developmental Care (CDeCLMU), Ludwig-Maximilians University, LMU Hospital, Munich, Germany
| |
Collapse
|
15
|
Yang W, Huang C, Wang W, Zhang B, Chen Y, Xie X. Bone mesenchymal stem cell-derived exosomes prevent hyperoxia-induced apoptosis of primary type II alveolar epithelial cells in vitro. PeerJ 2022; 10:e13692. [PMID: 36071827 PMCID: PMC9443791 DOI: 10.7717/peerj.13692] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/16/2022] [Indexed: 01/17/2023] Open
Abstract
Background The presence of alveolar epithelial type II cells (AECIIs) is one of the most important causes of bronchopulmonary dysplasia (BPD). Exosomes from bone mesenchymal stem cells (BMSCs) can reduce hyperoxia-induced damage and provide better results in terms of alveolar and pulmonary vascularization parameters than BMSCs. Currently, intervention studies using BMSC-derived exosomes on the signaling pathways regulating proliferation and apoptosis of alveolar epithelial cells under the condition of BPD have not been reported. This study investigated the effects of rat BMSC-derived exosomes on the proliferation and apoptosis of hyperoxia-induced primary AECIIs in vitro. Methods The isolated AECIIs were grouped as follows: normal control (21% oxygen), hyperoxia (85% oxygen), hyperoxia+exosome (20 µg/mL), hyperoxia+exosome+LY294002 (PI3K/Akt inhibitor, 20 µM), and hyperoxia+exosome+rapamycin (mTOR inhibitor, 5 nM). We used the PI3K/Akt inhibitor LY294002 and the mTOR inhibitor rapamycin to determine the roles of the PI3K/Akt and mTOR signaling pathways. The effects of BMSC-derived exosomes on AECII proliferation and apoptosis were assessed, respectively. Results Decreased levels of the antiapoptotic protein Bcl-2, the cell proliferation protein Ki67, p-PI3K, p-Akt, and p-mTOR, as well as increased levels of AECII apoptosis and the proapoptotic protein Bax in the hyperoxia group were observed. Notably, Sprague Dawley rat BMSC-derived exosomes could reverse the effect of hyperoxia on AECII proliferation. However, the application of LY294002 and rapamycin inhibited the protective effects of BMSC-derived exosomes. Conclusion Our findings revealed that BMSC-derived exosomes could regulate the expression of apoptosis-related proteins likely via the PI3K/Akt/mTOR signaling pathway, thereby preventing hyperoxia-induced AECII apoptosis.
Collapse
Affiliation(s)
- Wei Yang
- Department of Pediatrics, The Second Affiliated Hospital of Shenzhen University (The People’s Hospital of Baoan Shenzhen), Shenzhen, China
| | - Chao Huang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University (The People’s Hospital of Baoan Shenzhen), Shenzhen, China
| | - Wenjian Wang
- Department of Respiratory Medicine, Shenzhen Children’s Hospital, Shenzhen, China
| | - Baozhu Zhang
- Department of Oncology, The Second Affiliated Hospital of Shenzhen University (The People’s Hospital of Baoan Shenzhen), Shenzhen, China
| | - Yunbin Chen
- Department of Pediatrics, Guangdong Women’s and Children’s Hospital, Guangzhou, China
| | - Xinlin Xie
- Department of Pediatrics, The Second Affiliated Hospital of Shenzhen University (The People’s Hospital of Baoan Shenzhen), Shenzhen, China
| |
Collapse
|
16
|
Manochkumar J, Singh A, Efferth T, Ramamoorthy S. Untapping the protective role of carotenoids against respiratory diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154286. [PMID: 35820304 DOI: 10.1016/j.phymed.2022.154286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/15/2022] [Accepted: 06/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Recent studies revealed a substantial role of carotenoids to treat respiratory diseases. This review aimed to give an updated overview of the investigational evidence on the preventive properties of carotenoids against respiratory diseases both in vitro and in vivo along with their pathophysiology and mechanisms of action. HYPOTHESIS Carotenoids as a potential therapeutic class of bioactive compounds to treat respiratory diseases. RESULTS Carotenoids such as β-carotene, lycopene, crocin, bixin, lutein, and astaxanthin show beneficial effects against chronic lung diseases (e.g., asthma, emphysema, fibrosis, COPD, acute lung injury, and lung cancer). Moreover, in vitro and in vivo studies also supported the preventive role of carotenoids. These carotenoids showed a beneficial role by activation of the NRF2/HO-1 pathway and inhibition of the NF-кB, MAPK, JAK/STAT-3, and PI3K/AKT pathways. Additionally, epidemiological studies also showed that dietary intake of carotenoids lowers the risk of lung diseases. CONCLUSION Carotenoids may be used as drugs or can be given in combination with other drugs to prevent and treat respiratory diseases. Although in vitro and in vivo results are encouraging, further well-conducted randomized clinical trials are required to approve carotenoids as drug candidates.
Collapse
Affiliation(s)
- Janani Manochkumar
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014 Tamil Nadu, India
| | - Anuma Singh
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014 Tamil Nadu, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Germany
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014 Tamil Nadu, India.
| |
Collapse
|
17
|
Herminghaus A, Kozlov AV, Szabó A, Hantos Z, Gylstorff S, Kuebart A, Aghapour M, Wissuwa B, Walles T, Walles H, Coldewey SM, Relja B. A Barrier to Defend - Models of Pulmonary Barrier to Study Acute Inflammatory Diseases. Front Immunol 2022; 13:895100. [PMID: 35874776 PMCID: PMC9300899 DOI: 10.3389/fimmu.2022.895100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Andrey V. Kozlov
- L Boltzmann Institute for Traumatology in Cooperation with AUVA and Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department of Human Pathology , IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Zoltán Hantos
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Mahyar Aghapour
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Bianka Wissuwa
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, Magdeburg University Medicine, Magdeburg, Germany
| | - Heike Walles
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- *Correspondence: Borna Relja,
| |
Collapse
|
18
|
Zhou O, You J, Xu X, Liu J, Qiu H, Hao C, Zou W, Wu W, Fu Z, Tian D, Zou L. Microvesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Enhance Alveolar Type II Cell Proliferation and Attenuate Lung Inflammation in a Rat Model of Bronchopulmonary Dysplasia. Stem Cells Int 2022; 2022:8465294. [PMID: 35795773 PMCID: PMC9252687 DOI: 10.1155/2022/8465294] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/06/2022] [Accepted: 06/09/2022] [Indexed: 01/08/2023] Open
Abstract
Although it is known that exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSCs) alleviate hyperoxic lung injury of bronchopulmonary dysplasia (BPD) in animal models, the role of microvesicles (MVs) derived from hUCMSCs in BPD is poorly defined. Furthermore, antenatal inflammation has been linked to high risk of BPD in preterm infants. The purpose of this study was to explore whether MVs derived from hUCMSCs can preserve lung structure and function in an antenatal lipopolysaccharide- (LPS-) induced BPD rat model and to clarify the underlying mechanism. We demonstrate that antenatal LPS induced alveolar simplification, altered lung function, and dysregulated pulmonary vasculature, which restored by hUCMSCs and MVs treatment. Furthermore, MVs were large vesicles with a diameter of 100-900 nanometers and mostly uptaken by alveolar epithelial type II cells (AT2) and macrophages. Compared with the LPS-exposed group, MVs restored the AT2 cell number and SP-C expression in vivo and promoted the proliferation of AT2 cells in vitro. MVs also restored the level of IL-6 and IL-10 in lung homogenate. Additionally, PTEN/AKT and MAPK pathways were associated with the protection of MVs. Taken together, this study suggests MVs derived from hUCMSCs improve lung architecture and function in an antenatal LPS-induced BPD rat model by promoting AT2 cell proliferation and attenuating lung inflammation; thus, MVs provide a promising therapeutic vehicle for BPD treatment.
Collapse
Affiliation(s)
- Ou Zhou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Jingyi You
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Xiaochuan Xu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Jiang Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Huijun Qiu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Chang Hao
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Wenjing Zou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Wenjie Wu
- Department of Pediatrics, Chongqing Youyoubaobei Women and Children's Hospital, Chongqing 401122, China
| | - Zhou Fu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Daiyin Tian
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Lin Zou
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
- Center of Clinical Molecular Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
- Clinical Research Unit, Children's Hospital of Shanghai Jiaotong University, Shanghai 200062, China
| |
Collapse
|
19
|
Kosik K, Sowińska A, Seremak-Mrozikiewicz A, Abu-Amara JA, Al-Saad SR, Karbowski LM, Gryczka K, Kurzawińska G, Szymankiewicz-Bręborowicz M, Drews K, Szpecht D. Polymorphisms of fibronectin-1 (rs3796123; rs1968510; rs10202709; rs6725958; and rs35343655) are not associated with bronchopulmonary dysplasia in preterm infants. Mol Cell Biochem 2022; 477:1645-1652. [PMID: 35230604 DOI: 10.1007/s11010-022-04397-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/16/2022] [Indexed: 11/27/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that mainly affects premature newborns. Many different factors, increasingly genetic, are involved in the pathogenesis of BPD. The aim of the study is to investigate the possible influence of fibronectin SNP on the occurrence of BPD. The study included 108 infants born between 24 and 32 weeks of gestation. BPD was diagnosed based on the National Institutes of Health Consensus definition. The 5 FN1 gene polymorphisms assessed in the study were the following: rs3796123; rs1968510; rs10202709; rs6725958; and rs35343655. BPD developed in 30 (27.8%) out of the 108 preterm infants. Incidence of BPD was higher in infants with lower APGAR scores and low birthweight. Investigation did not confirm any significant prevalence for BPD development in any genotypes and alleles of FN1. Further studies should be performed to confirm the role of genetic factors in etiology and pathogenesis of BPD.
