1
|
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
|
2
|
Bahrami R, Golshan-Tafti M, Dastgheib SA, Alijanpour K, Yeganegi M, Lookzadeh MH, Mirjalili SR, Azizi S, Aghasipour M, Shiri A, Noorishadkam M, Neamatzadeh H. A Comprehensive Consolidation of Data on the Relationship Between Surfactant Protein-B (SFTPB) Polymorphisms and Susceptibility to Bronchopulmonary Dysplasia. Fetal Pediatr Pathol 2024; 43:436-454. [PMID: 39245635 DOI: 10.1080/15513815.2024.2400145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND This meta-analysis aims to evaluate the potential link between common variations in the Surfactant Protein-B (SFTPB) gene and the risk of bronchopulmonary dysplasia (BPD) in preterm neonates. METHODS All pertinent articles published prior to February 1, 2024, in PubMed, Web of Science, EMBASE, CNKI, and Scopus databases were reviewed. RESULTS Nineteen case-control studies involving 1149 BPD cases and 1845 non-BPD controls, were analyzed. Combined data indicated a significant link between SFTPB -18 A > C and Intron 4 VNTR polymorphisms with increased BPD susceptibility, while the 1580 C > T polymorphism provides a protective impact on BPD initiation. CONCLUSIONS Pooled data indicated a significant association between SFTPB -18 A > C and Intron 4 VNTR polymorphisms with increased BPD risk, whereas the 1580 C > T polymorphism confers protection. These findings suggest a genetic susceptibility to BPD, underscoring the complex interplay of different genetic elements in its development.
Collapse
Affiliation(s)
- Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamran Alijanpour
- General Practitioner, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Yeganegi
- Department of Obstetrics and Gynecology, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mohamad Hosein Lookzadeh
- Mother and Newborn Health Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Reza Mirjalili
- Mother and Newborn Health Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sepideh Azizi
- Shahid Akbarabadi Cilinical Research Development Unit, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Aghasipour
- Department of Cancer Biology, College of Medicine, University of Cincinnati, OH, USA
| | - Amirmasoud Shiri
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Noorishadkam
- Mother and Newborn Health Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Mother and Newborn Health Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
3
|
Gandhi CK, Depicolzuane LC, Chen C, Roberts CM, Sicher N, Johnson Wegerson K, Thomas NJ, Wu R, Floros J. Association of SNP-SNP interactions of surfactant protein genes with severity of respiratory syncytial virus infection in children. Physiol Genomics 2024; 56:691-697. [PMID: 39222066 PMCID: PMC11495184 DOI: 10.1152/physiolgenomics.00045.2024] [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: 04/19/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The severity of respiratory syncytial virus (RSV) may be linked to host genetic susceptibility. Surfactant protein (SP) genetic variants have been associated with RSV severity, but the impact of single-nucleotide polymorphism (SNP)-SNP interactions remains unexplored. Therefore, we used a novel statistical model to investigate the association of SNP-SNP interactions of SFTP genes with RSV severity in two- and three-interaction models. We analyzed available genotype and clinical data from prospectively enrolled 405 children diagnosed with RSV, categorizing them into moderate or severe RSV groups. Using Wang's statistical model, we studied significant associations of SNP-SNP interactions with RSV severity in a case-control design. We observed, first, association of three interactions with increased risk of severe RSV in a two-SNP model. One intragenic interaction was between SNPs of SFTPA2, and the other two were intergenic, involving SNPs of hydrophilic and hydrophobic SPs alone. We also observed, second, association of 22 interactions with RSV severity in a three-SNP model. Among these, 20 were unique, with 12 and 10 interactions associated with increased or decreased risk of RSV severity, respectively, and included at least one SNP of either SFTPA1 or SFTPA2. All interactions were intergenic except one, among SNPs of SFTPA1. The remaining interactions were either among SNPs of hydrophilic SPs alone (n = 8) or among SNPs of both hydrophilic or hydrophobic SPs (n = 11). Our findings indicate that SNPs of all SFTPs may contribute to genetic susceptibility to RSV severity. However, the predominant involvement of SFTPA1 and/or SFTPA2 SNPs in these interactions underscores their significance in RSV severity.NEW & NOTEWORTHY Although surfactant protein (SP) genetic variants are associated with respiratory syncytial virus (RSV) severity, the impact of single-nucleotide polymorphism (SNP)-SNP interactions of SP genes remained unexplored. Using advanced statistical models, we uncovered 22 SNP-SNP interactions associated with RSV severity, with notable involvement of SFTPA1 and SFTPA2 SNPs. This highlights the comprehensive role of all SPs in genetic susceptibility to RSV severity, shedding light on potential avenues for targeted interventions.
Collapse
Affiliation(s)
- Chintan K Gandhi
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Lynnlee C Depicolzuane
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Chixiang Chen
- Department of Public Health Science, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Catherine M Roberts
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Natalie Sicher
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Katelyn Johnson Wegerson
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Neal J Thomas
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Rongling Wu
- Department of Public Health Science, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| | - Joanna Floros
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
- Department of Obstetrics and Gynecology, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States
| |
Collapse
|
4
|
Cao S, Li H, Xin J, Jin Z, Zhang Z, Li J, Zhu Y, Su L, Huang P, Jiang L, Du M, Christiani DC. Identification of genetic profile and biomarkers involved in acute respiratory distress syndrome. Intensive Care Med 2024; 50:46-55. [PMID: 37922010 PMCID: PMC11167213 DOI: 10.1007/s00134-023-07248-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/08/2023] [Indexed: 11/05/2023]
Abstract
PURPOSE The purpose of this study was to profile genetic causal factors of acute respiratory distress syndrome (ARDS) and early predict patients at high ARDS risk. METHODS We performed a phenome-wide Mendelian Randomization analysis through summary statistics of an ARDS genome-wide association study (1250 cases and 1583 controls of European ancestry) and 33,150 traits. Transcriptomic data from human blood and lung tissues of a preclinical mouse model were used to validate biomarkers, which were further used to construct a prediction model and nomogram. RESULTS A total of 1736 traits, including 1223 blood RNA, 159 plasma proteins, and 354 non-gene phenotypes (classified by Biochemistry, Anthropometry, Disease, Nutrition and Habit, Immunology, and Treatment), exhibited a potentially causal relationship with ARDS development, which were accessible through a user-friendly interface platform called CARDS (Causal traits for Acute Respiratory Distress Syndrome). Regarding candidate blood RNA, four genes were validated, namely TMEM176B, SLC2A5, CDC45, and VSIG8, showing differential expression in blood of ARDS patients compared to controls, as well as dynamic expression in mouse lung tissues. Importantly, the addition of four blood genes and five immune cell proportions significantly improved the prediction performance of ARDS development, with 0.791 of the area under the curve from receiver-operator characteristic, compared to 0.725 for the basic model consisting of Acute Physiology and Chronic Health Evaluation (APACHE) III Score, sex, body mass index, bacteremia, and sepsis. A model-based nomogram was also developed for the clinical practice. CONCLUSION This study identifies a wide range of ARDS relevant factors and develops a promising prediction model, enhancing early clinical management and intervention for ARDS development.
Collapse
Affiliation(s)
- Shurui Cao
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Huiqin Li
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China
| | - Junyi Xin
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenghao Jin
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengyu Zhang
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiawei Li
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yukun Zhu
- School of Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Li Su
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
| | - Peipei Huang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Jiang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mulong Du
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, Jiangsu, China.
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Abstract
Pulmonary surfactant is a critical component of lung function in healthy individuals. It functions in part by lowering surface tension in the alveoli, thereby allowing for breathing with minimal effort. The prevailing thinking is that low surface tension is attained by a compression-driven squeeze-out of unsaturated phospholipids during exhalation, forming a film enriched in saturated phospholipids that achieves surface tensions close to zero. A thorough review of past and recent literature suggests that the compression-driven squeeze-out mechanism may be erroneous. Here, we posit that a surfactant film enriched in saturated lipids is formed shortly after birth by an adsorption-driven sorting process and that its composition does not change during normal breathing. We provide biophysical evidence for the rapid formation of an enriched film at high surfactant concentrations, facilitated by adsorption structures containing hydrophobic surfactant proteins. We examine biophysical evidence for and against the compression-driven squeeze-out mechanism and propose a new model for surfactant function. The proposed model is tested against existing physiological and pathophysiological evidence in neonatal and adult lungs, leading to ideas for biophysical research, that should be addressed to establish the physiological relevance of this new perspective on the function of the mighty thin film that surfactant provides.
