1
|
Kelchtermans J, March ME, Hakonarson H, McGrath-Morrow SA. Phenotype wide association study links bronchopulmonary dysplasia with eosinophilia in children. Sci Rep 2024; 14:21391. [PMID: 39271728 PMCID: PMC11399246 DOI: 10.1038/s41598-024-72348-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common complication of preterm birth. Despite this, genetic drivers of BPD are poorly understood. The objective of this study is to better understand the impact of single nucleotide polymorphisms (SNPs) previously associated with BPD by examining associations with other phenotypes. We drew pediatric subjects from the biorepository at the Center for Applied Genomics to identify associations between these SNPs and 2,146 imputed phenotypes. Methylation data, external cohorts, and in silico validation methods were used to corroborate significant associations. We identified 60 SNPs that were previously associated with BPD. We found a significant association between rs3771150 and rs3771171 and mean eosinophil percentage in a European cohort of 6,999 patients and replicated this in external cohorts. Both SNPs were also associated with asthma, COPD and FEV1/FVC ratio. These SNPs displayed associations with methylation probes and were functionally linked to ST2 (IL1RL1) levels in blood and lung tissue. Our findings support a genetic justification for the epidemiological link between BPD and asthma. Given the well-established link between ST2 and type 2 inflammation in asthma, these findings provide a rationale for future studies exploring the role of type 2 inflammation in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Jelte Kelchtermans
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA, 19104, USA.
| | - Michael E March
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA, 19104, USA
| | - Sharon A McGrath-Morrow
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA, 19104, USA
| |
Collapse
|
2
|
Sikdar O, Harris C, Greenough A. Improving early diagnosis of bronchopulmonary dysplasia. Expert Rev Respir Med 2024; 18:283-294. [PMID: 38875260 DOI: 10.1080/17476348.2024.2367584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
INTRODUCTION Bronchopulmonary disease (BPD) is associated with long-term neurodevelopmental and cardiorespiratory complications, often requiring significant use of resources. To reduce this healthcare burden, it is essential that those at high risk of BPD are identified early so that strategies are introduced to prevent disease progression. Our aim was to discuss potential methods for improving early diagnosis in the first week after birth. AREAS COVERED A narrative review was undertaken. The search strategy involved searching PubMed, Embase and Cochrane databases from 1967 to 2024. The results of potential biomarkers and imaging modes are discussed. Furthermore, the value of scoring systems is explored. EXPERT OPINION BPD occurs as a result of disruption to pulmonary vascular and alveolar development, thus abnormal levels of factors regulating those processes are promising avenues to explore with regard to early detection of high-risk infants. Data from twin studies suggests genetic factors can be attributed to 82% of the observed difference in moderate to severe BPD, but large genome-wide studies have yielded conflicting results. Comparative studies are required to determine which biomarker or imaging mode may most accurately diagnose early BPD development. Models which include the most predictive factors should be evaluated going forward.
Collapse
Affiliation(s)
- Oishi Sikdar
- Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Christopher Harris
- Neonatal Intensive Care Centre, King's College Hospital NHS Foundation Trust, Denmark Hill, London, UK
| | - Anne Greenough
- Department of Women and Children's Health, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
3
|
Wozniak PS, Makhoul L, Botros MM. Bronchopulmonary dysplasia in adults: Exploring pathogenesis and phenotype. Pediatr Pulmonol 2024; 59:540-551. [PMID: 38050796 DOI: 10.1002/ppul.26795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/06/2023]
Abstract
This review highlights both the longstanding impact of bronchopulmonary dysplasia (BPD) on the health of adult survivors of prematurity and the pressing need for prospective, longitudinal studies of this population. Conservatively, there are an estimated 1,000,000 survivors of BPD in the United States alone. Unfortunately, most of the available literature regarding outcomes of lung disease due to prematurity naturally focuses on pediatric patients in early or middle childhood, and the relative amount of literature on adult survivors is scant. As the number of adult survivors of BPD continues to increase, it is essential that both adult and pediatric pulmonologists have a comprehensive understanding of the pathophysiology and underlying disease process, including the molecular signaling pathways and pro-inflammatory modulators that contribute to the pathogenesis of BPD. We summarize the most common presenting symptoms for adults with BPD and identify the critical challenges adult pulmonologists face in managing the care of survivors of prematurity. Specifically, these challenges include the wide variability of the clinical presentation of adult patients, comorbid cardiopulmonary complications, and the paucity of longitudinal data available on these patients. Adult survivors of BPD have even required lung transplantation, indicating the high burden of morbidity that can result from premature birth and subsequent lung injury. In addition, we analyze the disparate symptoms and management approach to adults with "old" BPD versus "new" BPD. The aim of this review is to assist pulmonologists in understanding the underlying pathophysiology of BPD and to improve clinical recognition of this increasingly common pulmonary disease.
Collapse
Affiliation(s)
- Phillip S Wozniak
- Department of Internal Medicine, Kansas City, Missouri, USA
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri, USA
- University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Lara Makhoul
- University of Missouri Kansas City School of Medicine, Kansas City, Missouri, USA
| | - Mena M Botros
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Houston Methodist Hospital, Houston, Texas, USA
| |
Collapse
|
4
|
Heathfield LJ, Martin LJ, van der Heyde Y, Molefe I, Ramesar R. Clinical exome sequencing elucidates underlying cause of death in sudden unexpected death of infants: two case reports. Int J Legal Med 2024; 138:693-700. [PMID: 37482595 PMCID: PMC10861614 DOI: 10.1007/s00414-023-03065-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Sudden unexpected death in infants (SUDI) is a traumatic event for families, and unfortunately its occurrence remains high in many parts of the world. Whilst cause of death is resolved for most cases, others remain undetermined following postmortem investigations. There has been a recognition of the role of genetic testing in unexplained cases, where previous studies have demonstrated the resolution of cases through DNA analyses. Here we present two case reports of SUDI cases admitted to Salt River Mortuary, South Africa, and show that underlying causes of death were determined for both infants using clinical exome sequencing. The first infant was heterozygous for a variant (rs148175795) in COL6A3, which suggested a bronchopulmonary dysplasia phenotype. This hypothesis led to finding of a second candidate variant in DMP1 (rs142880465), which may contribute towards a digenic/polygenic mechanism of a more severe phenotype. Histological analysis of retained tissue sections showed an asphyxial mechanism of death, where bronchiolar muscle weakness from an underlying bronchopulmonary dysplasia may have contributed to the asphyxia by affecting respiration. In the second infant, a homozygous variant (rs201340753) was identified in MASP1, which was heterozygous in each parent, highlighting the value of including parental DNA in genetic studies. Whilst mannose-binding lectin deficiency could not be assessed, it is plausible that this variant may have acted in combination with other risk factors within the triple-risk model to result in sudden death. These results may have genetic implications for family members, and represent possible new candidate variants for molecular autopsies.
Collapse
Affiliation(s)
- Laura Jane Heathfield
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Lorna Jean Martin
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Yolande van der Heyde
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Itumeleng Molefe
- Division of Forensic Medicine and Toxicology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Raj Ramesar
- MRC/UCT Research Unit for Genomic and Precision Medicine, Division of Human Genetics, Institute of Infectious Diseases and Molecular Medicine, Department of Pathology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
5
|
Kim J, Villarreal M, Arya S, Hernandez A, Moreira A. Bridging the Gap: Exploring Bronchopulmonary Dysplasia through the Lens of Biomedical Informatics. J Clin Med 2024; 13:1077. [PMID: 38398389 PMCID: PMC10889493 DOI: 10.3390/jcm13041077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD), a chronic lung disease predominantly affecting premature infants, poses substantial clinical challenges. This review delves into the promise of biomedical informatics (BMI) in reshaping BPD research and care. We commence by highlighting the escalating prevalence and healthcare impact of BPD, emphasizing the necessity for innovative strategies to comprehend its intricate nature. To this end, we introduce BMI as a potent toolset adept at managing and analyzing extensive, diverse biomedical data. The challenges intrinsic to BPD research are addressed, underscoring the inadequacies of conventional approaches and the compelling need for data-driven solutions. We subsequently explore how BMI can revolutionize BPD research, encompassing genomics and personalized medicine to reveal potential biomarkers and individualized treatment strategies. Predictive analytics emerges as a pivotal facet of BMI, enabling early diagnosis and risk assessment for timely interventions. Moreover, we examine how mobile health technologies facilitate real-time monitoring and enhance patient engagement, ultimately refining BPD management. Ethical and legal considerations surrounding BMI implementation in BPD research are discussed, accentuating issues of privacy, data security, and informed consent. In summation, this review highlights BMI's transformative potential in advancing BPD research, addressing challenges, and opening avenues for personalized medicine and predictive analytics.
Collapse
Affiliation(s)
- Jennifer Kim
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| | - Mariela Villarreal
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| | - Shreyas Arya
- Division of Neonatal-Perinatal Medicine, Dayton Children’s Hospital, Dayton, OH 45404, USA
| | - Antonio Hernandez
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| | - Alvaro Moreira
- Division of Neonatology, Department of Pediatrics, University of Texas Health San Antonio, San Antonio, TX 78229, USA; (J.K.); (M.V.); (A.H.)
| |
Collapse
|
6
|
Brumbaugh JE, Vohr BR, Bell EF, Bann CM, Travers CP, McGowan EC, Harmon HM, Carlo WA, Duncan AF, Hintz SR. Early-Life Outcomes in Relation to Social Determinants of Health for Children Born Extremely Preterm. J Pediatr 2023; 259:113443. [PMID: 37105408 PMCID: PMC10468025 DOI: 10.1016/j.jpeds.2023.113443] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/25/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVE To characterize the relationships between social determinants of health (SDOH) and outcomes for children born extremely preterm. STUDY DESIGN This is a cohort study of infants born at 22-26 weeks of gestation in National Institute of Child Health and Human Development Neonatal Research Network centers (2006-2017) who survived to discharge. Infants were classified by 3 maternal SDOH: education, insurance, and race. Outcomes included postmenstrual age (PMA) at discharge, readmission, neurodevelopmental impairment (NDI), and death postdischarge. Regression analyses adjusted for center, perinatal characteristics, neonatal morbidity, ethnicity, and 2 SDOH (eg, group comparisons by education adjusted for insurance and race). RESULTS Of 7438 children, 5442 (73%) had at least 1 risk-associated SDOH. PMA at discharge was older (adjusted mean difference 0.37 weeks, 95% CL 0.06, 0.68) and readmission more likely (aOR 1.27, 95% CL 1.12, 1.43) for infants whose mothers had public/no insurance vs private. Neither PMA at discharge nor readmission varied by education or race. NDI was twice as likely (aOR 2.36, 95% CL 1.86, 3.00) and death 5 times as likely (aOR 5.22, 95% CL 2.54, 10.73) for infants with 3 risk-associated SDOH compared with those with none. CONCLUSIONS Children born to mothers with public/no insurance were older at discharge and more likely to be readmitted than those born to privately insured mothers. NDI and death postdischarge were more common among children exposed to multiple risk-associated SDOH at birth compared with those not exposed. Addressing disparities due to maternal education, insurance coverage, and systemic racism are potential intervention targets to improve outcomes for children born preterm.
