1
|
Collada A, Cruz A, Pérez-Gil J. Studying the interfacial activity and structure of pulmonary surfactant complexes. Chem Phys Lipids 2025; 266:105459. [PMID: 39581437 DOI: 10.1016/j.chemphyslip.2024.105459] [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: 09/10/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Pulmonary surfactant (PS) is a membranous complex that coats the respiratory air-liquid interface in air-breathing animal lungs. Its main function is to minimize the surface tension at the end of expiration, what is needed for preventing alveolar collapse. Although the tension reduction capabilities of surfactant depend on the formation of air-exposed phospholipid-enriched monolayers, the interfacial surfactant films are far from simple monolayers. Surfactant surface films are dynamically interconnected to continuously secreted newly synthetized material thanks to the action of a pair of very hydrophobic proteins, termed SP-B and SP-C, which are responsible to modulate the biophysical behavior of the complex. Other proteins in the system, such as the hydrophilic SP-A and SP-D, are integrated into different surfactant structures but participate primarily in the immune defense of the lung. In spite of countless studies on the structure and chemico-physical properties of surfactant membranes, the full complexity of surfactant three-dimensional structure is far from being completely understood. Here we review some of the most useful techniques that have allowed the characterization of the PS system along the years to develop the current models interpreting surfactant structure-function relationships.
Collapse
Affiliation(s)
- Ainhoa Collada
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| | - Antonio Cruz
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain.
| | - Jesús Pérez-Gil
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain.
| |
Collapse
|
2
|
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
|
3
|
Aghaei M, Talari FS, Mollahosseini A, Keramati M. Validation of a high-performance liquid chromatography method for determining lysophosphatidylcholine content in bovine pulmonary surfactant medication. Biomed Chromatogr 2024; 38:e5926. [PMID: 38881378 DOI: 10.1002/bmc.5926] [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: 01/24/2024] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
Pulmonary surfactant replacement therapy is a promising improvement in neonatal care for infants with respiratory distress syndrome. Lysophosphatidylcholine (LPC) is an undesirable component that can hinder surfactant proteins from enhancing the adsorption of surfactant lipids to balance surface tensions by creating a saturated coating on the interior of the lungs. A novel normal-phase liquid chromatography method utilizing UV detection and non-toxic solvents was developed and validated for the first time to analyze LPC in the complex matrix of pulmonary surfactant medication. The analytical method validation included evaluation of system suitability, repeatability, intermediate precision, linearity, accuracy, limit of detection (LOD), limit of quantification (LOQ), stability and robustness. The method yielded detection and quantification limits of 4.4 and 14.5 μg/ml, respectively. The calibration curve was modified linearly within the LOQ to 1.44 mg/ml range, with a determination coefficient of 0.9999 for standards and 0.9997 for sample solutions. Given the lack of reliable published data on LPC analysis in pulmonary surfactant medications, this newly developed method demonstrates promising results and offers advantages of HPLC methodology, including simplicity, accuracy, specificity, sensitivity and an exceptionally low LOD and LOQ. These attributes contribute to considering this achievement as an innovative method.
Collapse
Affiliation(s)
- Mahsa Aghaei
- ARC Bioassay (Iran Food and Drug Administration Accredited QC Laboratory of Biopharmaceutical Products), Tehran, Iran
- Department of Chemistry, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Faezeh Shirgaei Talari
- ARC Bioassay (Iran Food and Drug Administration Accredited QC Laboratory of Biopharmaceutical Products), Tehran, Iran
| | - Afsaneh Mollahosseini
- Research Laboratory of Spectrometry & Micro and Nano Extraction, Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Malihe Keramati
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
4
|
Tang K, Cui X. A Review on Investigating the Interactions between Nanoparticles and the Pulmonary Surfactant Monolayer with Coarse-Grained Molecular Dynamics Method. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:11829-11842. [PMID: 38809819 DOI: 10.1021/acs.langmuir.4c00909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Pulmonary drug delivery has garnered significant attention due to its targeted local lung action, minimal toxic side effects, and high drug utilization. However, the physicochemical properties of inhaled nanoparticles (NPs) used as drug carriers can influence their interactions with the pulmonary surfactant (PS) monolayer, potentially altering the fate of the NPs and impairing the biophysical function of the PS monolayer. Thus, the objective of this review is to summarize how the physicochemical properties of NPs affect their interactions with the PS monolayer. Initially, the definition and properties of NPs, as well as the composition and characteristics of the PS monolayer, are introduced. Subsequently, the coarse-grained molecular dynamics (CGMD) simulation method for studying the interactions between NPs and the PS monolayer is presented. Finally, the implications of the hydrophobicity, size, shape, surface charge, surface modification, and aggregation of NPs on their interactions with the PS monolayer and on the composition of biomolecular corona are discussed. In conclusion, gaining a deeper understanding of the effects of the physicochemical properties of NPs on their interactions with the PS monolayer will contribute to the development of safer and more effective nanomedicines for pulmonary drug delivery.
