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
|
CLYMAN RONALDI. Patent Ductus Arteriosus in the Preterm Infant. AVERY'S DISEASES OF THE NEWBORN 2024:716-726.e11. [DOI: 10.1016/b978-0-323-82823-9.00048-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Abstract
Bronchopulmonary dysplasia (BPD) is a multi-factorial disease that results from multiple clinical factors, including lung immaturity, mechanical ventilation, oxidative stress, pulmonary congestion due to increasing cardiac blood shunting, nutritional and immunological factors. Twin studies have indicated that susceptibility to BPD can be strongly inherited in some settings. Studies have reported associations between common genetic variants and BPD in preterm infants. Recent genomic studies have highlighted a potential role for molecular pathways involved in inflammation and lung development in affected infants. Rare mutations in genes encoding the lipid transporter ATP-binding cassette, sub-family A, member 3 (ABCA3 gene) which is involved in surfactant synthesis in alveolar type II cells, as well as surfactant protein B (SFTPB) and C (SFTPC) can also result in severe form of neonatal-onset interstitial lung diseases and may also potentially affect the course of BPD. This chapter summarizes the current state of knowledge on the genetics of BPD.
Collapse
Affiliation(s)
- Pascal M Lavoie
- Division of Neonatology, Department of Pediatrics, University of British Columbia, Vancouver, Canada; BC Children's Hospital Research Institute, Vancouver, Canada.
| | - Jonathan H Rayment
- BC Children's Hospital Research Institute, Vancouver, Canada; Division of Respiratory Medicine, Department of Pediatrics, University of British Columbia, Vancouver, Canada; Division of Respiratory Medicine, BC Children's Hospital, Vancouver, Canada
| |
Collapse
|
4
|
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
|
5
|
Ovalı F. Molecular and Mechanical Mechanisms Regulating Ductus Arteriosus Closure in Preterm Infants. Front Pediatr 2020; 8:516. [PMID: 32984222 PMCID: PMC7477801 DOI: 10.3389/fped.2020.00516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Failure of ductus arteriosus closure after preterm birth is associated with significant morbidities. Ductal closure requires and is regulated by a complex interplay of molecular and mechanical mechanisms with underlying genetic factors. In utero patency of the ductus is maintained by low oxygen tension, high levels of prostaglandins, nitric oxide and carbon monoxide. After birth, ductal closure occurs first by functional closure, followed by anatomical remodeling. High oxygen tension and decreased prostaglandin levels mediated by numerous factors including potassium channels, endothelin-1, isoprostanes lead to the contraction of the ductus. Bradykinin and corticosteroids also induce ductal constriction by attenuating the sensitivity of the ductus to PGE2. Smooth muscle cells of the ductus can sense oxygen through a mitochondrial network by the role of Rho-kinase pathway which ends up with increased intracellular calcium levels and contraction of myosin light chains. Anatomical closure of the ductus is also complex with various mechanisms such as migration and proliferation of smooth muscle cells, extracellular matrix production, endothelial cell proliferation which mediate cushion formation with the interaction of blood cells. Regulation of vessel walls is affected by retinoic acid, TGF-β1, notch signaling, hyaluronan, fibronectin, chondroitin sulfate, elastin, and vascular endothelial cell growth factor (VEGF). Formation of the platelet plug facilitates luminal remodeling by the obstruction of the constricted ductal lumen. Vasa vasorum are more pronounced in the term ductus but are less active in the preterm ductus. More than 100 genes are effective in the prostaglandin pathway or in vascular smooth muscle development and structure may affect the patency of ductus. Hemodynamic changes after birth including fluid load and flow characteristics as well as shear forces within the ductus also stimulate closure. Current pharmacological treatment for the closure of a patent ductus is based on the blockage of the prostaglandin pathway mainly through COX or POX inhibition, albeit with some limitations and side effects. Further research for new agents aiming ductal closure should focus on a clear understanding of vascular biology of the ductus.
Collapse
Affiliation(s)
- Fahri Ovalı
- Division of Neonatology, Department of Pediatrics, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
6
|
Lewis TR, Shelton EL, Van Driest SL, Kannankeril PJ, Reese J. Genetics of the patent ductus arteriosus (PDA) and pharmacogenetics of PDA treatment. Semin Fetal Neonatal Med 2018; 23:232-238. [PMID: 29510900 PMCID: PMC6098727 DOI: 10.1016/j.siny.2018.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Patent ductus arteriosus (PDA) is a frequent, complex, and difficult to treat clinical syndrome among preterm infants in the neonatal intensive care unit. In addition to known clinical risk factors, there are emerging data about genetic predisposition to PDA in both animal and human models. Clinical response and toxicity from drugs used to treat PDA are highly variable. Developmental and genetic aspects of pharmacokinetics and pharmacodynamics influence exposure and response to pharmacologic therapies. Given the variable efficacy and toxicity of known drug therapies, novel therapeutic targets for PDA treatment offer the promise of precision medicine. This review addresses the known genetic contributions to prolonged ductal patency, variability in response to drug therapy for PDA, and potential novel drug targets for future PDA treatment discovery.
