1
|
Isasi-Campillo M, Rangel-Arranz P, García-Ortega L, Pérez-Gil J. Role of N-glycosylation of surfactant protein SP-B N in lipid and SP-B interacting properties. Implications in disease. Am J Physiol Lung Cell Mol Physiol 2025; 328:L700-L715. [PMID: 40214202 DOI: 10.1152/ajplung.00350.2024] [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: 10/28/2024] [Revised: 01/05/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
SP-BN is an independent protein derived from the precursor of pulmonary surfactant protein B (SP-B), a critical component of the pulmonary surfactant (PS), the membrane-based system that coats the alveolar air-liquid interface and is essential for both respiratory mechanics and innate defense. In humans, a single-nucleotide polymorphism (SNP) defining hSP-BN glycosylation has been associated with propensity to certain respiratory diseases, but molecular studies in this regard are scarce. Previous studies with the murine SP-BN, nonglycosylated, have suggested a role for this protein in lipid transfer during PS biogenesis. This study focuses on the structural and functional characterization of both glycosylated and nonglycosylated human SP-BN protein variants to elucidate the impact of N-glycosylation. Recombinant proteins (hSP-BN, glycosylated, and hSP-BN-T73I, nonglycosylated) were produced in Pichia pastoris and purified to homogeneity. The structural characterization confirmed the main features of hSP-BN as a member of the SAPLIP protein family: mainly α-helical, a propensity to dimerization and a high stability. Interestingly, N-glycosylation did not significantly affect hSP-BN structure. Regarding lipid interactions, both hSP-BN variants were able to bind and perturb membranes in lipid vesicles with a PS-like composition at acidic, but not neutral pH, which is relevant given the acidification during PS biogenesis. Remarkably, N-glycosylation impaired the synergistic effect of hSP-BN and mature SP-B to promote lipid mixing/transfer activity. These results support the joint action of both proteins in PS biogenesis and, more importantly, suggest that this combined activity affected with the SNP-induced glycosylation of hSP-BN could be behind certain PS defects acquired during biogenesis causing some susceptibility to respiratory diseases.NEW & NOTEWORTHY The impact of N-glycosylation on the structure and function of human SP-BN protein has been studied. Homogeneous production of glycosylated hSP-BN and nonglycosylated hSP-BN-T73I was achieved in Pichia pastoris. Structural characterization and lipid interaction properties at acidic pH revealed no significant differences due to glycosylation. N-glycosylation impairs the synergistic action of hSP-BN and SP-B in lipid transfer/mixing activity. N-glycosylation of hSP-BN could impair PS biogenesis, in agreement with its potential involvement in respiratory disease.
Collapse
Affiliation(s)
- Miriam Isasi-Campillo
- Biochemistry and Molecular Biology Department, Complutense University, Madrid, Spain
- Research Institute Hospital 12 de Octubre (Imas12), Madrid, Spain
| | - Paula Rangel-Arranz
- Biochemistry and Molecular Biology Department, Complutense University, Madrid, Spain
| | - Lucía García-Ortega
- Biochemistry and Molecular Biology Department, Complutense University, Madrid, Spain
- Research Institute Hospital 12 de Octubre (Imas12), Madrid, Spain
| | - Jesús Pérez-Gil
- Biochemistry and Molecular Biology Department, Complutense University, Madrid, Spain
- Research Institute Hospital 12 de Octubre (Imas12), Madrid, Spain
| |
Collapse
|
2
|
Fotook Kiaei SZ, Schwartz DA. Genetic underpinning of idiopathic pulmonary fibrosis: the role of mucin. Expert Rev Respir Med 2025:1-12. [PMID: 39912527 DOI: 10.1080/17476348.2025.2464035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/27/2024] [Accepted: 02/04/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by progressive scarring and reduced survival. The development of IPF is influenced by rare and common genetic variants, cigarette smoking, aging, and environmental exposures. Among the two dozen genetic contributors, the MUC5B promoter variant (rs35705950) is the dominant risk factor, increasing the risk of both familial and sporadic IPF and accounting for nearly 50% of the genetic predisposition to the disease. AREAS COVERED This review provides an expert perspective on the genetic underpinnings of IPF rather than a systematic analysis, emphasizing key insights into its genetic basis. The articles referenced in this review were identified through targeted searches in PubMed, Scopus, and Web of Science for studies published between 2000 and 2023, prioritizing influential research on the genetic factors contributing to IPF. Search terms included 'idiopathic pulmonary fibrosis,' 'genetics,' 'MUC5B,' 'telomere dysfunction,' and 'surfactant proteins.' The selection of studies was guided by the authors' expertise, focusing on the most relevant publications. EXPERT OPINION The identification of genetic variants not only highlights the complexity of IPF but also offers potential for earlier diagnosis and personalized treatment strategies targeting specific genetic pathways, ultimately aiming to improve patient outcomes.
Collapse
Affiliation(s)
| | - David A Schwartz
- Department of Medicine, University of Colorado Denver, School of Medicine, Aurora, CO, USA
| |
Collapse
|
3
|
Behrouzi A, Sakhaee F, Ghazanfari Jajin M, Ahmadi I, Anvari E, Sotoodehnejadnematalahi F, Fateh A. The surfactant protein B polymorphisms (rs7316 and rs1130866) and their correlation with disease progression of COVID-19. Cytokine 2024; 184:156775. [PMID: 39368228 DOI: 10.1016/j.cyto.2024.156775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/15/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND It is critical to examine the pathogenic pathways in coronavirus disease 2019 (COVID-19) that resulted in the development of severe lung injury. Surfactant protein B (SFTPB) is a vital component for sustaining life and serves pivotal functions in the host's defensive mechanisms and alveolar surface tension reduction. Our study aimed to determine the effect of SFTPB rs7316 and rs1130866 variants on the course of disease in COVID-19 patients. METHODS The study cohort comprised 3,184 individuals diagnosed with COVID-19. We employed the RFLP approach to determine the variations of the SFTPB genes. RESULTS SFTPB rs7316 did not exhibit a statistically significant correlation with COVID-19 mortality across different inheritance models. But, after making more changes for SARS-CoV-2 variants, it was found that there was a strong link between the TT and TC genotypes of SFTPB rs7316 and death rates, especially for the Delta variant. Furthermore, our study's findings indicate a significant association between the SFTPB rs1130866 G allele and an elevated risk of mortality in COVID-19 across all variants of SARS-CoV-2. CONCLUSIONS The use of the SFTPB rs1130866 marker has the potential to facilitate the prediction of COVID-19 severity. On the other hand, for SFTPB rs7316, this kind of prediction seems to depend on the particular SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Amir Behrouzi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Sakhaee
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | | | - Iraj Ahmadi
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | - Enayat Anvari
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran
| | | | - Abolfazl Fateh
- Department of Physiology, School of Medicine, Ilam University of Medical Science, Ilam, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
4
|
Hassan M, Ali AS, Zubairi ABS, Padhani ZA, Kirmani S, Ahmad H, Fatmi Z, Das JK. Gene polymorphisms and risk of idiopathic pulmonary fibrosis: a systematic review and meta-analysis. Monaldi Arch Chest Dis 2024. [PMID: 39480160 DOI: 10.4081/monaldi.2024.2952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) has been widely hypothesized to occur as a result of an interplay between a nexus of environmental and genetic risk factors. However, not much is known about the genetic aspect of this disease. The objective of this review was to identify the genetic polymorphisms associated with the risk of developing IPF. We searched PubMed, EBSCO CINAHL Plus, Web of Science, and Wiley Cochrane Library databases for studies on risk factors of IPF published between March 2000 and November 2023. Studies with an IPF diagnosis based only on the American Thoracic Society and the European Respiratory Society guidelines were included. Thirty-one case-control studies were included with 3997 IPF and 20,925 non-IPF subjects. Two of the studies enrolled biopsy-proven IPF patients; 13 studies diagnosed IPF on the basis of clinical and high-resolution computed tomography (HRCT) findings; and 14 studies diagnosed based on both biopsy and clinical and HRCT findings. 16 studies with MUC5B rs35705950, IL-4 rs2243250, IL-4 rs2070874, and tumor necrosis factor α (TNFα)-308 were eligible for meta-analysis. The allele contrast model (T versus G) for MUC5B rs35705950 revealed statistically significant association of T allele with the risk of IPF [odds ratio (OR) 3.84, 95% confidence interval (CI) 3.20 to 4.61, adjusted p<0.0001), as was the allele contrast model for Asian (OR 2.83, 95% CI 1.51 to 5.32, adjusted p=0.009) and Caucasian (OR 4.11, 95% CI 3.56 to 4.75, adjusted p<0.0001). The allele contrast models for IL-4 rs2243250, IL-4 rs2070874, and TNFα-308 did not demonstrate any significant association with IPF. This review suggests an association of MUC5B rs35705950 T allele with the risk of developing IPF. To our knowledge, this study is the first to aggregate several genetic polymorphisms associated with IPF.
Collapse
Affiliation(s)
- Maryam Hassan
- Department of Medicine, Aga Khan University Hospital, Karachi
| | | | - Ali Bin Sarwar Zubairi
- Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan; Department of Medicine, Southern Illinois University School of Medicine, Springfield, IL
| | - Zahra Ali Padhani
- Faculty of Health and Medical Sciences, School of Public Health, University of Adelaide
| | - Salman Kirmani
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi
| | - Huzaifa Ahmad
- Department of Medicine, Aga Khan University Hospital, Karachi
| | - Zafar Fatmi
- Department of Community Health Sciences, Aga Khan University Hospital, Karachi
| | - Jai K Das
- Department of Pediatrics and Child Health, Aga Khan University Hospital, Karachi; Institute of Global Health and Development, Aga Khan University, Karachi
| |
Collapse
|
5
|
Bermudez J, Nathan N, Coiffard B, Roux A, Hirschi S, Degot T, Bunel V, Le Pavec J, Macey J, Le Borgne A, Legendre M, Cottin V, Thomas PA, Borie R, Reynaud-Gaubert M. Outcome of lung transplantation for adults with interstitial lung disease associated with genetic disorders of the surfactant system. ERJ Open Res 2023; 9:00240-2023. [PMID: 38020562 PMCID: PMC10658627 DOI: 10.1183/23120541.00240-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/31/2023] [Indexed: 12/01/2023] Open
Abstract
Background Interstitial lung disease associated with genetic disorders of the surfactant system is a rare entity in adults that can lead to lung transplantation. Our objective was to describe the outcome of these patients after lung transplantation. Methods We conducted a retrospective, multicentre study, on adults who underwent lung transplantation for such disease in the French lung transplant centres network, from 1997 to 2018. Results 20 patients carrying mutations in SFTPA1 (n=5), SFTPA2 (n=7) or SFTPC (n=8) were included. Median interquartile range (IQR) age at diagnosis was 45 (40-48) years, and median (IQR) age at lung transplantation was 51 (45-54) years. Median overall survival after transplantation was 8.6 years. Two patients had a pre-transplant history of lung cancer, and two developed post-transplant lung cancer. Female gender and a body mass index <25 kg·m-2 were significantly associated with a better prognosis, whereas transplantation in high emergency was associated with a worst prognosis. Conclusions Lung transplantation in adults with interstitial lung disease associated with genetic disorders of surfactant system may be a valid therapeutic option. Our data suggest that these patients may have a good prognosis. Immunosuppressive protocol was not changed for these patients, and close lung cancer screening is needed before and after transplantation.