Collapse
Affiliation(s)
- Kailiang Tang
- School of Aerospace Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xinguang Cui
- School of Aerospace Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
5
|
Bastani MN, Jalilian S. Unraveling the enigma: The emerging significance of pulmonary surfactant proteins in predicting, diagnosing, and managing COVID-19. Immun Inflamm Dis 2024; 12:e1302. [PMID: 38860749 PMCID: PMC11165688 DOI: 10.1002/iid3.1302] [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: 01/11/2024] [Revised: 04/23/2024] [Accepted: 05/19/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Severe cases of COVID-19 often lead to the development of acute respiratory syndrome, a critical condition believed to be caused by the harmful effects of SARS-CoV-2 on type II alveolar cells. These cells play a crucial role in producing pulmonary surfactants, which are essential for proper lung function. Specifically focusing on surfactant proteins, including Surfactant protein A (SP-A), Surfactant protein B, Surfactant protein C, and Surfactant protein D (SP-D), changes in the levels of pulmonary surfactants may be a significant factor in the pathological changes seen in COVID-19 infection. OBJECTIVE This study aims to gain insights into surfactants, particularly their impacts and changes during COVID-19 infection, through a comprehensive review of current literature. The study focuses on the function of surfactants as prognostic markers, diagnostic factors, and essential components in the management and treatment of COVID-19. FINDING In general, pulmonary surfactants serve to reduce the surface tension at the gas-liquid interface, thereby significantly contributing to the regulation of respiratory mechanics. Additionally, these surfactants play a crucial role in the innate immune system within the pulmonary microenvironment. Within the spectrum of COVID-19 infections, a compelling association is observed, characterized by elevated levels of SP-D and SP-A across a range of manifestations from mild to severe pneumonia. The sudden decline in respiratory function observed in COVID-19 patients may be attributed to the decreased synthesis of surfactants by type II alveolar cells. CONCLUSION Collectin proteins such as SP-A and SP-D show promise as biomarkers, offering potential avenues for predicting and monitoring pulmonary alveolar injury in the context of COVID-19. This clarification enhances our understanding of the molecular complexities contributing to respiratory complications in severe COVID-19 cases, providing a foundation for targeted therapeutic approaches using surfactants and refined clinical management strategies.