Collapse
Affiliation(s)
- Fred Possmayer
- Department of Biochemistry, Western University, London, Ontario N6A 3K7, Canada
- Department of Obstetrics/Gynaecology, Western University, London, Ontario N6A 3K7, Canada
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manon, Honolulu, Hawaii 96822, United States
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96826, United States
| | - Ruud A W Veldhuizen
- Department of Physiology & Pharmacology, Western University, London, Ontario N6A 5C1, Canada
- Department of Medicine, Western University, London, Ontario N6A 3K7, Canada
- Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| |
Collapse
|
6
|
Kruckow KL, Zhao K, Bowdish DME, Orihuela CJ. Acute organ injury and long-term sequelae of severe pneumococcal infections. Pneumonia (Nathan) 2023; 15:5. [PMID: 36870980 PMCID: PMC9985869 DOI: 10.1186/s41479-023-00110-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is a major public health problem, as it is a main cause of otitis media, community-acquired pneumonia, bacteremia, sepsis, and meningitis. Acute episodes of pneumococcal disease have been demonstrated to cause organ damage with lingering negative consequences. Cytotoxic products released by the bacterium, biomechanical and physiological stress resulting from infection, and the corresponding inflammatory response together contribute to organ damage accrued during infection. The collective result of this damage can be acutely life-threatening, but among survivors, it also contributes to the long-lasting sequelae of pneumococcal disease. These include the development of new morbidities or exacerbation of pre-existing conditions such as COPD, heart disease, and neurological impairments. Currently, pneumonia is ranked as the 9th leading cause of death, but this estimate only considers short-term mortality and likely underestimates the true long-term impact of disease. Herein, we review the data that indicates damage incurred during acute pneumococcal infection can result in long-term sequelae which reduces quality of life and life expectancy among pneumococcal disease survivors.
Collapse
Affiliation(s)
- Katherine L Kruckow
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kevin Zhao
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Dawn M E Bowdish
- McMaster Immunology Research Centre and the Firestone Institute for Respiratory Health, McMaster University, Hamilton, Canada
| | - Carlos J Orihuela
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
Emery L, Kane E, Anderson-Fears K, Liu D, Floros J, Gandhi CK. Association of surfactant protein A2 with acute respiratory failure in children. Pediatr Int 2023; 65:e15672. [PMID: 37888536 PMCID: PMC10617656 DOI: 10.1111/ped.15672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/23/2023] [Accepted: 09/06/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Interactions among single nucleotide polymorphisms (SNPs) of surfactant protein (SP) are associated with acute respiratory failure (ARF) and its short-term outcome, pulmonary dysfunction at discharge (PDAD) in children. However, genetic association studies using individual SNPs have not been conducted before. We hypothesize that SP genetic variants are associated with pediatric ARF and its short-term complications by themselves. METHODS We used available genotype and clinical data in the Floros biobank consisting of 248 children aged ≤24 months with ARF; 86 developed PDAD. A logistic regression analysis was performed for each of the 14 selected SNPs, SP-A1 and SP-A2 genotypes. A p-value less than the Bonferroni correction threshold was considered significant. A likelihood ratio test was done to compare two models (one with demographic data and another with genetic variants). RESULTS Before Bonferroni correction, female sex is associated with a decreased risk of ARF. Black race and the rs721917 of the SFTPD are associated with increased risk of ARF. After Bonferroni correction, the 1A0 1A1 genotype of SFTPA2 was associated with decreased risk of ARF. The likelihood ratio test showed that the model of the genotype information with demographic data was a better fit to predict ARF risk. None of the SP SNPs and SP-A1, SP-A2 genotypes were associated with PDAD. CONCLUSION Our results indicate that SNPs and genotypes of SPs involved in innate immunity and host defense play an important role in ARF and, in the future, may be used as biomarkers.
Collapse
Affiliation(s)
- Lucy Emery
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Elizabeth Kane
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Keenan Anderson-Fears
- Department of Public Health Science, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Dajiang Liu
- Department of Public Health Science, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Joanna Floros
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Obstetrics & Gynecology, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Chintan K Gandhi
- Department of Pediatrics, The Pennsylvania State College of Medicine, Hershey, Pennsylvania, United States of America
| |
Collapse
|
8
|
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
|
9
|
AYBEK SD, ATEŞ Ö, SEZER SONDAŞ S, GÜL A, TAKÇI Ş, ALTINTAŞ SEYYAH B. Association between surfactant protein B gene locus and acute bronchiolitis in infants. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1124468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Purpose: The aim of this study was to investigate whether there is a relationship between surfactant protein B (SFTPB) C1580T polymorphism and acute bronchiolitis.
Materials and Methods: The study analyzed the allele frequency and genotype distribution for the SFTPB C1580T polymorphism using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique in 103 acute bronchiolitis infants and 102 healthy infants.
Results: The results showed no association between SFTPB C1580T polymorphism and clinical characteristics of acute bronchiolitis. The distribution of the CT genotype was higher in acute bronchiolitis infants (43%) than in healthy subjects (39%) and distribution of the TT genotype was found lower in acute bronchiolitis infants (38%) than in healthy subjects (41%). No significant differences in genotype distribution and allele frequency for the SFTPB C1580T polymorphism were found between case group and control group
Conclusion: SFTPB C1580T gene polymorphism plays no important role in susceptibility to acute bronchiolitis. Further work on the relevance of SFTPB C1580T polymorphism in larger cohorts will require validating our results.
Collapse
Affiliation(s)
| | | | | | - Ali GÜL
- TOKAT GAZİOSMANPAŞA ÜNİVERSİTESİ
| | | | | |
Collapse
|
10
|
Battaglini D, Al-Husinat L, Normando AG, Leme AP, Franchini K, Morales M, Pelosi P, Rocco PR. Personalized medicine using omics approaches in acute respiratory distress syndrome to identify biological phenotypes. Respir Res 2022; 23:318. [PMID: 36403043 PMCID: PMC9675217 DOI: 10.1186/s12931-022-02233-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
In the last decade, research on acute respiratory distress syndrome (ARDS) has made considerable progress. However, ARDS remains a leading cause of mortality in the intensive care unit. ARDS presents distinct subphenotypes with different clinical and biological features. The pathophysiologic mechanisms of ARDS may contribute to the biological variability and partially explain why some pharmacologic therapies for ARDS have failed to improve patient outcomes. Therefore, identifying ARDS variability and heterogeneity might be a key strategy for finding effective treatments. Research involving studies on biomarkers and genomic, metabolomic, and proteomic technologies is increasing. These new approaches, which are dedicated to the identification and quantitative analysis of components from biological matrixes, may help differentiate between different types of damage and predict clinical outcome and risk. Omics technologies offer a new opportunity for the development of diagnostic tools and personalized therapy in ARDS. This narrative review assesses recent evidence regarding genomics, proteomics, and metabolomics in ARDS research.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Lou'i Al-Husinat
- Department of Clinical Medical Sciences, Faculty of Medicine, Yarmouk University, P.O. Box 566, Irbid, 21163, Jordan
| | - Ana Gabriela Normando
- Brazilian Biosciences National Laboratory, LNBio, Brazilian Center for Research in Energy and Materials, CNPEM, Campinas, Brazil
| | - Adriana Paes Leme
- Brazilian Biosciences National Laboratory, LNBio, Brazilian Center for Research in Energy and Materials, CNPEM, Campinas, Brazil
| | - Kleber Franchini
- Brazilian Biosciences National Laboratory, LNBio, Brazilian Center for Research in Energy and Materials, CNPEM, Campinas, Brazil
| | - Marcelo Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia Rm Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Cooney AL, Wambach JA, Sinn PL, McCray PB. Gene Therapy Potential for Genetic Disorders of Surfactant Dysfunction. Front Genome Ed 2022; 3:785829. [PMID: 35098209 PMCID: PMC8798122 DOI: 10.3389/fgeed.2021.785829] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary surfactant is critically important to prevent atelectasis by lowering the surface tension of the alveolar lining liquid. While respiratory distress syndrome (RDS) is common in premature infants, severe RDS in term and late preterm infants suggests an underlying genetic etiology. Pathogenic variants in the genes encoding key components of pulmonary surfactant including surfactant protein B (SP-B, SFTPB gene), surfactant protein C (SP-C, SFTPC gene), and the ATP-Binding Cassette transporter A3 (ABCA3, ABCA3 gene) result in severe neonatal RDS or childhood interstitial lung disease (chILD). These proteins play essential roles in pulmonary surfactant biogenesis and are expressed in alveolar epithelial type II cells (AEC2), the progenitor cell of the alveolar epithelium. SP-B deficiency most commonly presents in the neonatal period with severe RDS and requires lung transplantation for survival. SFTPC mutations act in an autosomal dominant fashion and more commonly presents with chILD or idiopathic pulmonary fibrosis than neonatal RDS. ABCA3 deficiency often presents as neonatal RDS or chILD. Gene therapy is a promising option to treat monogenic lung diseases. Successes and challenges in developing gene therapies for genetic disorders of surfactant dysfunction include viral vector design and tropism for target cell types. In this review, we explore adeno-associated virus (AAV), lentiviral, and adenoviral (Ad)-based vectors as delivery vehicles. Both gene addition and gene editing strategies are compared to best design treatments for lung diseases resulting from pathogenic variants in the SFTPB, SFTPC, and ABCA3 genes.