Collapse
Affiliation(s)
- Julien Bermudez
- Department of Respiratory Medicine and Lung Transplantation, Assistance Publique – Hôpitaux de Marseille, Hôpital Nord; Aix-Marseille Université, Marseille, France
| | - Nadia Nathan
- Assistance Publique – Hôpitaux de Paris (APHP) – Sorbonne Université, Inserm Childhood Genetic Disorders and Reference Center for Rare Lung Diseases, Armand Trousseau Hospital, Paris, France
- APHP – Sorbonne Université, Pediatric Pulmonology Department and Reference Center for Rare Lung Diseases RespiRare, Armand Trousseau Hospital, Paris, France
| | - Benjamin Coiffard
- Department of Respiratory Medicine and Lung Transplantation, Assistance Publique – Hôpitaux de Marseille, Hôpital Nord; Aix-Marseille Université, Marseille, France
| | - Antoine Roux
- Department of Pulmonary Medicine, Foch Hospital, Suresnes, France
| | - Sandrine Hirschi
- Respiratory Medicine and Lung Transplantation, Strasbourg University Hospital, Strasbourg, France
| | - Tristan Degot
- Respiratory Medicine and Lung Transplantation, Strasbourg University Hospital, Strasbourg, France
| | - Vincent Bunel
- AP-HP, Bichat Claude Bernard Hospital, Pulmonology B and Lung Transplant Department, INSERM 1152, Paris, France
| | - Jérôme Le Pavec
- Service de Pneumologie et de Transplantation Pulmonaire, Hôpital Marie-Lannelongue, Groupe hospitalier Paris-Saint Joseph, Le Plessis-Robinson, France
- Université Paris–Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre, France
- UMR_S 999, Université Paris–Sud, INSERM, Hôpital Marie Lannelongue, Le Plessis Robinson, France
| | - Julie Macey
- Respiratory Medicine and Cystic Fibrosis Center, University Hospital Center of Bordeaux, Bordeaux, France
| | - Aurélie Le Borgne
- Service de Pneumologie, Pôle des voies respiratoires, Hôpital Larrey, CHU Toulouse, Toulouse, France
| | - Marie Legendre
- Assistance Publique – Hôpitaux de Paris (APHP) – Sorbonne Université, Inserm Childhood Genetic Disorders and Reference Center for Rare Lung Diseases, Armand Trousseau Hospital, Paris, France
- UF de Génétique moléculaire, APHP, Sorbonne Université, Hôpital Armand-Trousseau, Paris, France
| | - Vincent Cottin
- Department of Respiratory Medicine, National Coordinating Reference Center for Rare Pulmonary Diseases, Louis Pradel Hospital, Hospices Civils de Lyon; IVPC, INRAE, Claude Bernard University Lyon 1, member of ERN-LUNG, Lyons, France
| | - Pascal-Alexandre Thomas
- Department of Thoracic Surgery, North Hospital, Aix-Marseille University, CNRS, INSERM, CRCM, AP-HM, Chemin des Bourrely, Marseille, France
| | - Raphaël Borie
- Service de Pneumologie A, APHP, Hôpital Bichat, Université de Paris and INSERM U1152, Paris, France
| | - Martine Reynaud-Gaubert
- Department of Respiratory Medicine and Lung Transplantation, Assistance Publique – Hôpitaux de Marseille, Hôpital Nord; Aix-Marseille Université, Marseille, France
| |
Collapse
|
6
|
Sharma NS, Patel K, Sari E, Shankar S, Gastanadui MG, Moncada-Giraldo D, Soto-Vazquez Y, Stacks D, Hecker L, Dsouza K, Banday M, O’Neill E, Benson P, Payne G, Margaroli C, Gaggar A. Active transcription in the vascular bed characterizes rapid progression in idiopathic pulmonary fibrosis. J Clin Invest 2023; 133:e165976. [PMID: 37384419 PMCID: PMC10425209 DOI: 10.1172/jci165976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Affiliation(s)
- Nirmal S. Sharma
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- West Roxbury VA Medical Center, Boston, Massachusetts, USA
| | - Kapil Patel
- Department of Medicine, University of South Florida, Tampa, Florida, USA
| | - Ezgi Sari
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Shruti Shankar
- Department of Medicine, University of South Florida, Tampa, Florida, USA
| | - Maria G. Gastanadui
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Yixel Soto-Vazquez
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Delores Stacks
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Louise Hecker
- Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Kevin Dsouza
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mudassir Banday
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Edward O’Neill
- Department of Medicine, University of South Florida, Tampa, Florida, USA
| | - Paul Benson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gregory Payne
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham VA Medical Center, Birmingham, Alabama, USA
- Lung Health Center, Program in Protease and Matrix Biology, and Gregory Fleming James CF Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Camilla Margaroli
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Birmingham VA Medical Center, Birmingham, Alabama, USA
- Lung Health Center, Program in Protease and Matrix Biology, and Gregory Fleming James CF Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
7
|
Bouma F, Nyberg F, Olin AC, Carlsen HK. Genetic susceptibility to airway inflammation and exposure to short-term outdoor air pollution. Environ Health 2023; 22:50. [PMID: 37386634 DOI: 10.1186/s12940-023-00996-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 06/02/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Air pollution is a large environmental health hazard whose exposure and health effects are unequally distributed among individuals. This is, at least in part, due to gene-environment interactions, but few studies exist. Thus, the current study aimed to explore genetic susceptibility to airway inflammation from short-term air pollution exposure through mechanisms of gene-environment interaction involving the SFTPA, GST and NOS genes. METHODS Five thousand seven hundred two adults were included. The outcome measure was fraction of exhaled nitric oxide (FeNO), at 50 and 270 ml/s. Exposures were ozone (O3), particulate matter < 10 µm (PM10), and nitrogen dioxide (NO2) 3, 24, or 120-h prior to FeNO measurement. In the SFTPA, GST and NOS genes, 24 single nucleotide polymorphisms (SNPs) were analyzed for interaction effects. The data were analyzed using quantile regression in both single-and multipollutant models. RESULTS Significant interactions between SNPs and air pollution were found for six SNPs (p < 0.05): rs4253527 (SFTPA1) with O3 and NOx, rs2266637 (GSTT1) with NO2, rs4795051 (NOS2) with PM10, NO2 and NOx, rs4796017 (NOS2) with PM10, rs2248814 (NOS2) with PM10 and rs7830 (NOS3) with NO2. The marginal effects on FeNO for three of these SNPs were significant (per increase of 10 µg/m3):rs4253527 (SFTPA1) with O3 (β: 0.155, 95%CI: 0.013-0.297), rs4795051 (NOS2) with PM10 (β: 0.073, 95%CI: 0.00-0.147 (single pollutant), β: 0.081, 95%CI: 0.004-0.159 (multipollutant)) and NO2 (β: -0.084, 95%CI: -0.147; -0.020 (3 h), β: -0.188, 95%CI: -0.359; -0.018 (120 h)) and rs4796017 (NOS2) with PM10 (β: 0.396, 95%CI: 0.003-0.790). CONCLUSIONS Increased inflammatory response from air pollution exposure was observed among subjects with polymorphisms in SFTPA1, GSTT1, and NOS genes, where O3 interacted with SFTPA1 and PM10 and NO2/NOx with the GSTT1 and NOS genes. This provides a basis for the further exploration of biological mechanisms as well as the identification of individuals susceptible to the effects of outdoor air pollution.
Collapse
Affiliation(s)
- Femke Bouma
- Department of Occupational and Environmental Health, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 16A, BOX 414, 40530, Gothenburg, Sweden
| | - Fredrik Nyberg
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg University, Gothenburg, Sweden
| | - Anna-Carin Olin
- Department of Occupational and Environmental Health, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 16A, BOX 414, 40530, Gothenburg, Sweden
| | - Hanne Krage Carlsen
- Department of Occupational and Environmental Health, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 16A, BOX 414, 40530, Gothenburg, Sweden.
| |
Collapse
|
8
|
Wang S, Rao W, Hoffman A, Lin J, Li J, Lin T, Liew AA, Vincent M, Mertens TCJ, Karmouty-Quintana H, Crum CP, Metersky ML, Schwartz DA, Davies PJA, Stephan C, Jyothula SSK, Sheshadri A, Suarez EE, Huang HJ, Engelhardt JF, Dickey BF, Parekh KR, McKeon FD, Xian W. Cloning a profibrotic stem cell variant in idiopathic pulmonary fibrosis. Sci Transl Med 2023; 15:eabp9528. [PMID: 37099633 PMCID: PMC10794039 DOI: 10.1126/scitranslmed.abp9528] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 02/28/2023] [Indexed: 04/28/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, irreversible, and rapidly fatal interstitial lung disease marked by the replacement of lung alveoli with dense fibrotic matrices. Although the mechanisms initiating IPF remain unclear, rare and common alleles of genes expressed in lung epithelia, combined with aging, contribute to the risk for this condition. Consistently, single-cell RNA sequencing (scRNA-seq) studies have identified lung basal cell heterogeneity in IPF that might be pathogenic. We used single-cell cloning technologies to generate "libraries" of basal stem cells from the distal lungs of 16 patients with IPF and 10 controls. We identified a major stem cell variant that was distinguished from normal stem cells by its ability to transform normal lung fibroblasts into pathogenic myofibroblasts in vitro and to activate and recruit myofibroblasts in clonal xenografts. This profibrotic stem cell variant, which was shown to preexist in low quantities in normal and even fetal lungs, expressed a broad network of genes implicated in organ fibrosis and showed overlap in gene expression with abnormal epithelial signatures identified in previously published scRNA-seq studies of IPF. Drug screens highlighted specific vulnerabilities of this profibrotic variant to inhibitors of epidermal growth factor and mammalian target of rapamycin signaling as prospective therapeutic targets. This profibrotic stem cell variant in IPF was distinct from recently identified profibrotic stem cell variants in chronic obstructive pulmonary disease and may extend the notion that inappropriate accrual of minor and preexisting stem cell variants contributes to chronic lung conditions.
Collapse
Affiliation(s)
- Shan Wang
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Wei Rao
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Ashley Hoffman
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Jennifer Lin
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Justin Li
- AccuraScience, Johnston, IA 50131, USA
| | - Tao Lin
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Audrey-Ann Liew
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | | | - Tinne C. J. Mertens
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Christopher P. Crum
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02215, USA
| | - Mark L. Metersky
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - David A. Schwartz
- Departments of Medicine and Microbiology and Immunology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | - Clifford Stephan
- Texas A&M Health Institute of Biotechnology, Houston, TX 77030, USA
| | - Soma S. K. Jyothula
- Lung Transplant Center at Memorial Hermann-Texas Medical Center, Houston, TX 77030, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Erik Eddie Suarez
- Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Howard J. Huang
- Department of Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - John F. Engelhardt
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Burton F. Dickey
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kalpaj R. Parekh
- Department of Surgery, Division of Cardiothoracic Surgery, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Frank D. McKeon
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| | - Wa Xian
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, TX 77003, USA
| |
Collapse
|
9
|
Schiefermeier-Mach N, Heinrich L, Lechner L, Perkhofer S. Regulation of Surfactant Protein Gene Expression by Aspergillus fumigatus in NCl-H441 Cells. Microorganisms 2023; 11:microorganisms11041011. [PMID: 37110432 PMCID: PMC10143823 DOI: 10.3390/microorganisms11041011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Aspergillus fumigatus is an opportunistic fungal pathogen that causes serious lung diseases in immunocompromised patients. The lung surfactant produced by alveolar type II and Clara cells in the lungs is an important line of defense against A. fumigatus. The surfactant consists of phospholipids and surfactant proteins (SP-A, SP-B, SP-C and SP-D). The binding to SP-A and SP-D proteins leads to the agglutination and neutralization of lung pathogens as well as the modulation of immune responses. SP-B and SP-C proteins are essential for surfactant metabolism and can modulate the local immune response; however, the molecular mechanisms remain unclear. We investigated changes in the SP gene expression in human lung NCI-H441 cells infected with conidia or treated with culture filtrates obtained from A. fumigatus. To further identify fungal cell wall components that may affect the expression of SP genes, we examined the effect of different A. fumigatus mutant strains, including dihydroxynaphthalene (DHN)-melanin-deficient ΔpksP, galactomannan (GM)-deficient Δugm1 and galactosaminogalactan (GAG)-deficient Δgt4bc strains. Our results show that the tested strains alter the mRNA expression of SP, with the most prominent and consistent downregulation of the lung-specific SP-C. Our findings also suggest that secondary metabolites rather than the membrane composition of conidia/hyphae inhibit SP-C mRNA expression in NCI-H441 cells.
Collapse
Affiliation(s)
- Natalia Schiefermeier-Mach
- Research and Innovation Unit, Health University of Applied Sciences Tyrol/FH Gesundheit Tirol, 6020 Innsbruck, Austria
| | - Lea Heinrich
- Research and Innovation Unit, Health University of Applied Sciences Tyrol/FH Gesundheit Tirol, 6020 Innsbruck, Austria
| | - Lukas Lechner
- Research and Innovation Unit, Health University of Applied Sciences Tyrol/FH Gesundheit Tirol, 6020 Innsbruck, Austria
| | - Susanne Perkhofer
- Research and Innovation Unit, Health University of Applied Sciences Tyrol/FH Gesundheit Tirol, 6020 Innsbruck, Austria
| |
Collapse
|
10
|
Fishchuk L, Rossokha Z, Pokhylko V, Cherniavska Y, Popova O, Vershyhora V, Kovtun S, Gorovenko N. SFTPB (rs11130866) and NR3C1 (rs41423247) gene variants as potential clinical biomarkers for personalized treatment strategy selection in patients with severe COVID-19 pneumonia. Respir Investig 2023; 61:103-109. [PMID: 36460583 PMCID: PMC9663752 DOI: 10.1016/j.resinv.2022.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/13/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Exploring the pathogenetic mechanisms behind severe lung damage in COVID-19 is crucial. In this study, we decided to focus on two molecular markers that affect surfactant metabolism and lung development: the surfactant protein B (SFTPB) and the glucocorticoid receptor (NR3C1) genes. The aim of our study was to determine the effect of SFTPB (rs11130866) and NR3C1 (rs41423247) gene variants on the course of the disease in patients with COVID-19, and the treatment measures they required. METHODS The study group included 58 patients with a diagnosis of severe "viral COVID-19 pneumonia." Determination of SFTPB and NR3C1 gene variants was performed using the PCR-RFLP method. RESULTS Our results indicate that the presence of the SFTPB gene CC genotype increases the risk of developing acute respiratory distress syndrome in patients with COVID-19 (χ2 = 4.03, p = 0.045, OR = 3.90 [1.19-12.78]). However, patients with the SFTPB gene TT genotype required respiratory support for a shorter period of time. Patients with the NR3C1 gene CC genotype underwent a longer glucocorticoid therapy. Moreover, for patients with the CC genotype, a longer stay in the intensive care unit was detected before lethal outcome. CONCLUSIONS The obtained results confirm the influence of the SFTPB (rs11130866) and NR3C1 (rs41423247) gene variants on the therapy, course, and severity of the disease in patients with COVID-19. Of course, these results require further study, analysis, and larger, complex, systematic research.