Collapse
Affiliation(s)
- Mohammad Navid Bastani
- Department of Medical Virology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Shahram Jalilian
- Department of Medical Virology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
6
|
Barlang LA, Deimel I, Mohl BP, Blaurock C, Balkema-Buschmann A, Weinbender K, Hess B, Obernolte H, Merkel OM, Popp A. Distribution and suitability of pulmonary surfactants as a vehicle for topically applied antibodies in healthy and SARS-CoV-2 infected rodent lungs. Eur J Pharm Sci 2024; 196:106744. [PMID: 38471595 DOI: 10.1016/j.ejps.2024.106744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024]
Abstract
The use of natural pulmonary surfactants (PS) as a drug delivery vehicle for biologics is a more recent therapeutic modality. Herein, we tested different contents of PS regarding their physicochemical properties under stress conditions. The PS content of 12.25 mg/ml (Formulation B) showed desired properties such as an isotonic osmolality ∼300 mOsm/kg and an acceptable viscosity of 8.61 cSt, being lower than in commercially available PS solutions. Formulation B passed the specifications of surface lowering capacities of >80 % total lung capacity and physiologically desired formulation properties were independent of the antibody used in the composition. The identified formulation showed the capability of significantly increasing the oxygen saturation in ex vivo isolated perfused rat lungs, compared to a control and up to 30 min post lavage. In the in vivo setting, we showed that intratracheal administration of a human mAB with and without pulmonary surfactant led to higher amounts of delivered antibody within the alveolar tissue compared to intravenous administration. The antibody with the PS formulation remained longer in the alveolar tissues than the antibody without the PS formulation. Further, SARS-CoV-2 infected Golden Syrian hamsters showed that the intranasally applied antibody reached the site of infection in the alveoli and could be detected in the alveolar region 24 h after the last administration. With this work, we demonstrated that pulmonary surfactants can be used as a pulmonary drug delivery mechanism for antibodies and may subsequently improve the antibody efficacy by increasing the residence time at the desired site of action in the alveolar tissue.
Collapse
Affiliation(s)
- Lea-Adriana Barlang
- Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany; Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5‑13, 8133 Munich, Germany; Quantitative, Translational & ADME Sciences, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany.
| | - Isabelle Deimel
- Biologics Drug Product Development Department, AbbVie Deutschland GmbH & Co.KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Björn-Patrick Mohl
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald- Insel Riems, Germany
| | - Claudia Blaurock
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald- Insel Riems, Germany
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493 Greifswald- Insel Riems, Germany
| | - Kristina Weinbender
- Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Brian Hess
- Quality Control Laboratories, AbbVie Inc. Illinois, USA
| | - Helena Obernolte
- Department of Preclinical Pharmacology and In Vitro Toxicology, Fraunhofer ITEM, Nikolai-Fuchs-Straße 1, 30625 Hannover, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5‑13, 8133 Munich, Germany
| | - Andreas Popp
- Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| |
Collapse
|
7
|
Kunchala SR, van Dijk A, Veldhuizen EJA, Donnellan SC, Haagsman HP, Orgeig S. Avian surfactant protein (SP)-A2 first arose in an early tetrapod before the divergence of amphibians and gradually lost the collagen domain. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 139:104582. [PMID: 36306971 DOI: 10.1016/j.dci.2022.104582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/20/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
The air-liquid interface of the mammalian lung is lined with pulmonary surfactants, a mixture of specific proteins and lipids that serve a dual purpose-enabling air-breathing and protection against pathogens. In mammals, surfactant proteins A (SP-A) and D (SP -D) are involved in innate defence of the lung. Birds seem to lack the SP-D gene, but possess SP-A2, an additional SP-A-like gene. Here we investigated the evolution of the SP-A and SP-D genes using computational gene prediction, homology, simulation modelling and phylogeny with published avian and other vertebrate genomes. PCR was used to confirm the identity and expression of SP-A analogues in various tissue homogenates of zebra finch and turkey. In silico analysis confirmed the absence of SP-D-like genes in all 47 published avian genomes. Zebra finch and turkey SP-A1 and SP-A2 sequences, confirmed by PCR of lung homogenates, were compared with sequenced and in silico predicted vertebrate homologs to construct a phylogenetic tree. The collagen domain of avian SP-A1, especially that of zebra finch, was dramatically shorter than that of mammalian SP-A. Amphibian and reptilian genomes also contain avian-like SP-A2 protein sequences with a collagen domain. NCBI Gnomon-predicted avian and alligator SP-A2 proteins all lacked the collagen domain completely. Both avian SP-A1 and SP-A2 sequences form separate clades, which are most closely related to their closest relatives, the alligators. The C-terminal carbohydrate recognition domain (CRD) of zebra finch SP-A1 was structurally almost identical to that of rat SP-A. In fact, the CRD of SP-A is highly conserved among all the vertebrates. Birds retained a truncated version of mammalian type SP-A1 as well as a non-collagenous C-type lectin, designated SP-A2, while losing the large collagenous SP-D lectin, reflecting their evolutionary trajectory towards a unidirectional respiratory system. In the context of zoonotic infections, how these evolutionary changes affect avian pulmonary surface protection is not clear.
