1
|
Casey A, Fiorino EK, Wambach J. Innovations in Childhood Interstitial and Diffuse Lung Disease. Clin Chest Med 2024; 45:695-715. [PMID: 39069332 PMCID: PMC11366208 DOI: 10.1016/j.ccm.2024.04.002] [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] [Indexed: 07/30/2024]
Abstract
Children's interstitial and diffuse lung diseases (chILDs) are a heterogenous and diverse group of lung disorders presenting during childhood. Infants and children with chILD disorders present with respiratory signs and symptoms as well as diffuse lung imaging abnormalities. ChILD disorders are associated with significant health care resource utilization and high morbidity and mortality. The care of patients with chILD has been improved through multidisciplinary care, multicenter collaboration, and the establishment of patient research networks in the United Stated and abroad. This review details past and current innovations in the diagnosis and clinical care of children with chILD.
Collapse
Affiliation(s)
- Alicia Casey
- Department of Pediatrics, Division of Pulmonary Medicine, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115, USA.
| | - Elizabeth K Fiorino
- Department of Science Education and Pediatrics, Donald and Barabara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Jennifer Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Jain KG, Liu Y, Zhao R, Muire PJ, Zhang J, Zang QS, Ji HL. Humanized L184Q Mutated Surfactant Protein C Gene Alters Alveolar Type 2 Epithelial Cell Fate. Int J Mol Sci 2024; 25:8723. [PMID: 39201410 PMCID: PMC11354303 DOI: 10.3390/ijms25168723] [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: 06/13/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Alveolar type 2 epithelial (AT2) cells synthesize surfactant protein C (SPC) and repair an injured alveolar epithelium. A mutated surfactant protein C gene (SftpcL184Q, Gene ID: 6440) in newborns has been associated with respiratory distress syndrome and pulmonary fibrosis. However, the underlying mechanisms causing Sftpc gene mutations to regulate AT2 lineage remain unclear. We utilized three-dimensional (3D) feeder-free AT2 organoids in vitro to simulate the alveolar epithelium and compared AT2 lineage characteristics between WT (C57BL/6) and SftpcL184Q mutant mice using colony formation assays, immunofluorescence, flow cytometry, qRT-PCR, and Western blot assays. The AT2 numbers were reduced significantly in SftpcL184Q mice. Organoid numbers and colony-forming efficiency were significantly attenuated in the 3D cultures of primary SftpcL184Q AT2 cells compared to those of WT mice. Podoplanin (PDPN, Alveolar type 1 cell (AT1) marker) expression and transient cell count was significantly increased in SftpcL184Q organoids compared to in the WT mice. The expression levels of CD74, heat shock protein 90 (HSP90), and ribosomal protein S3A1 (RPS3A1) were not significantly different between WT and SftpcL184Q AT2 cells. This study demonstrated that humanized SftpcL184Q mutation regulates AT2 lineage intrinsically. This regulation is independent of CD74, HSP90, and RPS3A1 pathways.
Collapse
Affiliation(s)
- Krishan G. Jain
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Yang Liu
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Runzhen Zhao
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
| | - Preeti J. Muire
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
- Department of Orthopedics and Rehabilitation, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
- Infectious Diseases and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL 60153, USA;
- Departments of Pathology and Radiation Oncology, Loyola University Medical Center, Maywood, IL 60153, USA
| | - Qun Sophia Zang
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
| | - Hong-Long Ji
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA; (K.G.J.); (R.Z.); (Q.S.Z.)
- Burn and Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA;
- Infectious Diseases and Immunology Research Institute, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
3
|
Ikrin AN, Moskalenko AM, Mukhamadeev RR, de Abreu MS, Kolesnikova TO, Kalueff AV. The emerging complexity of molecular pathways implicated in mouse self-grooming behavior. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110840. [PMID: 37580009 DOI: 10.1016/j.pnpbp.2023.110840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/29/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Rodent self-grooming is an important complex behavior, and its deficits are translationally relevant to a wide range of neuropsychiatric disorders. Here, we analyzed a comprehensive dataset of 227 genes whose mutations are known to evoke aberrant self-grooming in mice. Using these genes, we constructed the network of their established protein-protein interactions (PPI), yielding several distinct molecular clusters related to postsynaptic density, the Wnt signaling, transcription factors, neuronal cell cycle, NOS neurotransmission, microtubule regulation, neuronal differentiation/trafficking, neurodevelopment and mitochondrial function. Utilizing further bioinformatics analyses, we also identified novel central ('hub') proteins within these clusters, whose genes may also be implicated in aberrant self-grooming and other repetitive behaviors in general. Untangling complex molecular pathways of this important behavior using in silico approaches contributes to our understanding of related neurological disorders, and may suggest novel potential targets for their pharmacological or gene therapy.
Collapse
Affiliation(s)
- Aleksey N Ikrin
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Anastasia M Moskalenko
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Radmir R Mukhamadeev
- Graduate Program in Bioinformatics and Genomics, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Murilo S de Abreu
- Moscow Institute of Science and Technology, Dolgoprudny 197028, Russia.
| | - Tatiana O Kolesnikova
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Allan V Kalueff
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 194021, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny 197758, Russia; Neuroscience Group, Ural Federal University, Ekaterinburg 620002, Russia; Laboratory of Translational Biopsychiatry, Scientific Research Institute of Neurosciences and Medicine, Novosibirsk 630117, Russia.
| |
Collapse
|
4
|
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
|
5
|
Abdel Megeid AK, Refeat MM, Ashaat EA, El-Kamah G, El-Saiedi SA, Elfalaki MM, El Ruby MO, Amr KS. Correlating SFTPC gene variants to interstitial lung disease in Egyptian children. J Genet Eng Biotechnol 2022; 20:117. [PMID: 35939165 PMCID: PMC9360361 DOI: 10.1186/s43141-022-00399-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 07/10/2022] [Indexed: 01/04/2023]
Abstract
Background Interstitial lung disease (ILD) is a broad heterogeneous group of lung disorders that is characterized by inflammation of the lungs. Surfactant dysfunction disorders are a rare form of ILD diseases that result from mutations in surfactant protein C gene (SFTPC) with prevalence of approximately 1/1.7 million births. SFTPC patients are presented with clinical manifestations of ILD ranging from fatal respiratory failure of newborn to chronic respiratory problems in children. In the current study, we aimed to investigate the spectrum of SFTPC genetic variants as well as the correlation of the SFTPC gene mutations with ILD disease in twenty unrelated Egyptian children with diffuse lung disease and suspected surfactant dysfunction using Sanger sequencing. Results Sequencing of SFTPC gene revealed five variants: c.42+35G>A (IVS1+35G>A) (rs8192340) and c.43-21T>C (IVS1-21T>C) (rs13248346) in intron 1, c.436-8C>G (IVS4-8C>G) (rs2070687) in intron 4, c.413C>A p.T138N (rs4715) in exon 4, and c.557G>Ap.S186N (rs1124) in exon 5. Conclusion The present study confirms the association of detecting variants of SFTPC with surfactant dysfunction disorders.