Collapse
Affiliation(s)
- Liliia Fishchuk
- Department of Genetic Diagnostics, State Institute of Genetic and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine; State Institution "Reference-centre for Molecular Diagnostic of Public Health Ministry of Ukraine", Kyiv, Ukraine.
| | - Zoia Rossokha
- State Institution "Reference-centre for Molecular Diagnostic of Public Health Ministry of Ukraine", Kyiv, Ukraine
| | - Valeriy Pokhylko
- Department of Pediatrics No 1 with Propedeutics and Neonatology, Poltava State Medical University, Poltava, Ukraine
| | - Yuliia Cherniavska
- Department of Pediatrics No 1 with Propedeutics and Neonatology, Poltava State Medical University, Poltava, Ukraine
| | - Olena Popova
- State Institution "Reference-centre for Molecular Diagnostic of Public Health Ministry of Ukraine", Kyiv, Ukraine
| | - Viktoriia Vershyhora
- State Institution "Reference-centre for Molecular Diagnostic of Public Health Ministry of Ukraine", Kyiv, Ukraine
| | - Serhii Kovtun
- Poltava Regional Clinical Infectious Diseases Hospital of Poltava Regional Council, Poltava, Ukraine
| | - Nataliia Gorovenko
- Department of Genetic Diagnostics, State Institute of Genetic and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
11
|
Axelsson GT, Gudmundsson G, Pratte KA, Aspelund T, Putman RK, Sanders JL, Gudmundsson EF, Hatabu H, Gudmundsdottir V, Gudjonsson A, Hino T, Hida T, Hobbs BD, Cho MH, Silverman EK, Bowler RP, Launer LJ, Jennings LL, Hunninghake GM, Emilsson V, Gudnason V. The Proteomic Profile of Interstitial Lung Abnormalities. Am J Respir Crit Care Med 2022; 206:337-346. [PMID: 35438610 PMCID: PMC9890263 DOI: 10.1164/rccm.202110-2296oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Rationale: Knowledge on biomarkers of interstitial lung disease is incomplete. Interstitial lung abnormalities (ILAs) are radiologic changes that may present in its early stages. Objectives: To uncover blood proteins associated with ILAs using large-scale proteomics methods. Methods: Data from two prospective cohort studies, the AGES-Reykjavik (Age, Gene/Environment Susceptibility-Reykjavik) study (N = 5,259) for biomarker discovery and the COPDGene (Genetic Epidemiology of COPD) study (N = 4,899) for replication, were used. Blood proteins were measured using DNA aptamers, targeting more than 4,700 protein analytes. The association of proteins with ILAs and ILA progression was assessed with regression modeling, as were associations with genetic risk factors. Adaptive Least Absolute Shrinkage and Selection Operator models were applied to bootstrap data samples to discover sets of proteins predictive of ILAs and their progression. Measurements and Main Results: Of 287 associations, SFTPB (surfactant protein B) (odds ratio [OR], 3.71 [95% confidence interval (CI), 3.20-4.30]; P = 4.28 × 10-67), SCGB3A1 (Secretoglobin family 3A member 1) (OR, 2.43 [95% CI, 2.13-2.77]; P = 8.01 × 10-40), and WFDC2 (WAP four-disulfide core domain protein 2) (OR, 2.42 [95% CI, 2.11-2.78]; P = 4.01 × 10-36) were most significantly associated with ILA in AGES-Reykjavik and were replicated in COPDGene. In AGES-Reykjavik, concentrations of SFTPB were associated with the rs35705950 MUC5B (mucin 5B) promoter polymorphism, and SFTPB and WFDC2 had the strongest associations with ILA progression. Multivariate models of ILAs in AGES-Reykjavik, ILAs in COPDGene, and ILA progression in AGES-Reykjavik had validated areas under the receiver operating characteristic curve of 0.880, 0.826, and 0.824, respectively. Conclusions: Novel, replicated associations of ILA, its progression, and genetic risk factors with numerous blood proteins are demonstrated as well as machine-learning-based models with favorable predictive potential. Several proteins are revealed as potential markers of early fibrotic lung disease.
Collapse
Affiliation(s)
- Gisli Thor Axelsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| | - Gunnar Gudmundsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Department of Respiratory Medicine, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Thor Aspelund
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| | | | | | | | - Hiroto Hatabu
- Department of Radiology, and,Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Valborg Gudmundsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| | | | - Takuya Hino
- Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tomoyuki Hida
- Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts;,Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Brian D. Hobbs
- Pulmonary and Critical Care Division,,Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Michael H. Cho
- Pulmonary and Critical Care Division,,Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Edwin K. Silverman
- Pulmonary and Critical Care Division,,Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Russell P. Bowler
- National Jewish Health, Denver, Colorado;,School of Medicine, University of Colorado, Aurora, Colorado
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, Maryland; and
| | - Lori L. Jennings
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Gary M. Hunninghake
- Pulmonary and Critical Care Division,,Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| |
Collapse
|
12
|
Depicolzuane LC, Roberts CM, Thomas NJ, Anderson-Fears K, Liu D, Barbosa JPP, Souza FR, Pimentel AS, Floros J, Gandhi CK. Hydrophilic But Not Hydrophobic Surfactant Protein Genetic Variants Are Associated With Severe Acute Respiratory Syncytial Virus Infection in Children. Front Immunol 2022; 13:922956. [PMID: 35903101 PMCID: PMC9317530 DOI: 10.3389/fimmu.2022.922956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection-related hospitalization in the first year of life. Surfactant dysfunction is central to pathophysiologic mechanisms of various pulmonary diseases including RSV. We hypothesized that RSV severity is associated with single nucleotide polymorphisms (SNPs) of surfactant proteins (SPs). We prospectively enrolled 405 RSV-positive children and divided them into moderate and severe RSV disease. DNA was extracted and genotyped for sixteen specific SP gene SNPs. SP-A1 and A2 haplotypes were assigned. The association of RSV severity with SP gene SNPs was investigated by multivariate logistic regression. A likelihood ratio test was used to test the goodness of fit between two models (one with clinical and demographic data alone and another that included genetic variants). p ≤ 0.05 denotes statistical significance. A molecular dynamics simulation was done to determine the impact of the SFTPA2 rs1965708 on the SP-A behavior under various conditions. Infants with severe disease were more likely to be younger, of lower weight, and exposed to household pets and smoking, as well as having co-infection on admission. A decreased risk of severe RSV was associated with the rs17886395_C of the SFTPA2 and rs2243639_A of the SFTPD, whereas an increased risk was associated with the rs1059047_C of the SFTPA1. RSV severity was not associated with SNPs of SFTPB and SFTPC. An increased risk of severe RSV was associated with the 1A0 genotype of SFTPA2 in its homozygous or heterozygous form with 1A3. A molecular dynamic simulation study of SP-A variants that differ in amino acid 223, an important amino acid change (Q223K) between 1A0 and 1A3, showed no major impact on the behavior of these two variants except for higher thermodynamic stability of the K223 variant. The likelihood ratio test showed that the model with multi-allelic variants along with clinical and demographic data was a better fit to predict RSV severity. In summary, RSV severity was associated with hydrophilic (but not with hydrophobic) SPs gene variants. Collectively, our findings show that SP gene variants may play a key role in RSV infection and have a potential role in prognostication.
Collapse
Affiliation(s)
- Lynnlee C. Depicolzuane
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, PA, United States
| | - Catherine M. Roberts
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, PA, United States
| | - Neal J. Thomas
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, PA, United States
| | - Keenan Anderson-Fears
- Department of Public Health Science, The Pennsylvania State College of Medicine, Hershey, PA, United States
| | - Dajiang Liu
- Department of Public Health Science, The Pennsylvania State College of Medicine, Hershey, PA, United States
| | | | - Felipe Rodrigues Souza
- Departamento de Química, Pontifícia Universidade Católica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - André Silva Pimentel
- Departamento de Química, Pontifícia Universidade Católica do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joanna Floros
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, PA, United States
- Department of Obstetrics & Gynecology, The Pennsylvania State College of Medicine, Hershey, PA, United States
- *Correspondence: Joanna Floros, ; Chintan K. Gandhi,
| | - Chintan K. Gandhi
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, PA, United States
- *Correspondence: Joanna Floros, ; Chintan K. Gandhi,
| |
Collapse
|
13
|
Abbasi A, Chen C, Gandhi CK, Wu R, Pardo A, Selman M, Floros J. Single Nucleotide Polymorphisms (SNP) and SNP-SNP Interactions of the Surfactant Protein Genes Are Associated With Idiopathic Pulmonary Fibrosis in a Mexican Study Group; Comparison With Hypersensitivity Pneumonitis. Front Immunol 2022; 13:842745. [PMID: 35720392 PMCID: PMC9201215 DOI: 10.3389/fimmu.2022.842745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/09/2022] [Indexed: 01/12/2023] Open
Abstract
Surfactant proteins (SPs) are important for normal lung function and innate immunity of the lungs and their genes have been identified with significant genetic variability. Changes in quantity or quality of SPs due to genetic mutations or natural genetic variability may alter their functions and contribute to the host susceptibility for particular diseases. Alternatively, SP single nucleotide polymorphisms (SNPs) can serve as markers to identify disease risk or response to therapies, as shown for other genes in a number of other studies. In the current study, we evaluated associations of SFTP SNPs with idiopathic pulmonary fibrosis (IPF) by studying novel computational models where the epistatic effects (dominant, additive, recessive) of SNP-SNP interactions could be evaluated, and then compared the results with a previously published hypersensitivity pneumonitis (HP) study where the same novel models were used. Mexican Hispanic patients (IPF=84 & HP=75) and 194 healthy control individuals were evaluated. The goal was to identify SP SNPs and SNP-SNP interactions that associate with IPF as well as SNPs and interactions that may be unique to each of these interstitial diseases or common between them. We observed: 1) in terms of IPF, i) three single SFTPA1 SNPs to associate with decreased IPF risk, ii) three SFTPA1 haplotypes to associate with increased IPF risk, and iii) a number of three-SNP interactions to associate with IPF susceptibility. 2) Comparison of IPF and HP, i) three SFTPA1 and one SFTPB SNP associated with decreased risk in IPF but increased risk in HP, and one SFTPA1 SNP associated with decreased risk in both IPF and HP, ii) a number of three-SNP interactions with the same or different effect pattern associated with IPF and/or HP susceptibility, iii) one of the three-SNP interactions that involved SNPs of SFTPA1, SFTPA2, and SFTPD, with the same effect pattern, was associated with a disease-specific outcome, a decreased and increased risk in HP and IPF, respectively. This is the first study that compares the SP gene variants in these two phenotypically similar diseases. Our findings indicate that SNPs of all SFTPs may play an important role in the genetic susceptibility to IPF and HP. Importantly, IPF and HP share some SP genetic variants, suggesting common pathophysiological mechanisms and pathways regarding surfactant biogenesis, but also some differences, highlighting the diverse underlying pathogenic mechanisms between an inflammatory-driven fibrosis (HP) and an epithelial-driven fibrosis (IPF). Alternatively, the significant SNPs identified here, along with SNPs of other genes, could serve as markers to distinguish these two devastating diseases.
Collapse
Affiliation(s)
- Ata Abbasi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.,Department of Pathology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Chixiang Chen
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rongling Wu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Moises Selman
- Unidad de Investigación, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosio Villegas", Mexico City, Mexico
| | - Joanna Floros
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
14
|
Gandhi CK, Thomas NJ, Meixia Y, Spear D, Fu C, Zhou S, Wu R, Keim G, Yehya N, Floros J. SNP–SNP Interactions of Surfactant Protein Genes in Persistent Respiratory Morbidity Susceptibility in Previously Healthy Children. Front Genet 2022; 13:815727. [PMID: 35401703 PMCID: PMC8989419 DOI: 10.3389/fgene.2022.815727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/21/2022] [Indexed: 01/10/2023] Open
Abstract
We studied associations of persistent respiratory morbidity (PRM) at 6 and 12 months after acute respiratory failure (ARF) in previously healthy children with single-nucleotide polymorphisms (SNPs) of surfactant protein (SP) genes. Of the 250 enrolled subjects, 155 and 127 were followed at 6 and 12 months after an ARF episode, respectively. Logistic regression analysis and SNP–SNP interaction models were used. We found that 1) in the multivariate analysis, an increased risk at 6 and 12 months was associated with rs1124_A and rs4715_A of SFTPC, respectively; 2) in a single SNP model, increased and decreased risks of PRM at both timepoints were associated with rs1124 of SFTPC and rs721917 of SFTPD, respectively; an increased risk at 6 months was associated with rs1130866 of SFTPB and rs4715 of SFTPC, and increased and decreased risks at 12 months were associated with rs17886395 of SFTPA2 and rs2243639 of SFTPD, respectively; 3) in a two-SNP model, PRM susceptibility at both timepoints was associated with a number of intergenic interactions between SNPs of the studied SP genes. An increased risk at 12 months was associated with one intragenic (rs1965708 and rs113645 of SFTPA2) interaction; 4) in a three-SNP model, decreased and increased risks at 6 and 12 months, respectively, were associated with an interaction among rs1130866 of SFTPB, rs721917 of SFTPD, and rs1059046 of SFTPA2. A decreased risk at 6 months was associated with an interaction among the same SNPs of SFTPB and SFTPD and the rs1136450 of SFTPA1. The findings revealed that SNPs of all SFTPs appear to play a role in long-term outcomes of ARF survivors and may serve as markers for disease susceptibility.