Collapse
Affiliation(s)
- Srinivasa Reddy Kunchala
- Centre for Cancer Diagnostics and Therapeutics, UniSA Cancer Research Institute, UniSA Clinical and Health Sciences, University of South Australia, SA, 5001, Australia
| | - Albert van Dijk
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Edwin J A Veldhuizen
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | | | - Henk P Haagsman
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Sandra Orgeig
- Centre for Cancer Diagnostics and Therapeutics, UniSA Cancer Research Institute, UniSA Clinical and Health Sciences, University of South Australia, SA, 5001, Australia.
| |
Collapse
|
8
|
Harder JW, Ma J, Alard P, Sokoloski KJ, Mathiowitz E, Furtado S, Egilmez NK, Kosiewicz MM. Male microbiota-associated metabolite restores macrophage efferocytosis in female lupus-prone mice via activation of PPARγ/LXR signaling pathways. J Leukoc Biol 2023; 113:41-57. [PMID: 36822162 DOI: 10.1093/jleuko/qiac002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Systemic lupus erythematosus development is influenced by both sex and the gut microbiota. Metabolite production is a major mechanism by which the gut microbiota influences the immune system, and we have previously found differences in the fecal metabolomic profiles of lupus-prone female and lupus-resistant male BWF1 mice. Here we determine how sex and microbiota metabolite production may interact to affect lupus. Transcriptomic analysis of female and male splenocytes showed genes that promote phagocytosis were upregulated in BWF1 male mice. Because patients with systemic lupus erythematosus exhibit defects in macrophage-mediated phagocytosis of apoptotic cells (efferocytosis), we compared splenic macrophage efferocytosis in vitro between female and male BWF1 mice. Macrophage efferocytosis was deficient in female compared to male BWF1 mice but could be restored by feeding male microbiota. Further transcriptomic analysis of the genes upregulated in male BWF1 mice revealed enrichment of genes stimulated by PPARγ and LXR signaling. Our previous fecal metabolomics analyses identified metabolites in male BWF1 mice that can activate PPARγ and LXR signaling and identified one in particular, phytanic acid, that is a very potent agonist. We show here that treatment of female BWF1 splenic macrophages with phytanic acid restores efferocytic activity via activation of the PPARγ and LXR signaling pathways. Furthermore, we found phytanic acid may restore female BWF1 macrophage efferocytosis through upregulation of the proefferocytic gene CD36. Taken together, our data indicate that metabolites produced by BWF1 male microbiota can enhance macrophage efferocytosis and, through this mechanism, could potentially influence lupus progression.
Collapse
Affiliation(s)
- James W Harder
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Jing Ma
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Pascale Alard
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Kevin J Sokoloski
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Edith Mathiowitz
- Department of Medical Science and Engineering, Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Stacia Furtado
- Department of Medical Science and Engineering, Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Michele M Kosiewicz
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| |
Collapse
|
9
|
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
|
10
|
The lung surfactant activity probed with molecular dynamics simulations. Adv Colloid Interface Sci 2022; 304:102659. [PMID: 35421637 DOI: 10.1016/j.cis.2022.102659] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 03/18/2022] [Accepted: 03/31/2022] [Indexed: 01/17/2023]
Abstract
The surface of pulmonary alveolar subphase is covered with a mixture of lipids and proteins. This lung surfactant plays a crucial role in lung functioning. It shows a complex phase behavior which can be altered by the interaction with third molecules such as drugs or pollutants. For studying multicomponent biological systems, it is of interest to couple experimental approach with computational modelling yielding atomic-scale information. Simple two, three, or four-component model systems showed to be useful for getting more insight in the interaction between lipids, lipids and proteins or lipids and proteins with drugs and impurities. These systems were studied theoretically using molecular dynamic simulations and experimentally by means of the Langmuir technique. A better understanding of the structure and behavior of lung surfactants obtained from this research is relevant for developing new synthetic surfactants for efficient therapies, and may contribute to public health protection.