Collapse
Affiliation(s)
- Ashley L. Cooney
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Ashley L. Cooney,
| | - Jennifer A. Wambach
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, United States
| | - Patrick L. Sinn
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
| | - Paul B. McCray
- Department of Pediatrics, The University of Iowa, Iowa City, IA, United States
- Pappajohn Biomedical Institute and the Center for Gene Therapy, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
15
|
Giannini HM, Meyer NJ. Genetics of Acute Respiratory Distress Syndrome: Pathways to Precision. Crit Care Clin 2021; 37:817-834. [PMID: 34548135 DOI: 10.1016/j.ccc.2021.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Clinical risk factors alone fail to fully explain acute respiratory distress syndrome (ARDS) risk or ARDS death, suggesting that individual risk factors contribute. The goals of genomic ARDS studies include better mechanistic understanding, identifying dysregulated pathways that may be amenable to pharmacologic targeting, using genomic causal inference techniques to find measurable traits with meaning, and deconvoluting ARDS heterogeneity by proving reproducible subpopulations that may share a unique biology. This article discusses the latest advances in ARDS genomics, provides historical perspective, and highlights some of the ways that the coronavirus disease 2019 (COVID-19) pandemic is accelerating genomic ARDS research.
Collapse
Affiliation(s)
- Heather M Giannini
- University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 5038 Gates Building, Philadelphia, PA 19104, USA
| | - Nuala J Meyer
- University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 5038 Gates Building, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Regulatory Roles of Human Surfactant Protein B Variants on Genetic Susceptibility to Pseudomonas Aeruginosa Pneumonia-Induced Sepsis. Shock 2021; 54:507-519. [PMID: 31851120 DOI: 10.1097/shk.0000000000001494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Surfactant protein B (SP-B) is essential for life and plays critical roles in host defense and lowering alveolar surface tension. A single-nucleotide polymorphism (SNP rs1130866) of human SP-B (hSP-B) alters the N-linked glycosylation, thus presumably affecting SP-B function. This study has investigated the regulatory roles of hSP-B genetic variants on lung injury in pneumonia-induced sepsis. METHODS Wild-type (WT) FVB/NJ and humanized transgenic SP-B-T and SP-B-C mice (expressing either hSP-B C or T allele without mouse SP-B gene) were infected intratracheally with 50 μL (4 × 10 colony-forming units [CFUs]/mouse) Pseudomonas aeruginosa Xen5 or saline, and then killed 24 or 48 h after infection. Bacterial dynamic growths were monitored from 0 to 48 h postinfection by in vivo imaging. Histopathological, cellular, and molecular changes of lung tissues and bronchoalveolar lavage fluid (BALF) were analyzed. Surface tension of surfactants was determined with constrained drop surfactometry. RESULTS SP-B-C mice showed higher bioluminescence and CFUs, increased inflammation and mortality, the higher score of lung injury, and reduced numbers of lamellar bodies in type II cells compared with SP-B-T or WT (P < 0.05). Minimum surface tension increased dramatically in infected mice (P < 0.01) with the order of SP-B-C > SP-B-T > WT. Levels of multiple cytokines in the lung of infected SP-B-C were higher than those of SP-B-T and WT (P < 0.01). Furthermore, compared with SP-B-T or WT, SP-B-C exhibited lower SP-B, higher NF-κB and NLRP3 inflammasome activation, and higher activated caspase-3. CONCLUSIONS hSP-B variants differentially regulate susceptibility through modulating the surface activity of surfactant, cell death, and inflammatory signaling in sepsis.
Collapse
|
17
|
M Mansour S, N Shamma R, A Ahmed K, A Sabry N, Esmat G, A Mahmoud A, Maged A. Safety of inhaled ivermectin as a repurposed direct drug for treatment of COVID-19: A preclinical tolerance study. Int Immunopharmacol 2021; 99:108004. [PMID: 34333358 PMCID: PMC8299187 DOI: 10.1016/j.intimp.2021.108004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Introduction SARS-CoV-2 replication in cell cultures has been shown to be inhibited by ivermectin. However, ivermectin's low aqueous solubility and bioavailability hinders its application in COVID-19 treatment. Also, it has been suggested that best outcomes for this medication can be achieved via direct administration to the lung. Objectives This study aimed at evaluating the safety of a novel ivermectin inhalable formulation in rats as a pre-clinical step. Methods Hydroxy propyl-β-cyclodextrin (HP-β-CD) was used to formulate readily soluble ivermectin lyophilized powder. Adult male rats were used to test lung toxicity for ivermectin-HP-β-CD formulations in doses of 0.05, 0.1, 0.2, 0.4 and 0.8 mg/kg for 3 successive days. Results The X-ray diffraction for lyophilized ivermectin-HP-β-CD revealed its amorphous structure that increased drug aqueous solubility 127-fold and was rapidly dissolved within 5 s in saline. Pulmonary administration of ivermectin-HP-β-CD in doses of 0.2, 0.4 and 0.8 mg/kg showed dose-dependent increase in levels of TNF-α, IL-6, IL-13 and ICAM-1 as well as gene expression of MCP-1, protein expression of PIII-NP and serum levels of SP-D paralleled by reduction in IL-10. Moreover, lungs treated with ivermectin (0.2 mg/kg) revealed mild histopathological alterations, while severe pulmonary damage was seen in rats treated with ivermectin at doses of 0.4 and 0.8 mg/kg. However, ivermectin-HP-β-CD formulation administered in doses of 0.05 and 0.1 mg/kg revealed safety profiles. Conclusion The safety of inhaled ivermectin-HP-β-CD formulation is dose-dependent. Nevertheless, use of low doses (0.05 and 0.1 mg/kg) could be considered as a possible therapeutic regimen in COVID-19 cases.
Collapse
Affiliation(s)
- Suzan M Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt; Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Rehab N Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Kawkab A Ahmed
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt
| | - Gamal Esmat
- Department of Endemic Medicine and Hepatogastroenterology, Faculty of Medicine, Cairo University, Egypt
| | - Azza A Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| | - Amr Maged
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt; Pharmaceutical Factory, Faculty of Pharmacy, Future University in Egypt, Cairo, Egypt
| |
Collapse
|
18
|
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: 23] [Impact Index Per Article: 5.8] [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
|
19
|
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
|
20
|
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
|
21
|
Weiskirchen R. Severity of Coronavirus Disease 2019 (COVID-19): Does Surfactant Matter? Front Microbiol 2020; 11:1905. [PMID: 32982999 PMCID: PMC7479844 DOI: 10.3389/fmicb.2020.01905] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023] Open
|
22
|
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: 7] [Impact Index Per Article: 1.4] [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
|
23
|
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
|
24
|
Lynn H, Sun X, Casanova N, Gonzales-Garay M, Bime C, Garcia JGN. Genomic and Genetic Approaches to Deciphering Acute Respiratory Distress Syndrome Risk and Mortality. Antioxid Redox Signal 2019; 31:1027-1052. [PMID: 31016989 PMCID: PMC6939590 DOI: 10.1089/ars.2018.7701] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Acute respiratory distress syndrome (ARDS) is a severe, highly heterogeneous critical illness with staggering mortality that is influenced by environmental factors, such as mechanical ventilation, and genetic factors. Significant unmet needs in ARDS are addressing the paucity of validated predictive biomarkers for ARDS risk and susceptibility that hamper the conduct of successful clinical trials in ARDS and the complete absence of novel disease-modifying therapeutic strategies. Recent Advances: The current ARDS definition relies on clinical characteristics that fail to capture the diversity of disease pathology, severity, and mortality risk. We undertook a comprehensive survey of the available ARDS literature to identify genes and genetic variants (candidate gene and limited genome-wide association study approaches) implicated in susceptibility to developing ARDS in hopes of uncovering novel biomarkers for ARDS risk and mortality and potentially novel therapeutic targets in ARDS. We further attempted to address the well-known health disparities that exist in susceptibility to and mortality from ARDS. Critical Issues: Bioinformatic analyses identified 201 ARDS candidate genes with pathway analysis indicating a strong predominance in key evolutionarily conserved inflammatory pathways, including reactive oxygen species, innate immunity-related inflammation, and endothelial vascular signaling pathways. Future Directions: Future studies employing a system biology approach that combines clinical characteristics, genomics, transcriptomics, and proteomics may allow for a better definition of biologically relevant pathways and genotype-phenotype connections and result in improved strategies for the sub-phenotyping of diverse ARDS patients via molecular signatures. These efforts should facilitate the potential for successful clinical trials in ARDS and yield a better fundamental understanding of ARDS pathobiology.