Collapse
Affiliation(s)
- Chintan K. Gandhi
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Neal J. Thomas
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Ye Meixia
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Debbie Spear
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chenqi Fu
- Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Shouhao Zhou
- Public Health Science, 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
| | - Garrett Keim
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Nadir Yehya
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
- *Correspondence: Joanna Floros,
| |
Collapse
|
15
|
Kou L, Kou P, Luo G, Wei S. Progress of Statin Therapy in the Treatment of Idiopathic Pulmonary Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6197219. [PMID: 35345828 PMCID: PMC8957418 DOI: 10.1155/2022/6197219] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/24/2022] [Indexed: 11/18/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a type of interstitial lung disease (ILD) characterized by the proliferation of fibroblasts and aberrant accumulation of extracellular matrix. These changes are accompanied by structural destruction of the lung tissue and the progressive decline of pulmonary function. In the past few decades, researchers have investigated the pathogenesis of IPF and sought a therapeutic approach for its treatment. Some studies have shown that the occurrence of IPF is related to pulmonary inflammatory injury; however, its specific etiology and pathogenesis remain unknown, and no effective treatment, with the exception of lung transplantation, has been identified yet. Several basic science and clinical studies in recent years have shown that statins, the traditional lipid-lowering drugs, exert significant antifibrotic effects, which can delay the progression of IPF and impairment of pulmonary function. This article is aimed at summarizing the current understanding of the pathogenesis of IPF, the progress of research on the use of statins in IPF models and clinical trials, and its main molecular targets.
Collapse
Affiliation(s)
- Leiya Kou
- Department of Respiratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, China
- Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Pei Kou
- Department of Medical Record, Wuhan No. 1 Hospital, Wuhan 430022, China
| | - Guangwei Luo
- Department of Respiratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, China
| | - Shuang Wei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital Tongji Medical College Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
16
|
Floros J, Thorenoor N, Tsotakos N, Phelps DS. Human Surfactant Protein SP-A1 and SP-A2 Variants Differentially Affect the Alveolar Microenvironment, Surfactant Structure, Regulation and Function of the Alveolar Macrophage, and Animal and Human Survival Under Various Conditions. Front Immunol 2021; 12:681639. [PMID: 34484180 PMCID: PMC8415824 DOI: 10.3389/fimmu.2021.681639] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
The human innate host defense molecules, SP-A1 and SP-A2 variants, differentially affect survival after infection in mice and in lung transplant patients. SP-A interacts with the sentinel innate immune cell in the alveolus, the alveolar macrophage (AM), and modulates its function and regulation. SP-A also plays a role in pulmonary surfactant-related aspects, including surfactant structure and reorganization. For most (if not all) pulmonary diseases there is a dysregulation of host defense and inflammatory processes and/or surfactant dysfunction or deficiency. Because SP-A plays a role in both of these general processes where one or both may become aberrant in pulmonary disease, SP-A stands to be an important molecule in health and disease. In humans (unlike in rodents) SP-A is encoded by two genes (SFTPA1 and SFTPA2) and each has been identified with extensive genetic and epigenetic complexity. In this review, we focus on functional, structural, and regulatory differences between the two SP-A gene-specific products, SP-A1 and SP-A2, and among their corresponding variants. We discuss the differential impact of these variants on the surfactant structure, the alveolar microenvironment, the regulation of epithelial type II miRNome, the regulation and function of the AM, the overall survival of the organism after infection, and others. Although there have been a number of reviews on SP-A, this is the first review that provides such a comprehensive account of the differences between human SP-A1 and SP-A2.
Collapse
Affiliation(s)
- Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Biochemistry & Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Nikolaos Tsotakos
- School of Science, Engineering, and Technology, The Pennsylvania State University, Harrisburg, PA, United States
| | - David S Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
17
|
Marchioni A, Tonelli R, Cerri S, Castaniere I, Andrisani D, Gozzi F, Bruzzi G, Manicardi L, Moretti A, Demurtas J, Baroncini S, Andreani A, Cappiello GF, Busani S, Fantini R, Tabbì L, Samarelli AV, Clini E. Pulmonary Stretch and Lung Mechanotransduction: Implications for Progression in the Fibrotic Lung. Int J Mol Sci 2021; 22:ijms22126443. [PMID: 34208586 PMCID: PMC8234308 DOI: 10.3390/ijms22126443] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 12/18/2022] Open
Abstract
Lung fibrosis results from the synergic interplay between regenerative deficits of the alveolar epithelium and dysregulated mechanisms of repair in response to alveolar and vascular damage, which is followed by progressive fibroblast and myofibroblast proliferation and excessive deposition of the extracellular matrix. The increased parenchymal stiffness of fibrotic lungs significantly affects respiratory mechanics, making the lung more fragile and prone to non-physiological stress during spontaneous breathing and mechanical ventilation. Given their parenchymal inhomogeneity, fibrotic lungs may display an anisotropic response to mechanical stresses with different regional deformations (micro-strain). This behavior is not described by the standard stress–strain curve but follows the mechano-elastic models of “squishy balls”, where the elastic limit can be reached due to the excessive deformation of parenchymal areas with normal elasticity that are surrounded by inelastic fibrous tissue or collapsed induration areas, which tend to protrude outside the fibrous ring. Increasing evidence has shown that non-physiological mechanical forces applied to fibrotic lungs with associated abnormal mechanotransduction could favor the progression of pulmonary fibrosis. With this review, we aim to summarize the state of the art on the relation between mechanical forces acting on the lung and biological response in pulmonary fibrosis, with a focus on the progression of damage in the fibrotic lung during spontaneous breathing and assisted ventilatory support.
Collapse
Affiliation(s)
- Alessandro Marchioni
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41125 Modena, Italy
- Correspondence:
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Ivana Castaniere
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Dario Andrisani
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41125 Modena, Italy
| | - Filippo Gozzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
- Clinical and Experimental Medicine PhD Program, University of Modena Reggio Emilia, 41125 Modena, Italy
| | - Giulia Bruzzi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Linda Manicardi
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Antonio Moretti
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Jacopo Demurtas
- Primary Care Department USL Toscana Sud Est-Grosseto, 58100 Grosseto, Italy;
| | - Serena Baroncini
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Alessandro Andreani
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Gaia Francesca Cappiello
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Stefano Busani
- University Hospital of Modena, Anesthesiology Unit, University of Modena Reggio Emilia, 41124 Modena, Italy;
| | - Riccardo Fantini
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Luca Tabbì
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena and Reggio Emilia, 41125 Modena, Italy; (A.M.); (S.C.); (I.C.); (D.A.); (F.G.); (G.B.); (L.M.); (A.M.); (A.V.S.); (E.C.)
- University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, 41125 Modena, Italy; (S.B.); (A.A.); (G.F.C.); (R.F.); (L.T.)
| |
Collapse
|
18
|
Sun S, Huang C, Leng D, Chen C, Zhang T, Lei KC, Zhang XD. Gene fusion of IL7 involved in the regulation of idiopathic pulmonary fibrosis. Ther Adv Respir Dis 2021; 15:1753466621995045. [PMID: 33878985 PMCID: PMC8064517 DOI: 10.1177/1753466621995045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a rare form of immune-mediated interstitial lung disease characterized by progressive pulmonary fibrosis and scarring. The pathogenesis of IPF is still unclear. Gene fusion events exist universally during transcription and show alternated patterns in a variety of lung diseases. Therefore, the comprehension of the function of gene fusion in IPF might shed light on IPF pathogenesis research and facilitate treatment development. Methods: In this study, we included 91 transcriptome datasets from the National Center for Biotechnology Information (NCBI), including 52 IPF patients and 39 healthy controls. We detected fusion events in these datasets and probed gene fusion-associated differential gene expression and functional pathways. To obtain robust results, we corrected the batch bias across different projects. Results: We identified 1550 gene fusion events in all transcriptomes and studied the possible impacts of IL7 = AC083837.1 gene fusion. The two genes locate adjacently in chromosome 8 and share the same promoters. Their fusion is associated with differential expression of 282 genes enriched in six Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and 35 functional gene sets. Gene ontology (GO) enrichment analysis shows that IL7 = AC083837.1 gene fusion is associated with the enrichment of 187 gene sets. The co-expression network of interleukin-7 (IL7) indicates that decreased IL7 expression is associated with many pathways that regulate IPF progress. Conclusion: Based on the results, we conclude that IL7 = AC083837.1 gene fusion might exacerbate fibrosis in IPF via enhancing activities of natural killer cell-mediated cytotoxicity, skin cell apoptosis, and vessel angiogenesis, the interaction of which contributes to the development of fibrosis and the deterioration of respiratory function of IPF patients. Our work unveils the possible roles of gene fusion in regulating IPF and demonstrates that gene fusion investigation is a valid approach in probing immunologic mechanisms and searching potential therapeutic targets for treating IPF. The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Shixue Sun
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chen Huang
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Dongliang Leng
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Chang Chen
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Teng Zhang
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Kuan Cheok Lei
- CRDA, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China
| | - Xiaohua Douglas Zhang
- CRDA, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| |
Collapse
|
19
|
Gallob F, Brcic L, Eidenhammer S, Rumpp F, Nerlich A, Popper H. Senescence and autophagy in usual interstitial pneumonia of different etiology. Virchows Arch 2021; 478:497-506. [PMID: 32851507 PMCID: PMC7973921 DOI: 10.1007/s00428-020-02917-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/15/2020] [Accepted: 08/19/2020] [Indexed: 01/25/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease with a dismal prognosis. Currently, the causing agent(s) are poorly understood. Recent data suggest that senescence and autophagy might play a role in its development, as well as changes in metabolism due to hypoxic conditions. In this study, the expression of senescence markers in 23 cases of usual interstitial pneumonia (UIP)/IPF and UIP/chronic autoimmune diseases (UIP/AuD) was investigated. The status of autophagy was evaluated with respect to either antiinflammatory or antihypoxia function. Formalin-fixed paraffin-embedded tissues of UIP were selected for immunohistochemistry with antibodies for p21, p16, and β-galactosidase (senescence); for LC3, SIRT1, MAP1S, and pAMKα (autophagy); and for LDH and GLUT1 (metabolism). Epithelial cells in cystic remodeled areas of UIP stained for p16 and p21, p16 being more specific compared with p21. Myofibroblasts were negative in all cases. An upregulation of all four autophagy markers was seen not only in epithelia within remodeled areas and proliferating myofibroblasts, but also in bronchial epithelia and pneumocytes. Upregulated autophagy points to a compensatory mechanism for hypoxia; therefore, LDH and GLUT1 were investigated. Their expression was present in epithelia within cystic remodeling and in myofibroblasts. The cells within the remodeled areas stained for cytokeratin 5, but coexpressed TTF1, confirming their origin from basal cells of bronchioles. Within this population, senescent cells arise. Our results indicated that autophagy in UIP very likely helps cells to survive in hypoxic condition. By phagocytosis of cellular debris, they supplement their need for nutrition, and by upregulating LDH and GLUT1, they compensate for local hypoxia.
Collapse
Affiliation(s)
- Florian Gallob
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Sylvia Eidenhammer
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Florian Rumpp
- Pulmonology Department, Clinics Munich-Bogenhausen, Munich, Germany
| | - Andreas Nerlich
- Pathology Department, Clinics Munich-Bogenhausen, Munich, Germany
| | - Helmut Popper
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria.
| |
Collapse
|
20
|
Gandhi CK, Chen C, Amatya S, Yang L, Fu C, Zhou S, Wu R, Buendía-Roldan I, Selman M, Pardo A, Floros J. SNP and Haplotype Interaction Models Reveal Association of Surfactant Protein Gene Polymorphisms With Hypersensitivity Pneumonitis of Mexican Population. Front Med (Lausanne) 2021; 7:588404. [PMID: 33469544 PMCID: PMC7813780 DOI: 10.3389/fmed.2020.588404] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/07/2020] [Indexed: 01/21/2023] Open
Abstract
Background: Hypersensitivity pneumonitis (HP) is an interstitial lung disease caused by inhalation of common environmental organic particles. Surfactant proteins (SPs) play a role in innate immunity and surfactant function. We hypothesized that single nucleotide polymorphisms (SNPs) or haplotypes of the SP genes associate with HP. Methods: Seventy-five HP patients caused by avian antigen and 258 controls, asymptomatic antigen exposed and non-exposed were enrolled. SNP association was performed using logistic regression analysis and SNP-SNP interaction models. Results: Based on odds ratio, regression analyses showed association of (a) rs7316_G, 1A3 (protective) compared to antigen exposed; (b) male sex, smoking, rs721917_T and rs1130866_T (protective) compared to non-exposed controls with HP; (c) compared to antigen exposed, 25 interactions associated with HP in a three-SNP model; (d) compared to non-exposed, (i) rs1136451 associated with increased, whereas rs1136450 and rs1130866 associated with lower HP risk, (ii) 97 interactions associated with HP in a three-SNP model. The majority of SNP-SNP interactions associated with increased HP risk involved SNPs of the hydrophilic SPs, whereas, the majority of interactions associated with lower HP risk involved SNPs of both hydrophilic and hydrophobic SPs; (e) haplotypes of SP genes associated with HP risk. Conclusions: The complexity of SNPs interactions of the SFTP genes observed indicate that the lung inflammatory response to avian antigens is modulated by a complex gene interplay rather than by single SNPs.