Collapse
|
11
|
Lettau M, Timm S, Dittmayer C, Lopez-Rodriguez E, Ochs M. The ultrastructural heterogeneity of lung surfactant revealed by serial section electron tomography: Insights into the 3D architecture of human tubular myelin. Am J Physiol Lung Cell Mol Physiol 2022; 322:L873-L881. [PMID: 35438000 DOI: 10.1152/ajplung.00020.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Weibel's hypothetical 3D model in 1966 provided first ultrastructural details into tubular myelin (TM), a unique, complex surfactant subtype found in the hypophase of the alveolar lining layer. Although initial descriptions by electron microscopy (EM) were already published in the 1950s, a uniform morphological differentiation from other intraalveolar surfactant subtypes is still missing and potential structure-function relationships remain enigmatic. Technical developments in volume EM methods now allow a more detailed reinvestigation. To address unanswered ultrastructural questions, we analyzed ultrathin sections of humanized SP-A1/SP-A2 co-expressing mouse as well as human lung samples by conventional transmission EM. We combined these 2D information with 3D analysis of single- and dual-axis electron tomography of serial sections for high z-resolution (in a range of a few nm) and extended volumes of up to 1 µm total z-information. This study reveals that TM constitutes a heterogeneous surfactant organization mainly comprised of distorted parallel membrane planes with local intersections, which are distributed all over the TM substructure. These intersecting membrane planes form, among other various polygons, the well-known 2D "lattice", respectively 3D quadratic tubules, which in many analyzed spots of human alveoli appear to be less abundant than also observed non-concentric 3D lamellae. The additional application of serial section electron tomography to conventional transmission EM demonstrates a high heterogeneity of TM membrane networks, which indicates dynamic transformations between its substructures. Our method provides an ideal basis for further in and ex vivo structural analyses of surfactant under various conditions at nanometer scale.
Collapse
Affiliation(s)
- Marie Lettau
- Institute of Functional Anatomy, Charité , Berlin, Germany
| | - Sara Timm
- Core Facility Electron Microscopy, Charité , Berlin, Germany
| | | | | | - Matthias Ochs
- Institute of Functional Anatomy, Charité , Berlin, Germany.,German Center for Lung Research, Berlin, Germany
| |
Collapse
|
12
|
Phelps DS, Chinchilli VM, Yang L, Shearer D, Weisz J, Zhang X, Floros J. The alveolar macrophage toponome of female SP-A knockout mice differs from that of males before and after SP-A1 rescue. Sci Rep 2022; 12:5039. [PMID: 35322074 PMCID: PMC8943067 DOI: 10.1038/s41598-022-08114-2] [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: 07/30/2021] [Accepted: 02/16/2022] [Indexed: 01/05/2023] Open
Abstract
Using the Toponome Imaging System (TIS), a serial immunostainer, we studied the patterns of expression of multiple markers in alveolar macrophages (AM) from female mice lacking surfactant protein A (SP-A knockouts; KO) after "rescue" with exogenous SP-A1. We also used a 7-marker subset to compare with AM from males. AM were harvested 18 h after intrapharyngeal SP-A1 or vehicle, attached to slides, and subjected to serial immunostaining for 12 markers. Expression of the markers in each pixel of the image was analyzed both in the whole image and in individual selected cells. The marker combination in each pixel is referred to as a combinatorial molecular phenotype (CMP). A subset of antibodies was used to compare AM from male mice to the females. We found: (a) extensive AM heterogeneity in females by CMP analysis and by clustering analysis of CMPs in single cells; (b) AM from female KO mice respond to exogenous SP-A1 by increasing CMP phenotypic diversity and perhaps enhancing their potential innate immune capabilities; and (c) comparison of male and female AM responses to SP-A1 revealed that males respond more vigorously than females and clustering analysis was more effective in distinguishing males from females rather than treated from control.