Collapse
Affiliation(s)
- Heather Lynn
- Department of Physiological Sciences and University of Arizona, Tucson, Arizona.,Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Xiaoguang Sun
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Nancy Casanova
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | | | - Christian Bime
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Joe G N Garcia
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| |
Collapse
|
25
|
Gomez JL, Himes BE, Kaminski N. Precision Medicine in Critical Illness: Sepsis and Acute Respiratory Distress Syndrome. PRECISION IN PULMONARY, CRITICAL CARE, AND SLEEP MEDICINE 2019. [PMCID: PMC7120471 DOI: 10.1007/978-3-030-31507-8_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Sepsis and the acute respiratory distress syndrome (ARDS) each cause substantial morbidity and mortality. In contrast to other lung diseases, the entire course of disease in these syndromes is measured in days to weeks rather than months to years, which raises unique challenges in achieving precision medicine. We review advances in sepsis and ARDS resulting from omics studies, including those involving genome-wide association, gene expression, targeted proteomics, and metabolomics approaches. We focus on promising evidence of biological subtypes in both sepsis and ARDS that consistently display high risk for death. In sepsis, a gene expression signature with dysregulated adaptive immune signaling has evidence for a differential response to systemic steroid therapy, whereas in ARDS, a hyperinflammatory pattern identified in plasma using targeted proteomics responded more favorably to randomized interventions including high positive end-expiratory pressure, volume conservative fluid therapy, and simvastatin therapy. These early examples suggest heterogeneous biology that may be challenging to detect by clinical factors alone and speak to the promise of a precision approach that targets the right treatment at the right time to the right patient.
Collapse
Affiliation(s)
- Jose L. Gomez
- Assistant Professor Pulmonary, Critical Care and Sleep Medicine Section, Department of Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Blanca E. Himes
- Assistant Professor of Informatics, Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Naftali Kaminski
- Boehringer-Ingelheim Endowed, Professor of Internal Medicine, Chief of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT USA
| |
Collapse
|
26
|
Xi J, Wang L, Yan C, Song J, Song Y, Chen J, Zhu Y, Chen Z, Jin C, Ding J, Zhao C. The Cancer Genome Atlas dataset-based analysis of aberrantly expressed genes by GeneAnalytics in thymoma associated myasthenia gravis: focusing on T cells. J Thorac Dis 2019; 11:2315-2323. [PMID: 31372268 DOI: 10.21037/jtd.2019.06.01] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background Myasthenia gravis (MG) is a group of autoimmune disease which could be accompanied by thymoma. Many differences have been observed between thymoma-associated MG (TAMG) and non-MG thymoma (NMG). However, the molecular difference between them remained unknown. This study aimed to explore the differentially expressed genes (DEGs) between the two categories and to elucidate the possible pathogenesis of TAMG further. Methods DEGs were calculated using the RNA-Sequencing data from 11 TAMG and 10 NMG in The Cancer Genome Atlas (TCGA) database. GeneAnalytics was performed to characterize the associations between DEGs and tissues and cells, diseases, gene ontology (GO) terms, pathways, phenotypes, and drug/compounds, respectively. Genes related to T cells were sorted out using LifeMapDiscovery Cells and Tissues Database. Genes directly related to the phenotype of autoimmune diseases that were identified by VarElect were validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Results The expression level of 169 genes showed a significant difference between the two groups, with 94 up-regulated and 75 down-regulated. Overexpression of six genes (ATM, SFTPB, ANKRD55, BTLA, CCR7, TNFRSF25), which are expressed in T cells and directly related to autoimmune disease through VarElect, was identified. The overexpression of soluble BTLA (sBTLA) (P=0.027), CCR7 (P=0.0018), TNFRSF25 (P=0.0013) and ANKRD55 (P=0.0026) was validated by RT-qPCR in thymoma tissues from our center. Conclusions Overexpression of sBTLA, CCR7, TNFRSF25 and ANKRD55 was identified and validated by RT-qPCR, which could partly explain the underlying pathogenesis in TAMG.
Collapse
Affiliation(s)
- Jianying Xi
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Liang Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chong Yan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Song
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yang Song
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ji Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongjun Zhu
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhiming Chen
- Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Chun Jin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Jianyong Ding
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Department of Neurology, Jing'an District Centre Hospital of Shanghai, Fudan University, Shanghai 200040, China
| |
Collapse
|
27
|
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
|
28
|
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: 35] [Impact Index Per Article: 5.0] [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
|
29
|
|
30
|
Sorensen GL. Surfactant Protein D in Respiratory and Non-Respiratory Diseases. Front Med (Lausanne) 2018; 5:18. [PMID: 29473039 PMCID: PMC5809447 DOI: 10.3389/fmed.2018.00018] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/19/2018] [Indexed: 12/16/2022] Open
Abstract
Surfactant protein D (SP-D) is a multimeric collectin that is involved in innate immune defense and expressed in pulmonary, as well as non-pulmonary, epithelia. SP-D exerts antimicrobial effects and dampens inflammation through direct microbial interactions and modulation of host cell responses via a series of cellular receptors. However, low protein concentrations, genetic variation, biochemical modification, and proteolytic breakdown can induce decomposition of multimeric SP-D into low-molecular weight forms, which may induce pro-inflammatory SP-D signaling. Multimeric SP-D can decompose into trimeric SP-D, and this process, and total SP-D levels, are partly determined by variation within the SP-D gene, SFTPD. SP-D has been implicated in the development of respiratory diseases including respiratory distress syndrome, bronchopulmonary dysplasia, allergic asthma, and chronic obstructive pulmonary disease. Disease-induced breakdown or modifications of SP-D facilitate its systemic leakage from the lung, and circulatory SP-D is a promising biomarker for lung injury. Moreover, studies in preclinical animal models have demonstrated that local pulmonary treatment with recombinant SP-D is beneficial in these diseases. In recent years, SP-D has been shown to exert antimicrobial and anti-inflammatory effects in various non-pulmonary organs and to have effects on lipid metabolism and pro-inflammatory effects in vessel walls, which enhance the risk of atherosclerosis. A common SFTPD polymorphism is associated with atherosclerosis and diabetes, and SP-D has been associated with metabolic disorders because of its effects in the endothelium and adipocytes and its obesity-dampening properties. This review summarizes and discusses the reported genetic associations of SP-D with disease and the clinical utility of circulating SP-D for respiratory disease prognosis. Moreover, basic research on the mechanistic links between SP-D and respiratory, cardiovascular, and metabolic diseases is summarized. Perspectives on the development of SP-D therapy are addressed.
Collapse
Affiliation(s)
- Grith L Sorensen
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
31
|
Reilly JP, Christie JD, Meyer NJ. Fifty Years of Research in ARDS. Genomic Contributions and Opportunities. Am J Respir Crit Care Med 2017; 196:1113-1121. [PMID: 28481621 PMCID: PMC5694838 DOI: 10.1164/rccm.201702-0405cp] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
Clinical factors alone poorly explain acute respiratory distress syndrome (ARDS) risk and ARDS outcome. In the search for individual factors that may influence ARDS risk, the past 20 years have witnessed the identification of numerous genes and genetic variants that are associated with ARDS. The field of ARDS genomics has cycled from candidate gene association studies to bias-free approaches that identify new candidates, and increasing effort is made to understand the functional consequences that may underlie significant associations. More recently, methodologies of causal inference are being applied to maximize the information gained from genetic associations. Although challenges of sample size, both recognized and unrecognized phenotypic heterogeneity, and the paucity of early ARDS lung tissue limit some applications of the rapidly evolving field of genomic investigation, ongoing genetic research offers unique contributions to elucidating ARDS pathogenesis and the paradigm of precision ARDS medicine.