Collapse
Affiliation(s)
- Tamorah R Lewis
- Department of Pediatrics, Children's Mercy Hospitals & Clinics, University of Missouri, Kansas City School of Medicine, Kansas City, MO, USA
| | - Elaine L Shelton
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara L Van Driest
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Prince J Kannankeril
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeff Reese
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt and Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
7
|
Jilling T, Ambalavanan N, Cotten CM, Martin CA, Maheshwari A, Schibler K, Levy J, Page GP. Surgical necrotizing enterocolitis in extremely premature neonates is associated with genetic variations in an intergenic region of chromosome 8. Pediatr Res 2018; 83:943-953. [PMID: 29538362 PMCID: PMC6053310 DOI: 10.1038/pr.2018.33] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 02/08/2018] [Indexed: 12/02/2022]
Abstract
BackgroundTwin studies suggest that genetic factors may account for up to 50% increased risk for necrotizing enterocolitis (NEC), but genome-wide association studies for NEC are lacking.MethodsGenotyping was done on Illumina BeadChip, followed by analysis using PLINK with logistic regression under an additive model.ResultsAmong 751 extremely-low-birth-weight (<1,000 g, >401 g) neonates, 30 had surgical NEC. Two hundred and sixty-one single-nucleotide polymorphisms (SNPs) showed association with NEC at P<0.05, of which 35 were significant at P<10-7. Minor allele(s) in a cluster of SNPs spanning a 43-kb region of chromosome 8 (8q23.3) conferred an odds ratio of 4.72 (95% confidence interval (CI): 2.51-8.88) for elevated risk of NEC. Two smaller clusters on chromosome 14 and chromosome 11 exhibited P values of 10-7-10-8. The chromosome 8 cluster is in an intergenic region between CUB and Sushi multiple domains 3 (-1.43 Mb) and trichorhinophalangeal syndrome I (+542 kb). RNA sequencing in this region identified a potential novel open-reading frame corresponding to a long interspersed element-1 retrotransposable element.ConclusionGenetic variation in an intergenic region of chromosome 8 is associated with increased risk for NEC with a mechanism that is yet to be identified.
Collapse
MESH Headings
- Chromosomes, Human, Pair 8
- Cohort Studies
- DNA, Intergenic
- Enterocolitis, Necrotizing/diagnosis
- Enterocolitis, Necrotizing/genetics
- Enterocolitis, Necrotizing/surgery
- Female
- Gene Frequency
- Genetic Predisposition to Disease
- Genetic Variation
- Genome-Wide Association Study
- Genotype
- Humans
- Infant
- Infant, Extremely Low Birth Weight
- Infant, Newborn
- Infant, Newborn, Diseases
- Infant, Premature
- Long Interspersed Nucleotide Elements
- Male
- Oligonucleotide Array Sequence Analysis
- Phenotype
- Polymorphism, Single Nucleotide
- Respiration, Artificial
- Sequence Analysis, RNA
- Signal Transduction
Collapse
Affiliation(s)
| | | | | | | | | | - Kurt Schibler
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | | | | | | |
Collapse
|
8
|
Moonen RM, Cavallaro G, Huizing MJ, González-Luis GE, Mosca F, Villamor E. Association between the p.Thr1406Asn polymorphism of the carbamoyl-phosphate synthetase 1 gene and necrotizing enterocolitis: A prospective multicenter study. Sci Rep 2016; 6:36999. [PMID: 27833157 PMCID: PMC5105130 DOI: 10.1038/srep36999] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/20/2016] [Indexed: 12/04/2022] Open
Abstract
The p.Thr1406Asn (rs1047891) polymorphism of the carbamoyl-phosphate synthetase 1 (CPS1) gene has been linked to functional consequences affecting the downstream availability of the nitric oxide precursor L-arginine. L-arginine concentrations are decreased in preterm infants with necrotizing enterocolitis (NEC). In this multicenter prospective study, we investigated the association of the p.Thr1406Asn polymorphism with NEC in 477 preterm infants (36 cases of NEC) from 4 European neonatal intensive care units (Maastricht, Las Palmas de Gran Canaria, Mantova, and Milan). Allele and genotype frequencies of the p.Thr1406Asn polymorphism did not significantly differ between the infants with and without NEC. In contrast, the minor A-allele was significantly less frequent in the group of 64 infants with the combined outcome NEC or death before 34 weeks of corrected gestational age than in the infants without the outcome (0.20 vs. 0.31, P = 0.03). In addition, a significant negative association of the A-allele with the combined outcome NEC or death was found using the dominant (adjusted odds ratio, aOR: 0.54, 95% CI 0.29–0.99) and the additive (aOR 0.58, 95% CI 0.36–0.93) genetic models. In conclusion, our study provides further evidence that a functional variant of the CPS1 gene may contribute to NEC susceptibility.
Collapse
Affiliation(s)
- Rob M Moonen
- Department of Pediatrics, Zuyderland Medical Center Heerlen, 6130 MB, The Netherlands.,Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, 6202 AZ, The Netherlands
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, 20122, Italy
| | - Maurice J Huizing
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, 6202 AZ, The Netherlands
| | - Gema E González-Luis
- Department of Pediatrics, Hospital Universitario Materno-Infantil de Canarias, Las Palmas de Gran Canaria, 35016, Spain
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Università degli Studi di Milano, Milan, 20122, Italy
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Developmental Biology (GROW), Maastricht, 6202 AZ, The Netherlands
| |
Collapse
|
9
|
Abstract
Necrotising enterocolitis (NEC) is an uncommon, but devastating intestinal inflammatory disease that predominantly affects preterm infants. NEC is sometimes dubbed the spectre of neonatal intensive care units, as its onset is insidiously non-specific, and once the disease manifests, the damage inflicted on the baby's intestine is already disastrous. Subsequent sepsis and multi-organ failure entail a mortality of up to 65%. Development of effective treatments for NEC has stagnated, largely because of our lack of understanding of NEC pathogenesis. It is clear, however, that NEC is driven by a profoundly dysregulated immune system. NEC is associated with local increases in pro-inflammatory mediators, e.g. Toll-like receptor (TLR) 4, nuclear factor-κB, tumour necrosis factor, platelet-activating factor (PAF), interleukin (IL)-18, interferon-gamma, IL-6, IL-8 and IL-1β. Deficiencies in counter-regulatory mechanisms, including IL-1 receptor antagonist (IL-1Ra), TLR9, PAF-acetylhydrolase, transforming growth factor beta (TGF-β)1&2, IL-10 and regulatory T cells likely facilitate a pro-inflammatory milieu in the NEC-afflicted intestine. There is insufficient evidence to conclude a predominance of an adaptive Th1-, Th2- or Th17-response in the disease. Our understanding of the accompanying regulation of systemic immunity remains poor; however, IL-1Ra, IL-6, IL-8 and TGF-β1 show promise as biomarkers. Here, we chart the emerging immunological landscape that underpins NEC by reviewing the involvement and potential clinical implications of innate and adaptive immune mediators and their regulation in NEC.