Collapse
Affiliation(s)
- Katarzyna Kosik
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Anna Sowińska
- Department of Computer Science and Statistics, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | - Katarzyna Gryczka
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Grażyna Kurzawińska
- Department of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Krzysztof Drews
- Department of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Dawid Szpecht
- Department of Neonatology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
20
|
Chaubey S, Bhandari V. Stem cells in neonatal diseases: An overview. Semin Fetal Neonatal Med 2022; 27:101325. [PMID: 35367186 DOI: 10.1016/j.siny.2022.101325] [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: 10/18/2022]
Abstract
Preterm birth and its common complications are major causes of infant mortality and long-term morbidity. Despite great advances in understanding the pathogenesis of neonatal diseases and improvements in neonatal intensive care, effective therapies for the prevention or treatment for these conditions are still lacking. Stem cell (SC) therapy is rapidly emerging as a novel therapeutic tool for several diseases of the newborn with encouraging pre-clinical results that hold promise for translation to the bedside. The utility of different types of SCs in neonatal diseases is being explored. SC therapeutic efficacy is closely associated with its secretome-conditioned media and SC-derived extracellular vesicles, and a subsequent paracrine action in response to tissue injuries. In the current review, we summarize the pre-clinical and clinical studies of SCs and its secretome in diverse preterm and term birth-related diseases, thereby providing new insights for future therapies in neonatal medicine.
Collapse
Affiliation(s)
- Sushma Chaubey
- Department of Biomedical Engineering, Widener University, Chester, PA, 19013, USA.
| | - Vineet Bhandari
- Neonatology Research Laboratory, Department of Pediatrics, The Children's Regional Hospital at Cooper, Cooper Medical School of Rowan University, Suite Dorrance 755, One Cooper Plaza, Camden, NJ, 08103, USA.
| |
Collapse
|
21
|
Damianos A, Xu K, Kalin GT, Kalinichenko VV. Placental tissue stem cells and their role in neonatal diseases. Semin Fetal Neonatal Med 2022; 27:101322. [PMID: 34953760 DOI: 10.1016/j.siny.2021.101322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neonatal diseases such as hypoxic ischemic encephalopathy, diseases of prematurity and congenital disorders carry increased morbidity and mortality. Despite technological advancements, their incidence remains largely unabated. Stem cell (SC) interventions are novel therapies in the neonatal world. In pre-clinical models of neonatal diseases, SC applications have shown encouraging results. SC sources vary, with the bone marrow being the most utilized. However, the ability to harvest bone marrow SCs from neonates is limited. Placental-tissue derived SCs (PTSCs), provide an alternative and highly attractive source. Human placentas, the cornerstone of fetal survival, are abundant with such cells. Comparing to adult pools, PTSCs exhibit increased potency, decreased immunogenicity and stronger anti-inflammatory effects. Several types of PTSCs have been identified, with mesenchymal stem cells being the most utilized population. This review will focus on PTSCs and their pre-clinical and clinical applications in neonatology.
Collapse
Affiliation(s)
- Andreas Damianos
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Kui Xu
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gregory T Kalin
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Division of Neonatology and Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
22
|
The Aryl Hydrocarbon Receptor (AHR): A Novel Therapeutic Target for Pulmonary Diseases? Int J Mol Sci 2022; 23:ijms23031516. [PMID: 35163440 PMCID: PMC8836075 DOI: 10.3390/ijms23031516] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/30/2021] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a cytoplasmic transcription factor that is well-known for regulating xenobiotic metabolism. Studies in knockout and transgenic mice indicate that the AHR plays a vital role in the development of liver and regulation of reproductive, cardiovascular, hematopoietic, and immune homeostasis. In this focused review on lung diseases associated with acute injury and alveolar development, we reviewed and summarized the current literature on the mechanistic role(s) and therapeutic potential of the AHR in acute lung injury, chronic obstructive pulmonary disease, and bronchopulmonary dysplasia (BPD). Pre-clinical studies indicate that endogenous AHR activation is necessary to protect neonatal and adult lungs against hyperoxia- and cigarette smoke-induced injury. Our goal is to provide insight into the high translational potential of the AHR in the meaningful management of infants and adults with these lung disorders that lack curative therapies.
Collapse
|
23
|
David C, Frémond ML. Lung Inflammation in STING-Associated Vasculopathy with Onset in Infancy (SAVI). Cells 2022; 11:318. [PMID: 35159128 PMCID: PMC8834229 DOI: 10.3390/cells11030318] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/15/2022] Open
Abstract
STING-associated vasculopathy with onset in infancy (SAVI) is a type I interferonopathy caused by gain-of-function mutations in STING1 encoding stimulator of interferon genes (STING) protein. SAVI is characterized by severe inflammatory lung disease, a feature not observed in previously described type I interferonopathies i.e., Mendelian autoinflammatory disorders defined by constitutive activation of the type I interferon (IFN) pathway. Molecular defects in nucleic acid metabolism or sensing are central to the pathophysiology of these diseases, with such defects occurring at any step of the tightly regulated pathway of type I IFN production and signaling (e.g., exonuclease loss of function, RNA-DNA hybrid accumulation, constitutive activation of adaptor proteins such as STING). Among over 30 genotypes, SAVI and COPA syndrome, whose pathophysiology was recently linked to a constitutive activation of STING signaling, are the only type I interferonopathies presenting with predominant lung involvement. Lung disease is the leading cause of morbidity and mortality in these two disorders which do not respond to conventional immunosuppressive therapies and only partially to JAK1/2 inhibitors. In human silicosis, STING-dependent sensing of self-DNA following cell death triggered by silica exposure has been found to drive lung inflammation in mice and human models. These recent findings support a key role for STING and nucleic acid sensing in the homeostasis of intrinsic pulmonary inflammation. However, mechanisms by which monogenic defects in the STING pathway lead to pulmonary damages are not yet fully elucidated, and an improved understanding of such mechanisms is fundamental to improved future patient management. Here, we review the recent insights into the pathophysiology of SAVI and outline our current understanding of self-nucleic acid-mediated lung inflammation in humans.
Collapse
Affiliation(s)
- Clémence David
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Marie-Louise Frémond
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 Boulevard du Montparnasse, 75015 Paris, France
- Paediatric Immunology-Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP.Centre-Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| |
Collapse
|
24
|
Twisselmann N, Pagel J, Künstner A, Weckmann M, Hartz A, Glaser K, Hilgendorff A, Göpel W, Busch H, Herting E, Weinberg JB, Härtel C. Hyperoxia/Hypoxia Exposure Primes a Sustained Pro-Inflammatory Profile of Preterm Infant Macrophages Upon LPS Stimulation. Front Immunol 2021; 12:762789. [PMID: 34868007 PMCID: PMC8637891 DOI: 10.3389/fimmu.2021.762789] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/26/2021] [Indexed: 11/15/2022] Open
Abstract
Preterm infants are highly susceptible to sustained lung inflammation, which may be triggered by exposure to multiple environmental cues such as supplemental oxygen (O2) and infections. We hypothesized that dysregulated macrophage (MФ) activation is a key feature leading to inflammation-mediated development of bronchopulmonary dysplasia (BPD) in preterm infants. Therefore, we aimed to determine age-dependent differences in immune responses of monocyte-derived MФ comparing cord blood samples derived from preterm (n=14) and term (n=19) infants as well as peripheral blood samples from healthy adults (n=17) after lipopolysaccharide (LPS) exposure. Compared to term and adult MФ, LPS-stimulated preterm MФ showed an enhanced and sustained pro-inflammatory immune response determined by transcriptome analysis, cytokine release inducing a RORC upregulation due to T cell polarization of neonatal T cells, and TLR4 surface expression. In addition, a double-hit model was developed to study pulmonary relevant exposure factors by priming MФ with hyperoxia (O2 = 65%) or hypoxia (O2 = 3%) followed by lipopolysaccharide (LPS, 100ng/ml). When primed by 65% O2, subsequent LPS stimulation in preterm MФ led to an exaggerated pro-inflammatory response (e.g. increased HLA-DR expression and cytokine release) compared to LPS stimulation alone. Both, exposure to 65% or 3% O2 together with subsequent LPS stimulation, resulted in an exaggerated pro-inflammatory response of preterm MФ determined by transcriptome analysis. Downregulation of two major transcriptional factors, early growth response gene (Egr)-2 and growth factor independence 1 (Gfi1), were identified to play a role in the exaggerated pro-inflammatory response of preterm MФ to LPS insult after priming with 65% or 3% O2. Preterm MФ responses to LPS and hyperoxia/hypoxia suggest their involvement in excessive inflammation due to age-dependent differences, potentially mediated by downregulation of Egr2 and Gfi1 in the developing lung.