Collapse
Affiliation(s)
- John P. Reilly
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine
- Center for Translational Lung Biology, and
| | - Jason D. Christie
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine
- Center for Translational Lung Biology, and
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine
- Center for Translational Lung Biology, and
| |
Collapse
|
32
|
Vieira F, Kung JW, Bhatti F. Structure, genetics and function of the pulmonary associated surfactant proteins A and D: The extra-pulmonary role of these C type lectins. Ann Anat 2017; 211:184-201. [PMID: 28351530 DOI: 10.1016/j.aanat.2017.03.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 10/19/2022]
Abstract
The collectins family encompasses several collagenous Ca2+-dependent defense lectins that are described as pathogen recognition molecules. They play an important role in both adaptive and innate immunity. Surfactant proteins A and D are two of these proteins which were initially discovered in association with surfactant in the pulmonary system. The structure, immune and inflammatory functions, and genetic variations have been well described in relation to their roles, function and pathophysiology in the pulmonary system. Subsequently, these proteins have been discovered in a wide range of other organs and organ systems. The role of these proteins outside the pulmonary system is currently an active area of research. This review intends to provide a current overview of the genetics, structure and extra-pulmonary functions of the surfactant collectin proteins.
Collapse
Affiliation(s)
- Frederico Vieira
- Neonatal Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| | - Johannes W Kung
- Neonatal Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| | - Faizah Bhatti
- Neonatal Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Oklahoma Center for Neurosciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.
| |
Collapse
|
33
|
Liu J, Li G, Li L, Liu Z, Zhou Q, Wang G, Chen D. Surfactant protein-D (SP-D) gene polymorphisms and serum level as predictors of susceptibility and prognosis of acute kidney injury in the Chinese population. BMC Nephrol 2017; 18:67. [PMID: 28212617 PMCID: PMC5316147 DOI: 10.1186/s12882-017-0485-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/09/2017] [Indexed: 01/05/2023] Open
Abstract
Background Injury to the kidney epithelial barrier is a characteristic feature of acute kidney injury (AKI). Serum surfactant protein-D (SP-D), a known biomarker of damaged alveolar epithelium, is also secreted by renal tubular epithelial cells. Therefore, the aim of this study was to examine the possible association of SP-D with AKI susceptibility and prognosis. Methods In this study, 159 AKI patients and 120 healthy individuals were included. SP-D polymorphisms Thr11Met and Thr160Ala, AKI patient serum SP-D levels at days 1, 3 and 7 and urine KIM-1 levels in both AKI patients and controls were examined. The obtained results were correlated with the AKI stage, duration of renal replacement therapy (RRT) and prognosis. Results Serum SP-D level in AKI patients was higher than controls (p < 0.01). SP-D 11Thr/Thr genotype was more frequent in AKI patients than in controls (p < 0.01). Furthermore, AKI patients with SP-D 11Thr/Thr genotype had significantly higher serum SP-D levels (p < 0.05) compared to other genotypes. Serum SP-D levels corrected to the progression of AKI with a peak at day 3. Furthermore, the SP-D 11Thr/Thr genotype frequency and baseline serum SP-D level were higher in patients who subsequently died. Baseline serum SP-D levels positively correlated with the urine KIM-1 levels, AKI stage and RRT duration. Conclusion In our study, elevated serum SP-D was associated with worse AKI clinical outcomes and patients with SP-D 11Thr/Thr genotype were more susceptible to AKI. Collectively, these findings suggest that SP-D may be useful as a biomarker of AKI susceptibility and prognosis.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Guang Li
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Lianghai Li
- Department of Critical Care Medicine, Jingzhou Central Hospital, Jingzhou, Hubei, 434020, China
| | - Zhiyong Liu
- Department of Internal medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Qingshan Zhou
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Dechang Chen
- Department of Critical Care Medicine, Shanghai Ruijin Hospital Affiliated with Jiaotong University, Shanghai, China.
| |
Collapse
|
34
|
DIFFERENTIAL SUSCEPTIBILITY OF HUMAN SP-B GENETIC VARIANTS ON LUNG INJURY CAUSED BY BACTERIAL PNEUMONIA AND THE EFFECT OF A CHEMICALLY MODIFIED CURCUMIN. Shock 2016; 45:375-84. [PMID: 26863117 DOI: 10.1097/shk.0000000000000535] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Staphylococcus aureus is a common cause of nosocomial pneumonia frequently resulting in acute respiratory distress syndrome (ARDS). Surfactant protein B (SP-B) gene expresses two proteins involved in lowering surface tension and host defense. Genotyping studies demonstrate a significant association between human SP-B genetic variants and ARDS. Curcumins have been shown to attenuate host inflammation in many sepsis models. Our hypothesis is that functional differences of SP-B variants and treatment with curcumin (CMC2.24) modulate lung injury in bacterial pneumonia. Humanized transgenic mice, expressing either SP-B T or C allele without mouse SP-B gene, were used. Bioluminescent labeled S. aureus Xen 36 (50 μL) was injected intratracheally to cause pneumonia. Infected mice received daily CMC2.24 (40 mg/kg) or vehicle alone by oral gavage. Dynamic changes of bacteria were monitored using in vivo imaging system. Histological, cellular, and molecular indices of lung injury were studied in infected mice 48 h after infection. In vivo imaging analysis revealed total flux (bacterial number) was higher in the lung of infected SP-B-C mice compared with infected SP-B-T mice (P < 0.05). Infected SP-B-C mice demonstrated increased mortality, lung injury, apoptosis, and NF-κB expression compared with infected SP-B-T mice. Compared with controls, CMC2.24 treatment significantly reduced the following: mortality, total bacterial flux and lung tissue apoptosis, inflammatory cells, NF-κB expression (P < 0.05), and MMPs-2, -9, -12 activities (P < 0.05). We conclude that mice with SP-B-C allele are more susceptible to S. aureus pneumonia than mice with SP-B-T allele, and that CMC2.24 attenuates lung injury thus reducing mortality.
Collapse
|
35
|
Chen YJ, Wambach JA, DePass K, Wegner DJ, Chen SK, Zhang QY, Heins H, Cole FS, Hamvas A. Population-based frequency of surfactant dysfunction mutations in a native Chinese cohort. World J Pediatr 2016; 12:190-5. [PMID: 26547207 DOI: 10.1007/s12519-015-0047-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 07/14/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Rare mutations in surfactant-associated genes contribute to neonatal respiratory distress syndrome. The frequency of mutations in these genes in the Chinese population is unknown. METHODS We obtained blood spots from the Guangxi Neonatal Screening Center in Nanning, China that included Han (n=443) and Zhuang (n=313) ethnic groups. We resequenced all exons of the surfactant proteins-B (SFTPB), -C (SFTPC), and the ATP-binding cassette member A3 (ABCA3) genes and compared the frequencies of 5 common and all rare variants. RESULTS We found minor differences in the frequencies of the common variants in the Han and Zhuang cohorts. We did not find any rare mutations in SFTPB or SFTPC, but we found three ABCA3 mutations in the Han [minor allele frequency (MAF)=0.003] and 7 in the Zhuang (MAF=0.011) cohorts (P=0.10). The ABCA3 mutations were unique to each cohort; five were novel. The collapsed carrier rate of rare ABCA3 mutations in the Han and Zhuang populations combined was 1.3%, which is significantly lower than that in the United States (P<0.001). CONCLUSION The population-based frequency of mutations in ABCA3 in south China newborns is significantly lower than that in United States. The contribution of these rare ABCA3 mutations to disease burden in the south China population is still unknown.
Collapse
Affiliation(s)
- Yu-Jun Chen
- Division of Neonatology, Department of Pediatrics, the First Affiliated Hospital of Guangxi Medical University, Nanning, China.,Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Jennifer Anne Wambach
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Kelcey DePass
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Daniel James Wegner
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Shao-Ke Chen
- Department of Pediatrics, Guangxi Maternal and Child Health Hospital, Nanning, China
| | - Qun-Yuan Zhang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - Hillary Heins
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Francis Sessions Cole
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA
| | - Aaron Hamvas
- Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, USA. .,Division of Neonatology, Ann and Robert H. Lurie Children's Hospital, 225 E. Chicago Ave, Box No. 45, Chicago, IL, 60611, USA.
| |
Collapse
|
36
|
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
|
37
|
Ge L, Liu X, Chen R, Xu Y, Zuo YY, Cooney RN, Wang G. Differential susceptibility of transgenic mice expressing human surfactant protein B genetic variants to Pseudomonas aeruginosa induced pneumonia. Biochem Biophys Res Commun 2015; 469:171-5. [PMID: 26620227 DOI: 10.1016/j.bbrc.2015.11.089] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 11/20/2015] [Indexed: 12/17/2022]
Abstract
Surfactant protein B (SP-B) is essential for lung function. Previous studies have indicated that a SP-B 1580C/T polymorphism (SNP rs1130866) was associated with lung diseases including pneumonia. The SNP causes an altered N-linked glycosylation modification at Asn129 of proSP-B, e.g. the C allele with this glycosylation site but not in the T allele. This study aimed to generate humanized SP-B transgenic mice carrying either SP-B C or T allele without a mouse SP-B background and then examine functional susceptibility to bacterial pneumonia in vivo. A total of 18 transgenic mouse founders were generated by the DNA microinjection method. These founders were back-crossed with SP-B KO mice to eliminate mouse SP-B background. Four founder lines expressing similar SP-B levels to human lung were chosen for further investigation. After intratracheal infection with 50 μl of Pseudomonas aeruginosa solution (1 × 10(6) CFU/mouse) or saline in SP-B-C, SP-B-T mice the mice were sacrificed 24 h post-infection and tissues were harvested. Analysis of surfactant activity revealed differential susceptibility between SP-B-C and SP-B-T mice to bacterial infection, e.g. higher minimum surface tension in infected SP-B-C versus infected SP-B-T mice. These results demonstrate for the first time that human SP-B C allele is more susceptible to bacterial pneumonia than SP-B T allele in vivo.