Collapse
Affiliation(s)
- Jane E Brumbaugh
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN.
| | - Betty R Vohr
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Warren Albert Medical School of Brown University, Providence, RI
| | - Edward F Bell
- Department of Pediatrics, University of Iowa, Iowa City, IA
| | - Carla M Bann
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, NC
| | - Colm P Travers
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL
| | - Elisabeth C McGowan
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Warren Albert Medical School of Brown University, Providence, RI
| | - Heidi M Harmon
- Department of Pediatrics, University of Iowa, Iowa City, IA
| | - Waldemar A Carlo
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL
| | - Andrea F Duncan
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Susan R Hintz
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine and Lucile Packard Children's Hospital, Palo Alto, CA
| |
Collapse
|
7
|
Cho HY, Wang X, Campbell MR, Panduri V, Coviello S, Caballero MT, Bennett BD, Kleeberger SR, Polack FP, Ofman G, Bell DA. Prospective epigenome and transcriptome analyses of cord and peripheral blood from preterm infants at risk of bronchopulmonary dysplasia. Sci Rep 2023; 13:12262. [PMID: 37507442 PMCID: PMC10382533 DOI: 10.1038/s41598-023-39313-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/23/2023] [Indexed: 07/30/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a prevalent chronic lung disease of prematurity with limited treatment options. To uncover biomarkers of BPD risk, this study investigated epigenetic and transcriptomic signatures of prematurity at birth and during the neonatal period at day 14 and 28. Peripheral blood DNAs from preterm infants were applied to methylation arrays and cell-type composition was estimated by deconvolution. Covariate-adjusted robust linear regression elucidated BPD- and prolonged oxygen (≥ 14 days) exposure-associated CpGs. RNAs from cord and peripheral blood were sequenced, and differentially expressed genes (DEGs) for BPD or oxygen exposure were determined. Estimated neutrophil-lymphocyte ratios in peripheral blood at day 14 in BPD infants were significantly higher than nonBPD infants, suggesting an heightened inflammatory response in developing BPD. BPD-DEGs in cord blood indicated lymphopoiesis inhibition, altered Th1/Th2 responses, DNA damage, and organ degeneration. On day 14, BPD-associated CpGs were highly enriched in neutrophil activation, infection, and CD4 + T cell quantity, and BPD-DEGs were involved in DNA damage, cellular senescence, T cell homeostasis, and hyper-cytokinesis. On day 28, BPD-associated CpGs along with BPD-DEGs were enriched for phagocytosis, neurological disorder, and nucleotide metabolism. Oxygen supplementation markedly downregulated mitochondrial biogenesis genes and altered CpGs annotated to developmental genes. Prematurity-altered DNA methylation could cause abnormal lymphopoiesis, cellular assembly and cell cycle progression to increase BPD risk. Similar pathways between epigenome and transcriptome networks suggest coordination of the two in dysregulating leukopoiesis, adaptive immunity, and innate immunity. The results provide molecular insights into biomarkers for early detection and prevention of BPD.
Collapse
Affiliation(s)
- Hye-Youn Cho
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Xuting Wang
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Michelle R Campbell
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Vijayalakshmi Panduri
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | | | - Mauricio T Caballero
- Fundación INFANT, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Brian D Bennett
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Steven R Kleeberger
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA
| | - Fernando P Polack
- Fundación INFANT, Buenos Aires, Argentina
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Gaston Ofman
- Fundación INFANT, Buenos Aires, Argentina
- Section of Neonatal-Perinatal Medicine, Center for Pregnancy and Newborn Research, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Douglas A Bell
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Building 101, MD C3-03, 111 TW Alexander Dr., Research Triangle Park, NC, 27709, USA.
| |
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: 4] [Impact Index Per Article: 2.0] [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
|
Dauengauer-Kirlienė S, Domarkienė I, Pilypienė I, Žukauskaitė G, Kučinskas V, Matulevičienė A. Causes of preterm birth: Genetic factors in preterm birth and preterm infant phenotypes. J Obstet Gynaecol Res 2023; 49:781-793. [PMID: 36519629 DOI: 10.1111/jog.15516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
AIM The aim is to provide an overview of recent research on genetic factors that influence preterm birth in the context of neonatal phenotypic assessment. METHODS This is a nonsystematic review of the recent scientific literature. RESULTS Maternal and fetal genetic diversity and rare genome variants are linked with crucial immune response sites. In addition, more frequent in preterm neonates, de novo variants may lead to attention deficits, hyperactivity, autism spectrum disorders, and infertility of both sexes later in life. Environmental factors may also greatly burden fetal, and consequently, neonatal development and neurodevelopment through a failure in the fetal epigenome reprogramming process and even influence the initiation of spontaneous preterm pregnancy termination. Minimally invasive analysis of the transcription factors associated with preterm birth helps elucidate labor mechanisms and predict its timing. We also provide valuable summaries of genomic and transcriptomic factors that contribute to preterm birth. CONCLUSIONS Investigation of the human genome, epigenome, and transcriptome helps to identify molecular mechanisms linked with preterm delivery and premature newborn clinical appearance in early and late neonatal life and even predict developmental outcomes. Further studies are needed to fully understand the implications of genetic changes in preterm births. These data could be used to develop targeted interventions aimed at selecting the most effective individual treatment and rehabilitation plan.
Collapse
Affiliation(s)
- Svetlana Dauengauer-Kirlienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ingrida Domarkienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Ingrida Pilypienė
- Clinic of Children's Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Gabrielė Žukauskaitė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Vaidutis Kučinskas
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Aušra Matulevičienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
10
|
Luo X, Zhao M, Chen C, Lin F, Li X, Huang H, Dou L, Feng J, Xiao S, Liu D, He J, Yu J. Identification of genetic susceptibility in preterm newborns with bronchopulmonary dysplasia by whole-exome sequencing: BIVM gene may play a role. Eur J Pediatr 2023; 182:1707-1718. [PMID: 36757497 PMCID: PMC10167099 DOI: 10.1007/s00431-022-04779-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 02/10/2023]
Abstract
UNLABELLED Bronchopulmonary dysplasia (BPD) is a common chronic respiratory disease in preterm infants caused by multifactorial etiology. Genetic factors are involved in the occurrence of BPD, but studies have found that candidate genes have poor reproducibility and are influenced by ethnic heterogeneity; therefore, more exploration is still needed. We performed whole-exon sequencing in 34 preterm infants with BPD and 32 non-BPD control neonates. The data were analyzed and interpreted by Fisher difference comparison, PLINK and eQTL association analysis, KEGG and GO enrichment analysis, STRING tool, Cytoscape software, ProtParam tool, HOPE online software, and GEOR2 analysis on NCBI GEO dataset. BPD has a highly heterogeneity in different populations, and we found 35 genes overlapped with previous whole-exon sequencing studies, such as APOB gene. Arterial and epithelial cell development and energy metabolism pathways affect BPD. In this study, 24 key genes were identified, and BIVM rs3825519 mutation leads to prolonged assisted ventilation in patients with BPD. A novel DDAH1 mutation site (NM_012137: exon1: c.89 T > G: p.L30R) was found in 9 BPD patients. CONCLUSION BIVM gene rs3825519 mutation may play a role in the pathogenesis of BPD by affecting cilia movement, and the DDAH1 and APOB genes mutations may have a pathogenic role in BPD. WHAT IS KNOWN • Genetic factors are involved in the occurrence of bronchopulmonary dysplasia. • The candidate genes have poor reproducibility and are influenced by ethnic heterogeneity, therefore, more exploration is still needed. WHAT IS NEW • We identified the role of susceptible SNPs in BPD in Shenzhen, China, and identified 24 key genes that influence the pathogenesis of BPD, and also found 35 genes overlapped with previous whole exon sequencing studies, such as APOB gene. • We found that BIVM and DDAH1 genes may play a pathogenic role in the pathogenesis of BPD.
Collapse
Affiliation(s)
- Xi Luo
- Department of Neonatology, 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 Child Infection and Immunity, Chongqing Key Laboratory of Pediatrics, 136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 40014, China
| | - Min Zhao
- Department of Neonatology, 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 Child Infection and Immunity, Chongqing Key Laboratory of Pediatrics, 136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 40014, China
| | - Cheng Chen
- Department of Neonatology, Shenzhen Longgang District Maternity & Child Healthcare Hospital, Shenzhen, 518172, China
| | - Fengji Lin
- Department of Neonatology, Shenzhen Longgang District Maternity & Child Healthcare Hospital, Shenzhen, 518172, China
| | - Xiaodong Li
- Department of Neonatology, Huazhong University of Science and Technology Union Shenzhen Hospital (NanShan Hospital), Shenzhen, 518052, China
| | - Haiyun Huang
- Department of Neonatology, Huazhong University of Science and Technology Union Shenzhen Hospital (NanShan Hospital), Shenzhen, 518052, China
| | - Lei Dou
- Department of Neonatology, Southern University of Science and Technology Hospital, No. 6019 Liuxian Avenue, Xili Street, Nanshan District, Shenzhen, 518055, China
| | - Jinxing Feng
- Department of Neonatology, Shenzhen Children's Hospital, Shenzhen, 518031, China
| | - Shanqiu Xiao
- Department of Neonatology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, 518133, China
| | - Dong Liu
- Department of Neonatology, Shenzhen People's Hospital, Shenzhen, 518020, China
| | - Junli He
- Department of Neonatology, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Jialin Yu
- Department of Neonatology, 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 Child Infection and Immunity, Chongqing Key Laboratory of Pediatrics, 136 Zhongshan 2nd Road, Yuzhong District, Chongqing, 40014, China. .,Department of Neonatology, Southern University of Science and Technology Hospital, No. 6019 Liuxian Avenue, Xili Street, Nanshan District, Shenzhen, 518055, China.
| |
Collapse
|
11
|
Validation of disease-specific biomarkers for the early detection of bronchopulmonary dysplasia. Pediatr Res 2023; 93:625-632. [PMID: 35595912 PMCID: PMC9988689 DOI: 10.1038/s41390-022-02093-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/23/2022] [Accepted: 04/25/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To demonstrate and validate the improvement of current risk stratification for bronchopulmonary dysplasia (BPD) early after birth by plasma protein markers (sialic acid-binding Ig-like lectin 14 (SIGLEC-14), basal cell adhesion molecule (BCAM), angiopoietin-like 3 protein (ANGPTL-3)) in extremely premature infants. METHODS AND RESULTS Proteome screening in first-week-of-life plasma samples of n = 52 preterm infants <32 weeks gestational age (GA) on two proteomic platforms (SomaLogic®, Olink-Proteomics®) confirmed three biomarkers with significant predictive power: BCAM, SIGLEC-14, and ANGPTL-3. We demonstrate high sensitivity (0.92) and specificity (0.86) under consideration of GA, show the proteins' critical contribution to the predictive power of known clinical risk factors, e.g., birth weight and GA, and predicted the duration of mechanical ventilation, oxygen supplementation, as well as neonatal intensive care stay. We confirmed significant predictive power for BPD cases when switching to a clinically applicable method (enzyme-linked immunosorbent assay) in an independent sample set (n = 25, p < 0.001) and demonstrated disease specificity in different cohorts of neonatal and adult lung disease. CONCLUSION While successfully addressing typical challenges of clinical biomarker studies, we demonstrated the potential of BCAM, SIGLEC-14, and ANGPTL-3 to inform future clinical decision making in the preterm infant at risk for BPD. TRIAL REGISTRATION Deutsches Register Klinische Studien (DRKS) No. 00004600; https://www.drks.de . IMPACT The urgent need for biomarkers that enable early decision making and personalized monitoring strategies in preterm infants with BPD is challenged by targeted marker analyses, cohort size, and disease heterogeneity. We demonstrate the potential of the plasma proteins BCAM, SIGLEC-14, and ANGPTL-3 to identify infants with BPD early after birth while improving the predictive power of clinical variables, confirming the robustness toward proteome assays and proving disease specificity. Our comprehensive analysis enables a phase-III clinical trial that allows full implementation of the biomarkers into clinical routine to enable early risk stratification in preterms with BPD.