Collapse
Affiliation(s)
- Nele Twisselmann
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Julia Pagel
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany.,Department of Infectious Diseases and Microbiology, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Institute of Experimental Dermatology, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Markus Weckmann
- Department of Pediatrics Pneumology & Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany.,Airway Research Center North (ARCN) , Member of the German Center for Lung Research (DZL), Lübeck, Germany
| | - Annika Hartz
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Kirsten Glaser
- Center for Pediatric Research, Division of Neonatology, Department of Women's and Children's Health, University of Leipzig Medical Centre, Leipzig, Germany
| | - Anne Hilgendorff
- Center for Comprehensive Developmental Care (CDeCLMU), Member of the German Center for Lung Research (DZL), Hospital of the Ludwig-Maximilians University (LMU), CPC-M bioArchive, Munich, Germany
| | - Wolfgang Göpel
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Institute of Experimental Dermatology, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Egbert Herting
- Department of Pediatrics, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Jason B Weinberg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States.,Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States
| | - Christoph Härtel
- Department of Pediatrics, University of Würzburg, Würzburg, Germany
| |
Collapse
|
25
|
Schmiedl A, Wagener I, Jungen M, von Hörsten S, Stephan M. Lung development and immune status under chronic LPS exposure in rat pups with and without CD26/DPP4 deficiency. Cell Tissue Res 2021; 386:617-636. [PMID: 34606000 PMCID: PMC8595150 DOI: 10.1007/s00441-021-03522-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 09/05/2021] [Indexed: 11/26/2022]
Abstract
Dipeptidyl-peptidase IV (CD26), a multifactorial integral type II protein, is expressed in the lungs during development and is involved in inflammation processes. We tested whether daily LPS administration influences the CD26-dependent retardation in morphological lung development and induces alterations in the immune status. Newborn Fischer rats with and without CD26 deficiency were nebulized with 1 µg LPS/2 ml NaCl for 10 min from days postpartum (dpp) 3 to 9. We used stereological methods and fluorescence activated cell sorting (FACS) to determine morphological lung maturation and alterations in the pulmonary leukocyte content on dpp 7, 10, and 14. Daily LPS application did not change the lung volume but resulted in a significant retardation of alveolarization in both substrains proved by significantly lower values of septal surface and volume as well as higher mean free distances in airspaces. Looking at the immune status after LPS exposure compared to controls, a significantly higher percentage of B lymphocytes and decrease of CD4+CD25+ T cells were found in both subtypes, on dpp7 a significantly higher percentage of CD4 T+ cells in CD26+ pups, and a significantly higher percentage of monocytes in CD26- pups. The percentage of T cells was significantly higher in the CD26-deficient group on each dpp. Thus, daily postnatal exposition to low doses of LPS for 1 week resulted in a delay in formation of secondary septa, which remained up to dpp 14 in CD26- pups. The retardation was accompanied by moderate parenchymal inflammation and CD26-dependent changes in the pulmonary immune cell composition.
Collapse
Affiliation(s)
- Andreas Schmiedl
- Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), 30625, Hannover, Germany.
| | - Inga Wagener
- Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Meike Jungen
- Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg Str. 1, 30625, Hannover, Germany
| | - Stephan von Hörsten
- Department of Experimental Therapy University Hospital Erlangen and Preclinical Experimental Center (PETZ), Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Michael Stephan
- Clinic for Psychosomatics and Psychotherapy, Hannover Medical School, 30625, Hannover, Germany
| |
Collapse
|
26
|
Walweel K, Boon AC, See Hoe LE, Obonyo NG, Pedersen SE, Diab SD, Passmore MR, Hyslop K, Colombo SM, Bartnikowski NJ, Bouquet M, Wells MA, Black DM, Pimenta LP, Stevenson AK, Bisht K, Skeggs K, Marshall L, Prabhu A, James LN, Platts DG, Macdonald PS, McGiffin DC, Suen JY, Fraser JF. Brain stem death induces pro-inflammatory cytokine production and cardiac dysfunction in sheep model. Biomed J 2021; 45:776-787. [PMID: 34666219 PMCID: PMC9661508 DOI: 10.1016/j.bj.2021.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 08/12/2021] [Accepted: 10/07/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Organs procured following brain stem death (BSD) are the main source of organ grafts for transplantation. However, BSD is associated with inflammatory responses that may damage the organ and affect both the quantity and quality of organs available for transplant. Therefore, we aimed to investigate plasma and bronchoalveolar lavage (BAL) pro-inflammatory cytokine profiles and cardiovascular physiology in a clinically relevant 6-h ovine model of BSD. Methods Twelve healthy female sheep (37–42 Kg) were anaesthetized and mechanically ventilated prior to undergoing BSD induction and then monitored for 6 h. Plasma and BAL endothelin-1 and cytokines (IL-1β, 6, 8 and tumour necrosis factor alpha (TNF-α)) were assessed by ELISA. Differential white blood cell counts were performed. Cardiac function during BSD was also examined using echocardiography, and cardiac biomarkers (A-type natriuretic peptide and troponin I were measured in plasma. Results Plasma concentrations big ET-1, IL-6, IL-8, TNF-α and BAL IL-8 were significantly (p < 0.01) increased over baseline at 6 h post-BSD. Increased numbers of neutrophils were observed in the whole blood (3.1 × 109 cells/L [95% confidence interval (CI) 2.06–4.14] vs. 6 × 109 cells/L [95%CI 3.92–7.97]; p < 0.01) and BAL (4.5 × 109 cells/L [95%CI 0.41–9.41] vs. 26 [95%CI 12.29–39.80]; p = 0.03) after 6 h of BSD induction vs baseline. A significant increase in ANP production (20.28 pM [95%CI 16.18–24.37] vs. 78.68 pM [95%CI 53.16–104.21]; p < 0.0001) and cTnI release (0.039 ng/mL vs. 4.26 [95%CI 2.69–5.83] ng/mL; p < 0.0001), associated with a significant reduction in heart contractile function, were observed between baseline and 6 h. Conclusions BSD induced systemic pro-inflammatory responses, characterized by increased neutrophil infiltration and cytokine production in the circulation and BAL fluid, and associated with reduced heart contractile function in ovine model of BSD.
Collapse
Affiliation(s)
- K Walweel
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| | - A C Boon
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L E See Hoe
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - N G Obonyo
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia; Initiative to Develop African Research Leaders, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - S E Pedersen
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - S D Diab
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - M R Passmore
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - K Hyslop
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - S M Colombo
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia; University of Milan, Italy
| | | | - M Bouquet
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - M A Wells
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia; School of Medical Science, Griffith University, Australia
| | - D M Black
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L P Pimenta
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - A K Stevenson
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - K Bisht
- Mater Research Institute, University of Queensland, Australia
| | - K Skeggs
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia; Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
| | - L Marshall
- Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
| | - A Prabhu
- The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L N James
- Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
| | - D G Platts
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - P S Macdonald
- Cardiac Mechanics Research Laboratory, St. Vincent's Hospital and the Victor Chang Cardiac Research Institute, Victoria Street, Darlinghurst, Sydney, Australia
| | - D C McGiffin
- Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Melbourne, Australia
| | - J Y Suen
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| | - J F Fraser
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| |
Collapse
|
27
|
Mühlfeld C, Schulte H, Jansing JC, Casiraghi C, Ricci F, Catozzi C, Ochs M, Salomone F, Brandenberger C. Design-Based Stereology of the Lung in the Hyperoxic Preterm Rabbit Model of Bronchopulmonary Dysplasia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:4293279. [PMID: 34659632 PMCID: PMC8514964 DOI: 10.1155/2021/4293279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/03/2021] [Indexed: 11/17/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a complex condition frequently occurring in preterm newborns, and different animal models are currently used to mimic the pathophysiology of BPD. The comparability of animal models depends on the availability of quantitative data obtained by minimally biased methods. Therefore, the aim of this study was to provide the first design-based stereological analysis of the lungs in the hyperoxia-based model of BPD in the preterm rabbit. Rabbit pups were obtained on gestation day 28 (three days before term) by cesarean section and exposed to normoxic (21% O2, n = 8) or hyperoxic (95% O2, n = 8) conditions. After seven days of exposure, lung function testing was performed, and lungs were taken for stereological analysis. In addition, the ratio between pulmonary arterial acceleration and ejection time (PAAT/PAET) was measured. Inspiratory capacity and static compliance were reduced whereas tissue elastance and resistance were increased in hyperoxic animals compared with normoxic controls. Hyperoxic animals showed signs of pulmonary hypertension indicated by the decreased PAAT/PAET ratio. In hyperoxic animals, the number of alveoli and the alveolar surface area were reduced by one-third or by approximately 50% of control values, respectively. However, neither the mean linear intercept length nor the mean alveolar volume was significantly different between both groups. Hyperoxic pups had thickened alveolar septa and intra-alveolar accumulation of edema fluid and inflammatory cells. Nonparenchymal blood vessels had thickened walls, enlarged perivascular space, and smaller lumen in hyperoxic rabbits in comparison with normoxic ones. In conclusion, the findings are in line with the pathological features of human BPD. The stereological data may serve as a reference to compare this model with BPD models in other species or future therapeutic interventions.
Collapse
Affiliation(s)
- Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Research (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Henri Schulte
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
| | | | - Costanza Casiraghi
- Corporate R&D Preclinical Department, Chiesi Farmaceutici S.p.A, Via Palermo 26/a, 43122 Parma, Italy
| | - Francesca Ricci
- Corporate R&D Preclinical Department, Chiesi Farmaceutici S.p.A, Via Palermo 26/a, 43122 Parma, Italy
| | - Chiara Catozzi
- Corporate R&D Preclinical Department, Chiesi Farmaceutici S.p.A, Via Palermo 26/a, 43122 Parma, Italy
| | - Matthias Ochs
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Institute of Functional Anatomy, Charité-Universitätsmedizin Berlin, Philippstr. 11, 10115 Berlin, Germany
- German Center for Lung Research (DZL), Berlin, Germany
| | - Fabrizio Salomone
- Corporate R&D Preclinical Department, Chiesi Farmaceutici S.p.A, Via Palermo 26/a, 43122 Parma, Italy
| | - Christina Brandenberger
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Research (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
28
|
Devi U, Pandita A. Surfactant delivery via thin catheters: Methods, limitations, and outcomes. Pediatr Pulmonol 2021; 56:3126-3141. [PMID: 34379878 DOI: 10.1002/ppul.25599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 01/08/2023]
Abstract
Various less invasive surfactant administration strategies like surfactant replacement therapy via thin catheters, laryngeal mask airway, pharyngeal instillation, and nebulized surfactant are increasingly being practiced to avoid the harmful effects of endotracheal intubation and ventilation. Numerous studies have been done to study surfactant replacement via thin catheters whereas little data is available for other methods. However, there are variations in premedication policies, type of respiratory support used in these studies. Surfactant delivery using thin catheters has been reported to be associated with decrease in the need for mechanical ventilation (MV), duration of MV, bronchopulmonary dysplasia and neonatal mortality. With the current evidence, among all the available surfactant delivery methods, the one using thin catheters appears to be the most feasible and beneficial to improve clinical neonatal outcomes.