Collapse
Affiliation(s)
| | - Miral M Refeat
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Engy A Ashaat
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Ghada El-Kamah
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | | | | | - Mona O El Ruby
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Khalda S Amr
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| |
Collapse
|
6
|
Amatya S, Ye M, Yang L, Gandhi CK, Wu R, Nagourney B, Floros J. Single Nucleotide Polymorphisms Interactions of the Surfactant Protein Genes Associated With Respiratory Distress Syndrome Susceptibility in Preterm Infants. Front Pediatr 2021; 9:682160. [PMID: 34671583 PMCID: PMC8521105 DOI: 10.3389/fped.2021.682160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/06/2021] [Indexed: 11/14/2022] Open
Abstract
Background: Neonatal respiratory distress syndrome (RDS), due to surfactant deficiency in preterm infants, is the most common cause of respiratory morbidity. The surfactant proteins (SFTP) genetic variants have been well-studied in association with RDS; however, the impact of SNP-SNP (single nucleotide polymorphism) interactions on RDS has not been addressed. Therefore, this study utilizes a newer statistical model to determine the association of SFTP single SNP model and SNP-SNP interactions in a two and a three SNP interaction model with RDS susceptibility. Methods: This study used available genotype and clinical data in the Floros biobank at Penn State University. The patients consisted of 848 preterm infants, born <36 weeks of gestation, with 477 infants with RDS and 458 infants without RDS. Seventeen well-studied SFTPA1, SFTPA2, SFTPB, SFTPC, and SFTPD SNPs were investigated. Wang's statistical model was employed to test and identify significant associations in a case-control study. Results: Only the rs17886395 (C allele) of the SFTPA2 was associated with protection for RDS in a single-SNP model (Odd's Ratio 0.16, 95% CI 0.06-0.43, adjusted p = 0.03). The highest number of interactions (n = 27) in the three SNP interactions were among SFTPA1 and SFTPA2. The three SNP models showed intergenic and intragenic interactions among all SFTP SNPs except SFTPC. Conclusion: The single SNP model and SNP interactions using the two and three SNP interactions models identified SFTP-SNP associations with RDS. However, the large number of significant associations containing SFTPA1 and/or SFTPA2 SNPs point to the importance of SFTPA1 and SFTPA2 in RDS susceptibility.
Collapse
Affiliation(s)
- Shaili Amatya
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Meixia Ye
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Lili Yang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chintan K Gandhi
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rongling Wu
- Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Beth Nagourney
- Albert Einstein College of Medicine, New York, NY, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
7
|
Nefzi M, Wahabi I, Hadj Fredj S, Othmani R, Dabboubi R, Boussetta K, Fanen P, Messaoud T. Association analysis of the surfactant protein-C gene to childhood asthma. J Asthma 2020; 59:1-11. [PMID: 32962475 DOI: 10.1080/02770903.2020.1827419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES This study aims to describe the molecular variability in the SFTPC gene in a childhood chronic respiratory disease, asthma, in the Tunisian population and to identify the implications based on a case-control study of p.Thr138Asn (T138N) and p.Ser186Asn (S186N) variants. METHODS We used direct sequencing for the genotyping of the SFTPC gene within 101 asthmatic children. The study of T138N and S186N variants in 110 controls is conducted by the PCR-RFLP technique. RESULTS The molecular study revealed 26 variants including 24 intronic variations and 2 exonic variations (T138N and S186N) with respective frequencies of 16.8% and 18.3%. We conducted a case-control study of the two identified exonic variations. A different genotypic and allelic distribution between the two groups was noted. Only the T138N polymorphism showed a significant association with asthma disease (p < 1 0 -3). Statistical analysis elaborated four haplotypes with the following frequencies in patients vs controls: 138Thr-186Ser (79.5% vs 57.6%), 138Thr-186Asn (3.7% vs 7.8%), 138Asn-186Thr (2.2% vs 20.2%) and 138Asn-186Asn (14.6% vs 14.4%). A significant difference (p < 1 0 -3) was highlighted in haplotype distribution. The 138Asn-186Ser (OR [95%CI] = 0.14[0.04-0.54], p = 0.004, R2=0.93) and 138Thr-186Asn (OR [95%CI] = 0.35[0.12-0.54], p = 0.047, R2=0.88) haplotypes showed a negative association with asthma which may constitute a protective factor against the disease. CONCLUSION In Tunisia, this work constitutes the first report interested in the SFTPC gene and highlights the genetic variability of the SFTPC gene in asthma. Therefore, the case-controls analysis may be useful in the study of surfactant proteins dysfunction in chronic respiratory disease at an early age.