Collapse
|
12
|
Deolmi M, Decarolis NM, Motta M, Makrinioti H, Fainardi V, Pisi G, Esposito S. Early Origins of Chronic Obstructive Pulmonary Disease: Prenatal and Early Life Risk Factors. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2294. [PMID: 36767660 PMCID: PMC9915555 DOI: 10.3390/ijerph20032294] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
The main risk factor for chronic obstructive pulmonary disease (COPD) is active smoking. However, a considerable amount of people with COPD never smoked, and increasing evidence suggests that adult lung disease can have its origins in prenatal and early life. This article reviews some of the factors that can potentially affect lung development and lung function trajectories throughout the lifespan from genetics and prematurity to respiratory tract infections and childhood asthma. Maternal smoking and air pollution exposure were also analyzed among the environmental factors. The adoption of preventive strategies to avoid these risk factors since the prenatal period may be crucial to prevent, delay the onset or modify the progression of COPD lung disease throughout life.
Collapse
Affiliation(s)
- Michela Deolmi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy
| | - Nicola Mattia Decarolis
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy
| | - Matteo Motta
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy
| | - Heidi Makrinioti
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 01451, USA
| | - Valentina Fainardi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy
| | - Giovanna Pisi
- Cystic Fibrosis Unit, Pediatric Clinic, Az. Ospedaliera-Universitaria di Parma, Via Gramsci 14, 43124 Parma, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW To provide an update on the current understanding of the role of wingless/integrase-1 (Wnt) signaling in pediatric allergic asthma and other pediatric lung diseases. RECENT FINDINGS The Wnt signaling pathway is critical for normal lung development. Genetic and epigenetic human studies indicate a link between Wnt signaling and the development and severity of asthma in children. Mechanistic studies using animal models of allergic asthma demonstrate a key role for Wnt signaling in allergic airway inflammation and remodeling. More recently, data on bronchopulmonary dysplasia (BPD) pathogenesis points to the Wnt signaling pathway as an important regulator. SUMMARY Current data indicates that the Wnt signaling pathway is an important mediator in allergic asthma and BPD pathogenesis. Further studies are needed to characterize the roles of individual Wnt signals in childhood disease, and to identify potential novel therapeutic targets to slow or prevent disease processes.
Collapse
Affiliation(s)
- Nooralam Rai
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Jeanine D’Armiento
- Department of Anesthesiology, Medicine, and Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
14
|
Chung J, Iyengar A, Santry L, Swanson E, Davis JM, Volpe MV. Changes in respiratory management and the impact on bronchopulmonary dysplasia. Pediatr Pulmonol 2022; 57:2327-2334. [PMID: 35673713 DOI: 10.1002/ppul.26035] [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: 11/01/2021] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Noninvasive respiratory support has reduced the need for mechanical ventilation and surfactant administration in very premature neonates. We sought to determine how the increased use of noninvasive ventilation and less surfactant instillation has impacted the development of bronchopulmonary dysplasia (BPD) using four currently used BPD definitions. STUDY DESIGN This is a retrospective, single-center cohort study of neonates born at less than 28 weeks gestation between 2010 and 2018. A respiratory practice change (less surfactant and more noninvasive ventilation) occurred in 2014 following participation in the Surfactant Positive Airway Pressure and Pulse Oximetry trial. Therefore, patients were divided into two epochs to compare postnatal respiratory and clinical course and BPD outcomes across four currently relevant definitions (Vermont Oxford Network, National Institute of Child Health and Human Development, Canadian, and Neonatal Research Network). RESULTS Clinical and demographic variables were similar between epochs. Despite significant differences in maternal and infant characteristics and clinical course, the incidence of BPD was not significantly different between the two epochs regardless of the BPD definition utilized. There was a wide range in the incidence of BPD depending on the definition used. CONCLUSIONS Despite decreased use of surfactant administration and invasive mechanical ventilation between the two epochs, the incidence of BPD did not change and there was wide variation depending on the definition used. A better understanding of the risk factors associated with BPD and a consensus definition is urgently needed to: 1) more accurately compare various studies, 2) help facilitate the conduct of clinical trials, and 3) enhance the development of novel therapeutic interventions to improve outcome.
Collapse
Affiliation(s)
- Jane Chung
- Division of Pediatrics/Newborn Medicine, Tufts Children's Hospital, Boston, Massachusetts, USA.,Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Anjali Iyengar
- Division of Pediatrics/Newborn Medicine, Tufts Children's Hospital, Boston, Massachusetts, USA.,Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Laura Santry
- Division of Pediatrics/Newborn Medicine, Tufts Children's Hospital, Boston, Massachusetts, USA
| | - Eric Swanson
- Division of Pediatrics/Newborn Medicine, Tufts Children's Hospital, Boston, Massachusetts, USA
| | - Jonathan M Davis
- Division of Pediatrics/Newborn Medicine, Tufts Children's Hospital, Boston, Massachusetts, USA.,Tufts University School of Medicine, Boston, Massachusetts, USA
| | - MaryAnn V Volpe
- Division of Pediatrics/Newborn Medicine, Tufts Children's Hospital, Boston, Massachusetts, USA.,Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Chen H, Chen X, Hu L, Ye C, Zhang J, Cheng G, Yang L, Lu Y, Dong X, Zhou W. Rare-variant Collapsing Analyses Identified Risk Genes for Neonatal Acute Respiratory Distress Syndrome. Comput Struct Biotechnol J 2022; 20:5047-5053. [PMID: 36187926 PMCID: PMC9486038 DOI: 10.1016/j.csbj.2022.08.055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/18/2022] [Accepted: 08/27/2022] [Indexed: 11/03/2022] Open
Abstract
Background Results Conclusions
Collapse
|
16
|
Bronchopulmonary dysplasia and wnt pathway-associated single nucleotide polymorphisms. Pediatr Res 2022; 92:888-898. [PMID: 34853430 DOI: 10.1038/s41390-021-01851-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 10/04/2021] [Accepted: 11/02/2021] [Indexed: 11/08/2022]
Abstract
AIM Genetic variants contribute to the pathogenesis of bronchopulmonary dysplasia (BPD). The aim of this study is to evaluate the association of 45 SNPs with BPD susceptibility in a Turkish premature infant cohort. METHODS Infants with gestational age <32 weeks were included. Patients were divided into BPD or no-BPD groups according to oxygen need at 28 days of life, and stratified according to the severity of BPD. We genotyped 45 SNPs, previously identified as BPD risk factors, in 192 infants. RESULTS A total of eight SNPs were associated with BPD risk at allele level, two of which (rs4883955 on KLF12 and rs9953270 on CHST9) were also associated at the genotype level. Functional relationship maps suggested an interaction between five of these genes, converging on WNT5A, a member of the WNT pathway known to be implicated in BPD pathogenesis. Dysfunctional CHST9 and KLF12 variants may contribute to BPD pathogenesis through an interaction with WNT5A. CONCLUSIONS We suggest investigating the role of SNPs on different genes which are in relation with the Wnt pathway in BPD pathogenesis. We identified eight SNPs as risk factors for BPD in this study. In-silico functional maps show an interaction of the genes harboring these SNPs with the WNT pathway, supporting its role in BPD pathogenesis. TRIAL REGISTRATION NCT03467828. IMPACT It is known that genetic factors may contribute to the development of BPD in preterm infants. Further studies are required to identify specific genes that play a role in the BPD pathway to evaluate them as a target for therapeutic interventions. Our study shows an association of BPD predisposition with certain polymorphisms on MBL2, NFKBIA, CEP170, MAGI2, and VEGFA genes at allele level and polymorphisms on CHST9 and KLF12 genes at both allele and genotype level. In-silico functional mapping shows a functional relationship of these five genes with WNT5A, suggesting that Wnt pathway disruption may play a role in BPD pathogenesis.
Collapse
|
17
|
Bush A, Hilgendorff A. Editorial: Bronchopulmonary Dysplasia: Past, Current and Future Pathophysiologic Concepts and Their Contribution to Understanding Lung Disease. Front Med (Lausanne) 2022; 9:922631. [PMID: 35872795 PMCID: PMC9302436 DOI: 10.3389/fmed.2022.922631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Andrew Bush
- Imperial Centre for Paediatrics and Child Health, London, United Kingdom
- National Heart and Lung Institute, London, United Kingdom
- Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | - Anne Hilgendorff
- Center for Comprehensive Developmental Care (CDeC) at the Interdisciplinary Social Pediatric Center, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Ludwig-Maximilians University, Munich, Germany
- Institute for Lung Health and Immunology and Comprehensive Pneumology Center, Helmholtz Zentrum München, Munich, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- *Correspondence: Anne Hilgendorff
| |
Collapse
|
18
|
Gilfillan M, Bhandari V. Moving Bronchopulmonary Dysplasia Research from the Bedside to the Bench. Am J Physiol Lung Cell Mol Physiol 2022; 322:L804-L821. [PMID: 35437999 DOI: 10.1152/ajplung.00452.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although advances in the respiratory management of extremely preterm infants have led to improvements in survival, this progress has not yet extended to a reduction in the incidence of bronchopulmonary dysplasia (BPD). BPD is a complex multifactorial condition that primarily occurs due to disturbances in the regulation of normal pulmonary airspace and vascular development. Preterm birth and exposure to invasive mechanical ventilation also compromises large airway development, leading to significant morbidity and mortality. Although both predisposing and protective genetic and environmental factors have been frequently described in the clinical literature, these findings have had limited impact on the development of effective therapeutic strategies. This gap is likely because the molecular pathways that underlie these observations are yet not fully understood, limiting the ability of researchers to identify novel treatments that can preserve normal lung development and/or enhance cellular repair mechanisms. In this review article, we will outline various well-established clinical observations whilst identifying key knowledge gaps that need to be filled with carefully designed pre-clinical experiments. We will address these issues by discussing controversial topics in the pathophysiology, the pathology and the treatment of BPD, including an evaluation of existing animal models that have been used to answer important questions.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Division of Neonatology, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA
| | - Vineet Bhandari
- Division of Neonatology, The Children's Regional Hospital at Cooper/Cooper Medical School of Rowan University, Camden, NJ
| |
Collapse
|
19
|
Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 2022; 7:3. [PMID: 34980884 PMCID: PMC8724284 DOI: 10.1038/s41392-021-00762-6] [Citation(s) in RCA: 899] [Impact Index Per Article: 299.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis. The deregulation of Wnt/β-catenin signalling often leads to various serious diseases, including cancer and non-cancer diseases. Although many articles have reviewed Wnt/β-catenin from various aspects, a systematic review encompassing the origin, composition, function, and clinical trials of the Wnt/β-catenin signalling pathway in tumour and diseases is lacking. In this article, we comprehensively review the Wnt/β-catenin pathway from the above five aspects in combination with the latest research. Finally, we propose challenges and opportunities for the development of small-molecular compounds targeting the Wnt signalling pathway in disease treatment.
Collapse
|
20
|
Verification of immunology-related genetic associations in BPD supports ABCA3 and five other genes. Pediatr Res 2022; 92:190-198. [PMID: 34465876 PMCID: PMC9411063 DOI: 10.1038/s41390-021-01689-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 07/08/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inflammatory processes are key drivers of bronchopulmonary dysplasia (BPD), a chronic lung disease in preterm infants. In a large sample, we verify previously reported associations of genetic variants of immunology-related genes with BPD. METHODS Preterm infants with a gestational age ≤32 weeks from PROGRESS and the German Neonatal Network (GNN) were included. Through a consensus case/control definition, 278 BPD cases and 670 controls were identified. We identified 49 immunity-related genes and 55 single-nucleotide polymorphisms (SNPs) previously associated with BPD through a comprehensive literature survey. Additionally, a quantitative genetic association analysis regarding oxygen supplements, mechanical ventilation, and continuous positive air pressure (CPAP) was performed. RESULTS Five candidate SNPs were nominally associated with BPD-related phenotypes with effect directions not conflicting the original studies: rs11265269-CRP, rs1427793-NUAK1, rs2229569-SELL, rs1883617-VNN2, and rs4148913-CHST3. Four of these genes are involved in cell adhesion. Extending our analysis to all well-imputed SNPs of all candidate genes, the strongest association was rs45538638-ABCA3 with CPAP (p = 4.9 × 10-7, FDR = 0.004), an ABC transporter involved in surfactant formation. CONCLUSIONS Most of the previously reported associations could not be replicated. We found additional support for SNPs in CRP, NUAK1, SELL, VNN2, and ABCA3. Larger studies and meta-analyses are required to corroborate these findings. IMPACT Larger cohort for improved statistical power to detect genetic associations with bronchopulmonary dysplasia (BPD). Most of the previously reported genetic associations with BPD could not be replicated in this larger study. Among investigated immunological relevant candidate genes, additional support was found for variants in genes CRP, NUAK1, SELL, VNN2, and CHST3, four of them related to cell adhesion. rs45538638 is a novel candidate SNP in reported candidate gene ABC-transporter ABCA3. Results help to prioritize molecular candidate pathomechanisms in follow-up studies.