Collapse
Affiliation(s)
- Usha Devi
- Department of Neonatology, Chettinad Hospital and Research Institute, Kelambakkam, Tamil Nadu, India
| | - Aakash Pandita
- Department of Neonatology, SGPGIMS, Lucknow, Uttar Pradesh, India
| |
Collapse
|
29
|
Pioselli B, Salomone F, Mazzola G, Amidani D, Sgarbi E, Amadei F, Murgia X, Catinella S, Villetti G, De Luca D, Carnielli V, Civelli M. Pulmonary surfactant: a unique biomaterial with life-saving therapeutic applications. Curr Med Chem 2021; 29:526-590. [PMID: 34525915 DOI: 10.2174/0929867328666210825110421] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
Pulmonary surfactant is a complex lipoprotein mixture secreted into the alveolar lumen by type 2 pneumocytes, which is composed by tens of different lipids (approximately 90% of its entire mass) and surfactant proteins (approximately 10% of the mass). It is crucially involved in maintaining lung homeostasis by reducing the values of alveolar liquid surface tension close to zero at end-expiration, thereby avoiding the alveolar collapse, and assembling a chemical and physical barrier against inhaled pathogens. A deficient amount of surfactant or its functional inactivation is directly linked to a wide range of lung pathologies, including the neonatal respiratory distress syndrome. This paper reviews the main biophysical concepts of surfactant activity and its inactivation mechanisms, and describes the past, present and future roles of surfactant replacement therapy, focusing on the exogenous surfactant preparations marketed worldwide and new formulations under development. The closing section describes the pulmonary surfactant in the context of drug delivery. Thanks to its peculiar composition, biocompatibility, and alveolar spreading capability, the surfactant may work not only as a shuttle to the branched anatomy of the lung for other drugs but also as a modulator for their release, opening to innovative therapeutic avenues for the treatment of several respiratory diseases.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Sgarbi
- Preclinical R&D, Chiesi Farmaceutici, Parma. Italy
| | | | - Xabi Murgia
- Department of Biotechnology, GAIKER Technology Centre, Zamudio. Spain
| | | | | | - Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, Antoine Béclère Medical Center, APHP, South Paris University Hospitals, Paris, France; Physiopathology and Therapeutic Innovation Unit-U999, South Paris-Saclay University, Paris. France
| | - Virgilio Carnielli
- Division of Neonatology, G Salesi Women and Children's Hospital, Polytechnical University of Marche, Ancona. Italy
| | | |
Collapse
|
30
|
Chia WK, Cheah FC, Abdul Aziz NH, Kampan NC, Shuib S, Khong TY, Tan GC, Wong YP. A Review of Placenta and Umbilical Cord-Derived Stem Cells and the Immunomodulatory Basis of Their Therapeutic Potential in Bronchopulmonary Dysplasia. Front Pediatr 2021; 9:615508. [PMID: 33791258 PMCID: PMC8006350 DOI: 10.3389/fped.2021.615508] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a devastating lung disorder of preterm infants as a result of an aberrant reparative response following exposures to various antenatal and postnatal insults. Despite sophisticated medical treatment in this modern era, the incidence of BPD remains unabated. The current strategies to prevent and treat BPD have met with limited success. The emergence of stem cell therapy may be a potential breakthrough in mitigating this complex chronic lung disorder. Over the last two decades, the human placenta and umbilical cord have gained increasing attention as a highly potential source of stem cells. Placenta-derived stem cells (PDSCs) and umbilical cord-derived stem cells (UCDSCs) display several advantages such as immune tolerance and are generally devoid of ethical constraints, in addition to their stemness qualities. They possess the characteristics of both embryonic and mesenchymal stromal/stem cells. Recently, there are many preclinical studies investigating the use of these cells as therapeutic agents in neonatal disease models for clinical applications. In this review, we describe the preclinical and clinical studies using PDSCs and UCDSCs as treatment in animal models of BPD. The source of these stem cells, routes of administration, and effects on immunomodulation, inflammation and regeneration in the injured lung are also discussed. Lastly, a brief description summarized the completed and ongoing clinical trials using PDSCs and UCDSCs as therapeutic agents in preventing or treating BPD. Due to the complexity of BPD, the development of a safe and efficient therapeutic agent remains a major challenge to both clinicians and researchers.
Collapse
Affiliation(s)
- Wai Kit Chia
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Fook Choe Cheah
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nor Haslinda Abdul Aziz
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nirmala Chandralega Kampan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Salwati Shuib
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Teck Yee Khong
- Department of Pathology, SA Pathology, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Geok Chin Tan
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Yin Ping Wong
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
31
|
Zhu X, Wang F, Lei X, Dong W. Resveratrol alleviates alveolar epithelial cell injury induced by hyperoxia by reducing apoptosis and mitochondrial dysfunction. Exp Biol Med (Maywood) 2020; 246:596-606. [PMID: 33215523 DOI: 10.1177/1535370220975106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Bronchopulmonary dysplasia is a severe and long-term pulmonary disease in premature infants. Hyperoxia-induced acute lung injury plays a critical role in bronchopulmonary dysplasia. Resveratrol is a polyphenolic phytoalexin and a natural agonist of Sirtuin 1. Many studies have shown that resveratrol has a protective effect on hyperoxia-induced lung damage, but its specific protective mechanism is still not clear. Further exploration of the possible protective mechanism of resveratrol was the main goal of this study. In this study, human alveolar epithelial cells were used to establish a hyperoxia-induced acute lung injury cell model, and resveratrol (Res or R), the Sirtuin 1 activator SRT1720 (S) and the Sirtuin 1 inhibitor EX-527 (E) were administered to alveolar epithelial cells, which were then exposed to hyperoxia to investigate the role of Res in mitochondrial function and apoptosis. We divided human alveolar epithelial cells into the following groups: (1) the control group, (2) hyperoxia group, (3) hyperoxia+Res20 group, (4) hyperoxia+Res20+E5 group, (5) hyperoxia+Res20+E10 group, (6) hyperoxia+S2 group, (7) hyperoxia+S2+E5 group, and (8) hyperoxia+S2+E10 group. Hyperoxia-induced cell apoptosis and mitochondrial dysfunction were alleviated by Res and SRT1720. Res and SRT1720 upregulated Sirtuin 1, PGC-1α, NRF1, and TFAM but decreased the expression of acetyl-p53 in human alveolar epithelial cells that were exposed to hyperoxia. These findings revealed that Res may alleviated hyperoxia-induced mitochondrial dysfunction and apoptosis in alveolar epithelial cells through the SIRT1/PGC-1a signaling pathway. Thus, Sirtuin 1 upregulation plays an important role in lung protection.
Collapse
Affiliation(s)
- Xiaodan Zhu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Fan Wang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Xiaoping Lei
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.,Sichuan Clinical Research Center for Birth Defects, Luzhou 646000, China
| |
Collapse
|
32
|
Walweel K, Skeggs K, Boon AC, See Hoe LE, Bouquet M, Obonyo NG, Pedersen SE, Diab SD, Passmore MR, Hyslop K, Wood ES, Reid J, Colombo SM, Bartnikowski NJ, Wells MA, Black D, Pimenta LP, Stevenson AK, Bisht K, Marshall L, Prabhu DA, James L, Platts DG, Macdonald PS, McGiffin DC, Suen JY, Fraser JF. Endothelin receptor antagonist improves donor lung function in an ex vivo perfusion system. J Biomed Sci 2020; 27:96. [PMID: 33008372 PMCID: PMC7532654 DOI: 10.1186/s12929-020-00690-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/24/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND A lung transplant is the last resort treatment for many patients with advanced lung disease. The majority of donated lungs come from donors following brain death (BD). The endothelin axis is upregulated in the blood and lung of the donor after BD resulting in systemic inflammation, lung damage and poor lung graft outcomes in the recipient. Tezosentan (endothelin receptor blocker) improves the pulmonary haemodynamic profile; however, it induces adverse effects on other organs at high doses. Application of ex vivo lung perfusion (EVLP) allows the development of organ-specific hormone resuscitation, to maximise and optimise the donor pool. Therefore, we investigate whether the combination of EVLP and tezosentan administration could improve the quality of donor lungs in a clinically relevant 6-h ovine model of brain stem death (BSD). METHODS After 6 h of BSD, lungs obtained from 12 sheep were divided into two groups, control and tezosentan-treated group, and cannulated for EVLP. The lungs were monitored for 6 h and lung perfusate and tissue samples were processed and analysed. Blood gas variables were measured in perfusate samples as well as total proteins and pro-inflammatory biomarkers, IL-6 and IL-8. Lung tissues were collected at the end of EVLP experiments for histology analysis and wet-dry weight ratio (a measure of oedema). RESULTS Our results showed a significant improvement in gas exchange [elevated partial pressure of oxygen (P = 0.02) and reduced partial pressure of carbon dioxide (P = 0.03)] in tezosentan-treated lungs compared to controls. However, the lungs hematoxylin-eosin staining histology results showed minimum lung injuries and there was no difference between both control and tezosentan-treated lungs. Similarly, IL-6 and IL-8 levels in lung perfusate showed no difference between control and tezosentan-treated lungs throughout the EVLP. Histological and tissue analysis showed a non-significant reduction in wet/dry weight ratio in tezosentan-treated lung tissues (P = 0.09) when compared to control. CONCLUSIONS These data indicate that administration of tezosentan could improve pulmonary gas exchange during EVLP.