Collapse
Affiliation(s)
- Malek Nefzi
- Biochemistry Laboratory (LR00SP03), Children's Hospital, Tunis, Tunisia
| | - Imen Wahabi
- Biochemistry Laboratory (LR00SP03), Children's Hospital, Tunis, Tunisia
| | | | - Rym Othmani
- Biochemistry Laboratory (LR00SP03), Children's Hospital, Tunis, Tunisia
| | - Rym Dabboubi
- Biochemistry Laboratory (LR00SP03), Children's Hospital, Tunis, Tunisia
| | | | - Pascale Fanen
- Genetic Department, AP-HP, Henri Mondor Hospital, Creteil, France
| | - Taieb Messaoud
- Biochemistry Laboratory (LR00SP03), Children's Hospital, Tunis, Tunisia
| |
Collapse
|
8
|
Zhao JW, Jiao L, Guo MM, Zheng L, Wang XB, Gao SH, Ying BW, Ming L. SFTPC genetic polymorphisms are associated with tuberculosis susceptibility and clinical phenotype in a Western Chinese Han population. Exp Ther Med 2020; 20:100. [PMID: 32973949 PMCID: PMC7507020 DOI: 10.3892/etm.2020.9230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 08/19/2020] [Indexed: 02/05/2023] Open
Abstract
Tuberculosis (TB) is one of the most common infectious diseases globally. The surfactant protein C (SFTPC), which is involved in innate immunity and surfactant function in the lung, may contribute toward the progression of TB. The aim of the present study was to preliminarily investigate the possible association of single nucleotide polymorphisms (SNPs) in the SFTPC gene with TB susceptibility and clinical phenotypes in a Western Chinese Han population. The improved multiplex ligation detection reaction method was used to genotype 6 SNPs in SFTPC, in 900 patients with TB and 1,534 healthy control subjects. It was found that the A allele for rs1124 and the C allele for rs8192313 were associated with increased susceptibility to TB, P=0.024 and P=0.045, respectively. However, these two P-values were not significant following Bonferroni correction. In all samples, the haplotype [CGA], representing three SFTPC variants, was revealed to increase the risk of TB (P=0.001 and P=0.005, following Bonferroni correction). Furthermore, patients with the AA genotype for rs1124 and with the CC genotype for rs8192313 were associated with higher levels of C-reactive protein (P=0.001 and P=0.005, respectively). The results of the present study indicated that the SFTPC SNPs may increase the susceptibility to TB and the immune response of the host to Mycobacterium tuberculosis and may potentially be novel biomarkers for the pathogenesis of TB.
Collapse
Affiliation(s)
- Jun-Wei Zhao
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, P.R. China
| | - Lin Jiao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Man-Man Guo
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, P.R. China
| | - Lei Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, P.R. China
| | - Xue-Bin Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, P.R. China
| | - Shu-Hui Gao
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, P.R. China
| | - Bin-Wu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Liang Ming
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
9
|
Sehlmeyer K, Ruwisch J, Roldan N, Lopez-Rodriguez E. Alveolar Dynamics and Beyond - The Importance of Surfactant Protein C and Cholesterol in Lung Homeostasis and Fibrosis. Front Physiol 2020; 11:386. [PMID: 32431623 PMCID: PMC7213507 DOI: 10.3389/fphys.2020.00386] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Surfactant protein C (SP-C) is an important player in enhancing the interfacial adsorption of lung surfactant lipid films to the alveolar air-liquid interface. Doing so, surface tension drops down enough to stabilize alveoli and the lung, reducing the work of breathing. In addition, it has been shown that SP-C counteracts the deleterious effect of high amounts of cholesterol in the surfactant lipid films. On its side, cholesterol is a well-known modulator of the biophysical properties of biological membranes and it has been proven that it activates the inflammasome pathways in the lung. Even though the molecular mechanism is not known, there are evidences suggesting that these two molecules may interplay with each other in order to keep the proper function of the lung. This review focuses in the role of SP-C and cholesterol in the development of lung fibrosis and the potential pathways in which impairment of both molecules leads to aberrant lung repair, and therefore impaired alveolar dynamics. From molecular to cellular mechanisms to evidences in animal models and human diseases. The evidences revised here highlight a potential SP-C/cholesterol axis as target for the treatment of lung fibrosis.
Collapse
Affiliation(s)
- Kirsten Sehlmeyer
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
| | - Jannik Ruwisch
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
| | - Nuria Roldan
- Alveolix AG and ARTORG Center, University of Bern, Bern, Switzerland
| | - Elena Lopez-Rodriguez
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
- Institute of Functional Anatomy, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
10
|
Lin Z, Thorenoor N, Wu R, DiAngelo SL, Ye M, Thomas NJ, Liao X, Lin TR, Warren S, Floros J. Genetic Association of Pulmonary Surfactant Protein Genes, SFTPA1, SFTPA2, SFTPB, SFTPC, and SFTPD With Cystic Fibrosis. Front Immunol 2018; 9:2256. [PMID: 30333828 PMCID: PMC6175982 DOI: 10.3389/fimmu.2018.02256] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/11/2018] [Indexed: 01/03/2023] Open
Abstract
Surfactant proteins (SP) are involved in surfactant function and innate immunity in the human lung. Both lung function and innate immunity are altered in CF, and altered SP levels and genetic association are observed in Cystic Fibrosis (CF). We hypothesized that single nucleotide polymorphisms (SNPs) within the SP genes associate with CF or severity subgroups, either through single SNP or via SNP-SNP interactions between two SNPs of a given gene (intragenic) and/or between two genes (intergenic). We genotyped a total of 17 SP SNPs from 72 case-trio pedigree (SFTPA1 (5), SFTPA2 (4), SFTPB (4), SFTPC (2), and SFTPD (2)), and identified SP SNP associations by applying quantitative genetic principles. The results showed (a) Two SNPs, SFTPB rs7316 (p = 0.0083) and SFTPC rs1124 (p = 0.0154), each associated with CF. (b) Three intragenic SNP-SNP interactions, SFTPB (rs2077079, rs3024798), and SFTPA1 (rs1136451, rs1059057 and rs4253527), associated with CF. (c) A total of 34 intergenic SNP-SNP interactions among the 4 SP genes to be associated with CF. (d) No SNP-SNP interaction was observed between SFTPA1 or SFTPA2 and SFTPD. (e) Equal number of SNP-SNP interactions were observed between SFTPB and SFTPA1/SFTPA2 (n = 7) and SP-B and SFTPD (n = 7). (f) SFTPC exhibited significant SNP-SNP interactions with SFTPA1/SFTPA2 (n = 11), SFTPB (n = 4) and SFTPD (n = 3). (g) A single SFTPB SNP was associated with mild CF after Bonferroni correction, and several intergenic interactions that are associated (p < 0.01) with either mild or moderate/severe CF were observed. These collectively indicate that complex SNP-SNP interactions of the SP genes may contribute to the pulmonary disease in CF patients. We speculate that SPs may serve as modifiers for the varied progression of pulmonary disease in CF and/or its severity.