Collapse
|
10
|
Du JF, Liu TT, Wu H. [Risk factors for patent ductus arteriosus in early preterm infants: a case-control study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:15-19. [PMID: 26781406 PMCID: PMC7390094 DOI: 10.7499/j.issn.1008-8830.2016.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/30/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To investigate the risk factors for the occurrence of patent ductus arteriosus (PDA) and to provide a clinical basis for reducing the occurrence of PDA in early preterm infants. METHODS A total of 136 early preterm infants (gestational age≤32 weeks) who were hospitalized between January 2013 and December 2014 and diagnosed with hemodynamicalhy significant PDA (hs-PDA) were enrolled as the case group. Based on the matched case-control principle, 136 early preterm infants without hs-PDA were selected among those who were hospitalized within the same period at a ratio of 1:1 and enrolled as the control group. The two groups were matched for sex and gestational age. The basic information of neonates and maternal conditions during the pregnancy and perinatal periods were collected. Logistic regression analysis was performed to identify the risk factors for the development of PDA. RESULTS Univariate analysis showed that neonatal infectious diseases, neonatal respiratory distress syndrome, decreased platelet count within 24 hours after birth, and low birth weight were associated with the development of hs-PDA (P<0.05). Multivariate conditional logistic regression analysis revealed that neonatal infectious diseases (OR=2.368) and decreased platelet count within 24 hours after birth (OR=0.996) were independent risk factors for hs-PDA. CONCLUSIONS Neonatal infectious diseases and decreased platelet count within 24 hours after birth increase the risk of hs-PDA in early preterm infants.
Collapse
Affiliation(s)
- Jin-Feng Du
- First Hospital of Jilin University, Changchun 130000, China.
| | | | | |
Collapse
|
11
|
Du JF, Liu TT, Wu H. [Risk factors for patent ductus arteriosus in early preterm infants: a case-control study]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2016; 18:15-9. [PMID: 26781406 PMCID: PMC7390094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/30/2015] [Indexed: 08/01/2024]
Abstract
OBJECTIVE To investigate the risk factors for the occurrence of patent ductus arteriosus (PDA) and to provide a clinical basis for reducing the occurrence of PDA in early preterm infants. METHODS A total of 136 early preterm infants (gestational age≤32 weeks) who were hospitalized between January 2013 and December 2014 and diagnosed with hemodynamicalhy significant PDA (hs-PDA) were enrolled as the case group. Based on the matched case-control principle, 136 early preterm infants without hs-PDA were selected among those who were hospitalized within the same period at a ratio of 1:1 and enrolled as the control group. The two groups were matched for sex and gestational age. The basic information of neonates and maternal conditions during the pregnancy and perinatal periods were collected. Logistic regression analysis was performed to identify the risk factors for the development of PDA. RESULTS Univariate analysis showed that neonatal infectious diseases, neonatal respiratory distress syndrome, decreased platelet count within 24 hours after birth, and low birth weight were associated with the development of hs-PDA (P<0.05). Multivariate conditional logistic regression analysis revealed that neonatal infectious diseases (OR=2.368) and decreased platelet count within 24 hours after birth (OR=0.996) were independent risk factors for hs-PDA. CONCLUSIONS Neonatal infectious diseases and decreased platelet count within 24 hours after birth increase the risk of hs-PDA in early preterm infants.
Collapse
Affiliation(s)
- Jin-Feng Du
- First Hospital of Jilin University, Changchun 130000, China.
| | | | | |
Collapse
|
12
|
Abstract
The objective of this study is to review the candidate gene and genome-wide association studies relevant to bronchopulmonary dysplasia, and to discuss the emerging understanding of the complexities involved in genetic predisposition to bronchopulmonary dysplasia and its outcomes. Genetic factors contribute much of the variance in risk for BPD. Studies to date evaluating single or a few candidate genes have not been successful in yielding results that are replicated in GWAS, perhaps due to more stringent p-value thresholds. GWAS studies have identified only a single gene (SPOCK2) at genome-wide significance in a European White and African cohort, which was not replicated in two North American studies. Pathway gene-set analysis in a North American cohort confirmed involvement of known pathways of lung development and repair (e.g., CD44 and phosphorus oxygen lyase activity) and indicated novel molecules and pathways (e.g., adenosine deaminase and targets of miR-219) involved in genetic predisposition to BPD. The genetic basis of severe BPD is different from that of mild/moderate BPD, and the variants/pathways associated with BPD vary by race/ethnicity. A pilot study of whole exome sequencing identified hundreds of genes of interest, and indicated the overall feasibility as well as complexity of this approach. Better phenotyping of BPD by severity and pathophysiology, and careful analysis of race/ethnicity is required to gain a better understanding of the genetic basis of BPD. Future translational studies are required for the identification of potential genetic predispositions (rare variants and dysregulated pathways) by next-generation sequencing methods in individual infants (personalized genomics).