Collapse
Affiliation(s)
- K Walweel
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| | - K Skeggs
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - A C Boon
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L E See Hoe
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - M Bouquet
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - N G Obonyo
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,Initiative to Develop African Research Leaders, KEMRI-Wellcome, Trust Research Programme, Kilifi, Kenya
| | - S E Pedersen
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - S D Diab
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - M R Passmore
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - K Hyslop
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - E S Wood
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - J Reid
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - S M Colombo
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,University of Milan, Milan, Italy
| | | | - M A Wells
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,School of Medical Science, Griffith University, Brisbane, Australia
| | - D Black
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L P Pimenta
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - A K Stevenson
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - K Bisht
- Mater Research Institute-The University of Queensland, Woolloongabba, QLD, Australia
| | - L Marshall
- The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - D A Prabhu
- The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L James
- Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - D G Platts
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - P S Macdonald
- Cardiac Mechanics Research Laboratory, St. Vincent's Hospital and the Victor Chang Cardiac Research Institute, Victoria Street, Darlinghurst, Sydney, NSW, 2061, Australia
| | - D C McGiffin
- Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Melbourne, Australia
| | - J Y Suen
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| | - J F Fraser
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| |
Collapse
|
33
|
Zimmermann LJI, Kostenzer J, Mader S. Tackling bronchopulmonary dysplasia to improve preterm health: a call for family-centered care at World Prematurity Day 2020. Am J Physiol Lung Cell Mol Physiol 2020; 319:L867-L870. [PMID: 32936025 DOI: 10.1152/ajplung.00415.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Luc J I Zimmermann
- European Foundation for the Care of Newborn Infants, Munich, Germany.,Department of Pediatrics, School for Oncology and Developmental Biology (GROW), Maastricht UMC+, Maastricht, The Netherlands
| | - Johanna Kostenzer
- European Foundation for the Care of Newborn Infants, Munich, Germany
| | - Silke Mader
- European Foundation for the Care of Newborn Infants, Munich, Germany
| |
Collapse
|
34
|
Shrestha AK, Menon RT, El-Saie A, Barrios R, Reynolds C, Shivanna B. Interactive and independent effects of early lipopolysaccharide and hyperoxia exposure on developing murine lungs. Am J Physiol Lung Cell Mol Physiol 2020; 319:L981-L996. [PMID: 32901520 DOI: 10.1152/ajplung.00013.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH) is a chronic infantile lung disease that lacks curative therapies. Infants with BPD-associated PH are often exposed to hyperoxia and additional insults such as sepsis that contribute to disease pathogenesis. Animal models that simulate these scenarios are necessary to develop effective therapies; therefore, we investigated whether lipopolysaccharide (LPS) and hyperoxia exposure during saccular lung development cooperatively induce experimental BPD-PH in mice. C57BL/6J mice were exposed to normoxia or 70% O2 (hyperoxia) during postnatal days (PNDs) 1-5 and intraperitoneally injected with varying LPS doses or a vehicle on PNDs 3-5. On PND 14, we performed morphometry, echocardiography, and gene and protein expression studies to determine the effects of hyperoxia and LPS on lung development, vascular remodeling and function, inflammation, oxidative stress, cell proliferation, and apoptosis. LPS and hyperoxia independently and cooperatively affected lung development, inflammation, and apoptosis. Growth rate and antioxidant enzyme expression were predominantly affected by LPS and hyperoxia, respectively, while cell proliferation and vascular remodeling and function were mainly affected by combined exposure to LPS and hyperoxia. Mice treated with lower LPS doses developed adaptive responses and hyperoxia exposure did not worsen their BPD phenotype, whereas those mice treated with higher LPS doses displayed the most severe BPD phenotype when exposed to hyperoxia and were the only group that developed PH. Collectively, our data suggest that an additional insult such as LPS may be necessary for models utilizing short-term exposure to moderate hyperoxia to recapitulate human BPD-PH.
Collapse
Affiliation(s)
- Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Ahmed El-Saie
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Corey Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, Texas
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
35
|
Sucre JMS, Vickers KC, Benjamin JT, Plosa EJ, Jetter CS, Cutrone A, Ransom M, Anderson Z, Sheng Q, Fensterheim BA, Ambalavanan N, Millis B, Lee E, Zijlstra A, Königshoff M, Blackwell TS, Guttentag SH. Hyperoxia Injury in the Developing Lung Is Mediated by Mesenchymal Expression of Wnt5A. Am J Respir Crit Care Med 2020; 201:1249-1262. [PMID: 32023086 DOI: 10.1164/rccm.201908-1513oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Rationale: Bronchopulmonary dysplasia (BPD) is a leading complication of preterm birth that affects infants born in the saccular stage of lung development at <32 weeks of gestation. Although the mechanisms driving BPD remain uncertain, exposure to hyperoxia is thought to contribute to disease pathogenesis.Objectives: To determine the effects of hyperoxia on epithelial-mesenchymal interactions and to define the mediators of activated Wnt/β-catenin signaling after hyperoxia injury.Methods: Three hyperoxia models were used: A three-dimensional organotypic coculture using primary human lung cells, precision-cut lung slices (PCLS), and a murine in vivo hyperoxia model. Comparisons of normoxia- and hyperoxia-exposed samples were made by real-time quantitative PCR, RNA in situ hybridization, quantitative confocal microscopy, and lung morphometry.Measurements and Main Results: Examination of an array of Wnt ligands in the three-dimensional organotypic coculture revealed increased mesenchymal expression of WNT5A. Inhibition of Wnt5A abrogated the BPD transcriptomic phenotype induced by hyperoxia. In the PCLS model, Wnt5A inhibition improved alveolarization following hyperoxia exposure, and treatment with recombinant Wnt5a reproduced features of the BPD phenotype in PCLS cultured in normoxic conditions. Chemical inhibition of NF-κB with BAY11-7082 reduced Wnt5a expression in the PCLS hyperoxia model and in vivo mouse hyperoxia model, with improved alveolarization in the PCLS model.Conclusions: Increased mesenchymal Wnt5A during saccular-stage hyperoxia injury contributes to the impaired alveolarization and septal thickening observed in BPD. Precise targeting of Wnt5A may represent a potential therapeutic strategy for the treatment of BPD.
Collapse
Affiliation(s)
- Jennifer M S Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics.,Department of Cell and Developmental Biology, and
| | | | - John T Benjamin
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - Erin J Plosa
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | | | - Alissa Cutrone
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | | | - Benjamin A Fensterheim
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan Millis
- Department of Cell and Developmental Biology, and.,Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee
| | - Ethan Lee
- Department of Cell and Developmental Biology, and
| | | | - Melanie Königshoff
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Colorado; and
| | - Timothy S Blackwell
- Department of Cell and Developmental Biology, and.,Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.,Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | | |
Collapse
|
36
|
Bourguignon C, Vernisse C, Mianné J, Fieldès M, Ahmed E, Petit A, Vachier I, Bertrand TL, Assou S, Bourdin A, De Vos J. [Lung organoids]. Med Sci (Paris) 2020; 36:382-388. [PMID: 32356715 DOI: 10.1051/medsci/2020056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
As burden of chronic respiratory diseases is constantly increasing, improving in vitro lung models is essential in order to reproduce as closely as possible the complex pulmonary architecture, responsible for oxygen uptake and carbon dioxide clearance. The study of diseases that affect the respiratory system has benefited from in vitro reconstructions of the respiratory epithelium with inserts in air/liquid interface (2D) or in organoids able to mimic up to the arborescence of the respiratory tree (3D). Recent development in the fields of pluripotent stem cells-derived organoids and genome editing technologies has provided new insights to better understand pulmonary diseases and to find new therapeutic perspectives.