Collapse
Affiliation(s)
- Chintan K. Gandhi
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chixiang Chen
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Shaili Amatya
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Lili Yang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenqi Fu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Shouhao Zhou
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rongling Wu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Ivette Buendía-Roldan
- Unidad de Investigación, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Moisés Selman
- Unidad de Investigación, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Annie Pardo
- Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA, United States
- Department of Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
21
|
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
|
22
|
Thorenoor N, Phelps DS, Floros J. Differential Sex-Dependent Regulation of the Alveolar Macrophage miRNome of SP-A2 and co-ex (SP-A1/SP-A2) and Sex Differences Attenuation after 18 h of Ozone Exposure. Antioxidants (Basel) 2020; 9:antiox9121190. [PMID: 33260937 PMCID: PMC7768498 DOI: 10.3390/antiox9121190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Human SP-A1 and SP-A2, encoded by SFTPA1 and SFTPA2, and their genetic variants differentially impact alveolar macrophage (AM) functions and regulation, including the miRNome. We investigated whether miRNome differences previously observed between AM from SP-A2 and SP-A1/SP-A2 mice are due to continued qualitative differences or a delayed response of mice carrying a single gene. Methods: Human transgenic (hTG) mice, carrying SP-A2 or both SP-A genes, and SP-A-KO mice were exposed to filtered air (FA) or ozone (O3). AM miRNA levels, target gene expression, and pathways determined 18 h after O3 exposure. RESULTS: We found (a) differences in miRNome due to sex, SP-A genotype, and exposure; (b) miRNome of both sexes was largely downregulated by O3, and co-ex had fewer changed (≥2-fold) miRNAs than either group; (c) the number and direction of the expression of genes with significant changes in males and females in co-ex are almost the opposite of those in SP-A2; (d) the same pathways were found in the studied groups; and (e) O3 exposure attenuated sex differences with a higher number of genotype-dependent and genotype-independent miRNAs common in both sexes after O3 exposure. Conclusion: Qualitative differences between SP-A2 and co-ex persist 18 h post-O3, and O3 attenuates sex differences.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
- Department of Biochemistry & Molecular Biology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - David S. Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
- Department of Obstetrics & Gynecology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
- Correspondence:
| |
Collapse
|
23
|
Thorenoor N, S. Phelps D, Kala P, Ravi R, Floros Phelps A, M. Umstead T, Zhang X, Floros J. Impact of Surfactant Protein-A Variants on Survival in Aged Mice in Response to Klebsiella pneumoniae Infection and Ozone: Serendipity in Action. Microorganisms 2020; 8:microorganisms8091276. [PMID: 32825654 PMCID: PMC7570056 DOI: 10.3390/microorganisms8091276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 01/10/2023] Open
Abstract
Innate immune molecules, SP-A1 (6A2, 6A4) and SP-A2 (1A0, 1A3), differentially affect young mouse survival after infection. Here, we investigated the impact of SP-A variants on the survival of aged mice. hTG mice carried a different SP-A1 or SP-A2 variant and SP-A-KO were either infected with Klebsiella pneumoniae or exposed to filtered air (FA) or ozone (O3) prior to infection, and their survival monitored over 14 days. In response to infection alone, no gene- or sex-specific (except for 6A2) differences were observed; variant-specific survival was observed (1A0 > 6A4). In response to O3, gene-, sex-, and variant-specific survival was observed with SP-A2 variants showing better survival in males than females, and 1A0 females > 1A3 females. A serendipitous, and perhaps clinically important observation was made; mice exposed to FA prior to infection exhibited significantly better survival than infected alone mice. 1A0 provided an overall better survival in males and/or females indicating a differential role for SP-A genetics. Improved ventilation, as provided by FA, resulted in a survival of significant magnitude in aged mice and perhaps to a lesser extent in young mice. This may have clinical application especially within the context of the current pandemic.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence: (N.T.); (J.F.)
| | - David S. Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
| | - Padma Kala
- Independent Consultant, Upper Saddle River, NJ 07458, USA;
| | - Radhika Ravi
- Division of Anesthesia, Department of Surgery, Veterans Affairs New Jersey Health Care System, 385 Tremont Avenue, East Orange, NJ 07018, USA;
| | | | - Todd M. Umstead
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
| | - Xuesheng Zhang
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
- Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence: (N.T.); (J.F.)
| |
Collapse
|
24
|
Abstract
Pulmonary alveolar proteinosis (PAP) is a rare respiratory syndrome characterised by the accumulation of surfactant lipoproteins within the alveoli. According to various pathogenetic mechanisms and aetiologies, PAP is classified as primary, secondary or congenital. Primary PAP is led by a granulocyte–macrophage colony-stimulating factor (GM-CSF) signalling disruption; the autoimmune form is driven by the presence of anti GM-CSF autoantibodies and represents 90% of all the PAP cases; and the hereditary form is the result of mutations in genes encoding GM-CSF receptor. Secondary PAP is associated with various diseases causing a reduction in function and/or number of alveolar macrophages. Congenital PAP emerges as a consequence of corrupted surfactant production, due to mutations in surfactant proteins or lipid transporter, or mutations affecting lung development. The clinical manifestations are various, ranging from insidious onset to acute or progressive respiratory failure, including premature death within the first days of life in neonates with congenital surfactant production disorders. The diagnostic workup includes clinical and radiological assessment (respiratory function test, high-resolution chest computed tomography), laboratory tests (anti-GM-CSF autoantibodies dosage, GM-CSF serum level and GM-CSF signalling test), and genetic tests. Whole-lung lavage is the current gold standard of care of PAP; however, the therapeutic approach depends on the pathogenic form and disease severity, including GM-CSF augmentation strategies in autoimmune PAP and other promising new treatments. A concise educational review of pulmonary alveolar proteinosis (PAP), a rare respiratory syndrome with various and heterogeneous aetiologies, caused by the impairment of pulmonary surfactant clearance or by abnormal surfactant productionhttps://bit.ly/3aFpQm9
Collapse
Affiliation(s)
| | - Ilaria Campo
- Pneumology Unit, IRCCS Policlinico San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
25
|
King SD, Chen SY. Recent progress on surfactant protein A: cellular function in lung and kidney disease development. Am J Physiol Cell Physiol 2020; 319:C316-C320. [PMID: 32639871 DOI: 10.1152/ajpcell.00195.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pulmonary surfactant is a heterogeneous active surface complex made up of lipids and proteins. The major glycoprotein in surfactant is surfactant protein A (SP-A), which is released into the alveolar lumen from cytoplasmic lamellar bodies in type II alveolar epithelial cells. SP-A is involved in phospholipid absorption. SP-A together with other surfactant proteins and phospholipids prevent alveolar collapse during respiration by decreasing the surface tension of the air-liquid interface. Additionally, SP-A interacts with pathogens to prevent their propagation and regulate host immune responses. Studies in human and animal models have shown that deficiencies or mutations in surfactant components result in various lung or kidney pathologies, suggesting a role for SP-A in the development of lung and kidney diseases. In this mini-review, we discuss the current understanding of SP-A functions, recent findings of its dysfunction in specific lung and kidney pathologies, and how SP-A has been used as a biomarker to detect the outcome of lung diseases.
Collapse
Affiliation(s)
- Skylar D King
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Shi-You Chen
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri.,Department of Molecular Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
26
|
Liu L, Qin J, Guo T, Chen P, Ouyang R, Peng H, Luo H. Identification and functional characterization of a novel surfactant protein A2 mutation (p.N207Y) in a Chinese family with idiopathic pulmonary fibrosis. Mol Genet Genomic Med 2020; 8:e1393. [PMID: 32602668 PMCID: PMC7507553 DOI: 10.1002/mgg3.1393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/20/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a serious disorder with a high mortality rate worldwide. It is characterized by irreversible scarring of the lung parenchyma resulting from excessive collagen production by proliferating fibroblasts/myofibroblasts. Previous studies have revealed that mutations in surfactant protein‐related genes and telomerase complex genes are crucial underlying genetic factors. Methods In this study, we enrolled a family with IPF from the central southern region of China. Whole‐exome sequencing was employed to explore candidate genes in this family. Real‐time PCR and western blotting were used to study the functions of the identified mutations in vitro. Results A novel mutation (NM_001098668.4: c.619A>T; NP_001092138.1: p.N207Y) in surfactant protein A2 (SFTPA2,), having not been previously reported to be a mutation, was identified and co‐separated with all affected individuals in the IPF family. Functional research further revealed that the novel mutation affects the secretion of SFTPA2 protein and induces endoplasmic reticulum stress as well as apoptosis in A549 cells. Conclusion We are confident that this novel mutation (NM_001098668.4: c.619A>T; NP_001092138.1: p.N207Y) in SFTPA2 is the genetic mutation of the IPF family. Our study not only confirms the importance of SFTPA2 in IPF but also expands the spectrum of SFTPA2 mutations and contributes to the genetic diagnosis and counseling of IPF patients.
Collapse
Affiliation(s)
- Lv Liu
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jieli Qin
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ting Guo
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ping Chen
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ruoyun Ouyang
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hong Peng
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hong Luo
- Department of Respiratory Medicine, Diagnosis and Treatment Center of Respiratory Disease, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
27
|
Gandhi CK, Chen C, Wu R, Yang L, Thorenoor N, Thomas NJ, DiAngelo SL, Spear D, Keim G, Yehya N, Floros J. Association of SNP-SNP Interactions of Surfactant Protein Genes with Pediatric Acute Respiratory Failure. J Clin Med 2020; 9:jcm9041183. [PMID: 32326132 PMCID: PMC7231046 DOI: 10.3390/jcm9041183] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 12/19/2022] Open
Abstract
The hallmarks of pediatric acute respiratory failure (ARF) are dysregulated inflammation and surfactant dysfunction. The objective is to study association of surfactant protein (SP) genes’ single nucleotide polymorphisms (SNPs) with ARF and its morbidity: pulmonary dysfunction at discharge (PDAD), employing a single-, two-, and three-SNP interaction model. We enrolled 468 newborn controls and 248 children aged ≤ 24 months with ARF; 86 developed PDAD. Using quantitative genetic principles, we tested the association of SP genes SNPs with ARF and PDAD. We observed a dominant effect of rs4715 of the SFTPC on ARF risk. In a three-SNP model, we found (a) 34 significant interactions among SNPs of SFTPA1, SFTPA2, and SFTPC associated with ARF (p = 0.000000002–0.05); 15 and 19 of those interactions were associated with increased and decreased risk for ARF, respectively; (b) intergenic SNP–SNP interactions of both hydrophobic and hydrophilic SP genes associated with PDAD (p = 0.00002–0.03). The majority of intra- and intergenic interactions associated with ARF involve the SFTPA2 SNPs, whereas most of the intra- and intergenic interactions associated with PDAD are of SFTPA1 SNPs. We also observed a dominant effect of haplotypes GG of SFTPA1 associated with increased and AA of SFTPC associated with decreased ARF risk (p = 0.02). To the best of our knowledge, this is the first study showing an association of complex interactions of SP genes with ARF and PDAD. Our data indicate that SP genes polymorphisms may contribute to ARF pathogenesis and subsequent PDAD and/or may serve as markers for disease susceptibility in healthy children.
Collapse
Affiliation(s)
- Chintan K. Gandhi
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Chixiang Chen
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rongling Wu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Lili Yang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nithyananda Thorenoor
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Neal J. Thomas
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Susan L. DiAngelo
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Debbie Spear
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Garrett Keim
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nadir Yehya
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joanna Floros
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
- Department of Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence:
| |
Collapse
|
28
|
Yang Y, Li Q, Tan F, Zhang J, Zhu W. Mechanism of IL-8-induced acute lung injury through pulmonary surfactant proteins A and B. Exp Ther Med 2019; 19:287-293. [PMID: 31853301 PMCID: PMC6909794 DOI: 10.3892/etm.2019.8192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/01/2019] [Indexed: 12/03/2022] Open
Abstract
This study explored how interleukin-8 (IL-8) causes acute lung injury (ALI) through pulmonary surfactant protein A (SP-A) and surfactant protein B (SP-B). Serum was collected from 53 ALI patients and further 56 healthy subjects who underwent physical examination. The IL-8, SP-A, and SP-B levels were determined using enzyme-linked immunosorbent assay (ELISA). An ALI model was constructed using lipopolysaccharide (LSP)-induced normal A549 cells. siRNA was employed to interfere with the expression of IL-8, SP-A and SP-B. Western blot analysis was carried out to determine the protein levels, and MTT assay to determine the cell activity. In addition, co-immunoprecipitation (Co-IP) assay was used to verify the interaction between IL-8, SP-A and SP-B. ALI patients showed high expression of serum IL-8, and low expression of SP-A and SP-B, and IL-8 was negatively correlated with SP-A and SP-B, respectively. LSP-induced normal A549 cells showed increased expression of IL-8 and decreased expression of SP-A and SP-B. Silencing IL-8 led to increased expression levels of SP-A, SP-B and Bcl2, decreased expression levels of caspase-9, caspase-3, Bax, TNF-α, IL-17 and IL-1β, reduced cell apoptosis rate, and enhanced cell viability. Silencing SP-A and SP-B resulted in increased expression of IL-8, caspase-9, caspase-3, Bax, TNF-α, IL-17 and IL-1β, and decreased expression of Bcl2. Co-IP assay revealed that IL-8 could interact with SP-A and SP-B, respectively. IL-8 induces apoptosis by inhibiting SP-A and SP-B, and intensifies cellular inflammatory reaction, leading eventually to ALI.