Collapse
Affiliation(s)
- Zhenwu Lin
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Nithyananda Thorenoor
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Rongling Wu
- Public Health Science, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Susan L. DiAngelo
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Meixia Ye
- Public Health Science, College of Medicine, Pennsylvania State University, Hershey, PA, United States
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Neal J. Thomas
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Xiaojie Liao
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Tony R. Lin
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Stuart Warren
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
- Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
11
|
Chen YJ, Meyer J, Wambach JA, DePass K, Wegner DJ, Fan X, Zhang QY, Hillary H, Cole FS, Hamvas A. Gene variants of the phosphatidylcholine synthesis pathway do not contribute to RDS in the Chinese population. World J Pediatr 2018; 14:52-56. [PMID: 29411327 DOI: 10.1007/s12519-017-0109-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/03/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND To determine population-based prevalence and disease contribution of phosphatidylcholine synthetic pathway-associated gene variants in a native southern Chinese cohort. METHODS We used bloodspots from 2010 that were obtained from the Guangxi Neonatal Screening Center in Nannning China and included the Han (n = 443) and Zhuang (n = 313) ethnic groups. We sequenced the exons of cholinephosphate cytidylyltransferase (PCYT1B) lysophospholipid acyltransferase 1 (LPCAT1), and cholinephosphotransferase (CHPT1) genes, and analyzed both rare and common exonic variants. RESULTS We obtained five mutations (G199D, A299V, G434C, Y490C, L312S) with eight alleles in the three candidate genes. The collapsed minor allele frequency for candidate genes was not significantly different between the Han and Zhuang populations (0.0045 vs. 0.0064, respectively, P = 0.725). The combined Han and Zhuang pool collapsed carrier frequency of rare mutation allele was found to be 1.06%, which is much higher than previously reported for the Missouri population (0.1%). Further, we detected six exonic common variants (three in LPCAT1 and three in CHPT1), with three non-synonymous variants (F162S, F341L, M427K) among them. Two of the non-synonymous exonic variants (F341L, M427K) were not found in CHB; F341L was also not previously reported in exome sequencing project. CONCLUSIONS The population-based frequency of mutations in the phosphatidylcholine synthesis pathway-associated genes PCYT1B LPCAT1, CHPT1 is low in southern Chinese newborns and there is no evidence of contribution to population-based disease burden of respiratory distress syndrome. As a population-based study of rare mutations and common variants, this work is valuable in directing future research.
Collapse
Affiliation(s)
- Yu-Jun Chen
- Neonatology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Julia Meyer
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Jennifer A Wambach
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Kelcey DePass
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Daniel J Wegner
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Xin Fan
- Guangxi Maternal and Child Health Hospital, Nanning, China
| | - Qun-Yuan Zhang
- Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - Heins Hillary
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - F Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Aaron Hamvas
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA. .,Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, 225 E. Chicago Ave, Box #45, Chicago, IL, 60611, USA.
| |
Collapse
|
12
|
Puthusseri B, Marudamuthu A, Tiwari N, Fu J, Idell S, Shetty S. Regulation of p53-mediated changes in the uPA-fibrinolytic system and in lung injury by loss of surfactant protein C expression in alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L783-L796. [PMID: 28385810 DOI: 10.1152/ajplung.00291.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 03/21/2017] [Accepted: 04/03/2017] [Indexed: 01/24/2023] Open
Abstract
Pulmonary surfactant protein C (SP-C) expression by type II alveolar epithelial cells (AECs) is markedly reduced in diverse types of lung injuries and is often associated with AEC apoptosis. It is unclear whether loss of SP-C contributes to the increased p53 and urokinase-type plasminogen activator (uPA) system cross-talk and apoptosis of AECs. Therefore, we inhibited SP-C expression in human and murine AECs using lentivirus vector expressing shRNA and tested p53 and downstream changes in the uPA-fibrinolytic system. Inhibition of SP-C expression in AECs induced p53 and activated caspase-3, indicating AEC apoptosis. We also found that bleomycin or cigarette smoke exposure failed to inhibit SP-C expression or apoptosis in AECs in p53- and plasminogen activator inhibitor-1 (PAI-1)-deficient mice. Depletion of SP-C expression by lentiviral SP-C shRNA in PAI-1-deficient mice failed to induce p53 or apoptosis in AECs, whereas it increased both AEC p53 and apoptosis in wild-type and uPA-deficient mice. SP-C inhibition in AECs also increased in CXCL1 and CXCL2 and their receptor CXCR2 as well as ICAM-1 expression, which is indicative of a proinflammatory response. Overexpression of p53-binding 3'-UTR sequences in AECs inhibited PAI-1 induction while maintaining uPA and uPAR protein and mRNA expression. Furthermore, caveolin-1 expression and phosphorylation were increased in AECs, indicating an intricate link between caveolin-1 and Src kinase-mediated cell signaling and AEC apoptosis due to loss of SP-C expression through p53 and uPA system-mediated cross-talk. The role of uPA, PAI-1, and p53 in the regulation of AEC apoptosis after injury was also determined in knockout mice.