Collapse
Affiliation(s)
- Chloé Bourguignon
- IRMB, Univ Montpellier, CHU de Montpellier, Hôpital Saint Eloi, Inserm, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Charlotte Vernisse
- PhyMedExp, Univ Montpellier, CHU de Montpellier, Inserm, Montpellier, France
| | - Joffrey Mianné
- IRMB, Univ Montpellier, CHU de Montpellier, Hôpital Saint Eloi, Inserm, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Mathieu Fieldès
- IRMB, Univ Montpellier, CHU de Montpellier, Hôpital Saint Eloi, Inserm, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Engi Ahmed
- IRMB, Univ Montpellier, CHU de Montpellier, Hôpital Saint Eloi, Inserm, 80 avenue Augustin Fliche, 34295 Montpellier, France - Département de pneumologie, CHU de Montpellier, Montpellier, France
| | - Aurélie Petit
- PhyMedExp, Univ Montpellier, CHU de Montpellier, Inserm, Montpellier, France
| | - Isabelle Vachier
- PhyMedExp, Univ Montpellier, CHU de Montpellier, Inserm, Montpellier, France
| | | | - Said Assou
- IRMB, Univ Montpellier, CHU de Montpellier, Hôpital Saint Eloi, Inserm, 80 avenue Augustin Fliche, 34295 Montpellier, France
| | - Arnaud Bourdin
- PhyMedExp, Univ Montpellier, CHU de Montpellier, Inserm, Montpellier, France - Département de pneumologie, CHU de Montpellier, Montpellier, France
| | - John De Vos
- IRMB, Univ Montpellier, CHU de Montpellier, Hôpital Saint Eloi, Inserm, 80 avenue Augustin Fliche, 34295 Montpellier, France - Département d'ingénierie cellulaire et tissulaire, CHU de Montpellier, Montpellier, France
| |
Collapse
|
37
|
Menon RT, Shrestha AK, Barrios R, Reynolds C, Shivanna B. Tie-2 Cre-Mediated Deficiency of Extracellular Signal-Regulated Kinase 2 Potentiates Experimental Bronchopulmonary Dysplasia-Associated Pulmonary Hypertension in Neonatal Mice. Int J Mol Sci 2020; 21:ijms21072408. [PMID: 32244398 PMCID: PMC7177249 DOI: 10.3390/ijms21072408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 01/09/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH) is a significant lung morbidity of infants, and disrupted lung angiogenesis is a hallmark of this disease. We observed that extracellular signal-regulated kinases (ERK) 1/2 support angiogenesis in vitro, and hyperoxia activates ERK1/2 in fetal human pulmonary microvascular endothelial cells (HPMECs) and in neonatal murine lungs; however, their role in experimental BPD and PH is unknown. Therefore, we hypothesized that Tie2 Cre-mediated deficiency of ERK2 in the endothelial cells of neonatal murine lungs would potentiate hyperoxia-induced BPD and PH. We initially determined the role of ERK2 in in vitro angiogenesis using fetal HPMECs. To disrupt endothelial ERK2 signaling in the lungs, we decreased ERK2 expression by breeding ERK2flox/flox mice with Tie-Cre mice. One-day-old endothelial ERK2-sufficient (eERK2+/+) or –deficient (eERK2+/−) mice were exposed to normoxia or hyperoxia (FiO2 70%) for 14 d. We then performed lung morphometry, gene and protein expression studies, and echocardiography to determine the extent of inflammation, oxidative stress, and development of lungs and PH. The knockdown of ERK2 in HPMECs decreased in vitro angiogenesis. Hyperoxia increased lung inflammation and oxidative stress, decreased lung angiogenesis and alveolarization, and induced PH in neonatal mice; however, these effects were augmented in the presence of Tie2-Cre mediated endothelial ERK2 deficiency. Therefore, we conclude that endothelial ERK2 signaling is necessary to mitigate hyperoxia-induced experimental BPD and PH in neonatal mice. Our results indicate that endothelial ERK2 is a potential therapeutic target for the management of BPD and PH in infants.
Collapse
Affiliation(s)
- Renuka T. Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.M.); (A.K.S.)
| | - Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.M.); (A.K.S.)
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA;
| | - Corey Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; (R.T.M.); (A.K.S.)
- Correspondence: ; Tel.: +1-832-824-6474; Fax: +1-832-825-3204
| |
Collapse
|
38
|
Salaets T, Aertgeerts M, Gie A, Vignero J, de Winter D, Regin Y, Jimenez J, Vande Velde G, Allegaert K, Deprest J, Toelen J. Preterm birth impairs postnatal lung development in the neonatal rabbit model. Respir Res 2020; 21:59. [PMID: 32085773 PMCID: PMC7035772 DOI: 10.1186/s12931-020-1321-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/13/2020] [Indexed: 01/10/2023] Open
Abstract
Background Bronchopulmonary dysplasia continues to cause important respiratory morbidity throughout life, and new therapies are needed. The common denominator of all BPD cases is preterm birth, however most preclinical research in this area focusses on the effect of hyperoxia or mechanical ventilation. In this study we investigated if and how prematurity affects lung structure and function in neonatal rabbits. Methods Pups were delivered on either day 28 or day 31. For each gestational age a group of pups was harvested immediately after birth for lung morphometry and surfactant protein B and C quantification. All other pups were hand raised and harvested on day 4 for the term pups and day 7 for the preterm pups (same corrected age) for lung morphometry, lung function testing and qPCR. A subset of pups underwent microCT and dark field imaging on day 0, 2 and 4 for terms and on day 0, 3, 5 and 7 for preterms. Results Preterm pups assessed at birth depicted a more rudimentary lung structure (larger alveoli and thicker septations) and a lower expression of surfactant proteins in comparison to term pups. MicroCT and dark field imaging revealed delayed lung aeration in preterm pups, in comparison to term pups. Preterm birth led to smaller pups, with smaller lungs with a lower alveolar surface area on day 7/day 4. Furthermore, preterm birth affected lung function with increased tissue damping, tissue elastance and resistance and decreased dynamic compliance. Expression of vascular endothelial growth factor (VEGFA) was significantly decreased in preterm pups, however in the absence of structural vascular differences. Conclusions Preterm birth affects lung structure and function at birth, but also has persistent effects on the developing lung. This supports the use of a preterm animal model, such as the preterm rabbit, for preclinical research on BPD. Future research that focuses on the identification of pathways that are involved in in-utero lung development and disrupted by pre-term birth, could lead to novel therapeutic strategies for BPD.
Collapse
Affiliation(s)
- Thomas Salaets
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Margo Aertgeerts
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - André Gie
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - Janne Vignero
- Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Derek de Winter
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - Yannick Regin
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| | - Julio Jimenez
- Facultad de Medicina, Universidad del Desarollo, Clínica Alemana, Santiago de Chile, Chile
| | | | - Karel Allegaert
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium.,Department of Clinical Pharmacy, Erasmus MC Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Jan Deprest
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium.,Institute for Women's Health, University College London Hospital, London, UK
| | - Jaan Toelen
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
39
|
Menon RT, Shrestha AK, Reynolds CL, Barrios R, Caron KM, Shivanna B. Adrenomedullin Is Necessary to Resolve Hyperoxia-Induced Experimental Bronchopulmonary Dysplasia and Pulmonary Hypertension in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:711-722. [PMID: 32093901 DOI: 10.1016/j.ajpath.2019.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 10/29/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
Abstract
Bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH) is an infantile lung disease characterized by aberrant angiogenesis and impaired resolution of lung injury. Adrenomedullin (AM) signals through calcitonin receptor-like receptor and receptor activity-modifying protein 2 and modulates lung injury initiation. However, its role in lung injury resolution and the mechanisms by which it regulates angiogenesis remain unclear. Consequently, we hypothesized that AM resolves hyperoxia-induced BPD and PH via endothelial nitric oxide synthase (NOS3). AM-sufficient (ADM+/+) or -deficient (ADM+/-) mice were exposed to normoxia or hyperoxia through postnatal days (PNDs) 1 to 14, and the hyperoxia-exposed mice were allowed to recover in normoxia for an additional 56 days. Lung injury and development and PH were quantified at different time points. Human pulmonary microvascular endothelial cells were also used to examine the effects of AM signaling on the NOS3 pathway and angiogenesis. Lung blood vessels and NOS3 expression decreased and the extent of hyperoxia-induced BPD and PH increased in ADM+/- mice compared with ADM+/+ mice. Hyperoxia-induced apoptosis and PH resolved by PND14 and PND70, respectively, in ADM+/+ mice but not in ADM+/- mice. Knockdown of ADM, calcitonin receptor-like receptor, and receptor activity-modifying protein 2 in vitro decreased NOS3 expression, nitric oxide generation, and angiogenesis. Furthermore, NOS3 knockdown abrogated the angiogenic effects of AM. Collectively, these results indicate that AM resolves hyperoxic lung injury via NOS3.
Collapse
Affiliation(s)
- Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Corey L Reynolds
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, Texas
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
40
|
Morty RE. Using Experimental Models to Identify Pathogenic Pathways and Putative Disease Management Targets in Bronchopulmonary Dysplasia. Neonatology 2020; 117:233-239. [PMID: 32485712 DOI: 10.1159/000506989] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 11/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is a common and serious complication of preterm birth. Limited pharmacological and other medical interventions are currently available for the management of severely affected, very preterm infants. BPD can be modelled in preclinical studies using experimental animals, and experimental animal models have been extremely valuable in the development of hallmark clinical management strategies for BPD, including pulmonary surfactant replacement and single-course antenatal corticosteroids. A gradual move away from large animal models of BPD in favor of term-born rodents has facilitated the identification of a multitude of new mechanisms of normal and stunted lung development, but this has also potentially limited the utility of experimental animal models for the identification of pathogenic pathways and putative disease management targets in BPD. Indeed, more recent pharmacological interventions for the management of BPD that have been validated in randomized controlled trials have relied very little on preclinical data generated in experimental animal models. While rodent-based models of BPD have tremendous advantages in terms of the availability of genetic tools, they also have considerable drawbacks, including limited utility for studying breathing mechanics, gas exchange, and pulmonary hemodynamics; and they have a less relevant clinical context where lung prematurity and a background of infection are now rarely present in the pathophysiology under study. There is a pressing need to refine existing models to better recapitulate pathological processes at play in affected infants, in order to better evaluate new candidate pharmacological and other interventions for the management of 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
|
41
|
Effect of a new respiratory care bundle on bronchopulmonary dysplasia in preterm neonates. Eur J Pediatr 2020; 179:1833-1842. [PMID: 32488737 PMCID: PMC7266384 DOI: 10.1007/s00431-020-03694-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/15/2020] [Accepted: 05/16/2020] [Indexed: 02/03/2023]
Abstract
The development of devices that can fix the tidal volume in high-frequency oscillatory ventilation (HFOV) has allowed for a significant improvement in the management of HFOV. At our institution, this had led to the earlier use of HFOV and promoted a change in the treatment strategy involving the use of higher frequencies (above 15 Hz) and lower high-frequency tidal volumes (VThf). The purpose of this observational study was to assess how survival without bronchopulmonary dysplasia grades 2 and 3 (SF-BPD) is influenced by these modifications in the respiratory strategy applied to preterm infants (gestational age < 32 weeks at birth) who required mechanical ventilation (MV) in the first 3 days of life. We compared a baseline period (2012-2013) against a period in which this strategy had been fully implemented (2016-2017). A total of 182 patients were exposed to MV in the first 3 days of life being a higher proportion on HFOV at day 3 in the second period 79.5% (n 35) in 2016-2017 vs 55.4% (n 31) in 2012-2013. After adjusting for perinatal risk factors, the second period is associated with an increased rate of SF-BPD (OR 2.28; CI 95% 1.072-4.878); this effect is more evident in neonates born at a gestational age of less than 29 weeks (OR 4.87; 95% CI 1.9-12.48).Conclusions : The early use of HFOV combined with the use of higher frequencies and very low VT was associated with an increase in the study population's SF-BPD. What is Known: • High-frequency ventilation with volume guarantee improve ventilation stability and has been shown to reduce lung damage in animal models. What is New: • The strategy of an earlier use of high-frequency oscillatory ventilation combined with the use of higher frequencies and lower tidal volume is associated to an increase in survival without bronchopulmonary dysplasia in our population of preterm infants.