Collapse
Affiliation(s)
- Lin Ge
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Biochemistry and Molecular Biology, Tianjin Medical University, Tianjin, 300070, China
| | - Xinyu Liu
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Rimei Chen
- Department of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Yongan Xu
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Robert N Cooney
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Guirong Wang
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
38
|
Choi S, Camp SM, Dan A, Garcia JGN, Dudek SM, Leckband DE. A genetic variant of cortactin linked to acute lung injury impairs lamellipodia dynamics and endothelial wound healing. Am J Physiol Lung Cell Mol Physiol 2015; 309:L983-94. [PMID: 26361873 PMCID: PMC4628987 DOI: 10.1152/ajplung.00062.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 09/04/2015] [Indexed: 01/05/2023] Open
Abstract
Inflammatory mediators released in acute lung injury (ALI) trigger the disruption of interendothelial junctions, leading to loss of vascular barrier function, protein-rich pulmonary edema, and severe hypoxemia. Genetic signatures that predict patient recovery or disease progression are poorly defined, but recent genetic screening of ALI patients has identified an association between lung inflammatory disease and a single nucleotide polymorphism (SNP) in the gene for the actin-binding and barrier-regulatory protein cortactin. This study investigated the impact of this disease-linked cortactin variant on wound healing processes that may contribute to endothelial barrier restoration. A microfabricated platform was used to quantify wound healing in terms of gap closure speed, lamellipodia dynamics, and cell velocity. Overexpression of wild-type cortactin in endothelial cells (ECs) improved directional cell motility and enhanced lamellipodial protrusion length, resulting in enhanced gap closure rates. By contrast, the cortactin SNP impaired wound closure and cell locomotion, consistent with the observed reduction in lamellipodial protrusion length and persistence. Overexpression of the cortactin SNP in lung ECs mitigated the barrier-enhancing activity of sphingosine 1-phosphate. These findings suggest that this common cortactin variant may functionally contribute to ALI predisposition by impeding endothelial wound healing.
Collapse
Affiliation(s)
- Sangwook Choi
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, Illinois
| | - Sara M Camp
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Arkaprava Dan
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, Illinois
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, Arizona
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois Hospital and Health Sciences System, Chicago, Illinois; and
| | - Deborah E Leckband
- Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, Illinois; Department of Chemistry, University of Illinois, Urbana, Illinois
| |
Collapse
|
39
|
Abstract
The unique characteristics of pulmonary circulation and alveolar-epithelial capillary-endothelial barrier allow for maintenance of the air-filled, fluid-free status of the alveoli essential for facilitating gas exchange, maintaining alveolar stability, and defending the lung against inhaled pathogens. The hallmark of pathophysiology in acute respiratory distress syndrome is the loss of the alveolar capillary permeability barrier and the presence of protein-rich edema fluid in the alveoli. This alteration in permeability and accumulation of fluid in the alveoli accompanies damage to the lung epithelium and vascular endothelium along with dysregulated inflammation and inappropriate activity of leukocytes and platelets. In addition, there is uncontrolled activation of coagulation along with suppression of fibrinolysis and loss of surfactant. These pathophysiological changes result in the clinical manifestations of acute respiratory distress syndrome, which include hypoxemia, radiographic opacities, decreased functional residual capacity, increased physiologic deadspace, and decreased lung compliance. Resolution of acute respiratory distress syndrome involves the migration of cells to the site of injury and re-establishment of the epithelium and endothelium with or without the development of fibrosis. Most of the data related to acute respiratory distress syndrome, however, originate from studies in adults or in mature animals with very few studies performed in children or juvenile animals. The lack of studies in children is particularly problematic because the lungs and immune system are still developing during childhood and consequently the pathophysiology of pediatric acute respiratory distress syndrome may differ in significant ways from that seen in acute respiratory distress syndrome in adults. This article describes what is known of the pathophysiologic processes of pediatric acute respiratory distress syndrome as we know it today while also presenting the much greater body of evidence on these processes as elucidated by adult and animal studies. It is also our expressed intent to generate enthusiasm for larger and more in-depth investigations of the mechanisms of disease and repair specific to children in the years to come.
Collapse
|
40
|
To KKW, Zhou J, Song YQ, Hung IFN, Ip WCT, Cheng ZS, Chan ASF, Kao RYT, Wu AKL, Chau S, Luk WK, Ip MSM, Chan KH, Yuen KY. Surfactant protein B gene polymorphism is associated with severe influenza. Chest 2014; 145:1237-1243. [PMID: 24337193 DOI: 10.1378/chest.13-1651] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Surfactant proteins play a key role in alveolar stability. We examined whether single nucleotide polymorphisms (SNPs) related to the surfactant protein genes are associated with severe influenza. METHODS In the first cohort, 12 SNPs related to surfactant protein genes were compared between Chinese patients with severe and mild pandemic 2009 influenza A(H1N1) (A[H1N1]pdm09) infection who were matched for age, sex, and underlying risk conditions. The SNP rs1130866, which was significantly different between the two groups, was further genotyped in a second cohort of patients. Multivariate analysis was performed to control for confounding factors. The genotype frequencies were also compared with those of the general Han Chinese population. RESULTS This study consisted of 380 patients with A(H1N1)pdm09 infection. In the first cohort of 84 patients, the C allele of rs1130866, an SNP in the surfactant protein B gene (SFTPB), was significantly associated with severe disease (OR = 3.37, P = .0048), although the P value was .057 after Bonferroni correction. In the second cohort of 296 patients, the C/C genotype was confirmed in the univariate analysis to be associated with severe disease. Multivariate analysis of the second cohort showed that genotype C/C was an independent risk factor for severe A(H1N1)pdm09 infection (second cohort: OR = 2.087, P = .023). Compared to the general Han Chinese population, the C/C genotype was overrepresented in patients with severe A(H1N1)pdm09 infection (OR = 3.232, P = .00000056). CONCLUSIONS SFTPB polymorphism is associated with severe influenza. The role of SFTPB in influenza warrants further studies.
Collapse
Affiliation(s)
- Kelvin K W To
- State Key Laboratory for Emerging Infectious Diseases; Carol Yu Centre for Infection; Research Centre of Infection and Immunology; Department of Microbiology
| | - Jie Zhou
- Research Centre of Infection and Immunology; Department of Microbiology
| | | | - Ivan F N Hung
- Carol Yu Centre for Infection; Research Centre of Infection and Immunology; Department of Medicine, The University of Hong Kong, Pokfulam
| | | | | | | | - Richard Y T Kao
- State Key Laboratory for Emerging Infectious Diseases; Research Centre of Infection and Immunology; Department of Microbiology
| | - Alan K L Wu
- Department of Pathology, Pamela Youde Nethersole Eastern Hospital, Hong Kong Special Administrative Region
| | - Sandy Chau
- Department of Pathology, United Christian Hospital, Hong Kong Special Administrative Region
| | - Wei-Kwang Luk
- Department of Pathology, Tseung Kwan O Hospital, Hong Kong Special Administrative Region, China
| | - Mary S M Ip
- Department of Medicine, The University of Hong Kong, Pokfulam
| | - Kwok-Hung Chan
- Carol Yu Centre for Infection; State Key Laboratory for Emerging Infectious Diseases; Research Centre of Infection and Immunology; Department of Microbiology
| | - Kwok-Yung Yuen
- Department of Microbiology; Research Centre of Infection and Immunology; Carol Yu Centre for Infection; State Key Laboratory for Emerging Infectious Diseases.