Collapse
Affiliation(s)
- Bijesh Puthusseri
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Amarnath Marudamuthu
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Nivedita Tiwari
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Jian Fu
- Center for Research on Environmental Disease and Toxicology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Steven Idell
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; and
| | - Sreerama Shetty
- Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, Tyler, Texas; and
| |
Collapse
|
13
|
Fatahi N, Dalili H, Kalani M, Niknafs N, Shariat M, Tavakkoly-Bazzaz J, Amini E, Esmaeilnia Shirvani T, Hardani AK, Taheritafti R, Ghasemi-Fakhr N, Ghadami M, Nayeri F, Rashidi-Nezhad A. Association of SP-C gene codon 186 polymorphism (rs1124) and risk of RDS. J Matern Fetal Neonatal Med 2016; 30:2585-2589. [DOI: 10.1080/14767058.2016.1256994] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Neda Fatahi
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Ronash Medical Genetic Center, Tehran, Iran,
| | - Hosein Dalili
- Breast feeding Research Center, Tehran University of Medical Sciences, Tehran, Iran,
| | - Majid Kalani
- Akbarabadi Hospital, Iran University of Medical Sciences, Tehran, Iran,
| | - Nikoo Niknafs
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Breast feeding Research Center, Tehran University of Medical Sciences, Tehran, Iran,
| | - Mamak Shariat
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Breast feeding Research Center, Tehran University of Medical Sciences, Tehran, Iran,
| | - Javad Tavakkoly-Bazzaz
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,
| | - Elaheh Amini
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,
| | - Tahereh Esmaeilnia Shirvani
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Department of Pediatrics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,
| | - Amir kamal Hardani
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- School of Medicine, Ahwaz Jundishapur University of Medical Sciences, Ahwaz, Iran, and
| | - Roya Taheritafti
- Breast feeding Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Department of Pediatrics, Faculty of Medicine, Shahid Beheshti University of medical Sciences, Tehran, Iran
| | - Nasrin Ghasemi-Fakhr
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
| | - Mohsen Ghadami
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran,
| | - Fatemeh Nayeri
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Breast feeding Research Center, Tehran University of Medical Sciences, Tehran, Iran,
| | - Ali Rashidi-Nezhad
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran,
- Ronash Medical Genetic Center, Tehran, Iran,
| |
Collapse
|
14
|
Tian W, Chen X, Qin H, Wei Q, Zhang S, Tang S, Liao L, Zhang Y, Chen Y. The Haplotype TGGAG in the ABCA3 Gene Increases the Risk of Respiratory Distress Syndrome in Preterm Infants in Southern China. Pediatr Neonatol 2016; 57:188-94. [PMID: 26522252 DOI: 10.1016/j.pedneo.2015.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 08/30/2015] [Accepted: 09/16/2015] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Rare mutations in the ATP-binding cassette (ABC) transporter A3 (ABCA3) gene are associated with neonatal respiratory distress syndrome (RDS). The contribution of common single nucleotide polymorphisms (SNPs) to preterm RDS differs between ethnicities and remains unclear in Chinese infants. This study evaluated whether common SNPs and consequent haplotypes increase susceptibility to RDS in a population of preterm infants from the Guangxi Zhuang Autonomous Region of China. METHODS Using a tagging SNP (tSNP) strategy and real-time polymerase chain reaction, we genotyped four tSNPs (i.e., rs150929, rs4787273, rs11867129, and rs17135889) and one coding SNP (p.F353F) of the ABCA3 gene in preterm infants with RDS (n = 83) and without RDS (n = 83). We predicted the haplotypes. Minor allele frequencies (MAFs) and haplotype distributions were compared between the two groups. We analyzed correlations between the clinical data and the genotypes. RESULTS Seven haplotypes existed at a frequency of 0.01 or greater. The haplotype TGGAG was significantly more frequent in RDS infants than in non-RDS infants (p = 0.026; odds ratio 3.41; 95% confidence interval 1.088-10.685). The MAF of rs17135889 SNP, a crucial SNP of the haplotype TGGAG located in the transcription factor binding site of ABCA3, was significantly higher in RDS infants (p < 0.05); however, the Bonferroni correction test showed no significant difference (p > 0.05). No significant correlation existed between the rs17135889 genotypes (AG/GG) and any clinical characteristic (e.g., oxygen supplementation duration and hospitalization, requirement for ventilation, bronchopulmonary dysplasia complications, and mortality rate). CONCLUSION The TGGAG haplotype may be a risk factor for RDS in preterm infants in this Chinese population. Further study is needed with a larger sample size to verify the association between the rs17135889 SNP and increased risk of RDS in preterm infants, and to determine whether rs17135889 can be a reference in further population-based studies of ABCA3.
Collapse
Affiliation(s)
- Wenjun Tian
- Division of Neonatology, Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Xiuqi Chen
- Division of Neonatology, Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Huijuan Qin
- Division of Neonatology, Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Qiufen Wei
- Division of Neonatology, Department of Pediatrics, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi, China
| | - Shuying Zhang
- Division of Neonatology, Department of Pediatrics, Nanning Women and Children's Hospital, Guangxi, China
| | - Shangying Tang
- Division of Neonatology, Department of Pediatrics, Qinzhou Maternal and Child Health Hospital, Qinzhou, China
| | - Liangrong Liao
- Division of Neonatology, Department of Pediatrics, Eighth Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Yanming Zhang
- Division of Neonatology, Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Yujun Chen
- Division of Neonatology, Department of Pediatrics, First Affiliated Hospital of Guangxi Medical University, Guangxi, China.
| |
Collapse
|
15
|
Chen YJ, Wambach JA, DePass K, Wegner DJ, Chen SK, Zhang QY, Heins H, Cole FS, Hamvas A. Population-based frequency of surfactant dysfunction mutations in a native Chinese cohort. World J Pediatr 2016; 12:190-5. [PMID: 26547207 DOI: 10.1007/s12519-015-0047-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 07/14/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Rare mutations in surfactant-associated genes contribute to neonatal respiratory distress syndrome. The frequency of mutations in these genes in the Chinese population is unknown. METHODS We obtained blood spots from the Guangxi Neonatal Screening Center in Nanning, China that included Han (n=443) and Zhuang (n=313) ethnic groups. We resequenced all exons of the surfactant proteins-B (SFTPB), -C (SFTPC), and the ATP-binding cassette member A3 (ABCA3) genes and compared the frequencies of 5 common and all rare variants. RESULTS We found minor differences in the frequencies of the common variants in the Han and Zhuang cohorts. We did not find any rare mutations in SFTPB or SFTPC, but we found three ABCA3 mutations in the Han [minor allele frequency (MAF)=0.003] and 7 in the Zhuang (MAF=0.011) cohorts (P=0.10). The ABCA3 mutations were unique to each cohort; five were novel. The collapsed carrier rate of rare ABCA3 mutations in the Han and Zhuang populations combined was 1.3%, which is significantly lower than that in the United States (P<0.001). CONCLUSION The population-based frequency of mutations in ABCA3 in south China newborns is significantly lower than that in United States. The contribution of these rare ABCA3 mutations to disease burden in the south China population is still unknown.