Collapse
Affiliation(s)
- David S. Phelps
- grid.29857.310000 0001 2097 4281Penn State Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Vernon M. Chinchilli
- grid.29857.310000 0001 2097 4281Public Health Sciences, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Lili Yang
- grid.29857.310000 0001 2097 4281Penn State Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Debra Shearer
- grid.29857.310000 0001 2097 4281Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Judith Weisz
- grid.29857.310000 0001 2097 4281Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Xuesheng Zhang
- grid.29857.310000 0001 2097 4281Penn State Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Joanna Floros
- grid.29857.310000 0001 2097 4281Penn State Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA ,grid.29857.310000 0001 2097 4281Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| |
Collapse
|
13
|
A recipe for a good clinical pulmonary surfactant. Biomed J 2022; 45:615-628. [PMID: 35272060 PMCID: PMC9486245 DOI: 10.1016/j.bj.2022.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022] Open
Abstract
The lives of thousands premature babies have been saved along the last thirty years thanks to the establishment and consolidation of pulmonary surfactant replacement therapies (SRT). It took some time to close the gap between the identification of the biophysical and molecular causes of the high mortality associated with respiratory distress syndrome in very premature babies and the development of a proper therapy. Closing the gap required the elucidation of some key questions defining the structure–function relationships in surfactant as well as the particular role of the different molecular components assembled into the surfactant system. On the other hand, the application of SRT as part of treatments targeting other devastating respiratory pathologies, in babies and adults, is depending on further extensive research still required before enough amounts of good humanized clinical surfactants will be available. This review summarizes our current concepts on the compositional and structural determinants defining pulmonary surfactant activity, the principles behind the development of efficient natural animal-derived or recombinant or synthetic therapeutic surfactants, as well as a the most promising lines of research that are already opening new perspectives in the application of tailored surfactant therapies to treat important yet unresolved respiratory pathologies.
Collapse
|
14
|
Liu Q, Guan J, Song R, Zhang X, Mao S. Physicochemical properties of nanoparticles affecting their fate and the physiological function of pulmonary surfactants. Acta Biomater 2022; 140:76-87. [PMID: 34843949 DOI: 10.1016/j.actbio.2021.11.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/10/2021] [Accepted: 11/23/2021] [Indexed: 12/22/2022]
Abstract
Pulmonary drug delivery has drawn great attention due to its targeted local lung action, reduced side effects, and ease of administration. However, inhaled nanoparticles (NPs) could adsorb different pulmonary surfactants depending on their physicochemical properties, which may impair the physiological function of the pulmonary surfactants or alter the fate of the NPs. Thus, the objective of this review is to summarize how the physicochemical properties of NPs affecting the physiological function of pulmonary surfactants and their fate. First of all, the composition and characteristics of pulmonary surfactants, methods for studying pulmonary surfactant interaction with NPs are introduced. Thereafter, the influence of physicochemical properties of NPs on hydrophobic protein adsorption and strategies to decrease the interaction of NPs with pulmonary surfactants are discussed. Finally, the influence of physicochemical properties of NPs on lipids and hydrophilic protein adsorption and consequently their fate is described. In conclusion, a better understanding of the interaction of NPs with pulmonary surfactants will promote the faster development of safe and effective nanomedicine for pulmonary drug delivery. STATEMENT OF SIGNIFICANCE: Drug delivery carriers often face complex body fluid components after entering the human body. Pulmonary surfactants diffuse at the lung gas-liquid interface, and particles inevitably interact with pulmonary surfactants after pulmonary nanomedicine delivery. This review presents an overview of how the physicochemical properties of nanoparticles affecting their fate and physiological function of pulmonary surfactants. We believe that the information included in this review can provide important guiding for the development of safe and effective pulmonary delivery nanocarriers.