Collapse
Affiliation(s)
- Yinong Yang
- Medical School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Qing Li
- Nursing School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Feng Tan
- School of Stomatology, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Jun Zhang
- Medical School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Wu Zhu
- Medical School, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| |
Collapse
|
29
|
Udagawa C, Horinouchi H, Shiraishi K, Kohno T, Okusaka T, Ueno H, Tamura K, Ohe Y, Zembutsu H. Whole genome sequencing to identify predictive markers for the risk of drug-induced interstitial lung disease. PLoS One 2019; 14:e0223371. [PMID: 31584970 PMCID: PMC6777826 DOI: 10.1371/journal.pone.0223371] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/19/2019] [Indexed: 01/13/2023] Open
Abstract
Drug-induced interstitial lung disease (DIILD) is a serious side effect of chemotherapy in cancer patients with an extremely high mortality rate. In this study, to identify genetic variants with greater risk of DIILD, we carried out whole genome sequencing (WGS) of germline DNA samples from 26 patients who developed DIILD, and conducted a case-control association study between these 26 cases and general Japanese population controls registered in the integrative Japanese Genome Variation Database (iJGVD) as a screening study. The associations of 42 single nucleotide variants (SNVs) showing P < 0.0001 were further validated using an independent cohort of 18 DIILD cases as a replication study. A further combined analysis of the screening and replication studies showed a possible association of two SNVs, rs35198919 in intron 1 of the chromosome 22 open reading frame 34 (C22orf34) and rs12625311 in intron 1 of the teashirt zinc finger homeobox 2 (TSHZ2), with DIILD (Pcombined = 1.87 × 10−5 and 5.16 × 10−5, respectively). Furthermore, in a subgroup analysis of epidermal growth factor receptor (EGFR)–tyrosine kinase inhibitor (TKI)-induced interstitial lung disease (ILD), we observed seven candidate SNVs that were possibly associated with ILD (P < 0.00001). This is the first study to identify genetic markers for the risk of DIILD using WGS. Collectively, our novel findings indicate that these SNVs may be applicable for predicting the risk of DIILD in patients receiving chemotherapy.
Collapse
Affiliation(s)
- Chihiro Udagawa
- Division of Genetics, National Cancer Center Research Institute, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hitoshi Zembutsu
- Division of Genetics, National Cancer Center Research Institute, Tokyo, Japan
- Project for Development of Liquid Biopsy Diagnosis, Japanese Foundation for Cancer Research, Research Institute, Tokyo, Japan
- * E-mail:
| |
Collapse
|
30
|
Tachiwada T, Oda K, Tahara M, Sennari K, Nemoto K, Noguchi S, Kawanami T, Kido T, Yamaguchi H, Yatera K. Fatal Acute Exacerbation of Familial Interstitial Pneumonia Complicated with Dyskeratosis Congenita after Influenza Virus B Infection. Intern Med 2019; 58:2683-2687. [PMID: 31178485 PMCID: PMC6794170 DOI: 10.2169/internalmedicine.2413-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Dyskeratosis congenita (DC) is occasionally complicated in patients with familial interstitial pneumonia (FIP). However, there have been no reports of FIP patients with DC that develop acute exacerbation (AE). We herein report a FIP patient with DC that showed AE of FIP after influenza virus B infection. Although DC is a rare disease in clinical practice, physicians should keep in mind that FIP combined with DC has the potential to cause AE.
Collapse
Affiliation(s)
- Takashi Tachiwada
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Keishi Oda
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Masahiro Tahara
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Konomi Sennari
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Kazuki Nemoto
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Shingo Noguchi
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Toshinori Kawanami
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | - Takashi Kido
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| | | | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan
| |
Collapse
|
31
|
Thorenoor N, Kawasawa YI, Gandhi CK, Zhang X, Floros J. Differential Impact of Co-expressed SP-A1/SP-A2 Protein on AM miRNome; Sex Differences. Front Immunol 2019; 10:1960. [PMID: 31475015 PMCID: PMC6707024 DOI: 10.3389/fimmu.2019.01960] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/02/2019] [Indexed: 12/04/2022] Open
Abstract
In humans there are two surfactant protein A (SP-A) functional genes SFTPA1 and SFTPA2 encoding innate immune molecules, SP-A1 and SP-A2, respectively, with numerous genetic variants each. SP-A interacts and regulates many of the functions of alveolar macrophages (AM). It is shown that SP-A variants differ in their ability to regulate the AM miRNome in response to oxidative stress (OxS). Because humans have both SP-A gene products, we were interested to determine the combined effect of co-expressed SP-A1/SP-A2 (co-ex) in response to ozone (O3) induced OxS on AM miRNome. Human transgenic (hTG) mice, carrying both SP-A1/SP-A2 (6A2/1A0, co-ex) and SP-A- KO were utilized. The hTG and KO mice were exposed to filtered air (FA) or O3 and miRNA levels were measured after AM isolation with or without normalization to KO. We found: (i) The AM miRNome of co-ex males and females in response to OxS to be largely downregulated after normalization to KO, but after Bonferroni multiple comparison analysis only in females the AM miRNome remained significantly different compared to control (FA); (ii) The targets of the significantly changed miRNAs were downregulated in females and upregulated in males; (iii) Several of the validated mRNA targets were involved in pro-inflammatory response, anti-apoptosis, cell cycle, cellular growth and proliferation; (iv) The AM of SP-A2 male, shown, previously to have major effect on the male AM miRNome in response to OxS, shared similarities with the co-ex, namely in pathways involved in the pro-inflammatory response and anti-apoptosis but also exhibited differences with the cell-cycle, growth, and proliferation pathway being involved in co-ex and ROS homeostasis in SP-A2 male. We speculate that the presence of both gene products vs. single gene products differentially impact the AM responses in males and females in response to OxS.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Departments of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xuesheng Zhang
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
32
|
Albert RK, Smith B, Perlman CE, Schwartz DA. Is Progression of Pulmonary Fibrosis due to Ventilation-induced Lung Injury? Am J Respir Crit Care Med 2019; 200:140-151. [PMID: 31022350 PMCID: PMC6635778 DOI: 10.1164/rccm.201903-0497pp] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/22/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
| | - Bradford Smith
- Department of Bioengineering, University of Colorado, Aurora, Colorado; and
| | - Carrie E. Perlman
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | | |
Collapse
|
33
|
Trapnell BC, Nakata K, Bonella F, Campo I, Griese M, Hamilton J, Wang T, Morgan C, Cottin V, McCarthy C. Pulmonary alveolar proteinosis. Nat Rev Dis Primers 2019; 5:16. [PMID: 30846703 DOI: 10.1038/s41572-019-0066-3] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pulmonary alveolar proteinosis (PAP) is a syndrome characterized by the accumulation of alveolar surfactant and dysfunction of alveolar macrophages. PAP results in progressive dyspnoea of insidious onset, hypoxaemic respiratory failure, secondary infections and pulmonary fibrosis. PAP can be classified into different types on the basis of the pathogenetic mechanism: primary PAP is characterized by the disruption of granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling and can be autoimmune (caused by elevated levels of GM-CSF autoantibodies) or hereditary (due to mutations in CSF2RA or CSF2RB, encoding GM-CSF receptor subunits); secondary PAP results from various underlying conditions; and congenital PAP is caused by mutations in genes involved in surfactant production. In most patients, pathogenesis is driven by reduced GM-CSF-dependent cholesterol clearance in alveolar macrophages, which impairs alveolar surfactant clearance. PAP has a prevalence of at least 7 cases per million individuals in large population studies and affects men, women and children of all ages, ethnicities and geographical locations irrespective of socioeconomic status, although it is more-prevalent in smokers. Autoimmune PAP accounts for >90% of all cases. Management aims at improving symptoms and quality of life; whole-lung lavage effectively removes excessive surfactant. Novel pathogenesis-based therapies are in development, targeting GM-CSF signalling, immune modulation and cholesterol homeostasis.
Collapse
Affiliation(s)
- Bruce C Trapnell
- Translational Pulmonary Science Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Koh Nakata
- Bioscience Medical Research Center, Niigata University, Niigata, Japan
| | - Francesco Bonella
- Interstitial and Rare Lung Disease Unit, Pneumology Department, Ruhrlandklinik University Hospital, University of Essen, Essen, Germany
| | - Ilaria Campo
- Pneumology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Matthias Griese
- Pediatric Pneumology, University of Munich, German Center for Lung Research (DZL), Munich, Germany
| | - John Hamilton
- University of Melbourne, Parkville, Victoria, Australia
| | - Tisha Wang
- Department of Medicine, University of California, Los Angeles, CA, USA
| | - Cliff Morgan
- Department of Critical Care and Anaesthesia, Royal Brompton Hospital, London, UK
| | - Vincent Cottin
- National Reference Center for Rare Pulmonary Diseases, University of Lyon, Lyon, France
| | - Cormac McCarthy
- Department of Medicine, St. Vincent's University Hospital and University College Dublin, Dublin, Ireland
| |
Collapse
|
34
|
Doubková M, Staňo Kozubík K, Radová L, Pešová M, Trizuljak J, Pál K, Svobodová K, Réblová K, Svozilová H, Vrzalová Z, Pospíšilová Š, Doubek M. A novel germline mutation of the SFTPA1 gene in familial interstitial pneumonia. Hum Genome Var 2019; 6:12. [PMID: 30854216 PMCID: PMC6399245 DOI: 10.1038/s41439-019-0044-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 02/03/2019] [Accepted: 02/03/2019] [Indexed: 12/11/2022] Open
Abstract
Different genes related to alveolar stability have been associated with familial interstitial pneumonia (FIP). Here, we report a novel, rare SFTPA1 variant in a family with idiopathic interstitial pneumonia (IIP). We performed whole-exome sequencing on germline DNA samples from four members of one family; three of them showed signs of pulmonary fibrosis (idiopathic interstitial pneumonia) with autosomal-dominant inheritance. A heterozygous single nucleotide variant c.532 G > A in the SFTPA1 gene has been identified. This variant encodes the substitution p.(Val178Met), localized within the carbohydrate recognition domain of surfactant protein A and segregates with the genes causing idiopathic interstitial pneumonia. This rare variant has not been previously reported. We also analyzed the detected sequence variant in the protein structure in silico. The replacement of valine by the larger methionine inside the protein may cause a disruption in the protein structure. The c.532 G > A variant was further validated using Sanger sequencing of the amplicons, confirming the diagnosis in all symptomatic family members. Moreover, this variant was also found by Sanger sequencing in one other symptomatic family member and one young asymptomatic family member. The autosomal-dominant inheritance, the family history of IIP, and the evidence of a mutation occurring in part of the SFTPA1 gene all suggest a novel variant that causes FIP. Researchers have identified a novel mutation that causes inheritable and ultimately deadly scarring of the tiny air sacs in the lungs, the alveoli. In familial interstitial pneumonia (FIP), alveoli are scarred and stiffened by inflammation, not by bacterial infection as the word ‘pneumonia’ usually indicates. Michael Doubek at University Hospital and Central European Institute of Technology, Brno, Czech Republic and co-workers investigated a suspected case of FIP following the early death of one family member. Sequencing the genomes of other family members revealed that they shared a mutation in a protein that keeps alveoli moist, aiding oxygen absorption. Computer analysis showed that the mutation probably changed the protein’s shape, preventing it from functioning. Identifying mutations that cause FIP will help provide proactive treatment for family members who are at risk but not yet showing symptoms.
Collapse
Affiliation(s)
- Martina Doubková
- 1Department of Pneumology and Phtiseology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Kateřina Staňo Kozubík
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,3Department of Internal Medicine, Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Lenka Radová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michaela Pešová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jakub Trizuljak
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,3Department of Internal Medicine, Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Karol Pál
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Klára Svobodová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Kamila Réblová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Hana Svozilová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,3Department of Internal Medicine, Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Zuzana Vrzalová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,3Department of Internal Medicine, Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Šárka Pospíšilová
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,3Department of Internal Medicine, Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| | - Michael Doubek
- 2Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,3Department of Internal Medicine, Hematology and Oncology, University Hospital and Faculty of Medicine, Brno, Czech Republic
| |
Collapse
|
35
|
Surfactant protein A and D polymorphisms and methylprednisolone pharmacogenetics in donor lungs. J Thorac Cardiovasc Surg 2019; 157:2109-2117. [PMID: 30827536 DOI: 10.1016/j.jtcvs.2018.12.098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/17/2018] [Accepted: 12/30/2018] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Surfactant proteins A and D are important molecules involved in lung allograft innate immunity. Genetic polymorphisms of surfactant proteins A and D are associated with various lung diseases. In this study, surfactant protein A and D expression responses were investigated during pharmacogenetics upon methylprednisolone treatment as observed during lung transplantation. METHODS A human cell line (NCI-H441) and precision-cut lung slices from 16 human donors were incubated with methylprednisolone, and surfactant protein A1, surfactant protein A2, and surfactant protein D messenger RNA and surfactant protein A protein expression were assayed. Surfactant protein A1, A2, and D polymorphisms and surfactant protein A gene and protein expressions were determined. RESULTS In NCI-H441 cells, methylprednisolone treatment at 10-5 M and 10-6 M reduced surfactant protein A1 and surfactant protein A2 messenger RNA and surfactant protein A protein expression (P < .05). A pharmacogenetic relationship was observed in human donor precision-cut lung slices between the surfactant protein A2 (1Ax) variants: Surfactant protein A1, A2, and D messenger RNA expression were greater for 1A0 versus 1A1 (P < .05); surfactant protein A1/surfactant protein A2 genotype 6A26A2/1A01A0 (n = 5) showed greater surfactant protein A1, A2, and D messenger RNA expression and surfactant protein A protein expression compared with the other surfactant protein A1/surfactant protein A2 genotypes (n = 11) (P < .05). CONCLUSIONS The surfactant protein A genotype and methylprednisolone stimuli influence donor lung surfactant protein A and D expression. Lungs carrying the surfactant protein A2 variant 1A0 have a greater expression of surfactant protein A when treated with methylprednisolone. Surfactant protein A polymorphisms could be used to personalize immunosuppressive regimens.