Collapse
Affiliation(s)
- Yu-Jun Chen
- Division of Neonatology, Department of Pediatrics, the First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Jennifer Anne Wambach
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Kelcey DePass
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Daniel James Wegner
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Shao-Ke Chen
- Department of Pediatrics, Guangxi Maternal and Child Health Hospital, Nanning, China
| | - Qun-Yuan Zhang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - Hillary Heins
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Francis Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Aaron Hamvas
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA. .,Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, 225 E. Chicago Ave, Box No. 45, Chicago, IL, 60611, USA.
| |
Collapse
|
16
|
Shen CL, Zhang Q, Meyer Hudson J, Cole FS, Wambach JA. Genetic Factors Contribute to Risk for Neonatal Respiratory Distress Syndrome among Moderately Preterm, Late Preterm, and Term Infants. J Pediatr 2016; 172:69-74.e2. [PMID: 26935785 PMCID: PMC4876036 DOI: 10.1016/j.jpeds.2016.01.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 12/14/2015] [Accepted: 01/06/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To determine the genetic contribution to risk for respiratory distress syndrome (RDS) among moderately preterm, late preterm, and term infants (estimated gestational age ≥32 weeks) of African- and European-descent. STUDY DESIGN We reviewed clinical records for 524 consecutive twin pairs ≥32 weeks gestation. We identified pairs in which at least 1 twin had RDS (n = 225) and compared the concordance of RDS between monozygotic and dizygotic twins. Using mixed-effects logistic regression, we identified covariates that increased disease risk. We performed additive genetic, common environmental, and residual effects modeling to estimate genetic variance and used the ratio of genetic variance to total variance to estimate genetic contribution to RDS disease risk. RESULTS Monozygotic twins were more concordant for RDS than dizygotic twins (P = .0040). Estimated gestational age, European-descent, male sex, delivery by cesarean, and 5-minute Apgar score each independently increased risk for RDS. After adjusting for these covariates, genetic effects accounted for 58% (P = .0002) of the RDS disease risk variance for all twin pairs. CONCLUSIONS In addition to environmental factors, genetic factors may contribute to RDS risk among moderately preterm, late preterm, and term infants. Discovery of risk alleles may be important for prediction and management of RDS risk.
Collapse
Affiliation(s)
- Carol L Shen
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Qunyuan Zhang
- Center for Genome Sciences and Systems Biology, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO
| | - Julia Meyer Hudson
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - F Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jennifer A Wambach
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
17
|
Griese M, Lorenz E, Hengst M, Schams A, Wesselak T, Rauch D, Wittmann T, Kirchberger V, Escribano A, Schaible T, Baden W, Schulze J, Krude H, Aslanidis C, Schwerk N, Kappler M, Hartl D, Lohse P, Zarbock R. Surfactant proteins in pediatric interstitial lung disease. Pediatr Res 2016; 79:34-41. [PMID: 26375475 DOI: 10.1038/pr.2015.173] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 06/22/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Children's interstitial lung diseases (chILD) comprise a broad spectrum of diseases. Besides the genetically defined surfactant dysfunction disorders, most entities pathologically involve the alveolar surfactant region, possibly affecting the surfactant proteins SP-B and SP-C. Therefore, our objective was to determine the value of quantitation of SP-B and SP-C levels in bronchoalveolar lavage fluid (BALF) for the diagnosis of chILD. METHODS Levels of SP-B and SP-C in BALF from 302 children with chILD and in controls were quantified using western blotting. In a subset, single-nucleotide polymorphisms (SNPs) in the SFTPC promoter were genotyped by direct sequencing. RESULTS While a lack of dimeric SP-B was found only in the sole subject with hereditary SP-B deficiency, low or absent SP-C was observed not only in surfactant dysfunction disorders but also in patients with other diffuse parenchymal lung diseases pathogenetically related to the alveolar surfactant region. Genetic analysis of the SFTPC promoter showed association of a single SNP with SP-C level. CONCLUSION SP-B levels may be used for screening for SP-B deficiency, while low SP-C levels may point out diseases caused by mutations in TTF1, SFTPC, ABCA3, and likely in other genes involved in surfactant metabolism that remain to be identified. We conclude that measurement of levels of SP-B and SP-C was useful for the differential diagnosis of chILD, and for the precise molecular diagnosis, sequencing of the genes is necessary.
Collapse
Affiliation(s)
- Matthias Griese
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Elke Lorenz
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Meike Hengst
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Andrea Schams
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Traudl Wesselak
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Daniela Rauch
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Thomas Wittmann
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | | | - Amparo Escribano
- Hospital Clinico Universitario, Unidad Neumologia Infantil, Valencia, Spain
| | - Thomas Schaible
- Neonatology, University Children's Hospital, Mannheim, Germany
| | - Winfried Baden
- Pediatric Cardiology, University Children's Hospital, Tuebingen, Germany
| | - Johannes Schulze
- Children´s Hospital, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Heiko Krude
- Pediatric Endocrinology, Charite, Berlin, Germany
| | - Charalampos Aslanidis
- Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Nicolaus Schwerk
- Pediatric Pulmonology, Hannover Medical School, Hannover, Germany
| | - Matthias Kappler
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| | - Dominik Hartl
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany
| | - Peter Lohse
- Praxis für Humangenetik, CeGaT GmbH, Tübingen, Germany
| | - Ralf Zarbock
- Dr von Hauner Children´s Hospital, German Center for Lung Research (DZL), University of Munich, Munich, Germany
| |
Collapse
|
18
|
Children’s Interstitial and Diffuse Lung Disease. Progress and Future Horizons. Ann Am Thorac Soc 2015; 12:1451-7. [DOI: 10.1513/annalsats.201508-558ps] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
19
|
Dai Y, Wang QW, He S, Zhang Z, Gao C. Correlation of ECR1 A3650G Polymorphism with Neonatal Respiratory Distress Syndrome. Genet Test Mol Biomarkers 2015; 19:18-23. [PMID: 25494101 DOI: 10.1089/gtmb.2014.0192] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Ying Dai