Collapse
|
21
|
Dai D, Chen H, Dong X, Chen J, Mei M, Lu Y, Yang L, Wu B, Cao Y, Wang J, Zhou W, Qian L. Bronchopulmonary Dysplasia Predicted by Developing a Machine Learning Model of Genetic and Clinical Information. Front Genet 2021; 12:689071. [PMID: 34276789 PMCID: PMC8283015 DOI: 10.3389/fgene.2021.689071] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Background An early and accurate evaluation of the risk of bronchopulmonary dysplasia (BPD) in premature infants is pivotal in implementing preventive strategies. The risk prediction models nowadays for BPD risk that included only clinical factors but without genetic factors are either too complex without practicability or provide poor-to-moderate discrimination. We aim to identify the role of genetic factors in BPD risk prediction early and accurately. Methods Exome sequencing was performed in a cohort of 245 premature infants (gestational age <32 weeks), with 131 BPD infants and 114 infants without BPD as controls. A gene burden test was performed to find risk genes with loss-of-function mutations or missense mutations over-represented in BPD and severe BPD (sBPD) patients, with risk gene sets (RGS) defined as BPD-RGS and sBPD-RGS, respectively. We then developed two predictive models for the risk of BPD and sBPD by integrating patient clinical and genetic features. The performance of the models was evaluated using the area under the receiver operating characteristic curve (AUROC). Results Thirty and 21 genes were included in BPD-RGS and sBPD-RGS, respectively. The predictive model for BPD, which combined the BPD-RGS and basic clinical risk factors, showed better discrimination than the model that was only based on basic clinical features (AUROC, 0.915 vs. AUROC, 0.814, P = 0.013, respectively) in the independent testing dataset. The same was observed in the predictive model for sBPD (AUROC, 0.907 vs. AUROC, 0.826; P = 0.016). Conclusion This study suggests that genetic information contributes to susceptibility to BPD. The predictive model in this study, which combined BPD-RGS with basic clinical risk factors, can thus accurately stratify BPD risk in premature infants.
Collapse
Affiliation(s)
- Dan Dai
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Huiyao Chen
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China
| | - Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China
| | - Jinglong Chen
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Mei Mei
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China
| | - Yulan Lu
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China
| | - Lin Yang
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| | - Bingbing Wu
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Jin Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Wenhao Zhou
- Molecular Medical Center, Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China.,Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Liling Qian
- Division of Pulmonary Medicine, Children's Hospital of Fudan University, Shanghai, China.,Shanghai Key Laboratory of Birth Defects, Shanghai, China
| |
Collapse
|
22
|
Mereness JA, Mariani TJ. The critical role of collagen VI in lung development and chronic lung disease. Matrix Biol Plus 2021; 10:100058. [PMID: 34195595 PMCID: PMC8233475 DOI: 10.1016/j.mbplus.2021.100058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 01/20/2023] Open
Abstract
Type VI collagen (collagen VI) is an obligate extracellular matrix component found mainly in the basement membrane region of many mammalian tissues and organs, including skeletal muscle and throughout the respiratory system. Collagen VI is probably most recognized in medicine as the genetic cause of a spectrum of muscular dystrophies, including Ullrich Congenital Myopathy and Bethlem Myopathy. Collagen VI is thought to contribute to myopathy, at least in part, by mediating muscle fiber integrity by anchoring myoblasts to the muscle basement membrane. Interestingly, collagen VI myopathies present with restrictive respiratory insufficiency, thought to be due primarily to thoracic muscular weakening. Although it was recently recognized as one of the (if not the) most abundant collagens in the mammalian lung, there is a substantive knowledge gap concerning its role in respiratory system development and function. A few studies have suggested that collagen VI insufficiency is associated with airway epithelial cell survival and altered lung function. Our recent work suggested collagen VI may be a genomic risk factor for chronic lung disease in premature infants. Using this as motivation, we thoroughly assessed the role of collagen VI in lung development and in lung epithelial cell biology. Here, we describe the state-of-the-art for collagen VI cell and developmental biology within the respiratory system, and reveal its essential roles in normal developmental processes and airway epithelial cell phenotype and intracellular signaling.
Collapse
Affiliation(s)
- Jared A. Mereness
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, Department of Pediatrics, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Thomas J. Mariani
- Corresponding author. Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, 601 Elmwood Ave, Box 850, Rochester, NY 14642, USA.
| |
Collapse
|
23
|
Liu M, Iosef C, Rao S, Domingo-Gonzalez R, Fu S, Snider P, Conway SJ, Umbach GS, Heilshorn SC, Dewi RE, Dahl MJ, Null DM, Albertine KH, Alvira CM. Transforming Growth Factor-induced Protein Promotes NF-κB-mediated Angiogenesis during Postnatal Lung Development. Am J Respir Cell Mol Biol 2021; 64:318-330. [PMID: 33264084 PMCID: PMC7909333 DOI: 10.1165/rcmb.2020-0153oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022] Open
Abstract
Pulmonary angiogenesis is a key driver of alveolarization. Our prior studies showed that NF-κB promotes pulmonary angiogenesis during early alveolarization. However, the mechanisms regulating temporal-specific NF-κB activation in the pulmonary vasculature are unknown. To identify mechanisms that activate proangiogenic NF-κB signaling in the developing pulmonary vasculature, proteomic analysis of the lung secretome was performed using two-dimensional difference gel electrophoresis. NF-κB activation and angiogenic function was assessed in primary pulmonary endothelial cells (PECs) and TGFBI (transforming growth factor-β-induced protein)-regulated genes identified using RNA sequencing. Alveolarization and pulmonary angiogenesis was assessed in wild-type and Tgfbi null mice exposed to normoxia or hyperoxia. Lung TGFBI expression was determined in premature lambs supported by invasive and noninvasive respiratory support. Secreted factors from the early alveolar, but not the late alveolar or adult lung, promoted proliferation and migration in quiescent, adult PECs. Proteomic analysis identified TGFBI as one protein highly expressed by the early alveolar lung that promoted PEC migration by activating NF-κB via αvβ3 integrins. RNA sequencing identified Csf3 as a TGFBI-regulated gene that enhances nitric oxide production in PECs. Loss of TGFBI in mice exaggerated the impaired pulmonary angiogenesis induced by chronic hyperoxia, and TGFBI expression was disrupted in premature lambs with impaired alveolarization. Our studies identify TGFBI as a developmentally regulated protein that promotes NF-κB-mediated angiogenesis during early alveolarization by enhancing nitric oxide production. We speculate that dysregulation of TGFBI expression may contribute to diseases marked by impaired alveolar and vascular growth.
Collapse
Affiliation(s)
- Min Liu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Cristiana Iosef
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | - Shailaja Rao
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| | | | - Sha Fu
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- Liuyang People’s Hospital, Hunan, China
| | - Paige Snider
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Simon J. Conway
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana
| | - Gray S. Umbach
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
- University of Texas Southwestern Medical School, Dallas, Texas; and
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Ruby E. Dewi
- Department of Materials Science and Engineering, Stanford University, Stanford, California
| | - Mar J. Dahl
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Donald M. Null
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kurt H. Albertine
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Cristina M. Alvira
- Department of Pediatrics, Center for Excellence in Pulmonary Biology, and
| |
Collapse
|
24
|
Lingappan K, Savani RC. The Wnt Signaling Pathway and the Development of Bronchopulmonary Dysplasia. Am J Respir Crit Care Med 2020; 201:1174-1176. [PMID: 32101467 PMCID: PMC7233338 DOI: 10.1164/rccm.202002-0277ed] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Krithika Lingappan
- Division of Neonatal-Perinatal MedicineBaylor College of MedicineHouston, Texasand
| | - Rashmin C Savani
- Division of Neonatal-Perinatal MedicineUniversity of Texas Southwestern Medical CenterDallas, Texas
| |
Collapse
|
25
|
Hadchouel A, Franco-Montoya ML, Guerin S, Do Cruzeiro M, Lhuillier M, Ribeiro Baptista B, Boyer L, Lanone S, Delacourt C. Overexpression of Spock2 in mice leads to altered lung alveolar development and worsens lesions induced by hyperoxia. Am J Physiol Lung Cell Mol Physiol 2020; 319:L71-L81. [PMID: 32374670 DOI: 10.1152/ajplung.00191.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
SPARC/osteonectin, cwcv and kazal-like domains proteoglycan 2 (SPOCK2) was previously associated with genetic susceptibility to bronchopulmonary dysplasia in a French population of very preterm neonates. Its expression increases during lung development and is increased after exposure of rat pups to hyperoxia compared with controls bred in room air. To further investigate the role of SPOCK2 during lung development, we designed two mouse models, one that uses a specific anti-Spock2 antibody and one that reproduces the hyperoxia-induced Spock2 expression with a transgenic mouse model resulting in a conditional and lung-targeted overexpression of Spock2. When mice were bred under hyperoxic conditions, treatment with anti-Spock2 antibodies significantly improved alveolarization. Lung overexpression of Spock2 altered alveolar development in pups bred in room air and worsened hyperoxia-induced lesions. Neither treatment with anti-Spock2 antibody nor overexpression of Spock2 was associated with abnormal activation of matrix metalloproteinase-2. These two models did not alter the expression of known players in alveolar development. This study brings strong arguments for the deleterious role of SPOCK2 on lung alveolar development especially after lung injury, suggesting its role in bronchopulmonary dysplasia susceptibility. These effects are not mediated by a deregulation in metalloproteases activity and in expression of factors essential to normal alveolarization. The balance between types 1 and 2 epithelial alveolar cells may be involved.