Collapse
|
36
|
Thorenoor N, Umstead TM, Zhang X, Phelps DS, Floros J. Survival of Surfactant Protein-A1 and SP-A2 Transgenic Mice After Klebsiella pneumoniae Infection, Exhibits Sex-, Gene-, and Variant Specific Differences; Treatment With Surfactant Protein Improves Survival. Front Immunol 2018; 9:2404. [PMID: 30459763 PMCID: PMC6232836 DOI: 10.3389/fimmu.2018.02404] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/28/2018] [Indexed: 01/23/2023] Open
Abstract
Surfactant protein A (SP-A) is involved in lung innate host defense and surfactant-related functions. The human SFTPA1 and SFTPA2 genes encode SP-A1 and SP-2 proteins, and each gene has been identified with numerous genetic variants. SP-A1 and SP-A2 differentially enhance bacterial phagocytosis. Sex differences have been observed in pulmonary disease and in survival of wild type and SP-A knockout (KO) mice. The impact of human SP-A variants on survival after infection is unknown. In this study, we determined whether SP-A variants differentially affect survival of male and female mice infected with Klebsiella pneumoniae. Transgenic (TG) mice, where each carries a different human (h) SP-A1 (6A2, 6A4), SP-A2 (1A0, 1A3) variant or both variants SP-A1/SP-A2 (6A2/1A0, co-ex), and SP-A- KO, were utilized. The hTG and KO mice were infected intratracheally with K. pneumoniae bacteria, and groups of KO mice were treated with SP-A1 or SP-A2 either prior to and/or at the time of infection and survival for both experimental groups was monitored over 14 days. The binding of purified SP-A1 and SP-A2 proteins to phagocytic and non-phagocytic cells and expression of cell surface proteins in alveolar macrophages (AM) from SP-A1 and SP-A2 mice was examined. We observed gene-, variant-, and sex-specific (except for co-ex) differences with females showing better survival: (a) Gene-specific differences: co-ex = SP-A2 > SP-A1 > KO (both sexes); (b) Variant-specific survival co-ex (6A2/1A0) = 1A0 > 1A3 = 6A2 > 6A4 (both sexes); (c) KO mice treated with SPs (SP-A1 or SP-A2) proteins exhibit significantly (p < 0.05) better survival; (d) SP-A1 and SP-A2 differentially bind to phagocytic, but not to non-phagocytic cells, and AM from SP-A1 and SP-A2 hTG mice exhibit differential expression of cell surface proteins. Our results indicate that sex and SP-A genetics differentially affect survival after infection and that exogenous SP-A1/SP-A2 treatment significantly improves survival. We postulate that the differential SP-A1/SP-A2 binding to the phagocytic cells and the differential expression of cell surface proteins that bind SP-A by AM from SP-A1 and SP-A2 mice play a role in this process. These findings provide insight into the importance of sex and innate immunity genetics in survival following infection.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Todd M Umstead
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xuesheng Zhang
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David S Phelps
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
37
|
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
|
38
|
Increased Expiratory Computed Tomography Density Reveals Possible Abnormalities in Radiologically Preserved Lung Parenchyma in Idiopathic Pulmonary Fibrosis. Invest Radiol 2018; 53:45-51. [PMID: 28817393 DOI: 10.1097/rli.0000000000000405] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Idiopathic pulmonary fibrosis (IPF) is a progressive lethal chronic lung disease with unclear pathogenesis. Radiological hallmark is the pattern of usual interstitial pneumonia accentuated in peripheral and basal areas with otherwise preserved lung structure. One hypothesis is that alveolar collapse and consequent induration lead to fibrotic transformation of lung tissue. The aim of the study was to investigate normal-appearing tissue during expiration for signs of collapsibility and differences from other diseases or controls. MATERIALS AND METHODS We retrospectively assessed a total of 43 patients (15 IPFs, 13 chronic obstructive pulmonary diseases, and 15 controls) with nonenhanced computed tomography (CT) in inspiration and expiration, performed for routine clinical workup. Densitometry of visually unaffected lung tissue was conducted in all lung lobes with a region of interest of 15-mm in diameter on soft tissue kernel reconstruction (slice thickness, 1 mm) during inspiration and expiration. RESULTS One-factor analysis of variance analysis yielded significant difference in attenuation changes between inspiration and expiration of unaffected lung parenchyma among all subject groups in all lung lobes. For IPF patients, the highest differences in densities were observed in the lower lobes, which is the predominantly affected site of usual interstitial pneumonia. In the chronic obstructive pulmonary disease group, the density remained rather equal in the entire lung. CONCLUSIONS High CT attenuation changes between inspiration and expiration in IPF patients might suggest altered lung parenchyma in normal-appearing tissue on CT. Density changes during the respiratory cycle might be explained by alveolar collapse of radiologically unaffected lung tissue possibly preceding fibrosis. These results support the concept of alveolar collapse preceding lung fibrosis in IPF.
Collapse
|
39
|
Differential effects of innate immune variants of surfactant protein-A1 (SFTPA1) and SP-A2 (SFTPA2) in airway function after Klebsiella pneumoniae infection and sex differences. Respir Res 2018; 19:23. [PMID: 29394894 PMCID: PMC5797374 DOI: 10.1186/s12931-018-0723-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/16/2018] [Indexed: 01/15/2023] Open
Abstract
Background Surfactant Protein-A (SP-A) is a major protein component of surfactant and plays a role in surfactant-related functions and innate immunity. Human SP-A consists of two functional genes, SFTPA1 and SFTPA2, encoding SP-A1 and SP-A2 proteins, respectively and each is identified with numerous genetic variants. These differentially enhance bacterial phagocytosis, with SP-A2 variants being more effective than SP-A1. Methods Lung functions of humanized transgenic (hTG) mice that carry different SP-A1 and SP-A2 variants or both variants SP-A1/SP-A2 (6A2/1A0, co-ex), as well as SP-A knockout (KO), were studied. The animals were connected to a flexiVent system to obtain forced oscillation technique (FOT) measurements and the data were analyzed using various models. Lung function was assessed after infection (baseline) and following inhaled methacholine concentrations (0–50 mg/mL). Results Here, we investigated the role of SP-A variants on airway function after Klebsiella pneumoniae (Kp) infection (baseline) and following inhaled methacholine. We found that: 1) in the absence of methacholine no significant differences were observed between SP-A1 and SP-A2 variants and/or SP-A knockout (KO) except for sex differences in most of the parameters studied. 2) In response to methacholine, i) sex differences were observed that were reverse of those observed in the absence of methacholine; ii) SP-A2 (1A3) gene variant in males exhibited increased total and central airway resistance (Rrs and Rn) versus all other variants; iii) In females, SP-A2 (1A3) and SP-A1 (6A2) variants had similar increases in total and central airway resistance (Rrs and Rn) versus all other variants; iv) Allele-specific differences were observed, a) with SP-A2 (1A3) exhibiting significantly higher lung functions versus SP-A2 (1A0) in both sexes, except for Crs, and b) SP-A1 (6A2, 6A4) had more diverse changes in lung function in both sexes. Conclusion We conclude that, in response to infection and methacholine, SP-A variants differentially affect lung function and exhibit sex-specific differences consistent with previously reported findings of functional differences of SP-A variants. Thus, the observed changes in respiratory function mechanics provide insight into the role and importance of genetic variation of innate immune molecules, such as SP-A, on mechanical consequences of lung function after infection and inhaled substances.
Collapse
|
40
|
Abstract
Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive genetic disorder characterized by oculocutaneous albinism and a bleeding diathesis due to platelet dysfunction. More than 50% of cases worldwide are diagnosed on the Caribbean island of Puerto Rico. Genetic testing plays a growing role in diagnosis; however, not all patients with HPS have identified genetic mutations. In Puerto Rico, patients with HPS are often identified shortly after birth by their albinism, although the degree of hypopigmentation is highly variable. Ten subtypes have been described. Patients with HPS-1, HPS-2, and HPS-4 tend to develop pulmonary fibrosis in Puerto Rico; 100% of patients with HPS-1 develop HPS-PF. HPS-PF and idiopathic pulmonary fibrosis are considered similar entities (albeit with distinct causes) because both can show similar histological disease patterns. However, in contrast to idiopathic pulmonary fibrosis, HPS-PF manifests much earlier, often at 30-40 years of age. The progression of HPS-PF is characterized by the development of dyspnea and increasingly debilitating hypoxemia. No therapeutic interventions are currently approved by the U.S. Food and Drug Administration for the treatment of HPS and HPS-PF. However, the approval of two new antifibrotic drugs, pirfenidone and nintedanib, has prompted new interest in identifying drugs capable of reversing or halting the progression of HPS-PF. Thus, lung transplantation remains the only potentially life-prolonging treatment. At present, two clinical trials are recruiting patients with HPS-PF to identify biomarkers for disease progression. Advances in the diagnosis and management of these patients will require the establishment of multidisciplinary centers of excellence staffed by experts in this disease.
Collapse
|
41
|
Pradipkumar D, Gautham A, Gupta R, James P, Thangakunam B, Christopher D. Familial interstitial pulmonary fibrosis in two different families in India: A case series. Lung India 2017; 34:475-479. [PMID: 28869238 PMCID: PMC5592765 DOI: 10.4103/0970-2113.213824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
42
|
Tsitoura MEI, Stavrou EF, Maraziotis IA, Sarafidis K, Athanassiadou A, Dimitriou G. Surfactant Protein A and B Gene Polymorphisms and Risk of Respiratory Distress Syndrome in Late-Preterm Neonates. PLoS One 2016; 11:e0166516. [PMID: 27835691 PMCID: PMC5106092 DOI: 10.1371/journal.pone.0166516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 10/31/2016] [Indexed: 11/19/2022] Open
Abstract
Background and Objectives Newborns delivered late-preterm (between 340/7 and 366/7 weeks of gestation) are at increased risk of respiratory distress syndrome (RDS). Polymorphisms within the surfactant protein (SP) A and B gene have been shown to predispose to RDS in preterm neonates. The aim of this study was to investigate whether specific SP-A and/or SP-B genetic variants are also associated with RDS in infants born late-preterm. Methods This prospective cross-sectional study included 56 late-preterm infants with and 60 without RDS. Specific SP-A1/SP-A2 haplotypes and SP-B Ile131Thr polymorphic alleles were determined in blood specimens using polymerase-chain-reaction and DNA sequencing. Results The SP-A1 6A4 and the SP-A2 1A5 haplotypes were significantly overrepresented in newborns with RDS compared to controls (OR 2.86, 95%CI 1.20–6.83 and OR 4.68, 95%CI 1.28–17.1, respectively). The distribution of the SP-B Ile131Thr genotypes was similar between the two late-preterm groups. Overall, the SP-A1 6A4 or/and SP-A2 1A5 haplotype was present in 20 newborns with RDS (35.7%), resulting in a 4.2-fold (1.60–11.0) higher probability of RDS in carriers. Multivariable regression analysis revealed that the effect of SP-A1 6A4 and SP-A2 1A5 haplotypes was preserved when adjusting for known risk or protective factors, such as male gender, smaller gestational age, smaller weight, complications of pregnancy, and administration of antenatal corticosteroids. Conclusions Specific SP-A genetic variants may influence the susceptibility to RDS in late-preterm infants, independently of the effect of other perinatal factors.
Collapse
Affiliation(s)
- Maria-Eleni I. Tsitoura
- Neonatal Intensive Care Unit, Department of Pediatrics, Faculty of Medicine, University of Patras, Rio, Patras, Greece
- Department of General Biology, Faculty of Medicine, University of Patras, Rio, Patras, Greece
| | - Eleana F. Stavrou
- Department of General Biology, Faculty of Medicine, University of Patras, Rio, Patras, Greece
| | - Ioannis A. Maraziotis
- Neonatal Intensive Care Unit, Department of Pediatrics, Faculty of Medicine, University of Patras, Rio, Patras, Greece
| | - Kosmas Sarafidis
- First Department of Neonatology, Aristotle University of Thessaloniki, Ippokration General Hospital, Thessaloniki, Greece
| | - Aglaia Athanassiadou
- Department of General Biology, Faculty of Medicine, University of Patras, Rio, Patras, Greece
| | - Gabriel Dimitriou
- Neonatal Intensive Care Unit, Department of Pediatrics, Faculty of Medicine, University of Patras, Rio, Patras, Greece
- * E-mail:
| |
Collapse
|
43
|
Papaioannou AI, Kostikas K, Manali ED, Papadaki G, Roussou A, Spathis A, Mazioti A, Tomos I, Papanikolaou I, Loukides S, Chainis K, Karakitsos P, Griese M, Papiris S. Serum Levels of Surfactant Proteins in Patients with Combined Pulmonary Fibrosis and Emphysema (CPFE). PLoS One 2016; 11:e0157789. [PMID: 27337142 PMCID: PMC4919090 DOI: 10.1371/journal.pone.0157789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/03/2016] [Indexed: 01/05/2023] Open
Abstract
Introduction Emphysema and idiopathic pulmonary fibrosis (IPF) present either per se or coexist in combined pulmonary fibrosis and emphysema (CPFE). Serum surfactant proteins (SPs) A, B, C and D levels may reflect lung damage. We evaluated serum SP levels in healthy controls, emphysema, IPF, and CPFE patients and their associations to disease severity and survival. Methods 122 consecutive patients (31 emphysema, 62 IPF, and 29 CPFE) and 25 healthy controls underwent PFTs, ABG-measurements, 6MWT and chest HRCT. Serum levels of SPs were measured. Patients were followed-up for 1-year. Results SP-A and SP-D levels differed between groups (p = 0.006 and p<0.001 respectively). In post-hoc analysis, SP-A levels differed only between controls and CPFE (p<0.05) and CPFE and emphysema (p<0.05). SP-D differed between controls and IPF or CPFE (p<0.001 for both comparisons). In IPF SP-B correlated to pulmonary function while SP-A, correlated to the Composite Physiological Index (CPI). Controls current smokers had higher SP-A and SP-D levels compared to non-smokers (p = 0.026 and p = 0.023 respectively). SP-D levels were higher in CPFE patients with extended emphysema (p = 0.042). In patients with IPF, SP-B levels at the upper quartile of its range (≥26 ng/mL) presented a weak association with reduced survival (p = 0.05). Conclusion In conclusion, serum SP-A and SP-D levels were higher where fibrosis exists or coexists and related to disease severity, suggesting that serum SPs relate to alveolar damage in fibrotic lungs and may reflect either local overproduction or overleakage. The weak association between high levels of SP-B and survival needs further validation in clinical trials.