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng, Henan, People's Republic of China
| | - Qi-Wei Wang
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng, Henan, People's Republic of China
| | - Shu He
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng, Henan, People's Republic of China
| | - Zhao Zhang
- Department of Pediatrics, Huaihe Hospital, Henan University, Kaifeng, Henan, People's Republic of China
| | - Chao Gao
- Department of Medicine and Equipment, Huaihe Hospital, Henan University, Kaifeng, Henan, People's Republic of China
| |
Collapse
|
20
|
Wambach JA, Casey AM, Fishman MP, Wegner DJ, Wert SE, Cole FS, Hamvas A, Nogee LM. Genotype-phenotype correlations for infants and children with ABCA3 deficiency. Am J Respir Crit Care Med 2014; 189:1538-43. [PMID: 24871971 DOI: 10.1164/rccm.201402-0342oc] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Recessive mutations in the ATP-binding cassette transporter A3 (ABCA3) cause lethal neonatal respiratory failure and childhood interstitial lung disease. Most ABCA3 mutations are private. OBJECTIVES To determine genotype-phenotype correlations for recessive ABCA3 mutations. METHODS We reviewed all published and unpublished ABCA3 sequence and phenotype data from our prospective genetic studies of symptomatic infants and children at Washington and Johns Hopkins Universities. Mutations were classified based on their predicted disruption of protein function: frameshift and nonsense mutations were classified as "null," whereas missense, predicted splice site mutations, and insertion/deletions were classified as "other." We compared age of presentation and outcomes for the three genotypes: null/null, null/other, and other/other. MEASUREMENTS AND MAIN RESULTS We identified 185 infants and children with homozygous or compound heterozygous ABCA3 mutations and lung disease. All of the null/null infants presented with respiratory failure at birth compared with 75% of infants with null/other or other/other genotypes (P = 0.00011). By 1 year of age, all of the null/null infants had died or undergone lung transplantation compared with 62% of the null/other and other/other children (P < 0.0001). CONCLUSIONS Genotype-phenotype correlations exist for homozygous or compound heterozygous mutations in ABCA3. Frameshift or nonsense ABCA3 mutations are predictive of neonatal presentation and poor outcome, whereas missense, splice site, and insertion/deletions are less reliably associated with age of presentation and prognosis. Counseling and clinical decision making should acknowledge these correlations.
Collapse
Affiliation(s)
- Jennifer A Wambach
- 1 Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Naderi HM, Murray JC, Dagle JM. Single mutations in ABCA3 increase the risk for neonatal respiratory distress syndrome in late preterm infants (gestational age 34-36 weeks). Am J Med Genet A 2014; 164A:2676-8. [PMID: 25073622 DOI: 10.1002/ajmg.a.36660] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 05/28/2014] [Indexed: 11/12/2022]
Affiliation(s)
- Hodad M Naderi
- Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | | | | |
Collapse
|
22
|
Synonymous ABCA3 variants do not increase risk for neonatal respiratory distress syndrome. J Pediatr 2014; 164:1316-21.e3. [PMID: 24657120 PMCID: PMC4035386 DOI: 10.1016/j.jpeds.2014.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/23/2013] [Accepted: 02/06/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To determine whether synonymous variants in the adenosine triphosphate-binding cassette A3 transporter (ABCA3) gene increase the risk for neonatal respiratory distress syndrome (RDS) in term and late preterm infants of European and African descent. STUDY DESIGN Using next-generation pooled sequencing of race-stratified DNA samples from infants of European and African descent at ≥34 weeks gestation with and without RDS (n = 503), we scanned all exons of ABCA3, validated each synonymous variant with an independent genotyping platform, and evaluated race-stratified disease risk associated with common synonymous variants and collapsed frequencies of rare synonymous variants. RESULTS The synonymous ABCA3 variant frequency spectrum differs between infants of European descent and those of African descent. Using in silico prediction programs and statistical strategies, we found no potentially disruptive synonymous ABCA3 variants or evidence of selection pressure. Individual common synonymous variants and collapsed frequencies of rare synonymous variants did not increase disease risk in term and late-preterm infants of European or African descent. CONCLUSION In contrast to rare, nonsynonymous ABCA3 mutations, synonymous ABCA3 variants do not increase the risk for neonatal RDS among term and late-preterm infants of European or African descent.
Collapse
|
23
|
Bersani I, Speer CP, Kunzmann S. Surfactant proteins A and D in pulmonary diseases of preterm infants. Expert Rev Anti Infect Ther 2014; 10:573-84. [DOI: 10.1586/eri.12.34] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
24
|
Glasser SW, Maxfield MD, Ruetschilling TL, Akinbi HT, Baatz JE, Kitzmiller JA, Page K, Xu Y, Bao EL, Korfhagen TR. Persistence of LPS-induced lung inflammation in surfactant protein-C-deficient mice. Am J Respir Cell Mol Biol 2013; 49:845-54. [PMID: 23795648 DOI: 10.1165/rcmb.2012-0374oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Pulmonary surfactant protein-C (SP-C) gene-targeted mice (Sftpc(-/-)) develop progressive lung inflammation and remodeling. We hypothesized that SP-C deficiency reduces the ability to suppress repetitive inflammatory injury. Sftpc(+/+) and Sftpc(-/-) mice given three doses of bacterial LPS developed airway and airspace inflammation, which was more intense in the Sftpc(-/-) mice at 3 and 5 days after the final dose. Compared with Sftpc(+/+)mice, inflammatory injury persisted in the lungs of Sftpc(-/-) mice 30 days after the final LPS challenge. Sftpc(-/-) mice showed LPS-induced airway goblet cell hyperplasia with increased detection of Sam pointed Ets domain and FoxA3 transcription factors. Sftpc(-/-) type II alveolar epithelial cells had increased cytokine expression after LPS exposure relative to Sftpc(+/+) cells, indicating that type II cell dysfunction contributes to inflammatory sensitivity. Microarray analyses of isolated type II cells identified a pattern of enhanced expression of inflammatory genes consistent with an intrinsic low-level inflammation resulting from SP-C deficiency. SP-C-containing clinical surfactant extract (Survanta) or SP-C/phospholipid vesicles blocked LPS signaling through the LPS receptor (Toll-like receptor [TLR] 4/CD14/MD2) in human embryonic kidney 293T cells, indicating that SP-C blocks LPS-induced cytokine production by a TLR4-dependent mechanism. Phospholipid vesicles alone did not modify the TLR4 response. In vivo deficiency of SP-C leads to inflammation, increased cytokine production by type II cells, and persistent inflammation after repetitive LPS stimulation.