Collapse
Affiliation(s)
- Charitharth Vivek Lal
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, 176F Suite 9380, Women and Infants Center, 619 South 19th St, Birmingham, AL 35249-7335
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, 176F Suite 9380, Women and Infants Center, 619 South 19th St, Birmingham, AL 35249-7335.
| |
Collapse
|
13
|
Dani C, Poggi C. The role of genetic polymorphisms in antioxidant enzymes and potential antioxidant therapies in neonatal lung disease. Antioxid Redox Signal 2014; 21:1863-80. [PMID: 24382101 PMCID: PMC4203110 DOI: 10.1089/ars.2013.5811] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
SIGNIFICANCE Oxidative stress is involved in the development of newborn lung diseases, such as bronchopulmonary dysplasia and persistent pulmonary hypertension of the newborn. The activity of antioxidant enzymes (AOEs), which is impaired as a result of prematurity and oxidative injury, may be further affected by specific genetic polymorphisms or an unfavorable combination of more of them. RECENT ADVANCES Genetic polymorphisms of superoxide dismutase and catalase were recently demonstrated to be protective or risk factors for the main complications of prematurity. A lot of research focused on the potential of different antioxidant strategies in the prevention and treatment of lung diseases of the newborn, providing promising results in experimental models. CRITICAL ISSUES The effect of different genetic polymorphisms on protein synthesis and activity has been poorly detailed in the newborn, hindering to derive conclusive results from the observed associations with adverse outcomes. Therapeutic strategies that aimed at enhancing the activity of AOEs were poorly studied in clinical settings and partially failed to produce clinical benefits. FUTURE DIRECTIONS The clarification of the effects of genetic polymorphisms on the proteomics of the newborn is mandatory, as well as the assessment of a larger number of polymorphisms with a possible correlation with adverse outcome. Moreover, antioxidant treatments should be carefully translated to clinical settings, after further details on optimal doses, administration techniques, and adverse effects are provided. Finally, the study of genetic polymorphisms could help select a specific high-risk population, who may particularly benefit from targeted antioxidant strategies.
Collapse
Affiliation(s)
- Carlo Dani
- Section of Neonatology, Department of Neurosciences, Psychology, Drug Research and Child Health, Careggi University Hospital , Florence, Italy
| | | |
Collapse
|
14
|
Vucovich MM, Cotton RB, Shelton EL, Goettel JA, Ehinger NJ, Poole SD, Brown N, Wynn JL, Paria BC, Slaughter JC, Clark RH, Rojas MA, Reese J. Aminoglycoside-mediated relaxation of the ductus arteriosus in sepsis-associated PDA. Am J Physiol Heart Circ Physiol 2014; 307:H732-40. [PMID: 24993047 PMCID: PMC4187398 DOI: 10.1152/ajpheart.00838.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 06/27/2014] [Indexed: 11/22/2022]
Abstract
Sepsis is strongly associated with patency of the ductus arteriosus (PDA) in critically ill newborns. Inflammation and the aminoglycoside antibiotics used to treat neonatal sepsis cause smooth muscle relaxation, but their contribution to PDA is unknown. We examined whether: 1) lipopolysaccharide (LPS) or inflammatory cytokines cause relaxation of the ex vivo mouse DA; 2) the aminoglycosides gentamicin, tobramycin, or amikacin causes DA relaxation; and 3) newborn infants treated with aminoglycosides have an increased risk of symptomatic PDA (sPDA). Changes in fetal mouse DA tone were measured by pressure myography in response to LPS, TNF-α, IFN-γ, macrophage-inflammatory protein 2, IL-15, IL-13, CXC chemokine ligand 12, or three aminoglycosides. A clinical database of inborn patients of all gestations was analyzed for association between sPDA and aminoglycoside treatment. Contrary to expectation, neither LPS nor any of the inflammatory mediators caused DA relaxation. However, each of the aminoglycosides caused concentration-dependent vasodilation in term and preterm mouse DAs. Pretreatment with indomethacin and N-(G)-nitro-L-arginine methyl ester did not prevent gentamicin-induced DA relaxation. Gentamicin-exposed DAs developed less oxygen-induced constriction than unexposed DAs. Among 488,349 infants who met the study criteria, 40,472 (8.3%) had sPDA. Confounder-adjusted odds of sPDA were higher in gentamicin-exposed infants, <25 wk and >32 wk. Together, these findings suggest that factors other than inflammation contribute to PDA. Aminoglycoside-induced vasorelaxation and inhibition of oxygen-induced DA constriction support the paradox that antibiotic treatment of sepsis may contribute to DA relaxation. This association was also found in newborn infants, suggesting that antibiotic selection may be an important consideration in efforts to reduce sepsis-associated PDA.