Collapse
Affiliation(s)
- Andriana I. Papaioannou
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
- * E-mail:
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Effrosyni D. Manali
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Papadaki
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aneza Roussou
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris Spathis
- Department of Cytopathology, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Argyro Mazioti
- Department of Radiology, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Tomos
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ilias Papanikolaou
- Respiratory Medicine Department, “Corfu General Hospital”, Corfu, Greece
| | - Stelios Loukides
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriakos Chainis
- Respiratory Medicine Department, “Corfu General Hospital”, Corfu, Greece
| | - Petros Karakitsos
- Department of Cytopathology, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Matthias Griese
- Hauner Children’s University Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Spyros Papiris
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
44
|
Puxeddu E, Rogliani P. Prognostic scoring systems for clinical course and survival in idiopathic pulmonary fibrosis. World J Respirol 2016; 6:14-23. [DOI: 10.5320/wjr.v6.i1.14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/22/2015] [Accepted: 01/31/2016] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common and rapidly fatal among idiopathic interstitial pneumonias. Its clinical course is variable. A significant fraction of the population of patients display a slow disease course and can remain stable for years, while other patients show a rapid progressive course and may die within few months from diagnosis. For these reasons estimating prognosis of IPF patients is extremely difficult and has important clinical repercussions on optimal patients management including patients referral for lung transplantation. Several studies have tried to address this key point in the course of the two last decades analyzing different clinical, functional, radiological and biological variables. The purpose of this review is to assess relevant studies published on this subject and to examine the variety of prognostic predictors proposed along with staging systems.
Collapse
|
45
|
Abstract
Pulmonary surfactant is essential for life as it lines the alveoli to lower surface tension, thereby preventing atelectasis during breathing. Surfactant is enriched with a relatively unique phospholipid, termed dipalmitoylphosphatidylcholine, and four surfactant-associated proteins, SP-A, SP-B, SP-C, and SP-D. The hydrophobic proteins, SP-B and SP-C, together with dipalmitoylphosphatidylcholine, confer surface tension-lowering properties to the material. The more hydrophilic surfactant components, SP-A and SP-D, participate in pulmonary host defense and modify immune responses. Specifically, SP-A and SP-D bind and partake in the clearance of a variety of bacterial, fungal, and viral pathogens and can dampen antigen-induced immune function of effector cells. Emerging data also show immunosuppressive actions of some surfactant-associated lipids, such as phosphatidylglycerol. Conversely, microbial pathogens in preclinical models impair surfactant synthesis and secretion, and microbial proteinases degrade surfactant-associated proteins. Deficiencies of surfactant components are classically observed in the neonatal respiratory distress syndrome, where surfactant replacement therapies have been the mainstay of treatment. However, functional or compositional deficiencies of surfactant are also observed in a variety of acute and chronic lung disorders. Increased surfactant is seen in pulmonary alveolar proteinosis, a disorder characterized by a functional deficiency of the granulocyte-macrophage colony-stimulating factor receptor or development of granulocyte-macrophage colony-stimulating factor antibodies. Genetic polymorphisms of some surfactant proteins such as SP-C are linked to interstitial pulmonary fibrosis. Here, we briefly review the composition, antimicrobial properties, and relevance of pulmonary surfactant to lung disorders and present its therapeutic implications.
Collapse
|
46
|
Nathan N, Giraud V, Picard C, Nunes H, Dastot-Le Moal F, Copin B, Galeron L, De Ligniville A, Kuziner N, Reynaud-Gaubert M, Valeyre D, Couderc LJ, Chinet T, Borie R, Crestani B, Simansour M, Nau V, Tissier S, Duquesnoy P, Mansour-Hendili L, Legendre M, Kannengiesser C, Coulomb-L'Hermine A, Gouya L, Amselem S, Clement A. GermlineSFTPA1mutation in familial idiopathic interstitial pneumonia and lung cancer. Hum Mol Genet 2016; 25:1457-67. [DOI: 10.1093/hmg/ddw014] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/14/2016] [Indexed: 01/18/2023] Open
|
47
|
Aquino-Gálvez A, González-Ávila G, Pérez-Rodríguez M, Partida-Rodríguez O, Nieves-Ramírez M, Piña-Ramírez I, Ramírez-Martínez G, Castillejos-López M, Checa M, Ruiz V, Urrea F, Sommer B, Zúñiga J, Selman M. Analysis of heat shock protein 70 gene polymorphisms Mexican patients with idiopathic pulmonary fibrosis. BMC Pulm Med 2015; 15:129. [PMID: 26496868 PMCID: PMC4619986 DOI: 10.1186/s12890-015-0127-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/14/2015] [Indexed: 12/13/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease of unknown etiology. Genetic variation within different major histocompatibility complex (MHC) loci contributes to the susceptibility to IPF. The effect of 70 kDa heat shock proteins (HSP70) gene polymorphisms in the susceptibility to IPF is unknown. The aim of this study was to explore the association between HSP70 polymorphisms and IPF susceptibility in the Mexican population. Methods Four HSP70 single nucleotide polymorphisms (SNPs) were evaluated using real time PCR assays in 168 IPF patients and 205 controls: +2763 C>T of HSPA1L (rs2075800), +2437 of HSP HSPA1L A>G (rs2227956), +190 of HSPA1A G>C (rs1043618) and +1267 of HSPA1B G>A (rs1061581). Results The analysis of the recessive model revealed a significant decrease in the frequency of the genotype HSPA1B AA (rs1061581) in IPF patients (OR = 0.27, 95 % CI = 0.13–0.57, Pc = 0.0003) when compared to controls. Using a multivariate logistic regression analysis in a codominant model the HSPA1B (rs1061581) GA and AA genotypes were associated with a lower risk of IPF compared with GG (OR = 0.22, 95 % CI = 0.07–0.65; p = 0.006 and OR = 0.17, 95 % CI = 0.07–0.41; p = <0.001). Similarly, HSPA1L (rs2227956) AG genotype (OR = 0.34, 95 % CI = 0.12–0.99; p = 0.04) and the dominant model AG + GG genotypes were also associated with a lower risk of IPF (OR = 0.24, 95 % CI = 0.08–0.67; p = 0.007). In contrast, the HSPA1L (rs2075800) TT genotype was associated with susceptibility to IPF (OR = 2.52, 95 % CI = 1.32–4.81; p = 0.005). Conclusion Our findings indicate that HSPA1B (rs1061581), HSPA1L (rs2227956) and HSPA1 (rs1043618) polymorphisms are associated with a decreased risk of IPF. Electronic supplementary material The online version of this article (doi:10.1186/s12890-015-0127-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arnoldo Aquino-Gálvez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Georgina González-Ávila
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Martha Pérez-Rodríguez
- Hospital General de México, Universidad Nacional Autónoma de México, Laboratorio de Inmunología, Mexico, Mexico.
| | - Oswaldo Partida-Rodríguez
- Unidad de Investigación Médica en Inmunología, CMN S-XXI Instituto Mexicano del Seguro Social, Mexico, Mexico.
| | - Miriam Nieves-Ramírez
- Unidad de Investigación Médica en Inmunología, CMN S-XXI Instituto Mexicano del Seguro Social, Mexico, Mexico.
| | - Inocencio Piña-Ramírez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Gustavo Ramírez-Martínez
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Manuel Castillejos-López
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Marco Checa
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Victor Ruiz
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Francisco Urrea
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Bettina Sommer
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Joaquin Zúñiga
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| | - Moisés Selman
- Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas" Tlalpan 4502, Col. Sección XVI, 14080, Mexico, México.
| |
Collapse
|
48
|
Hambly N, Shimbori C, Kolb M. Molecular classification of idiopathic pulmonary fibrosis: personalized medicine, genetics and biomarkers. Respirology 2015; 20:1010-22. [PMID: 26109466 DOI: 10.1111/resp.12569] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/16/2015] [Accepted: 05/06/2015] [Indexed: 12/29/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive fibrotic lung disease associated with high morbidity and poor survival. Characterized by substantial disease heterogeneity, the diagnostic considerations, clinical course and treatment response in individual patients can be variable. In the past decade, with the advent of high-throughput proteomic and genomic technologies, our understanding of the pathogenesis of IPF has greatly improved and has led to the recognition of novel treatment targets and numerous putative biomarkers. Molecular biomarkers with mechanistic plausibility are highly desired in IPF, where they have the potential to accelerate drug development, facilitate early detection in susceptible individuals, improve prognostic accuracy and inform treatment recommendations. Although the search for candidate biomarkers remains in its infancy, attractive targets such as MUC5B and MPP7 have already been validated in large cohorts and have demonstrated their potential to improve clinical predictors beyond that of routine clinical practices. The discovery and implementation of future biomarkers will face many challenges, but with strong collaborative efforts among scientists, clinicians and the industry the ultimate goal of personalized medicine may be realized.
Collapse
Affiliation(s)
- Nathan Hambly
- Firestone Institute for Respiratory Health, Hamilton, ON, Canada.,St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Medicine, McMaster University Hamilton, Hamilton, ON, Canada
| | - Chiko Shimbori
- Firestone Institute for Respiratory Health, Hamilton, ON, Canada.,St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Medicine, McMaster University Hamilton, Hamilton, ON, Canada
| | - Martin Kolb
- Firestone Institute for Respiratory Health, Hamilton, ON, Canada.,St. Joseph's Healthcare, Hamilton, ON, Canada.,Department of Medicine, McMaster University Hamilton, Hamilton, ON, Canada
| |
Collapse
|
49
|
Furukawa H, Oka S, Shimada K, Tsuchiya N, Tohma S. Genetics of Interstitial Lung Disease: Vol de Nuit (Night Flight). CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2015; 9:1-7. [PMID: 26056507 PMCID: PMC4444491 DOI: 10.4137/ccrpm.s23283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 01/03/2023]
Abstract
Interstitial lung disease (ILD) is a chronic, progressive fibrotic lung disease with a dismal prognosis. ILD of unknown etiology is referred to as idiopathic interstitial pneumonia (IIP), which is sporadic in the majority of cases. ILD is frequently accompanied by rheumatoid arthritis (RA), systemic sclerosis (SSc), polymyositis/dermatomyositis (PM/DM), and other autoimmune diseases, and is referred to as collagen vascular disease-associated ILD (CVD-ILD). Susceptibility to ILD is influenced by genetic and environmental factors. Recent advances in radiographic imaging techniques such as high-resolution computed tomography (CT) scanning as well as high-throughput genomic analyses have provided insights into the genetics of ILD. These studies have repeatedly revealed an association between IIP (sporadic and familial) and a single nucleotide polymorphism (SNP) in the promoter region of the mucin 5B (MUC5B). HLA-DRB1*11 alleles have been reported to correlate with ILD in European patients with SSc, whereas in Japanese patients with RA, the HLA-DR2 serological group was identified. The aim of this review is to describe the genetic background of sporadic IIP, CVD-ILD, drug-induced-ILD (DI-ILD), pneumoconiosis, and hypersensitivity pneumonitis. The genetics of ILD is still in progress. However, this information will enhance the understanding of the pathogenesis of ILD and aid the identification of novel therapeutic targets for personalized medicine in future.
Collapse
Affiliation(s)
- Hiroshi Furukawa
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Japan
| | - Shomi Oka
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Japan
| | - Kota Shimada
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shigeto Tohma
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Japan
| |
Collapse
|
50
|
Goto H, Mitsuhashi A, Nishioka Y. Role of surfactant protein A in non-infectious lung diseases. THE JOURNAL OF MEDICAL INVESTIGATION 2015; 61:1-6. [PMID: 24705741 DOI: 10.2152/jmi.61.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Surfactant protein A (SP-A) is a large multimeric protein found in the airways and alveoli of the lungs. SP-A is a member of the collectin family of proteins, characterized by NH2-terminal collagen-like regions and COOH-terminal lectin domains. Although other surfactant proteins such as SP-B function to reduce surface tension in the lungs, SP-A as well as SP-D regulates the pulmonary immune response. To date, a number of studies have shown the immunoregulatory function of SP-A, mainly in the field of infectious diseases. By binding to a wide variety of pathogens, SP-A opsonizes and enhances pathogen uptake by phagocytes. In addition to the effect on pathogens, recent studies have shown that SP-A also modulates lung immune system in the area of non-infectious lung diseases. In this review, the potential role of SP-A in the multiple aspects of pulmonary host defense will be discussed, focusing mainly on non-infectious lung diseases such as acute and chronic pulmonary fibrosis and lung cancer. J. Med. Invest. 61: 1-6, February, 2014.
Collapse
Affiliation(s)
- Hisatsugu Goto
- Department of Respiratory Medicine and Rheumatology, Institute of Health Biosciences, the University of Tokushima Graduate School
| | | | | |
Collapse
|