Collapse
|
25
|
Glasser SW, Senft AP, Maxfield MD, Ruetschilling TL, Baatz JE, Page K, Korfhagen TR. Genetic replacement of surfactant protein-C reduces respiratory syncytial virus induced lung injury. Respir Res 2013; 14:19. [PMID: 23399055 PMCID: PMC3598668 DOI: 10.1186/1465-9921-14-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 02/08/2013] [Indexed: 12/31/2022] Open
Abstract
Background Individuals with deficiencies of pulmonary surfactant protein C (SP-C) develop interstitial lung disease (ILD) that is exacerbated by viral infections including respiratory syncytial virus (RSV). SP-C gene targeted mice (Sftpc -/-) lack SP-C, develop an ILD-like disease and are susceptible to infection with RSV. Methods In order to determine requirements for correction of RSV induced injury we have generated compound transgenic mice where SP-C expression can be induced on the Sftpc -/- background (SP-C/Sftpc -/-) by the administration of doxycycline (dox). The pattern of induced SP-C expression was determined by immunohistochemistry and processing by Western blot analysis. Tissue and cellular inflammation was measured following RSV infection and the RSV-induced cytokine response of isolated Sftpc +/+ and -/- type II cells determined. Results After 5 days of dox administration transgene SP-C mRNA expression was detected by RT-PCR in the lungs of two independent lines of bitransgenic SP-C/Sftpc -/- mice (lines 55.3 and 54.2). ProSP-C was expressed in the lung, and mature SP-C was detected by Western blot analysis of the lavage fluid from both lines of SP-C/Sftpc -/- mice. Induced SP-C expression was localized to alveolar type II cells by immunostaining with an antibody to proSP-C. Line 55.3 SP-C/Sftpc -/- mice were maintained on or off dox for 7 days and infected with 2.6x107 RSV pfu. On day 3 post RSV infection total inflammatory cell counts were reduced in the lavage of dox treated 55.3 SP-C/Sftpc -/- mice (p = 0.004). The percentage of neutrophils was reduced (p = 0.05). The viral titers of lung homogenates from dox treated 55.3 SP-C/Sftpc -/- mice were decreased relative to 55.3 SP-C/Sftpc -/- mice without dox (p = 0.01). The cytokine response of Sftpc -/- type II cells to RSV was increased over that of Sftpc +/+ cells. Conclusions Transgenic restoration of SP-C reduced inflammation and improved viral clearance in the lungs of SP-C deficient mice. The loss of SP-C in alveolar type II cells compromises their response to infection. These findings show that the restoration of SP-C in Sftpc -/- mice in response to RSV infection is a useful model to determine parameters for therapeutic intervention.
Collapse
Affiliation(s)
- Stephan W Glasser
- Cincinnati Children's Hospital Medical Center, Perinatal Institute, Division of Neonatology, Perinatal and Pulmonary Biology, MLC7029, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Wambach JA, Wegner DJ, DePass K, Heins H, Druley TE, Mitra RD, An P, Zhang Q, Nogee LM, Cole FS, Hamvas A. Single ABCA3 mutations increase risk for neonatal respiratory distress syndrome. Pediatrics 2012; 130:e1575-82. [PMID: 23166334 PMCID: PMC3507255 DOI: 10.1542/peds.2012-0918] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Neonatal respiratory distress syndrome (RDS) due to pulmonary surfactant deficiency is heritable, but common variants do not fully explain disease heritability. METHODS Using next-generation, pooled sequencing of race-stratified DNA samples from infants ≥34 weeks' gestation with and without RDS (n = 513) and from a Missouri population-based cohort (n = 1066), we scanned all exons of 5 surfactant-associated genes and used in silico algorithms to identify functional mutations. We validated each mutation with an independent genotyping platform and compared race-stratified, collapsed frequencies of rare mutations by gene to investigate disease associations and estimate attributable risk. RESULTS Single ABCA3 mutations were overrepresented among European-descent RDS infants (14.3% of RDS vs 3.7% of non-RDS; P = .002) but were not statistically overrepresented among African-descent RDS infants (4.5% of RDS vs 1.5% of non-RDS; P = .23). In the Missouri population-based cohort, 3.6% of European-descent and 1.5% of African-descent infants carried a single ABCA3 mutation. We found no mutations among the RDS infants and no evidence of contribution to population-based disease burden for SFTPC, CHPT1, LPCAT1, or PCYT1B. CONCLUSIONS In contrast to lethal neonatal RDS resulting from homozygous or compound heterozygous ABCA3 mutations, single ABCA3 mutations are overrepresented among European-descent infants ≥34 weeks' gestation with RDS and account for ~10.9% of the attributable risk among term and late preterm infants. Although ABCA3 mutations are individually rare, they are collectively common among European- and African-descent individuals in the general population.
Collapse
Affiliation(s)
| | | | | | | | - Todd E. Druley
- Division of Hematology and Oncology, the Edward Mallinckrodt Department of Pediatrics,,Center for Genome Sciences and Systems Biology, Department of Genetics
| | - Robi D. Mitra
- Center for Genome Sciences and Systems Biology, Department of Genetics
| | - Ping An
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, Missouri; and
| | - Qunyuan Zhang
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, Missouri; and
| | - Lawrence M. Nogee
- Division of Neonatal–Perinatal Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | |
Collapse
|
27
|
Lyra P, Diniz E, Abe-Sandes K, Angelo A, Machado T, Cardeal M. Surfactant protein B gene polymorphism in preterm babies with respiratory distress syndrome. Braz J Med Biol Res 2011; 44:66-72. [PMID: 21180884 DOI: 10.1590/s0100-879x2010007500147] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Accepted: 11/26/2010] [Indexed: 11/21/2022] Open
Affiliation(s)
- P.P.R. Lyra
- Universidade de São Paulo, Brasil; Universidade Federal da Bahia
| | | | | | | | | | | |
Collapse
|