| |
Collapse
|
41
|
Zhu B, Zheng F, Liu N, Zhu MH, Xie J, Ye JR, Zhang J, Jiang DQ, Yang C, Jiang Y. Diagnostic value of surfactant protein-a in severe acute pancreatitis-induced acute respiratory distress syndrome. Med Sci Monit 2014; 20:1728-34. [PMID: 25256693 PMCID: PMC4186216 DOI: 10.12659/msm.891272] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background The complexity of multiple-item criteria in acute respiratory distress syndrome (ARDS) often causes inconvenience for physicians in the management of patients with severe acute pancreatitis (SAP). We evaluated whether serum SP-A levels in the presence of diffuse alveolar damage (DAD) can be qualitatively assessed for diagnosis of SAP-induced ARDS. Material/Methods Eighty rats were randomly divided into 2 groups (n=40 each) – the sham-operated (SO) group and the SAP group – and then randomly subdivided into 4 subgroups in a time-course manner. Furthermore, rats in the SAP group were subdivided into the SAP induced-ARDS group (ARDS group) and the SAP without ARDS group (non-ARDS group) according to the diagnostic standard of ARDS. The diagnostic cut-off values of SP-A for SAP-induced ARDS were determined by the receiver operating characteristic curve (ROC). Results Serum SP-A levels in Baseline, SO group, SAP group, ARDS group, and non-ARDS group were 43.15±14.29, 51.91±16.99, 193.4±35.37, 198.0+29.73, and 185.7±43.21 ug/ml, respectively. The best cut-off value for the serum SP-A level for the diagnosis of SAP-induced ARDS was 150 ug/ml and the area under the ROC curve of SP-A was 0.88. The sensitivity, specificity, positive predictive value, negative predictive value, and accuracy of SP-A in the diagnosis of SAP-induced ARDS were 100.0%, 81.8%, 71.4%, 100.0%, and 87.5%, respectively. Conclusions Serum SP-A levels may allow the detection of SAP-induced ARDS and may help to support the clinical diagnosis of ARDS. The optimal serum SP-A cut-off value to discriminate SAP-induced ARDS and other groups (SO group and non-ARDS group) is around 150 ug/ml.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Critical Care Medicine, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Feng Zheng
- Department of Critical Care Medicine, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Ning Liu
- Department of Critical Care Medicine, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Ming-Hui Zhu
- Department of Critical Care Medicine, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Jun Xie
- Department of Pathology, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Ji-Ru Ye
- Department of Respiratory Medicine, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Jun Zhang
- Comprehensive Laboratory, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Dan-Qian Jiang
- Department of Hepatobiliary Surgery, Wujin Hospital, Changzhou, China (mainland)
| | - Chun Yang
- Department of Anesthesiology, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| | - Yong Jiang
- Department of Hepatobiliary Surgery, Third Affiliated Hospital of Soochow University, Changzhou, China (mainland)
| |
Collapse
|
42
|
Meyer NJ. Beyond single-nucleotide polymorphisms: genetics, genomics, and other 'omic approaches to acute respiratory distress syndrome. Clin Chest Med 2014; 35:673-84. [PMID: 25453417 DOI: 10.1016/j.ccm.2014.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This article summarizes the contributions of high-throughput genomic, proteomic, metabolomic, and gene expression investigations to the understanding of inherited or acquired risk for acute respiratory distress syndrome (ARDS). Although not yet widely applied to a complex trait like ARDS, these techniques are now routinely used to study a variety of disease states. Omic applications hold great promise for identifying novel factors that may contribute to ARDS pathophysiology or may be appropriate for further development as biomarkers or surrogates in clinical studies. Opportunities and challenges of different techniques are discussed, and examples of successful applications in non-ARDS fields are used to illustrate the potential use of each technique.
Collapse
Affiliation(s)
- Nuala J Meyer
- Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania, Perelman School of Medicine, 3600 Spruce Street, 5039 Maloney Building, Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
Mitra S, Wade MS, Sun X, Moldobaeva N, Flores C, Ma SF, Zhang W, Garcia JGN, Jacobson JR. GADD45a promoter regulation by a functional genetic variant associated with acute lung injury. PLoS One 2014; 9:e100169. [PMID: 24940746 PMCID: PMC4062486 DOI: 10.1371/journal.pone.0100169] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 05/22/2014] [Indexed: 01/28/2023] Open
Abstract
Rationale Growth arrest DNA damage inducible alpha (GADD45a) is a stress-induced gene we have shown to participate in the pathophysiology of ventilator-induced lung injury (VILI) via regulation of mechanical stress-induced Akt ubiquitination and phosphorylation. The regulation of GADD45a expression by mechanical stress and its relationship with acute lung injury (ALI) susceptibility and severity, however, remains unknown. Objectives We examined mechanical stress-dependent regulatory elements (MSRE) in the GADD45a promoter and the contribution of promoter polymorphisms in GADD45a expression and ALI susceptibility. Methods and Results Initial studies in GADD45a knockout and heterozygous mice confirmed the relationship of GADD45a gene dose to VILI severity. Human lung endothelial cells (EC) transfected with a luciferase vector containing the full length GADD45a promoter sequence (−771 to +223) demonstrated a >4 fold increase in GADD45a expression in response to 18% cyclic stretch (CS, 4 h) compared to static controls while specific promoter regions harboring CS-dependent MSRE were identified using vectors containing serial deletion constructs of the GADD45a promoter. In silico analyses of GADD45a promoter region (−371 to −133) revealed a potential binding site for specificity protein 1 (SP1), a finding supported by confirmed SP1 binding with the GADD45a promoter and by the significant attenuation of CS-dependent GADD45a promoter activity in response to SP1 silencing. Separately, case-control association studies revealed a significant association of a GADD45a promoter SNP at −589 (rs581000, G>C) with reduced ALI susceptibility. Subsequently, we found allelic variation of this SNP is associated with both differential GADD45a expression in mechanically stressed EC (18% CS, 4 h) and differential binding site of interferon regulatory factor 7 (IRF7) at this site. Conclusion These results strongly support a functional role for GADD45a in ALI/VILI and identify a specific gene variant that confers risk for ALI.
Collapse
Affiliation(s)
- Sumegha Mitra
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Michael S. Wade
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Xiaoguang Sun
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Nurgul Moldobaeva
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Carlos Flores
- Centro de Investigacion Biomedica en red Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Nuestra Senora de Candelaria, Tenerife, Spain
| | - Shwu-Fan Ma
- Division of Pulmonary, Critical Care and Sleep, University of Chicago, Chicago, Illinois, United States of America
| | - Wei Zhang
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Joe G. N. Garcia
- Arizona Health Sciences Center, University of Arizona, Tucson, Arizona, United States of America
| | - Jeffrey R. Jacobson
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
44
|
Genetic Polymorphisms of Surfactant Protein D rs2243639, Interleukin (IL)-1β rs16944 and IL-1RN rs2234663 in Chronic Obstructive Pulmonary Disease, Healthy Smokers, and Non-Smokers. Mol Diagn Ther 2014; 18:343-54. [DOI: 10.1007/s40291-014-0084-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
Allele-specific N-glycosylation delays human surfactant protein B secretion in vitro and associates with decreased protein levels in vivo. Pediatr Res 2013; 74:646-51. [PMID: 24002332 DOI: 10.1038/pr.2013.151] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 04/18/2013] [Indexed: 11/09/2022]
Abstract
BACKGROUND Surfactant protein B (SP-B) is essential for normal lung function, and decreased concentrations of SP-B have a deleterious effect on pulmonary outcome. SP-B levels may correlate with variations in the encoding gene (SFTPB). SFTPB single-nucleotide polymorphism Ile131Thr affects proSP-B N-glycosylation in humans and the glycosylated Thr variant associates with pulmonary diseases. METHODS We analyzed SP-B levels in amniotic fluid samples for associations with SFTPB polymorphisms and generated cell lines expressing either proSP-B/131Ile or proSP-B/131Thr for examining the effect of glycosylation on proSP-B secretion kinetics. To determine any transcription preference between Ile131Thr allelic variants, we used heterozygous human lungs for allelic expression imbalance assays. RESULTS Protein levels correlated with Ile131Thr genotype and the lowest SP-B levels were observed in Thr/Thr homozygotes. Our results suggest that Ile131Thr variation-dependent N-glycosylation associates with decreased levels of SP-B, which is secreted from fetal lung to amniotic fluid. Glycosylated proSP-B/131Thr was secreted from transfected cells at a lower rate than nonglycosylated proSP-B/131Ile. Expression levels of the mRNA variants were equal. Secretion of the glycosylated variant was thus delayed in vitro by a posttranscriptional mechanism. CONCLUSION These data support the hypothesis that proSP-B glycosylation due to Ile131Thr variation may have a causal role in genetic susceptibility to acute respiratory distress.