Collapse
|
42
|
Ruan Y, Dong W, Kang L, Lei X, Zhang R, Wang F, Zhu X. The Changes of Twist1 Pathway in Pulmonary Microvascular Permeability in a Newborn Rat Model of Hyperoxia-Induced Acute Lung Injury. Front Pediatr 2020; 8:190. [PMID: 32391293 PMCID: PMC7190807 DOI: 10.3389/fped.2020.00190] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 03/31/2020] [Indexed: 01/12/2023] Open
Abstract
Background: Bronchopulmonary dysplasia (BPD) is a chronic lung disease in preterm infants, which is characterized by alveolar and vascular dysplasia and increased vascular permeability. Hyperoxia is a critical factor in the pathogenesis of BPD, hyperoxia-induced acute lung injury (HALI) model has similar pathological manifestations as human BPD, therefore, may provide insight into the pathogenesis of human BPD. Studies have shown that Twist1 regulates pulmonary vascular permeability of LPS-induced lung injury through the Ang-Tie2 pathway. However, the effect of Twist1 pathway on vascular permeability in HALI has not been reported. Methods: We randomly exposed newborn rats to the room air or hyperoxia for 14 days. Lung histopathology, immunofluorescence, vascular permeability, mRNA and protein expression was assessed on day 1,7,14. Results: Our results verified that hyperoxia caused alveolar and vascular developmental disorders and increased pulmonary vascular permeability, which was consistent with previous findings. In hyperoxia-exposed rat lungs, the expressions of Twist1, Ang1, Tie1, Tie2, and pTie2 were significantly reduced, whereas the expression of Ang2 was significantly increased. Next, we observed a significant down-regulation of the Akt/Foxo1 pathway. Conclusion: In HALI, the pulmonary microvascular permeability was increased, accompanied by changes in Twist1-Tie2 pathway which combined to Angs, and downregulation of Tie1 and Akt/Foxo1 pathway.
Collapse
Affiliation(s)
- Ying Ruan
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lan Kang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoping Lei
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Zhang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Wang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaodan Zhu
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
43
|
Abstract
In the absence of effective interventions to prevent preterm births, improved survival of infants who are born at the biological limits of viability has relied on advances in perinatal care over the past 50 years. Except for extremely preterm infants with suboptimal perinatal care or major antenatal events that cause severe respiratory failure at birth, most extremely preterm infants now survive, but they often develop chronic lung dysfunction termed bronchopulmonary dysplasia (BPD; also known as chronic lung disease). Despite major efforts to minimize injurious but often life-saving postnatal interventions (such as oxygen, mechanical ventilation and corticosteroids), BPD remains the most frequent complication of extreme preterm birth. BPD is now recognized as the result of an aberrant reparative response to both antenatal injury and repetitive postnatal injury to the developing lungs. Consequently, lung development is markedly impaired, which leads to persistent airway and pulmonary vascular disease that can affect adult lung function. Greater insights into the pathobiology of BPD will provide a better understanding of disease mechanisms and lung repair and regeneration, which will enable the discovery of novel therapeutic targets. In parallel, clinical and translational studies that improve the classification of disease phenotypes and enable early identification of at-risk preterm infants should improve trial design and individualized care to enhance outcomes in preterm infants.
Collapse
|
44
|
Will JP, Hirani D, Thielen F, Klein F, Vohlen C, Dinger K, Dötsch J, Alejandre Alcázar MA. Strain-dependent effects on lung structure, matrix remodeling, and Stat3/Smad2 signaling in C57BL/6N and C57BL/6J mice after neonatal hyperoxia. Am J Physiol Regul Integr Comp Physiol 2019; 317:R169-R181. [PMID: 31067073 DOI: 10.1152/ajpregu.00286.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of preterm infants, characterized by lung growth arrest and matrix remodeling. Various animal models provide mechanistic insights in the pathogenesis of BPD. Since there is increasing evidence that genetic susceptibility modifies the response to lung injury, we investigated strain-dependent effects in hyperoxia (HYX)-induced lung injury of newborn mice. To this end, we exposed newborn C57BL/6N and C57BL/6J mice to 85% O2 (HYX) or normoxia (NOX; 21% O2) for 28 days, followed by lung excision for histological and molecular measurements. BL/6J-NOX mice exhibited a lower body and lung weight than BL/6N-NOX mice; hyperoxia reduced body weight in both strains and increased lung weight only in BL/6J-HYX mice. Quantitative histomorphometric analyses revealed reduced alveolar formation in lungs of both strains after HYX, but the effect was greater in BL/6J-HYX mice than BL/6N-HYX mice. Septal thickness was lower in BL/6J-NOX mice than BL/6N-NOX mice but increased in both strains after HYX. Elastic fiber density was significantly greater in BL/6J-HYX mice than BL/6N-HYX mice. Lungs of BL/6J-HYX mice were protected from changes in gene expression of fibrillin-1, fibrillin-2, fibulin-4, fibulin-5, and surfactant proteins seen in BL/6N-HYX mice. Finally, Stat3 was activated by HYX in both strains; in contrast, activation of Smad2 was markedly greater in lungs of BL/6N mice than BL/6J mice after HYX. In summary, we demonstrate strain-dependent differences in lung structure and matrix, alveolar epithelial cell markers, and Smad2 (transforming growth factor β) signaling in neonatal HYX-induced lung injury. Strain-dependent effects and genetic susceptibility need be taken into consideration for reproducibility and reliability of results in animal models.
Collapse
Affiliation(s)
- Johannes P Will
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Dharmesh Hirani
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Center of Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Thielen
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Fabian Klein
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Christina Vohlen
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Katharina Dinger
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Jörg Dötsch
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany
| | - Miguel A Alejandre Alcázar
- Department of Pediatrics, Translational Experimental Pediatrics, Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne , Cologne , Germany.,Center of Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
45
|
Das B, Jadotte MM, Mills J, Chan KC. Digital Subtraction Pulmonary Angiography in Children with Pulmonary Hypertension due to Bronchopulmonary Dysplasia. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2019; 7:medsci7020026. [PMID: 30743999 PMCID: PMC6410099 DOI: 10.3390/medsci7020026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 01/28/2019] [Accepted: 02/05/2019] [Indexed: 11/16/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common respiratory sequelae of prematurity and histopathologically features fewer, dysmorphic, pulmonary arteries. We present our experience with the digital subtraction pulmonary angiography (DSPA) findings of a segmental vascular filling abnormality in three children who were born at extreme prematurity and have pulmonary hypertension due to severe BPD. Our preliminary data suggest that DSPA may be useful in evaluating the severity of pulmonary vascular disease in children with BPD.
Collapse
Affiliation(s)
- Bibhuti Das
- Joe DiMaggio Children's Hospital Heart Institute, Memorial Healthcare System, 33021 Hollywood, FL, USA.
| | - Michelle-Marie Jadotte
- Joe DiMaggio Children's Hospital Heart Institute, Memorial Healthcare System, 33021 Hollywood, FL, USA.
| | - Jaime Mills
- Joe DiMaggio Children's Hospital Heart Institute, Memorial Healthcare System, 33021 Hollywood, FL, USA.
| | - Kak-Chen Chan
- Joe DiMaggio Children's Hospital Heart Institute, Memorial Healthcare System, 33021 Hollywood, FL, USA.
| |
Collapse
|
46
|
Shrestha AK, Bettini ML, Menon RT, Gopal VYN, Huang S, Edwards DP, Pammi M, Barrios R, Shivanna B. Consequences of early postnatal lipopolysaccharide exposure on developing lungs in mice. Am J Physiol Lung Cell Mol Physiol 2019; 316:L229-L244. [PMID: 30307313 PMCID: PMC6383495 DOI: 10.1152/ajplung.00560.2017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 10/03/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of infants that is characterized by interrupted lung development. Postnatal sepsis causes BPD, yet the contributory mechanisms are unclear. To address this gap, studies have used lipopolysaccharide (LPS) during the alveolar phase of lung development. However, the lungs of infants who develop BPD are still in the saccular phase of development, and the effects of LPS during this phase are poorly characterized. We hypothesized that chronic LPS exposure during the saccular phase disrupts lung development by mechanisms that promote inflammation and prevent optimal lung development and repair. Wild-type C57BL6J mice were intraperitoneally administered 3, 6, or 10 mg/kg of LPS or a vehicle once daily on postnatal days (PNDs) 3-5. The lungs were collected for proteomic and genomic analyses and flow cytometric detection on PND6. The impact of LPS on lung development, cell proliferation, and apoptosis was determined on PND7. Finally, we determined differences in the LPS effects between the saccular and alveolar lungs. LPS decreased the survival and growth rate and lung development in a dose-dependent manner. These effects were associated with a decreased expression of proteins regulating cell proliferation and differentiation and increased expression of those mediating inflammation. While the lung macrophage population of LPS-treated mice increased, the T-regulatory cell population decreased. Furthermore, LPS-induced inflammatory and apoptotic response and interruption of cell proliferation and alveolarization was greater in alveolar than in saccular lungs. Collectively, the data support our hypothesis and reveal several potential therapeutic targets for sepsis-mediated BPD in infants.