Collapse
Affiliation(s)
- Megan M Vucovich
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Robert B Cotton
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Jeremy A Goettel
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Noah J Ehinger
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Stanley D Poole
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Naoko Brown
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - James L Wynn
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - Bibhash C Paria
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Mario A Rojas
- Department of Pediatrics, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Jeff Reese
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee;
| |
Collapse
|
15
|
Markel TA, Engelstad H, Poindexter BB. Predicting disease severity of necrotizing enterocolitis: how to identify infants for future novel therapies. J Clin Neonatol 2014; 3:1-9. [PMID: 24741531 PMCID: PMC3982330 DOI: 10.4103/2249-4847.128717] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Necrotizing enterocolitis (NEC) remains a very devastating problem within the very low birth weight neonatal population. Several experimental therapies are being tested in animal models and soon may be ready for human trials. Despite this progress, we currently have no way to identify infants who would be optimal targets for therapy. Specifically, we are unable to predict which infants will progress to the more severe Bell's stage of disease that may necessitate surgery. Ideally, an algorithm could be constructed that would encompass multiple neonatal and maternal risk factors as well as potential biologic markers of disease so that these infants could be identified in a more timely fashion. This review summarizes the known risk factors and biomarkers of disease in hopes of stimulating clinical research to identify such an “early warning” NEC algorithm.
Collapse
Affiliation(s)
- Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Holly Engelstad
- Department of Pediatrics, Section of Neonatology, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brenda B Poindexter
- Department of Pediatrics, Section of Neonatology, Riley Hospital for Children at Indiana University Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Antioxidant strategies and respiratory disease of the preterm newborn: an update. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:721043. [PMID: 24803984 PMCID: PMC3996983 DOI: 10.1155/2014/721043] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/06/2014] [Indexed: 01/20/2023]
Abstract
Preterm newborns are challenged by an excessive oxidative burden, as a result of several perinatal stimuli, as intrauterine infections, resuscitation, mechanical ventilation, and postnatal complications, in the presence of immature antioxidant capacities. "Oxygen radical disease of neonatology" comprises a wide range of conditions sharing a common pathway of pathogenesis and includes bronchopulmonary dysplasia (BPD) and other main complications of prematurity. Antioxidant strategies may be beneficial in the prevention and treatment of oxidative stress- (OS-) related lung disease of the preterm newborn. Endotracheal supplementation or lung-targeted overexpression of superoxide dismutase was proved to reduce lung damage in several models; however, the supplementation in preterm newborn failed to reduce the risk of BPD, although long-term respiratory outcomes were improved. Also melatonin administration to small cohorts of preterm newborns suggested beneficial effects on lung OS. The possibility to identify single nucleotide polymorphism affecting the risk of BPD may help to identify specific populations with particularly high risk of OS-related diseases and may pose the basis for individually targeted treatments. Finally, surfactant replacement may lead to local anti-inflammatory and antioxidant effects, thanks to specific enzymatic and nonenzymatic antioxidants naturally present in animal surfactants.
Collapse
|
17
|
Devi SG, Kumar A, Kar P, Husain SA, Sharma S. Association of pregnancy outcome with cytokine gene polymorphisms in HEV infection during pregnancy. J Med Virol 2014; 86:1366-76. [DOI: 10.1002/jmv.23925] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Salam Gyaneshwori Devi
- Department of Obstetrics & Gynaecology; Maulana Azad Medical College and Lok Nayak Hospital; New Delhi India
| | - Ashok Kumar
- Department of Obstetrics & Gynaecology; Maulana Azad Medical College and Lok Nayak Hospital; New Delhi India
| | - Premashis Kar
- Department of Medicine; Maulana Azad Medical College and Lok Nayak Hospital; New Delhi India
| | | | - Shashi Sharma
- Department of Statistics and Epidemiology; Institute of Cytology and Preventive Oncology; Noida India
| |
Collapse
|
18
|
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease in infants. Its treatment imposes considerable healthcare burden and costs in the perinatal and early childhood period and patients are usually left with lifelong deficits in lung function. Evidence exists for different pathophysiologic pathways that can promote the structural changes that characterize BPD, including the impairment in alveolarization; however, there is increasing interest regarding heritable factors that may predispose very low birth weight infants to BPD. Our review focuses on recent publications that have investigated genetic factors that may potentially contribute to such reported heritability. These publications point us toward some possible genomic candidates for further study, but certainly do not identify any particular gene or gene pathway that would be inferred to be contributing substantially to the underlying etiology of BPD.
Collapse
Affiliation(s)
- Gary M. Shaw
- Department of Pediatrics of Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| | - Hugh M. O'Brodovich
- Department of Pediatrics of Stanford University School of Medicine, Palo Alto, CA, 94305, USA
| |
Collapse
|
19
|
Jo HS. Association between Respiratory Disorders and Candidate Genes in Korean Newborn Infants. NEONATAL MEDICINE 2013. [DOI: 10.5385/nm.2013.20.3.311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Heui Seung Jo
- Department of Pediatrics, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
20
|
Aghai ZH, Saslow JG, Mody K, Eydelman R, Bhat V, Stahl G, Pyon K, Bhandari V. IFN-γ and IP-10 in tracheal aspirates from premature infants: relationship with bronchopulmonary dysplasia. Pediatr Pulmonol 2013; 48:8-13. [PMID: 22431160 DOI: 10.1002/ppul.22540] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 01/11/2012] [Indexed: 11/10/2022]
Abstract
BACKGROUND Interferon-gamma (IFN-γ) and interferon-inducible protein of 10 kDa (IP-10) are potent inflammatory mediators and contribute to acute lung injury in adults. Recently, a potential role for IFN-γ and IP-10 in the pathogenesis of bronchopulmonary dysplasia (BPD) has been reported in animal models. OBJECTIVE To study the association between IFN-γ and IP-10 in tracheal aspirate (TA) and the development of BPD in premature infants. DESIGN/METHODS TA samples collected within 48 hr after birth from 79 mechanically ventilated premature neonates [gestational age (GA) <30 weeks (w), birth weight (BW) <1,250 g (g)] were analyzed. IFN-γ was measured in a subgroup of 38 infants by using a biochip multi-analyte immunoassay. The level of IP-10 was determined using a commercially available ELISA kit. Total protein in TA was measured by Bradford assay to correct for sampling related dilution. BPD was defined as the need of supplemental oxygen at 36 weeks postmenstrual age (PMA). RESULTS Twenty infants (GA 26.4 ± 1.9w, BW 860 ± 201 g) survived without BPD at 36 weeks PMA and 59 infants (GA 25.5 ± 1.5w, BW 751 ± 163 g) died before 36 weeks PMA or developed BPD. The mean IFN-γ level was higher in infants who died or developed BPD (9.7 ± 2.8 vs. 3.1 ± 1.1 pg/ml, P = 0.03). Similarly, the mean IP-10 level was higher in infants who died or developed BPD (63.4 ± 17.5 pg/ml) compared to those who survived without BPD (18.5 ± 7.5 pg/ml, P = 0.02). CONCLUSIONS Higher IFN-γ and IP-10 levels in TA samples are associated with the development of BPD or death in premature infants.