Collapse
|
46
|
Guan J, Liu XS, Xu YJ, Xu XL, Yang YS, Wang R. Relationship between the microsatellite D2S388-5 and D2S2232 polymorphisms and chronic obstructive pulmonary disease in the Chinese Kazakh population. Respirology 2013; 18:303-7. [PMID: 23088317 DOI: 10.1111/resp.12000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE Chronic obstructive pulmonary disease (COPD) is influenced by multiple genetic and environmental factors. The role of genetic susceptibility in the pathogenesis of COPD has recently gained more attention. The surface lung surfactant protein B plays an important role in COPD pathogenesis. Microsatellite DNA has been characterized in the surfactant protein B alleles D2S388-5 and D2S2232. The aim of this research was to investigate the distribution of the D2S388-5 and D2S2232 microsatellite polymorphisms in smokers of the Kazakh ethnic group in Xinjiang, China, with and without COPD to assess whether such polymorphisms are associated with COPD susceptibility. METHODS DNA was extracted from the blood of 197 smokers with COPD and 236 control smokers of Kazakh ethnicity. The smokers diagnosed with COPD were registered at the Department of Respiratory Medicine from four different hospitals. The control group was recruited at the medical examination centre from the same area. The polymorphisms of the D2S388-5 and D2S2232 microsatellite loci were measured by multiple short tandem repeat amplification using fluorescence-labelled polymerase chain reaction and capillary electrophoresis. RESULTS Nine alleles and 32 genotypes were identified in D2S388-5, while 9 alleles and 31 genotypes were identified in D2S2232. Both genotype distributions in control smokers were in accordance with Hardy-Weinberg equilibrium. The frequency of the 254 bp allele from the D2S388-5 locus was significantly higher in the COPD group versus the control (P < 0.001, odds ratio = 5.942). CONCLUSIONS D2S388-5 microsatellite polymorphism may be associated with susceptibility to COPD in Xinjiang Kazakhs.
Collapse
Affiliation(s)
- Jian Guan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
47
|
Meyer NJ. Future clinical applications of genomics for acute respiratory distress syndrome. THE LANCET RESPIRATORY MEDICINE 2013; 1:793-803. [PMID: 24461759 DOI: 10.1016/s2213-2600(13)70134-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute respiratory distress syndrome remains a substantial cause of morbidity and mortality in intensive care units, yet no specific pharmacotherapy has proven useful in reducing the duration of mechanical ventilation or improving survival. One factor that might hamper the development of treatment for acute respiratory distress syndrome is the heterogeneous nature of the population who present with the syndrome. In this Review, the potential of genomic approaches-genetic association, gene expression, metabolomic, proteomic, and systems biology applications-for the identification of molecular endotypes within acute respiratory distress syndrome and potentially for the prediction, diagnosis, prognosis, and treatment of this difficult disorder are discussed.
Collapse
Affiliation(s)
- Nuala J Meyer
- Department of Medicine Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Aramini B, Kim C, DiAngelo S, Petersen E, Lederer D, Shah L, Robbins H, Floros J, Arcasoy SM, Sonett JR, D’Ovidio F. Donor surfactant protein D (SP-D) polymorphisms are associated with lung transplant outcome. Am J Transplant 2013; 13:2130-6. [PMID: 23841811 PMCID: PMC3819598 DOI: 10.1111/ajt.12326] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 04/20/2013] [Accepted: 04/26/2013] [Indexed: 01/25/2023]
Abstract
Chronic lung allograft dysfunction (CLAD) is the major factor limiting long-term success of lung transplantation. Polymorphisms of surfactant protein D (SP-D), an important molecule within lung innate immunity, have been associated with various lung diseases. We investigated the association between donor lung SP-D polymorphisms and posttransplant CLAD and survival in 191 lung transplant recipients consecutively transplanted. Recipients were prospectively followed with routine pulmonary function tests. Donor DNA was assayed by pyrosequencing for SP-D polymorphisms of two single-nucleotide variations altering amino acids in the mature protein N-terminal domain codon 11 (Met(11) Thr), and in codon 160 (Ala(160) Thr) of the C-terminal domain. CLAD was diagnosed in 88/191 patients, and 60/191 patients have died. Recipients of allografts that expressed the homozygous Met(11) Met variant of aa11 had significantly greater freedom from CLAD development and better survival compared to those with the homozygous Thr(11) Th variant of aa11. No significant association was noted for SP-D variants of aa160. Lung allografts with the SP-D polymorphic variant Thr(11) Th of aa11 are associated with development of CLAD and reduced survival. The observed genetic differences of the donor lung, potentially with their effects on innate immunity, may influence the clinical outcomes after lung transplantation.
Collapse
Affiliation(s)
- B. Aramini
- Lung Transplant Program, Columbia University, New York, NY
| | - C. Kim
- Lung Transplant Program, Columbia University, New York, NY
| | - S. DiAngelo
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA
| | - E. Petersen
- Lung Transplant Program, Columbia University, New York, NY
| | - D.J. Lederer
- Lung Transplant Program, Columbia University, New York, NY
| | - L. Shah
- Lung Transplant Program, Columbia University, New York, NY
| | - H. Robbins
- Lung Transplant Program, Columbia University, New York, NY
| | - J. Floros
- Department of Obstetrics and Gynecology, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA
| | - S. M. Arcasoy
- Lung Transplant Program, Columbia University, New York, NY
| | - J. R. Sonett
- Lung Transplant Program, Columbia University, New York, NY
| | - F. D’Ovidio
- Lung Transplant Program, Columbia University, New York, NY,Corresponding author: Frank D’Ovidio,
| |
Collapse
|
49
|
Cai BH, Chang LW, Li WB, Liu W, Wang XJ, Mo LX, Zhao LX, Xu HT, Yang H. Association of surfactant protein B gene polymorphisms (C/A-18, C/T1580, intron 4 and A/G9306) and haplotypes with bronchopulmonary dysplasia in chinese han population. ACTA ACUST UNITED AC 2013; 33:323-328. [PMID: 23771654 PMCID: PMC7102067 DOI: 10.1007/s11596-013-1118-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Indexed: 12/25/2022]
Abstract
This study aimed to investigate the association between surfactant protein B (SP-B) polymorphisms and bronchopulmonary dysplasia (BPD) in Chinese Han infants. We performed a casecontrol study including 86 infants with BPD and 156 matched controls. Genotyping was performed by sequence specific primer-polymerase chain reaction (PCR) and haplotypes were reconstructed by the fastPHASE software. The results showed that significant differences were detected in the genotype distribution of C/A-18 and intron 4 polymorphisms of SP-B gene between cases and controls. No significant differences were detected in the genotype distribution of C/T1580 or A/G9306 between the two groups. Haplotype analysis revealed that the frequency of A-del-C-A haplotype was higher in case group (0.12 to 0.05, P=0.003), whereas the frequency of C-inv-C-A haplotype was higher in control group (0.19 to 0.05, P=0.000). In addition, a significant difference was observed in the frequency of C-inv-T-A haplotype between the two groups. It was concluded that the polymorphisms of SP-B intron 4 and C/A-18 could be associated with BPD in Chinese Han infants, and the del allele of intron 4 and A allele of C/A-18 might be used as markers of susceptibility in the disease. Haplotype analysis indicated that the gene-gene interactions would play an important part in determining susceptibility to BPD.
Collapse
Affiliation(s)
- Bao-huan Cai
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Li-wen Chang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wen-bin Li
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wei Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xi-juan Wang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Lu-xia Mo
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Ling-xia Zhao
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hong-tao Xu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Hui Yang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
50
|
Dechert RE, Haas CF, Ostwani W. Current knowledge of acute lung injury and acute respiratory distress syndrome. Crit Care Nurs Clin North Am 2013; 24:377-401. [PMID: 22920464 DOI: 10.1016/j.ccell.2012.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) continues to be a major cause of mortality in adult and pediatric critical care medicine. This article discusses the pulmonary sequelae associated with ALI and ARDS, the support of ARDS with mechanical ventilation, available adjunctive therapies, and experimental therapies currently being tested. It is hoped that further understanding of the fundamental biology, improved identification of the patient's inflammatory state, and application of therapies directed at multiple sites of action may ultimately prove beneficial for patients suffering from ALI/ARDS.
Collapse
Affiliation(s)
- Ronald E Dechert
- Department of Respiratory Care, University of Michigan Health System, 8-720 Mott Hospital, 1540 East Hospital Drive, SPC 4208, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|