Collapse
Affiliation(s)
- Amrit Kumar Shrestha
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Matthew L Bettini
- Section of Diabetes and Endocrinology, Department of Pediatrics, McNair Medical Institute, Baylor College of Medicine , Houston, Texas
| | - Renuka T Menon
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Vashisht Y N Gopal
- Department of Melanoma Medical Oncology and Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center , Houston, Texas
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine , Houston, Texas
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine , Houston, Texas
| | - Mohan Pammi
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital , Houston, Texas
| | - Binoy Shivanna
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
47
|
Bancalari E, Jain D. Bronchopulmonary Dysplasia: 50 Years after the Original Description. Neonatology 2019; 115:384-391. [PMID: 30974430 DOI: 10.1159/000497422] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 11/19/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is one of the few diseases in neonatal medicine that has continued to evolve since its first description about 50 years ago. Over these years, advancements in neonatal medicine such as antenatal steroids and exogenous surfactant therapy have significantly reduced neonatal mortality and lowered the limits of viability for preterm infants. Although the incidence of BPD continues to be high, especially in extremely low birth weight infants, the clinical picture has evolved into a milder disease with low mortality or significant morbidities. This new BPD is the result of complex interactions between altered alveolar and vascular development, injury by ante- and postnatal pathogenic factors, and reparative processes in the lung. There has been significant progress in our understanding of risk factors for BPD, but challenges persist in its definition, and in finding effective preventive strategies. There are promising developments with newer preventive interventions such as mesenchymal stem cells, exosomes, immunomodulators, and growth factors, but they are still in preclinical stage. The future challenges include finding ways to define BPD based on the severity of lung pathology, which can better predict long-term outcomes, development of early predictors of lung disease, and finding innovative and evidence-based preventive and management strategies.
Collapse
Affiliation(s)
- Eduardo Bancalari
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA,
| | - Deepak Jain
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
48
|
Kim I, Kim SS, Song JI, Yoon SH, Park GY, Lee YW. Association between vitamin D level at birth and respiratory morbidities in very-low-birth-weight infants. KOREAN JOURNAL OF PEDIATRICS 2018; 62:166-172. [PMID: 30360037 PMCID: PMC6528057 DOI: 10.3345/kjp.2018.06632] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 10/15/2018] [Indexed: 12/31/2022]
Abstract
Purpose This study aimed to evaluate vitamin D status at birth in very-low-birth-weight infants (VLBWIs: <1,500 g) and to determine the association between vitamin D level and respiratory morbidity. Methods A retrospective study was conducted at Soonchunhyang University Bucheon Hospital between November 2013 and November 2017. We collected blood samples and data on respiratory morbidity from 230 VLBWIs on the first day of life. Patients who were transferred to other hospitals (n=19), died before 36 weeks of gestational age (n=18), or whose blood samples were not collected immediately after birth (n=5) were excluded. Finally, 188 patients were enrolled. VLBWIs with different vitamin D levels were compared with respect to demographic features, maternal diseases, respiratory morbidities, and other neonatal diseases. Results The mean serum vitamin D level, as measured by 25-hydroxyvitamin D (25(OH)D), was 13.4± 9.3 ng/mL. The incidence of vitamin D deficiency (<20 ng/mL) was 79.8%, and 44.1% of preterm infants had severe vitamin D deficiency (<10 ng/mL). Logistic analysis shows that a low serum 25(OH)D level (<20 ng/mL) was a risk factor for respiratory distress syndrome (odds ratio [OR], 4.32; P=0.010) and bronchopulmonary dysplasia (OR, 4.11; P=0.035). Conclusion The results showed that 79.8% of preterm infants in this study had vitamin D deficiency at birth. Low vitamin D status was associated with respiratory morbidity, but the exact mechanism was unknown. Additional studies on the association between vitamin D level and neonatal morbidity are required.
Collapse
Affiliation(s)
- Ian Kim
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Sung Shin Kim
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Jee In Song
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Seock Hwa Yoon
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Ga Young Park
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Yong-Wha Lee
- Department of Laboratory Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| |
Collapse
|
49
|
Sucre JMS, Jetter CS, Loomans H, Williams J, Plosa EJ, Benjamin JT, Young LR, Kropski JA, Calvi CL, Kook S, Wang P, Gleaves L, Eskaros A, Goetzl L, Blackwell TS, Guttentag SH, Zijlstra A. Successful Establishment of Primary Type II Alveolar Epithelium with 3D Organotypic Coculture. Am J Respir Cell Mol Biol 2018; 59:158-166. [PMID: 29625013 PMCID: PMC6096337 DOI: 10.1165/rcmb.2017-0442ma] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 04/06/2018] [Indexed: 12/31/2022] Open
Abstract
Alveolar type II (AT2) epithelial cells are uniquely specialized to produce surfactant in the lung and act as progenitor cells in the process of repair after lung injury. AT2 cell injury has been implicated in several lung diseases, including idiopathic pulmonary fibrosis and bronchopulmonary dysplasia. The inability to maintain primary AT2 cells in culture has been a significant barrier in the investigation of pulmonary biology. We have addressed this knowledge gap by developing a three-dimensional (3D) organotypic coculture using primary human fetal AT2 cells and pulmonary fibroblasts. Grown on top of matrix-embedded fibroblasts, the primary human AT2 cells establish a monolayer and have direct contact with the underlying pulmonary fibroblasts. Unlike conventional two-dimensional (2D) culture, the structural and functional phenotype of the AT2 cells in our 3D organotypic culture was preserved over 7 days of culture, as evidenced by the presence of lamellar bodies and by production of surfactant proteins B and C. Importantly, the AT2 cells in 3D cocultures maintained the ability to replicate, with approximately 60% of AT2 cells staining positive for the proliferation marker Ki67, whereas no such proliferation is evident in 2D cultures of the same primary AT2 cells. This organotypic culture system enables interrogation of AT2 epithelial biology by providing a reductionist in vitro model in which to investigate the response of AT2 epithelial cells and AT2 cell-fibroblast interactions during lung injury and repair.
Collapse
Affiliation(s)
| | | | | | | | - Erin J. Plosa
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - John T. Benjamin
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - Lisa R. Young
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Division of Pulmonary Medicine, Department of Pediatrics, and
| | - Jonathan A. Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Carla L. Calvi
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Seunghyi Kook
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - Ping Wang
- Mildred Stahlman Division of Neonatology, Department of Pediatrics
| | - Linda Gleaves
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
| | - Adel Eskaros
- Program in Cancer Biology
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Laura Goetzl
- Department of Obstetrics and Gynecology, Temple University, Philadelphia, Pennsylvania; and
| | - Timothy S. Blackwell
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | | | - Andries Zijlstra
- Program in Cancer Biology
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
50
|
Hyperoxia Disrupts Extracellular Signal-Regulated Kinases 1/2-Induced Angiogenesis in the Developing Lungs. Int J Mol Sci 2018; 19:ijms19051525. [PMID: 29783779 PMCID: PMC5983575 DOI: 10.3390/ijms19051525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 11/17/2022] Open
Abstract
Hyperoxia contributes to the pathogenesis of bronchopulmonary dysplasia (BPD), a chronic lung disease of infants that is characterized by interrupted alveologenesis. Disrupted angiogenesis inhibits alveologenesis, but the mechanisms of disrupted angiogenesis in the developing lungs are poorly understood. In pre-clinical BPD models, hyperoxia increases the expression of extracellular signal-regulated kinases (ERK) 1/2; however, its effects on the lung endothelial ERK1/2 signaling are unclear. Further, whether ERK1/2 activation promotes lung angiogenesis in infants is unknown. Hence, we tested the following hypotheses: (1) hyperoxia exposure will increase lung endothelial ERK1/2 signaling in neonatal C57BL/6J (WT) mice and in fetal human pulmonary artery endothelial cells (HPAECs); (2) ERK1/2 inhibition will disrupt angiogenesis in vitro by repressing cell cycle progression. In mice, hyperoxia exposure transiently increased lung endothelial ERK1/2 activation at one week of life, before inhibiting it at two weeks of life. Interestingly, hyperoxia-mediated decrease in ERK1/2 activation in mice was associated with decreased angiogenesis and increased endothelial cell apoptosis. Hyperoxia also transiently activated ERK1/2 in HPAECs. ERK1/2 inhibition disrupted angiogenesis in vitro, and these effects were associated with altered levels of proteins that modulate cell cycle progression. Collectively, these findings support our hypotheses, emphasizing that the ERK1/2 pathway is a potential therapeutic target for BPD infants with decreased lung vascularization.
Collapse
|