Collapse
Affiliation(s)
- Zubair H Aghai
- Department of Pediatrics, Cooper University Hospital, Camden, New Jersey, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Genetic associations of surfactant protein D and angiotensin-converting enzyme with lung disease in preterm neonates. J Perinatol 2012; 32:349-55. [PMID: 21960125 PMCID: PMC3370386 DOI: 10.1038/jp.2011.104] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To replicate genetic associations with respiratory distress syndrome (RDS) and bronchopulmonary dysplasia (BPD) in genes related to surfactant deficiency, inflammation and infection, and the renin-angiotensin system. STUDY DESIGN We examined eight candidate genes for associations with RDS and BPD in 433 preterm birth (PTB-<37 weeks) infants (251 with RDS and 134 with BPD). Both case-control and family-based analyses were performed in preterm (<37 weeks) and very preterm birth (VPTB-<32 weeks) infants. RESULT We replicated a previous finding that rs1923537, a marker downstream of surfactant protein D (SFTPD) is associated with RDS in VPTB infants in that the T allele was overtransmitted from parents to offspring with RDS (P=8.4 × 10(-3)). We also observed the A allele of rs4351 in the angiotensin-converting enzyme (ACE) gene was overtransmitted from parents to VPTB offspring with BPD (P=9.8 × 10(-3)). CONCLUSION These results give further insight into the genetic risk factors for complex neonatal respiratory diseases and provide more evidence of the importance of SFTPD and ACE in the etiology of RDS and BPD, respectively.
Collapse
|
22
|
Abstract
The ductus arteriosus is a vital fetal structure designed to close shortly after birth. Although many physiologic and pharmacologic investigations have characterized the closure of this structure, genetic studies of persistent patency of the ductus arteriosus (patent ductus arteriosus, PDA) are relatively recent. Progress in the identification of specific genes associated with PDA is well behind that of many adult-onset diseases because of several reasons ranging from the lack of large biorepositories for this unique population to the belief that any genetic contribution to PDA is minimal. Viewing the PDA as a complex, developmentally influenced disease with both genetic and environmental risk factors has resulted in initial successes in some genetic studies. We will introduce several genetic approaches, which have been or are currently being applied to the study of PDA, that have been successful in identifying polymorphisms associated with adult diseases. Genetic investigations of PDA will be discussed with respect to heritability, in general, and to specific risk genes. Several animal models that have been used to study PDA-related genes will also be presented. Further advances in discovering genetic variation causing PDA will drive the more rational use of current therapies, and may help identify currently unknown targets for future therapeutic manipulation.
Collapse
Affiliation(s)
- Hanine Hajj
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
23
|
Stoller JZ, Demauro SB, Dagle JM, Reese J. Current Perspectives on Pathobiology of the Ductus Arteriosus. ACTA ACUST UNITED AC 2012; 8. [PMID: 23519783 DOI: 10.4172/2155-9880.s8-001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The ductus arteriosus (DA) shunts blood away from the lungs during fetal life, but at birth this shunt is no longer needed and the vessel rapidly constricts. Postnatal persistence of the DA, patent ductus arteriosus (PDA), is predominantly a detrimental condition for preterm infants but is simultaneously a condition required to maintain systemic blood flow for infants born with certain severe congenital heart defects. Although PDA in preterm infants is associated with significant morbidities, there is controversy regarding whether PDA is truly causative. Despite advances in our understanding of the pathobiology of PDA, the optimal treatment strategy for PDA in preterm infants is unclear. Here we review recent studies that have continued to elucidate the fundamental mechanisms of DA development and pathogenesis.
Collapse
Affiliation(s)
- Jason Z Stoller
- Department of Pediatrics, University of Pennsylvania School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
24
|
Wright CJ, Kirpalani H. Targeting inflammation to prevent bronchopulmonary dysplasia: can new insights be translated into therapies? Pediatrics 2011; 128:111-26. [PMID: 21646264 PMCID: PMC3124103 DOI: 10.1542/peds.2010-3875] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) frequently complicates preterm birth and leads to significant long-term morbidity. Unfortunately, few therapies are known to effectively prevent or treat BPD. Ongoing research has been focusing on potential therapies to limit inflammation in the preterm lung. In this review we highlight recent bench and clinical research aimed at understanding the role of inflammation in the pathogenesis of BPD. We also critically assess currently used therapies and promising developments in the field.