Collapse
Affiliation(s)
- Alice Hadchouel
- Service de Pneumologie et d'Allergologie Pédiatriques, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris, Paris, France.,Equipe 4, U955, Institut National de la Santé et de la Recherche Médicale, Créteil, France.,Université de Paris, Paris, France
| | - Marie-Laure Franco-Montoya
- Service de Pneumologie et d'Allergologie Pédiatriques, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Sophie Guerin
- Service de Pneumologie et d'Allergologie Pédiatriques, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris, Paris, France.,Equipe 4, U955, Institut National de la Santé et de la Recherche Médicale, Créteil, France
| | - Marcio Do Cruzeiro
- Homologous Recombination, Cochin Institute, Université de Paris, Paris, France
| | - Mickaël Lhuillier
- U1151, Institut National de la Santé et de la Recherche Médicale, Institut Necker-Enfants Malades, Université de Paris, Paris, France
| | - Bruno Ribeiro Baptista
- Equipe 4, U955, Institut National de la Santé et de la Recherche Médicale, Créteil, France
| | - Laurent Boyer
- Equipe 4, U955, Institut National de la Santé et de la Recherche Médicale, Créteil, France
| | - Sophie Lanone
- Equipe 4, U955, Institut National de la Santé et de la Recherche Médicale, Créteil, France
| | - Christophe Delacourt
- Service de Pneumologie et d'Allergologie Pédiatriques, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique - Hôpitaux de Paris, Paris, France.,Equipe 4, U955, Institut National de la Santé et de la Recherche Médicale, Créteil, France.,Université de Paris, Paris, France
| |
Collapse
|
26
|
Exome sequencing of extreme phenotypes in bronchopulmonary dysplasia. Eur J Pediatr 2020; 179:579-586. [PMID: 31848748 DOI: 10.1007/s00431-019-03535-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/19/2019] [Accepted: 11/24/2019] [Indexed: 01/08/2023]
Abstract
Bronchopulmonary dysplasia is the most common chronic respiratory disease in premature infants with growing evidence that genetic factors contribute largely to moderate and severe cases. We assessed by exome sequencing if rare genetic variants could account for extremely severe phenotypes. We selected 6 infants born very preterm with severe bronchopulmonary dysplasia and 8 very preterm born controls for exome sequencing. We filtered whole exome sequencing results to include only rare variants and selected variants and/or genes with variants that were present in at least 2 cases and absent in controls. We selected variants, all heterozygous, in 9 candidate genes, 7 with a putative role in lung development and 2 that displayed 3 variations in 3 different cases, independently of their potential role in lung development. Sequencing of 5 other severe cases for these variants did not replicate our results.Conclusion: In selected preterm born infants with severe bronchopulmonary dysplasia and controls, we failed to find any rare variant shared by several infants with an extremely severe phenotype. Our results are not consistent with the role of rare causative variants in bronchopulmonary dysplasia's development and argue for the highly polygenic nature of susceptibility of this disorder.What is Known:• Bronchopulmonary dysplasia is a multifactorial disease resulting from complex environmental and genetic interactions occurring in an immature lung.• It is not known whether rare genetic variants in coding regions could account for extreme phenotypes of the disease.What is New:• In a group of infants with an extreme phenotype of bronchopulmonary dysplasia and in comparison to controls, no common genetic variants were found, nor did variants that were select in other exome studies in this setting.• These results argue for the highly polygenic nature of susceptibility of bronchopulmonary dysplasia.
Collapse
|
27
|
Ryan FJ, Drew DP, Douglas C, Leong LEX, Moldovan M, Lynn M, Fink N, Sribnaia A, Penttila I, McPhee AJ, Collins CT, Makrides M, Gibson RA, Rogers GB, Lynn DJ. Changes in the Composition of the Gut Microbiota and the Blood Transcriptome in Preterm Infants at Less than 29 Weeks Gestation Diagnosed with Bronchopulmonary Dysplasia. mSystems 2019; 4:e00484-19. [PMID: 31662429 PMCID: PMC6819732 DOI: 10.1128/msystems.00484-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/09/2019] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a common chronic lung condition in preterm infants that results in abnormal lung development and leads to considerable morbidity and mortality, making BPD one of the most common complications of preterm birth. We employed RNA sequencing and 16S rRNA gene sequencing to profile gene expression in blood and the composition of the fecal microbiota in infants born at <29 weeks gestational age and diagnosed with BPD in comparison to those of preterm infants that were not diagnosed with BPD. 16S rRNA gene sequencing, performed longitudinally on 255 fecal samples collected from 50 infants in the first months of life, identified significant differences in the relative levels of abundance of Klebsiella, Salmonella, Escherichia/Shigella, and Bifidobacterium in the BPD infants in a manner that was birth mode dependent. Transcriptome sequencing (RNA-Seq) analysis revealed that more than 400 genes were upregulated in infants with BPD. Genes upregulated in BPD infants were significantly enriched for functions related to red blood cell development and oxygen transport, while several immune-related pathways were downregulated. We also identified a gene expression signature consistent with an enrichment of immunosuppressive CD71+ early erythroid cells in infants with BPD. Intriguingly, genes that were correlated in their expression with the relative abundances of specific taxa in the microbiota were significantly enriched for roles in the immune system, suggesting that changes in the microbiota might influence immune gene expression systemically.IMPORTANCE Bronchopulmonary dysplasia (BPD) is a serious inflammatory condition of the lung and is the most common complication associated with preterm birth. A large body of evidence now suggests that the gut microbiota can influence immunity and inflammation systemically; however, the role of the gut microbiota in BPD has not been evaluated to date. Here, we report that there are significant differences in the gut microbiota of infants born at <29 weeks gestation and subsequently diagnosed with BPD, which are particularly pronounced when infants are stratified by birth mode. We also show that erythroid and immune gene expression levels are significantly altered in BPD infants. Interestingly, we identified an association between the composition of the microbiota and immune gene expression in blood in early life. Together, these findings suggest that the composition of the microbiota may influence the risk of developing BPD and, more generally, may shape systemic immune gene expression.
Collapse
Affiliation(s)
- Feargal J Ryan
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Damian P Drew
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Chloe Douglas
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lex E X Leong
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Max Moldovan
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Miriam Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Naomi Fink
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anastasia Sribnaia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Irmeli Penttila
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Andrew J McPhee
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Neonatal Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Carmel T Collins
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Maria Makrides
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert A Gibson
- SAHMRI Women and Kids, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- School of Agriculture, Food, and Wine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Geraint B Rogers
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - David J Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
28
|
Respiratory Phenotypes for Preterm Infants, Children, and Adults: Bronchopulmonary Dysplasia and More. Ann Am Thorac Soc 2019; 15:530-538. [PMID: 29328889 DOI: 10.1513/annalsats.201709-756fr] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ongoing advancements in neonatal care since the late 1980s have led to increased numbers of premature infants surviving well beyond the neonatal period. As a result of increased survival, many individuals born preterm manifest chronic respiratory symptoms throughout infancy, childhood, and adult life. The archetypical respiratory disease of prematurity, bronchopulmonary dysplasia, is the second most common chronic pediatric respiratory disease after asthma. However, there are several commonly held misconceptions. These misconceptions include that bronchopulmonary dysplasia is rare, that bronchopulmonary dysplasia resolves within the first few years of life, and that bronchopulmonary dysplasia does not impact respiratory health in adult life. This focused review describes a spectrum of respiratory conditions that individuals born prematurely may experience throughout their lifespan. Specifically, this review provides quantitative estimates of the number of individuals with alveolar, airway, and vascular phenotypes associated with bronchopulmonary dysplasia, as well as non-bronchopulmonary dysplasia respiratory phenotypes such as airway malacia, obstructive sleep apnea, and control of breathing issues. Furthermore, this review illustrates what is known about the potential for progression and/or lack of resolution of these respiratory phenotypes in childhood and adult life. Recognizing the spectrum of respiratory phenotypes associated with individuals born preterm and providing comprehensive and personalized care to these individuals may help to modulate adverse respiratory outcomes in later life.
Collapse
|
29
|
Danopoulos S, Shiosaki J, Al Alam D. FGF Signaling in Lung Development and Disease: Human Versus Mouse. Front Genet 2019; 10:170. [PMID: 30930931 PMCID: PMC6423913 DOI: 10.3389/fgene.2019.00170] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/15/2019] [Indexed: 12/30/2022] Open
Abstract
Fibroblast growth factor 10 (FGF10) plays an important role in mouse lung development, injury, and repair. It is considered the main morphogen driving lung branching morphogenesis in rodents. While many studies have found FGF10 SNPs associated with COPD and branch variants in COPD smokers, there is no evidence of a causative role for FGF10 or these SNPs in human lung development and pediatric lung diseases. We and others have shown divergent roles for FGF10 in mouse lung development and early human lung development. Herein, we only review the existing literature on FGF signaling in human lung development and pediatric human lung diseases, comparing what is known in mouse lung to that in human lung.
Collapse
Affiliation(s)
- Soula Danopoulos
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Jessica Shiosaki
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Denise Al Alam
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
30
|
Preventing bronchopulmonary dysplasia: new tools for an old challenge. Pediatr Res 2019; 85:432-441. [PMID: 30464331 DOI: 10.1038/s41390-018-0228-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/12/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) is the most prevalent chronic lung disease in infants and presents as a consequence of preterm birth. Due to the lack of effective preventive and treatment strategies, BPD currently represents a major therapeutic challenge that requires continued research efforts at the basic, translational, and clinical levels. However, not all very low birth weight premature babies develop BPD, which suggests that in addition to known gestational age and intrauterine and extrauterine risk factors, other unknown factors must be involved in this disease's development. One of the main goals in BPD research is the early prediction of very low birth weight infants who are at risk of developing BPD in order to initiate the adequate preventive strategies. Other benefits of determining the risk of BPD include providing prognostic information and stratifying infants for clinical trial enrollment. In this article, we describe new opportunities to address BPD's complex pathophysiology by identifying prognostic biomarkers and develop novel, complex in vitro human lung models in order to develop effective therapies. These therapies for protecting the immature lung from injury can be developed by taking advantage of recent scientific progress in -omics, 3D organoids, and regenerative medicine.
Collapse
|
31
|
McGrath-Morrow SA, Collaco JM. Bronchopulmonary dysplasia: what are its links to COPD? Ther Adv Respir Dis 2019; 13:1753466619892492. [PMID: 31818194 PMCID: PMC6904782 DOI: 10.1177/1753466619892492] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/13/2019] [Indexed: 12/23/2022] Open
Abstract
Emerging evidence suggests that adverse early life events can affect long-term health trajectories throughout life. Preterm birth, in particular, is a significant early life event that affects approximately 10% of live births. Worldwide, prematurity is the number one cause of death in children less than 5 years of age and has been shown to disrupt normal lung development with lasting effects into adult life. Along with impaired lung development, interventions used to support gas exchange and other sequelae of prematurity can lead to the development of bronchopulmonary dysplasia (BPD). BPD is a chronic respiratory disease of infancy characterized by alveolar simplification, small airways disease, and pulmonary vascular changes. Although many survivors of BPD improve with age, survivors of BPD often have chronic lung disease characterized by airflow obstruction and intermittent pulmonary exacerbations. Long-term lung function trajectories as measured by FEV1 can be lower in children and adults with a history BPD. In this review, we discuss the epidemiology and manifestations of BPD and its long-term consequences throughout childhood and into adulthood. Available evidence suggests that disrupted lung development, genetic susceptibility and subsequent environment and infectious events that occur in prenatal and postnatal life likely increase the predisposition of children with BPD to develop early onset chronic obstructive pulmonary disease (COPD). The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Sharon A. McGrath-Morrow
- Eudowood Division of Pediatric Respiratory
Sciences, David M. Rubenstein Building, Suite 3075B, 200 North Wolfe Street,
Baltimore, MD, 21287-2533, USA
| | - Joseph M. Collaco
- Department of Pediatrics, Eudowood Division of
Respiratory Sciences, Johns Hopkins University School of Medicine,
Baltimore, MD, USA
| |
Collapse
|
32
|
High-Throughput Sequencing in Respiratory, Critical Care, and Sleep Medicine Research. An Official American Thoracic Society Workshop Report. Ann Am Thorac Soc 2019; 16:1-16. [PMID: 30592451 PMCID: PMC6812157 DOI: 10.1513/annalsats.201810-716ws] [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] [Indexed: 12/30/2022] Open
Abstract
High-throughput, "next-generation" sequencing methods are now being broadly applied across all fields of biomedical research, including respiratory disease, critical care, and sleep medicine. Although there are numerous review articles and best practice guidelines related to sequencing methods and data analysis, there are fewer resources summarizing issues related to study design and interpretation, especially as applied to common, complex, nonmalignant diseases. To address these gaps, a single-day workshop was held at the American Thoracic Society meeting in May 2017, led by the American Thoracic Society Section on Genetics and Genomics. The aim of this workshop was to review the design, analysis, interpretation, and functional follow-up of high-throughput sequencing studies in respiratory, critical care, and sleep medicine research. This workshop brought together experts in multiple fields, including genetic epidemiology, biobanking, bioinformatics, and research ethics, along with physician-scientists with expertise in a range of relevant diseases. The workshop focused on application of DNA and RNA sequencing research in common chronic diseases and did not cover sequencing studies in lung cancer, monogenic diseases (e.g., cystic fibrosis), or microbiome sequencing. Participants reviewed and discussed study design, data analysis and presentation, interpretation, functional follow-up, and reporting of results. This report summarizes the main conclusions of the workshop, specifically addressing the application of these methods in respiratory, critical care, and sleep medicine research. This workshop report may serve as a resource for our research community as well as for journal editors and reviewers of sequencing-based manuscript submissions in our research field.