Collapse
Affiliation(s)
- Clyde J. Wright
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; ,Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; and
| | - Haresh Kirpalani
- Division of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; ,Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; and ,Department of Clinical Epidemiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
25
|
Fanos V, Pusceddu M, Dessì A, Marcialis MA. Should we definitively abandon prophylaxis for patent ductus arteriosus in preterm new-borns? Clinics (Sao Paulo) 2011; 66:2141-9. [PMID: 22189742 PMCID: PMC3226612 DOI: 10.1590/s1807-59322011001200022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Accepted: 08/12/2011] [Indexed: 12/30/2022] Open
Abstract
Although the prophylactic administration of indomethacin in extremely low-birth weight infants reduces the frequency of patent ductus arteriosus and severe intraventricular hemorrhage, it does not appear to provide any long-term benefit in terms of survival without neurosensory and cognitive outcomes. Considering the increased drug-induced reduction in renal, intestinal, and cerebral blood flow, the use of prophylaxis cannot be routinely recommended in preterm neonates. However, a better understanding of the genetic background of each infant may allow for individualized prophylaxis using NSAIDs and metabolomics.
Collapse
Affiliation(s)
- Vassilios Fanos
- Neonatal Intensive Care Unit, Puericulture Institute And Neonatal Section, AOU University of Cagliari, Italy
| | | | | | | |
Collapse
|
26
|
Waleh N, Hodnick R, Jhaveri N, McConaghy S, Dagle J, Seidner S, McCurnin D, Murray JC, Ohls R, Clyman RI. Patterns of gene expression in the ductus arteriosus are related to environmental and genetic risk factors for persistent ductus patency. Pediatr Res 2010; 68:292-7. [PMID: 20581741 PMCID: PMC2940964 DOI: 10.1203/pdr.0b013e3181ed8609] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Three independent risk factors (immature gestation, absence of antenatal glucocorticoid exposure, and presence of the rs2817399(A) allele of the gene TFAP2B) are associated with patent ductus arteriosus (PDAs) that fail to close during prostaglandin inhibition. We hypothesized that these three factors may affect a common set of genes that increase the risk of persistent PDA after birth. We studied baboon ductus from term, preterm, and glucocorticoid-treated preterm fetuses and found that both immature gestation and absence of antenatal glucocorticoid exposure decreased RNA expression of calcium- and potassium-channel genes involved in oxygen-induced constriction, and phosphodiesterase genes (that modulate cAMP/cGMP signaling). Ductus obtained from second trimester human pregnancies were genotyped for TFAP2B polymorphisms. When present, the rs2817399(A) allele also was associated with decreased expression of calcium- and potassium-channel genes. In contrast, alleles of two other TFAP2B polymorphisms, rs2817419(G) and rs2635727(T), which are not related to the incidence of PDA after birth, had no effect on RNA expression. In conclusion, three calcium- and potassium-channel genes (CACNA1G/ alpha1G, CACNB 2/CaL-beta2, and KCNA2/ Kv1.2) were similarly affected by each of the PDA risk factors. We speculate that these channels may play a significant role in closing the preterm ductus during prostaglandin inhibition and may be potential targets for future pharmacologic manipulations.
Collapse
Affiliation(s)
- Nahid Waleh
- Pharmaceutical Discovery Division, SRI International, Menlo Park, California 94025, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW According to recent evidence, susceptibility to bronchopulmonary dysplasia (BPD) in preterm infants is predominantly inherited. The purpose of this review is to discuss current published genetic association studies in light of the accumulated knowledge in genomics. RECENT FINDINGS Major advances in the development of next-generation genotyping and sequencing platforms, statistical methodologies, inventories of functional outcome of some common genetic polymorphisms and large-scale cataloguing of genetic variability among many of the world's ethnic populations have greatly facilitated the study of polygenic conditions. For BPD, genetic-association studies have primarily focused on components of innate (e.g. first-line) immune and antioxidant defences, mechanisms of vascular and lung remodelling, and surfactant proteins. However, studies have been limited in sample size and therefore fraught with a high probability of false-positive and false-negative associations. Nonetheless, candidate gene associations have indicated some novel biological pathways and provide a conceptual framework for the design of larger, collaborative population-based studies. SUMMARY Although studies to date have not been able to identify reproducible genetic risk markers for BPD, they have directed us towards new, promising research avenues.
Collapse
|
28
|
Bhandari V, Zhou G, Bizzarro MJ, Buhimschi C, Hussain N, Gruen JR, Zhang H. Genetic contribution to patent ductus arteriosus in the premature newborn. Pediatrics 2009; 123:669-73. [PMID: 19171636 PMCID: PMC3161726 DOI: 10.1542/peds.2008-1117] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The most common congenital heart disease in the newborn population, patent ductus arteriosus, accounts for significant morbidity in preterm newborns. In addition to prematurity and environmental factors, we hypothesized that genetic factors play a significant role in this condition. OBJECTIVE The objective of this study was to quantify the contribution of genetic factors to the variance in liability for patent ductus arteriosus in premature newborns. PATIENTS AND METHODS A retrospective study (1991-2006) from 2 centers was performed by using zygosity data from premature twins born at < or =36 weeks' gestational age and surviving beyond 36 weeks' postmenstrual age. Patent ductus arteriosus was diagnosed by echocardiography at each center. Mixed-effects logistic regression was used to assess the effect of specific covariates. Latent variable probit modeling was then performed to estimate the heritability of patent ductus arteriosus, and mixed-effects probit modeling was used to quantify the genetic component. RESULTS We obtained data from 333 dizygotic twin pairs and 99 monozygotic twin pairs from 2 centers (Yale University and University of Connecticut). Data on chorioamnionitis, antenatal steroids, gestational age, body weight, gender, respiratory distress syndrome, patent ductus arteriosus, necrotizing enterocolitis, oxygen supplementation, and bronchopulmonary dysplasia were comparable between monozygotic and dizygotic twins. We found that gestational age, respiratory distress syndrome, and institution were significant covariates for patent ductus arteriosus. After controlling for specific covariates, genetic factors or the shared environment accounted for 76.1% of the variance in liability for patent ductus arteriosus. CONCLUSIONS Preterm patent ductus arteriosus is highly familial (contributed to by genetic and environmental factors), with the effect being mainly environmental, after controlling for known confounders.