Collapse
|
33
|
Parad RB, Winston AB, Kalish LA, Gupta M, Thompson I, Sheldon Y, Morey J, Van Marter LJ. Role of Genetic Susceptibility in the Development of Bronchopulmonary Dysplasia. J Pediatr 2018; 203:234-241.e2. [PMID: 30287068 PMCID: PMC8516345 DOI: 10.1016/j.jpeds.2018.07.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 07/17/2018] [Accepted: 07/27/2018] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To assess heritable contributions to bronchopulmonary dysplasia (BPD) risk in a twin cohort restricted to gestational age at birth <29 weeks. STUDY DESIGN A total of 250 twin pairs (192 dichorionic, 58 monochorionic) born <29 weeks gestational age with known BPD status were identified. Three statistical methods applicable to twin cohorts (χ2 test, intraclass correlations [ICCs], and ACE modeling [additive genetic or A, common environmental or C, and unique environmental or E components]) were applied. Heritability was estimated as percent variability from A. Identical methods were applied to a subcohort defined by zygosity and to an independent validation cohort. RESULTS χ2 analyses comparing whether neither, 1, or both of monochorionic (23, 19, 16) and dichorionic (88, 56, 48) twin pairs developed BPD revealed no difference. Although there was similarity in BPD outcome within both monochorionic and dichorionic twin pairs by ICC (monochorionic ICC = 0.34, 95% CI [0.08, 0.55]; dichorionic ICC = 0.39, 95% CI [0.25, 0.51]), monochorionic twins were not more likely than dichorionic twins to have the same outcome (P = .70). ACE modeling revealed no contribution of heritability to BPD risk (% A = 0.0%, 95% CI [0.0%, 43.1%]). Validation and zygosity based cohort results were similar. CONCLUSIONS Our analysis suggests that heritability is not a major contributor to BPD risk in preterm infants <29 weeks gestational age.
Collapse
Affiliation(s)
- Richard B Parad
- Brigham and Women's Hospital, Boston, MA; Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA.
| | | | - Leslie A Kalish
- Boston Children’s Hospital, Boston, MA,Harvard Medical School, Boston, MA
| | - Munish Gupta
- Boston Children’s Hospital, Boston, MA,Beth Israel Deaconess Medical Center, Boston, MA,Harvard Medical School, Boston, MA
| | | | | | | | - Linda J Van Marter
- Brigham and Women’s Hospital, Boston, MA,Boston Children’s Hospital, Boston, MA,Harvard Medical School, Boston, MA
| |
Collapse
|
34
|
Torgerson DG, Ballard PL, Keller RL, Oh SS, Huntsman S, Hu D, Eng C, Burchard EG, Ballard RA. Ancestry and genetic associations with bronchopulmonary dysplasia in preterm infants. Am J Physiol Lung Cell Mol Physiol 2018; 315:L858-L869. [PMID: 30113228 PMCID: PMC6295513 DOI: 10.1152/ajplung.00073.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 01/19/2023] Open
Abstract
Bronchopulmonary dysplasia in premature infants is a common and often severe lung disease with long-term sequelae. A genetic component is suspected but not fully defined. We performed an ancestry and genome-wide association study to identify variants, genes, and pathways associated with survival without bronchopulmonary dysplasia in 387 high-risk infants treated with inhaled nitric oxide in the Trial of Late Surfactant study. Global African genetic ancestry was associated with increased survival without bronchopulmonary dysplasia among infants of maternal self-reported Hispanic white race/ethnicity [odds ratio (OR) = 4.5, P = 0.01]. Admixture mapping found suggestive outcome associations with local African ancestry at chromosome bands 18q21 and 10q22 among infants of maternal self-reported African-American race/ethnicity. For all infants, the top individual variant identified was within the intron of NBL1, which is expressed in midtrimester lung and is an antagonist of bone morphogenetic proteins ( rs372271081 , OR = 0.17, P = 7.4 × 10-7). The protective allele of this variant was significantly associated with lower nitric oxide metabolites in the urine of non-Hispanic white infants ( P = 0.006), supporting a role in the racial differential response to nitric oxide. Interrogating genes upregulated in bronchopulmonary dysplasia lungs indicated association with variants in CCL18, a cytokine associated with fibrosis and interstitial lung disease, and pathway analyses implicated variation in genes involved in immune/inflammatory processes in response to infection and mechanical ventilation. Our results suggest that genetic variation related to lung development, drug metabolism, and immune response contribute to individual and racial/ethnic differences in respiratory outcomes following inhaled nitric oxide treatment of high-risk premature infants.
Collapse
Affiliation(s)
- Dara G Torgerson
- Department of Pediatrics, University of California , San Francisco, California
| | - Philip L Ballard
- Department of Pediatrics, University of California , San Francisco, California
| | - Roberta L Keller
- Department of Pediatrics, University of California , San Francisco, California
| | - Sam S Oh
- Department of Medicine, University of California , San Francisco, California
| | - Scott Huntsman
- Department of Medicine, University of California , San Francisco, California
| | - Donglei Hu
- Department of Medicine, University of California , San Francisco, California
| | - Celeste Eng
- Department of Medicine, University of California , San Francisco, California
| | - Esteban G Burchard
- Department of Medicine, University of California , San Francisco, California
- Department of Bioengineering and Therapeutic Sciences, University of California , San Francisco, California
| | - Roberta A Ballard
- Department of Pediatrics, University of California , San Francisco, California
| |
Collapse
|
35
|
Abstract
Bronchopulmonary Dysplasia (BPD) is a disorder with a multifactorial etiology and highly variable clinical phenotype. Several traditional biomarkers have been identified, but due to the complex disease phenotype, these biomarkers have low predictive accuracy for BPD. In recent years, newer technologies have facilitated the in-depth and unbiased analysis of 'big data' in delineating the diagnosis, pathogenesis, and mechanisms of diseases. Novel systems-biology based 'omic' approaches, including but not limited to genomics, microbiomics, proteomics, and metabolomics may help define the multiple cellular and humoral interactions that regulate normal as well as abnormal lung development and response to injury that are the hallmarks of BPD.
Collapse
Affiliation(s)
- Charitharth Vivek Lal
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Women and Infants Center, 176F Suite 9380, 619 South 19th Street, Birmingham, AL 35249-7335, United States.
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University, Philadelphia, PA, United States
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Women and Infants Center, 176F Suite 9380, 619 South 19th Street, Birmingham, AL 35249-7335, United States
| |
Collapse
|
36
|
Hamvas A, Feng R, Bi Y, Wang F, Bhattacharya S, Mereness J, Kaushal M, Cotten CM, Ballard PL, Mariani TJ. Exome sequencing identifies gene variants and networks associated with extreme respiratory outcomes following preterm birth. BMC Genet 2018; 19:94. [PMID: 30342483 PMCID: PMC6195962 DOI: 10.1186/s12863-018-0679-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 10/01/2018] [Indexed: 12/28/2022] Open
Abstract
Background Previous studies have identified genetic variants associated with bronchopulmonary dysplasia (BPD) in extremely preterm infants. However, findings with genome-wide significance have been rare, and not replicated. We hypothesized that whole exome sequencing (WES) of premature subjects with extremely divergent phenotypic outcomes could facilitate the identification of genetic variants or gene networks contributing disease risk. Results The Prematurity and Respiratory Outcomes Program (PROP) recruited a cohort of > 765 extremely preterm infants for the identification of markers of respiratory morbidity. We completed WES on 146 PROP subjects (85 affected, 61 unaffected) representing extreme phenotypes of early respiratory morbidity. We tested for association between disease status and individual common variants, screened for rare variants exclusive to either affected or unaffected subjects, and tested the combined association of variants across gene loci. Pathway analysis was performed and disease-related expression patterns were assessed. Marginal association with BPD was observed for numerous common and rare variants. We identified 345 genes with variants unique to BPD-affected preterm subjects, and 292 genes with variants unique to our unaffected preterm subjects. Of these unique variants, 28 (19 in the affected cohort and 9 in unaffected cohort) replicate a prior WES study of BPD-associated variants. Pathway analysis of sets of variants, informed by disease-related gene expression, implicated protein kinase A, MAPK and Neuregulin/epidermal growth factor receptor signaling. Conclusions We identified novel genes and associated pathways that may play an important role in susceptibility/resilience for the development of lung disease in preterm infants. Electronic supplementary material The online version of this article (10.1186/s12863-018-0679-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aaron Hamvas
- Department of Pediatrics, Northwestern University, Chicago, IL, USA. .,Ann and Robert H. Lurie Children's Hospital of Chicago and Northwestern University, Chicago, IL, USA.
| | - Rui Feng
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yingtao Bi
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Fan Wang
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Jared Mereness
- Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Madhurima Kaushal
- Center for Biomedical Informatics, Washington University, St. Louis, MO, USA
| | | | - Philip L Ballard
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Thomas J Mariani
- Department of Pediatrics, University of Rochester, Rochester, NY, USA. .,Division of Neonatology and Pediatric Molecular and Personalized Medicine Program University of Rochester Medical Center, 601 Elmwood Ave, Box 850, Rochester, NY, 14642, USA.
| | | |
Collapse
|
37
|
Abstract
Blood vessels are essential for blood circulation but also control organ growth, homeostasis, and regeneration, which has been attributed to the release of paracrine signals by endothelial cells. Endothelial tubules are associated with specialised mesenchymal cells, termed pericytes, which help to maintain vessel wall integrity. Here we identify pericytes as regulators of epithelial and endothelial morphogenesis in postnatal lung. Mice lacking expression of the Hippo pathway components YAP and TAZ in pericytes show defective alveologenesis. Mutant pericytes are present in normal numbers but display strongly reduced expression of hepatocyte growth factor leading to impaired activation of the c-Met receptor, which is expressed by alveolar epithelial cells. YAP and TAZ are also required for expression of angiopoietin-1 by pulmonary pericytes, which also controls hepatocyte growth factor expression and thereby alveologenesis in an autocrine fashion. These findings establish that pericytes have important, organ-specific signalling properties and coordinate the behavior of epithelial and vascular cells during lung morphogenesis. Pericytes surround endothelial tubules and help maintain the integrity of blood vessels. Here the authors show that pericytes regulate lung morphogenesis via paracrine signalling controlled by components of the Hippo pathway.
Collapse
|
38
|
Yee M, Cohen ED, Domm W, Porter GA, McDavid AN, O’Reilly MA. Neonatal hyperoxia depletes pulmonary vein cardiomyocytes in adult mice via mitochondrial oxidation. Am J Physiol Lung Cell Mol Physiol 2018; 314:L846-L859. [PMID: 29345197 PMCID: PMC6008126 DOI: 10.1152/ajplung.00409.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Supplemental oxygen given to preterm infants has been associated with permanently altering postnatal lung development. Now that these individuals are reaching adulthood, there is growing concern that early life oxygen exposure may also promote cardiovascular disease through poorly understood mechanisms. We previously reported that adult mice exposed to 100% oxygen between postnatal days 0 and 4 develop pulmonary hypertension, defined pathologically by capillary rarefaction, dilation of arterioles and veins, cardiac failure, and a reduced lifespan. Here, Affymetrix Gene Arrays are used to identify early transcriptional changes that take place in the lung before pulmonary capillary rarefaction. We discovered neonatal hyperoxia reduced expression of cardiac muscle genes, including those involved in contraction, calcium signaling, mitochondrial respiration, and vasodilation. Quantitative RT-PCR, immunohistochemistry, and genetic lineage mapping using Myh6CreER; Rosa26RmT/mG mice revealed this reflected loss of pulmonary vein cardiomyocytes. The greatest loss of cadiomyocytes was seen within the lung followed by a graded loss beginning at the hilum and extending into the left atrium. Loss of these cells was seen by 2 wk of age in mice exposed to ≥80% oxygen and was attributed, in part, to reduced proliferation. Administering mitoTEMPO, a scavenger of mitochondrial superoxide during neonatal hyperoxia prevented loss of these cells. Since pulmonary vein cardiomyocytes help pump oxygen-rich blood out of the lung, their early loss following neonatal hyperoxia may contribute to cardiovascular disease seen in these mice, and perhaps in people who were born preterm.
Collapse
Affiliation(s)
- Min Yee
- 1Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - Ethan David Cohen
- 2Department of Medicine, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - William Domm
- 1Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - George A. Porter
- 1Department of Pediatrics, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | - Andrew N. McDavid
- 3Biostatistics and Computational Biology, School of Medicine and Dentistry, The University of Rochester, Rochester, New York
| | | |
Collapse
|
39
|
Sucre JMS, Deutsch GH, Jetter CS, Ambalavanan N, Benjamin JT, Gleaves LA, Millis BA, Young LR, Blackwell TS, Kropski JA, Guttentag SH. A Shared Pattern of β-Catenin Activation in Bronchopulmonary Dysplasia and Idiopathic Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:853-862. [PMID: 29355514 PMCID: PMC5866104 DOI: 10.1016/j.ajpath.2017.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/14/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
Wnt/β-catenin signaling is necessary for normal lung development, and abnormal Wnt signaling contributes to the pathogenesis of both bronchopulmonary dysplasia (BPD) and idiopathic pulmonary fibrosis (IPF), fibrotic lung diseases that occur during infancy and aging, respectively. Using a library of human normal and diseased human lung samples, we identified a distinct signature of nuclear accumulation of β-catenin phosphorylated at tyrosine 489 and epithelial cell cytosolic localization of β-catenin phosphorylated at tyrosine 654 in early normal lung development and fibrotic lung diseases BPD and IPF. Furthermore, this signature was recapitulated in murine models of BPD and IPF. Image analysis of immunofluorescence colocalization demonstrated a consistent pattern of elevated nuclear phosphorylated β-catenin in the lung epithelium and surrounding mesenchyme in BPD and IPF, closely resembling the pattern observed in 18-week fetal lung. Nuclear β-catenin phosphorylated at tyrosine 489 associated with an increased expression of Wnt target gene AXIN2, suggesting that the observed β-catenin signature is of functional significance during normal development and injury repair. The association of specific modifications of β-catenin during normal lung development and again in response to lung injury supports the widely held concept that repair of lung injury involves the recapitulation of developmental programs. Furthermore, these observations suggest that β-catenin phosphorylation has potential as a therapeutic target for the treatment and prevention of both BPD and IPF.
Collapse
Affiliation(s)
- Jennifer M S Sucre
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee.
| | - Gail H Deutsch
- Department of Pathology, Seattle Children's Hospital and University of Washington, Seattle, Washington
| | - Christopher S Jetter
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | | | - John T Benjamin
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Linda A Gleaves
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Cell Imaging Shared Resource, Vanderbilt University, Nashville, Tennessee; Vanderbilt Biophotonics Center, Vanderbilt University, Nashville, Tennessee
| | - Lisa R Young
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Division of Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Division of Pulmonary Medicine, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee; Nashville Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Susan H Guttentag
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
40
|
Hasan SU, Potenziano J, Konduri GG, Perez JA, Van Meurs KP, Walker MW, Yoder BA. Effect of Inhaled Nitric Oxide on Survival Without Bronchopulmonary Dysplasia in Preterm Infants: A Randomized Clinical Trial. JAMA Pediatr 2017; 171:1081-1089. [PMID: 28973344 PMCID: PMC5710365 DOI: 10.1001/jamapediatrics.2017.2618] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IMPORTANCE Bronchopulmonary dysplasia (BPD) occurs in approximately 40% of infants born at younger than 30 weeks' gestation and is associated with adverse pulmonary and neurodevelopmental outcomes. OBJECTIVE To test whether administration of inhaled nitric oxide to preterm infants requiring positive pressure respiratory support on postnatal days 5 to 14 improves the rate of survival without BPD. DESIGN, SETTING, AND PARTICIPANTS This intent-to-treat study was a randomized clinical trial performed at 33 US and Canadian neonatal intensive care units. Participants included 451 neonates younger than 30 weeks' gestation with birth weight less than 1250 g receiving mechanical ventilation or positive pressure respiratory support on postnatal days 5 to 14. Enrollment spanned from December 23, 2009, to April 23, 2012, and neurodevelopmental outcome studies were completed by April 4, 2014. INTERVENTIONS Placebo (nitrogen) or inhaled nitric oxide initiated at 20 ppm was decreased to 10 ppm between 72 and 96 hours after starting treatment and then to 5 ppm on day 10 or 11. Infants remained on the 5-ppm dose until completion of therapy (24 days). MAIN OUTCOMES AND MEASURES The primary outcome was the rate of survival without BPD at 36 weeks' postmenstrual age (PMA). Secondary outcomes included BPD severity, postnatal corticosteroid use, respiratory support, survival, and neurodevelopmental outcomes at 18 to 24 months' PMA. RESULTS In total, 222 infants (52.3% male [n = 116]) received placebo, and 229 infants (50.2% male [n = 115]) received inhaled nitric oxide. Their mean (SD) gestation was 25.6 (1.5) vs 25.6 (1.4) weeks, and their mean (SD) birth weight was 750 (164) vs 724 (160) g. Survival without BPD at 36 weeks' PMA was similar between the placebo and inhaled nitric oxide groups (31.5% [n = 70] vs 34.9% [n = 80]) (odds ratio, 1.17; 95% CI, 0.79-1.73). Rates for severe BPD (26.6% [55 of 207] vs 20.5% [43 of 210]) and postnatal corticosteroid use for BPD (41.0% [91 of 222] vs 41.5% [95 of 229]) and the mean (SD) days of positive pressure respiratory support (55 [40] vs 54 [42]), oxygen therapy (88 [41] vs 91 [59]), and hospitalization (105 [37] vs 108 [54]) were equivalent between the 2 groups. No differences in the incidence of common morbidities were observed. Respiratory outcomes on discharge to home, at 1 year, and at age 18 to 24 months' PMA and neurodevelopmental assessments at 18 to 24 months' PMA did not differ between groups. CONCLUSIONS AND RELEVANCE Inhaled nitric oxide, initiated at 20 ppm on postnatal days 5 to 14 to high-risk preterm infants and continued for 24 days, appears to be safe but did not improve survival without BPD at 36 weeks' PMA or respiratory and neurodevelopmental outcomes at 18 to 24 months' PMA. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00931632.
Collapse
Affiliation(s)
- Shabih U. Hasan
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Alberta Children’s Hospital Research Institute, Calgary, Alberta, Canada
| | | | | | - Jose A. Perez
- Department of Neonatology, Winnie Palmer Hospital, Pediatrix Medical Group, Orlando, Florida
| | - Krisa P. Van Meurs
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California,Lucile Packard Children’s Hospital Stanford, Palo Alto, California
| | - M. Whit Walker
- Division of Neonatology, Department of Pediatrics, University of South Carolina School of Medicine, Greenville
| | - Bradley A. Yoder
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City
| | | |
Collapse
|
41
|
Pulmonary hypertension associated with bronchopulmonary dysplasia in preterm infants. J Reprod Immunol 2017; 124:21-29. [PMID: 29035757 DOI: 10.1016/j.jri.2017.09.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 09/11/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Bronchopulmonary dysplasia (BPD) and BPD-associated pulmonary hypertension (BPD-PH) are chronic inflammatory cardiopulmonary diseases with devastating short- and long-term consequences for infants born prematurely. The immature lungs of preterm infants are ill-prepared to achieve sufficient gas exchange, thus usually necessitating immediate commencement of respiratory support and oxygen supplementation. These therapies are life-saving, but they exacerbate the tissue damage that is inevitably inflicted on a preterm lung forced to perform gas exchange. Together, air-breathing and necessary therapeutic interventions disrupt normal lung development by aggravating pulmonary inflammation and vascular remodelling, thus frequently precipitating BPD and PH via an incompletely understood pathogenic cascade. BPD and BPD-PH share common risk factors, such as low gestational age at birth, fetal growth restriction and perinatal maternal inflammation; however, these risk factors are not unique to BPD or BPD-PH. Occurring in 17-24% of BPD patients, BPD-PH substantially worsens the morbidity and mortality attributable to BPD alone, thus darkening their outlook; for example, BPD-PH entails a mortality of up to 50%. The absence of a safe and effective therapy for BPD and BPD-PH renders neonatal cardiopulmonary disease an area of urgent unmet medical need. Besides the need to develop new therapeutic strategies, a major challenge for clinicians is the lack of a reliable method for identifying babies at risk of developing BPD and BPD-PH. In addition to discussing current knowledge on pathophysiology, diagnosis and treatment of BPD-PH, we highlight emerging biomarkers that could enable clinicians to predict disease-risk and also optimise treatment of BPD-PH in our tiniest patients.
Collapse
|
42
|
Surate Solaligue DE, Rodríguez-Castillo JA, Ahlbrecht K, Morty RE. Recent advances in our understanding of the mechanisms of late lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1101-L1153. [PMID: 28971976 DOI: 10.1152/ajplung.00343.2017] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of lung development is to generate an organ of gas exchange that provides both a thin gas diffusion barrier and a large gas diffusion surface area, which concomitantly generates a steep gas diffusion concentration gradient. As such, the lung is perfectly structured to undertake the function of gas exchange: a large number of small alveoli provide extensive surface area within the limited volume of the lung, and a delicate alveolo-capillary barrier brings circulating blood into close proximity to the inspired air. Efficient movement of inspired air and circulating blood through the conducting airways and conducting vessels, respectively, generates steep oxygen and carbon dioxide concentration gradients across the alveolo-capillary barrier, providing ideal conditions for effective diffusion of both gases during breathing. The development of the gas exchange apparatus of the lung occurs during the second phase of lung development-namely, late lung development-which includes the canalicular, saccular, and alveolar stages of lung development. It is during these stages of lung development that preterm-born infants are delivered, when the lung is not yet competent for effective gas exchange. These infants may develop bronchopulmonary dysplasia (BPD), a syndrome complicated by disturbances to the development of the alveoli and the pulmonary vasculature. It is the objective of this review to update the reader about recent developments that further our understanding of the mechanisms of lung alveolarization and vascularization and the pathogenesis of BPD and other neonatal lung diseases that feature lung hypoplasia.
Collapse
Affiliation(s)
- David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
43
|
Mahlman M, Karjalainen MK, Huusko JM, Andersson S, Kari MA, Tammela OKT, Sankilampi U, Lehtonen L, Marttila RH, Bassler D, Poets CF, Lacaze-Masmonteil T, Danan C, Delacourt C, Palotie A, Muglia LJ, Lavoie PM, Hadchouel A, Rämet M, Hallman M. Genome-wide association study of bronchopulmonary dysplasia: a potential role for variants near the CRP gene. Sci Rep 2017; 7:9271. [PMID: 28839172 PMCID: PMC5571168 DOI: 10.1038/s41598-017-08977-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/20/2017] [Indexed: 11/28/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD), the main consequence of prematurity, has a significant heritability, but little is known about predisposing genes. The aim of this study was to identify gene loci predisposing infants to BPD. The initial genome-wide association study (GWAS) included 174 Finnish preterm infants of gestational age 24–30 weeks. Thereafter, the most promising single-nucleotide polymorphisms (SNPs) associated with BPD were genotyped in both Finnish (n = 555) and non-Finnish (n = 388) replication cohorts. Finally, plasma CRP levels from the first week of life and the risk of BPD were assessed. SNP rs11265269, flanking the CRP gene, showed the strongest signal in GWAS (odds ratio [OR] 3.2, p = 3.4 × 10−6). This association was nominally replicated in Finnish and French African populations. A number of other SNPs in the CRP region, including rs3093059, had nominal associations with BPD. During the first week of life the elevated plasma levels of CRP predicted the risk of BPD (OR 3.4, p = 2.9 × 10–4) and the SNP rs3093059 associated nominally with plasma CRP levels. Finally, SNP rs11265269 was identified as a risk factor of BPD (OR 1.8, p = 5.3 × 10−5), independently of the robust antenatal risk factors. As such, in BPD, a potential role for variants near CRP gene is proposed.