Collapse
Affiliation(s)
- Vineet Bhandari
- Yale University School of Medicine, Department of Pediatrics, 333 Cedar St, PO Box 208064, New Haven, CT 06520-8064, USA.
| | - Gongfu Zhou
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut
| | - Matthew J. Bizzarro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Catalin Buhimschi
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut
| | - Naveed Hussain
- Division of Neonatology, University of Connecticut Health Center, Farmington, Connecticut
| | - Jeffrey R. Gruen
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut,Department of Genetics, Yale University School of Medicine, New Haven, Connecticut,Department of Investigative Medicine, Yale University School of Medicine, New Haven, Connecticut,Department of Yale Child Health Research Center, Yale University School of Medicine, New Haven, Connecticut
| | - Heping Zhang
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
29
|
Lin PW, Nasr TR, Stoll BJ. Necrotizing enterocolitis: recent scientific advances in pathophysiology and prevention. Semin Perinatol 2008; 32:70-82. [PMID: 18346530 DOI: 10.1053/j.semperi.2008.01.004] [Citation(s) in RCA: 202] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality among infants in the neonatal intensive care unit. Here we review the epidemiology and pathophysiology of NEC, with an emphasis on the latest research findings and potential areas for future research. NEC continues to be one of the most devastating and unpredictable diseases affecting premature infants. Despite decades of research, the pathogenesis of this disease remains unclear, and prevention and treatment strategies are limited. Hopefully, future studies aimed at understanding premature intestinal defenses, commensal or probiotic bacterial influences, and possible genetic predisposition will lead to the improvement of prevention and treatment strategies.
Collapse
Affiliation(s)
- Patricia W Lin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.
| | | | | |
Collapse
|
30
|
Abstract
Bronchopulmonary dysplasia (BPD) is a common perinatal complication of very low birth weight preterm infants with a significant risk of long-term disability and morbidity. While clinical conditions such as prematurity and mechanical ventilation are its major risk factors, studies suggest that there is an individual susceptibility to BPD. This comprehensive review summarizes data collected about the implication of genetic polymorphisms in BPD and in its risk factors. Some studies have directly related the risk of BPD to genotype. Indeed, carrier states of genetic variants of cytokines (IFNgamma T+874A), adhesion molecules (L-selectin-Pro213Ser), elements of renin-angiotensin system (ACE-I/D), antioxidant enzymes (GST-P1 Val105Ile), and surfactant proteins (SPA1, SPB intron 4) has been identified as risk factors to BPD. Other studies investigated the role of genotype in BPD risk factors. Premature birth has been linked to carrier states of genetic variants with an impact on immune status (such as IL-6 G(-174)C, MBL2 54G/A, VEGF G+405C, HSP72 A+1267G genes) and matrix metalloproteases. Fetal inflammatory response syndrome, a major determinant of BPD is also affected by genotype (including LTalpha A+250G). Disturbed intrauterine lung development and vascularization may also contribute to BPD; these processes may be impaired in the presence of some rare genetic mutations. Furthermore, there is also a genetic component in the susceptibility to other perinatal adaptational disturbances such as respiratory distress syndrome that are associated with an increased need for mechanical ventilation, and, hence, with lung damage. The genetic variants presented in this article may help to identify infants at risk for BPD.
Collapse
Affiliation(s)
- Géza Bokodi
- Ist Department of Pediatrics, Semmelweis University, Budapest, Hungary.
| | | | | | | | | |
Collapse
|
31
|
Hallman M, Marttila R, Pertile R, Ojaniemi M, Haataja R. Genes and environment in common neonatal lung disease. Neonatology 2007; 91:298-302. [PMID: 17575473 DOI: 10.1159/000101345] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Respiratory distress syndrome (RDS) and bronchopulmonary dysplasia (BPD) are common, serious lung diseases in preterm infants. Polymorphism of the genes involved in basic lung function and alveolar stability, lung differentiation and pulmonary host defense may influence the risk. Natural selection has refined the genes responsible for cardiopulmonary adaptation and resistance against pneumonia in term and near-term infants. Before the era of antibiotics, however, virtually all very preterm infants died of asphyxia, respiratory failure or infections. Today, the degree of prematurity plays a dominant role in susceptibility to serious lung disease. In addition, genetic polymorphism and constitution modulate the risk of RDS and BPD that have different, partly overlapping predisposition. According to twin studies, the genetic impact on the risk of RDS and BPD among preterm and very preterm infants is 35-65%. Individual disease genes generally have low penetrance. Large-scale genetic studies are required as part of neonatal and perinatal research in order to learn about the risk factors and to investigate pharmacogenetics. The aim in the future is to individualize therapies.
Collapse
Affiliation(s)
- Mikko Hallman
- Department of Pediatrics, Biocenter Oulu, University of Oulu, Oulu, Finland.
| | | | | | | | | |
Collapse
|