Collapse
Affiliation(s)
- Mari Mahlman
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland. .,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.
| | - Minna K Karjalainen
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Johanna M Huusko
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.,Perinatal Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sture Andersson
- Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - M Anneli Kari
- Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Outi K T Tammela
- Tampere University Hospital, Tampere University, and Center of Pediatric Child Health, Tampere, Finland
| | - Ulla Sankilampi
- Department of Pediatrics, Kuopio University Hospital, Kuopio, Finland
| | - Liisa Lehtonen
- Turku University Hospital, and the University of Turku, Turku, Finland
| | - Riitta H Marttila
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Dirk Bassler
- Department of Neonatology, University Hospital Zurich, and University of Zurich, Zurich, Switzerland
| | - Christian F Poets
- Department of Neonatology, Tuebingen University Hospital, Tuebingen, Germany
| | | | - Claude Danan
- Inserm, U955, Créteil, France.,CRB, CHI-Creteil, France.,Department of neonatology, CHI-Creteil, Creteil, France
| | - Christophe Delacourt
- Inserm, U955, Créteil, France.,AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Paris, France.,Université Paris-Descartes, Paris, France
| | - Aarno Palotie
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.,The Stanley Center for Psychiatric Research, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland.,Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Louis J Muglia
- Perinatal Institute, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Vancouver Canada, Vancouver, Canada
| | - Alice Hadchouel
- Inserm, U955, Créteil, France.,AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Paris, France.,Université Paris-Descartes, Paris, France
| | - Mika Rämet
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland.,BioMediTech Institute and Faculty of Medical and Life Sciences, University of Tampere, Tampere, Finland
| | - Mikko Hallman
- PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
44
|
Royce SG, Nold MF, Bui C, Donovan C, Lam M, Lamanna E, Rudloff I, Bourke JE, Nold-Petry CA. Airway Remodeling and Hyperreactivity in a Model of Bronchopulmonary Dysplasia and Their Modulation by IL-1 Receptor Antagonist. Am J Respir Cell Mol Biol 2017; 55:858-868. [PMID: 27482635 DOI: 10.1165/rcmb.2016-0031oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic disease of extreme prematurity that has serious long-term consequences including increased asthma risk. We earlier identified IL-1 receptor antagonist (IL-1Ra) as a potent inhibitor of murine BPD induced by combining perinatal inflammation (intraperitoneal LPS to pregnant dams) and exposure of pups to hyperoxia (fraction of inspired oxygen = 0.65). In this study, we determined whether airway remodeling and hyperresponsiveness similar to asthma are evident in this model, and whether IL-1Ra is protective. During 28-day exposure to air or hyperoxia, pups received vehicle or 10 mg/kg IL-1Ra by daily subcutaneous injection. Lungs were then prepared for histology and morphometry of alveoli and airways, or for real-time PCR, or inflated with agarose to prepare precision-cut lung slices to visualize ex vivo intrapulmonary airway contraction and relaxation by phase-contrast microscopy. In pups reared under normoxic conditions, IL-1Ra treatment did not affect alveolar or airway structure or airway responses. Pups reared in hyperoxia developed a severe BPD-like lung disease, with fewer, larger alveoli, increased subepithelial collagen, and increased expression of α-smooth muscle actin and cyclin D1. After hyperoxia, methacholine elicited contraction with similar potency but with an increased maximum reduction in lumen area (air, 44%; hyperoxia, 89%), whereas dilator responses to salbutamol were maintained. IL-1Ra treatment prevented hyperoxia-induced alveolar disruption and airway fibrosis but, surprisingly, not the increase in methacholine-induced airway contraction. The current study is the first to demonstrate ex vivo airway hyperreactivity caused by systemic maternal inflammation and postnatal hyperoxia, and it reveals further preclinical mechanistic insights into IL-1Ra as a treatment targeting key pathophysiological features of BPD.
Collapse
Affiliation(s)
- Simon G Royce
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Marcel F Nold
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Christine Bui
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Chantal Donovan
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Maggie Lam
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Emma Lamanna
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Ina Rudloff
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Jane E Bourke
- 1 Biomedicine Discovery Institute, Department of Pharmacology
| | - Claudia A Nold-Petry
- 2 Ritchie Centre, Hudson Institute of Medical Research, and.,3 Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
45
|
Naumburg E, Söderström L, Huber D, Axelsson I. Risk factors for pulmonary arterial hypertension in children and young adults. Pediatr Pulmonol 2017; 52:636-641. [PMID: 27801982 DOI: 10.1002/ppul.23633] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/05/2016] [Accepted: 10/05/2016] [Indexed: 01/29/2023]
Abstract
OBJECTIVES Pulmonary hypertension (PH) has been linked to preterm birth explained by congenital heart defects and pulmonary diseases. WORKING HYPOTHESIS Other factors may influence the risk of PH among adolescences and children born premature. STUDY DESIGN This national registry-based study assess risk of PH following premature birth adjusted for known risk factors. PATIENT-SUBJECT SELECTION AND METHODOLOGY All cases born 1993-2010, identified by diagnostic codes applicable to PH and retrieved from the Swedish Registry of Congenital Heart Disease (N = 67). Six controls were randomly selected and matched to each case by year of birth and hospital by the Swedish Medical Birth Register (N = 402). Maternal and infant data related to preterm birth, pulmonary diseases, and congenital defects were retrieved. The association between preterm birth and pulmonary hypertension was calculated by conditional logistic regression taking into account potential confounding factors. RESULTS One third of the cases and seven percent of the controls were born preterm in our study. Preterm birth was associated with PH, OR = 8.46 (95%CI 2.97-24.10) (P < 0.0001) even after adjusting for confounding factors. Other factors, such as acute pulmonary diseases, congenital heart defects, congenital diaphragm herniation, and chromosomal disorders were also associated with PH in the multivariate analysis. CONCLUSIONS Children and young adults born preterm are known to have an increased risk of PH, previously explained by congenital heart defects and pulmonary diseases. By adjusting for such factors, our study indicates that new factors may play a role in the risk of developing PH among children born preterm. Pediatr Pulmonol. 2017;52:636-641. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Estelle Naumburg
- Department of Clinical Science, Paediatrics, Umeå University, Umeå, Sweden.,Östersund Hospital, Unit of Research, Education and Development, Östersund, Sweden
| | - Lars Söderström
- Östersund Hospital, Unit of Research, Education and Development, Östersund, Sweden
| | - Daniel Huber
- Östersund Hospital, Unit of Research, Education and Development, Östersund, Sweden
| | - Inge Axelsson
- Östersund Hospital, Unit of Research, Education and Development, Östersund, Sweden
| |
Collapse
|
46
|
Li J, Oehlert J, Snyder M, Stevenson DK, Shaw GM. Fetal de novo mutations and preterm birth. PLoS Genet 2017; 13:e1006689. [PMID: 28388617 PMCID: PMC5384656 DOI: 10.1371/journal.pgen.1006689] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/14/2017] [Indexed: 01/11/2023] Open
Abstract
Preterm birth (PTB) affects ~12% of pregnancies in the US. Despite its high mortality and morbidity, the molecular etiology underlying PTB has been unclear. Numerous studies have been devoted to identifying genetic factors in maternal and fetal genomes, but so far few genomic loci have been associated with PTB. By analyzing whole-genome sequencing data from 816 trio families, for the first time, we observed the role of fetal de novo mutations in PTB. We observed a significant increase in de novo mutation burden in PTB fetal genomes. Our genomic analyses further revealed that affected genes by PTB de novo mutations were dosage sensitive, intolerant to genomic deletions, and their mouse orthologs were likely developmentally essential. These genes were significantly involved in early fetal brain development, which was further supported by our analysis of copy number variants identified from an independent PTB cohort. Our study indicates a new mechanism in PTB occurrence independently contributed from fetal genomes, and thus opens a new avenue for future PTB research. Preterm birth is a prevalent pregnancy condition and leads to substantial morbidity and mortality. Its genetic association has been well observed, but the underlying etiology remains unclear. Current research has been focused on identifying risk factors in maternal genomes. In this study, we tested an unexplored hypothesis that preterm birth could be independently influenced by fetal genomes. We analyzed fetal de novo mutations, those not inherited from parents, from 816 trio families, and found preterm infants tended to have increased de novo mutation rates compared to infants born at term. Importantly, we also observed that these preterm-associated de novo mutations preferentially affect dosage sensitive genes that are essential in embryonic development, and these affected genes are involved in early fetal brain development. Overall, our study for the first time showed the fetal genetic contribution to preterm birth, and suggested abnormal fetal development as a potential cause for preterm birth.
Collapse
Affiliation(s)
- Jingjing Li
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA, California, United States of America
- Department of Genetics, Center for Genomics and Personalized Medicine Stanford University, School of Medicine, Stanford, CA, California, United States of America
| | - John Oehlert
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA, California, United States of America
| | - Michael Snyder
- Department of Genetics, Center for Genomics and Personalized Medicine Stanford University, School of Medicine, Stanford, CA, California, United States of America
| | - David K. Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA, California, United States of America
| | - Gary M. Shaw
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Abstract
The pathogenesis of Bronchopulmonary Dysplasia (BPD) is multifactorial and the clinical phenotype of BPD is extremely variable. Predicting BPD is difficult, as it is a disease with a clinical operational definition but many clinical phenotypes and endotypes. Most biomarkers studied over the years have low predictive accuracy, and none are currently used in routine clinical care or shown to be useful for predicting longer-term respiratory outcome. Targeted cellular and humoral biomarkers and novel systems biology 'omic' based approaches including genomic and microbiomic analyses are described in this review.
Collapse
|
48
|
Bush A, Bolton CE. Longer Term Sequelae of Prematurity: The Adolescent and Young Adult. RESPIRATORY OUTCOMES IN PRETERM INFANTS 2017. [DOI: 10.1007/978-3-319-48835-6_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
49
|
|
50
|
Ota C, Baarsma HA, Wagner DE, Hilgendorff A, Königshoff M. Linking bronchopulmonary dysplasia to adult chronic lung diseases: role of WNT signaling. Mol Cell Pediatr 2016; 3:34. [PMID: 27718180 PMCID: PMC5055515 DOI: 10.1186/s40348-016-0062-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most common chronic lung diseases in infants caused by pre- and/or postnatal lung injury. BPD is characterized by arrested alveolarization and vascularization due to extracellular matrix remodeling, inflammation, and impaired growth factor signaling. WNT signaling is a critical pathway for normal lung development, and its altered signaling has been shown to be involved in the onset and progression of incurable chronic lung diseases in adulthood, such as chronic obstructive pulmonary disease (COPD) or idiopathic pulmonary fibrosis (IPF). In this review, we summarize the impact of WNT signaling on different stages of lung development and its potential contribution to developmental lung diseases, especially BPD, and chronic lung diseases in adulthood.
Collapse
Affiliation(s)
- Chiharu Ota
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.
| | - Hoeke A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.,The Perinatal Center, Campus Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|