Collapse
|
15
|
Autilio C. Techniques to evaluate surfactant activity for a personalized therapy of RDS neonates. Biomed J 2021; 44:671-677. [PMID: 34758409 PMCID: PMC8847822 DOI: 10.1016/j.bj.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/28/2021] [Accepted: 11/01/2021] [Indexed: 02/06/2023] Open
Abstract
According to both European and American Guidelines, preterm neonates have to be treated by nasal continuous air pressure (CPAP) early in the delivery room. The administration of surfactant should be reserved only for babies with respiratory distress syndrome (RDS) with increased oxygen requirement, according to different thresholds of FiO2. However, these oxygenation thresholds do not fully take into consideration the lung physiopathology and mechanics or the lung surfactant biology of RDS neonates. Since surfactant replacement therapy (SRT) seems to be more effective if it is initiated within the first 3 hours after birth, the use of a reliable bench-to-bedside biological test able to predict as soon as possible the necessity of SRT will help optimise individualised therapies and personalise the actual collective strategy used to treat RDS neonates. With this in mind, in the present review several quantitative and qualitative biological tests to assess the surfactant status in RDS neonates are introduced as potential candidates for the early prediction of SRT requirement, summarising the state-of-the-art in the evaluation of surfactant activity.
Collapse
Affiliation(s)
- Chiara Autilio
- Department of Biochemistry and Molecular Biology and Research Institute ``Hospital 12 de Octubre (imas12)'', Faculty of Biology, Complutense University, Jose Antonio Novais 12, Madrid, Spain; Clinical Pathology and Microbiology Unit, "San Carlo" Hospital, Potenza, Italy.
| |
Collapse
|
16
|
Floros J, Thorenoor N, Tsotakos N, Phelps DS. Human Surfactant Protein SP-A1 and SP-A2 Variants Differentially Affect the Alveolar Microenvironment, Surfactant Structure, Regulation and Function of the Alveolar Macrophage, and Animal and Human Survival Under Various Conditions. Front Immunol 2021; 12:681639. [PMID: 34484180 PMCID: PMC8415824 DOI: 10.3389/fimmu.2021.681639] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
The human innate host defense molecules, SP-A1 and SP-A2 variants, differentially affect survival after infection in mice and in lung transplant patients. SP-A interacts with the sentinel innate immune cell in the alveolus, the alveolar macrophage (AM), and modulates its function and regulation. SP-A also plays a role in pulmonary surfactant-related aspects, including surfactant structure and reorganization. For most (if not all) pulmonary diseases there is a dysregulation of host defense and inflammatory processes and/or surfactant dysfunction or deficiency. Because SP-A plays a role in both of these general processes where one or both may become aberrant in pulmonary disease, SP-A stands to be an important molecule in health and disease. In humans (unlike in rodents) SP-A is encoded by two genes (SFTPA1 and SFTPA2) and each has been identified with extensive genetic and epigenetic complexity. In this review, we focus on functional, structural, and regulatory differences between the two SP-A gene-specific products, SP-A1 and SP-A2, and among their corresponding variants. We discuss the differential impact of these variants on the surfactant structure, the alveolar microenvironment, the regulation of epithelial type II miRNome, the regulation and function of the AM, the overall survival of the organism after infection, and others. Although there have been a number of reviews on SP-A, this is the first review that provides such a comprehensive account of the differences between human SP-A1 and SP-A2.
Collapse
Affiliation(s)
- Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Biochemistry & Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Nikolaos Tsotakos
- School of Science, Engineering, and Technology, The Pennsylvania State University, Harrisburg, PA, United States
| | - David S Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
17
|
Pioselli B, Salomone F, Mazzola G, Amidani D, Sgarbi E, Amadei F, Murgia X, Catinella S, Villetti G, De Luca D, Carnielli V, Civelli M. Pulmonary surfactant: a unique biomaterial with life-saving therapeutic applications. Curr Med Chem 2021; 29:526-590. [PMID: 34525915 DOI: 10.2174/0929867328666210825110421] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
Pulmonary surfactant is a complex lipoprotein mixture secreted into the alveolar lumen by type 2 pneumocytes, which is composed by tens of different lipids (approximately 90% of its entire mass) and surfactant proteins (approximately 10% of the mass). It is crucially involved in maintaining lung homeostasis by reducing the values of alveolar liquid surface tension close to zero at end-expiration, thereby avoiding the alveolar collapse, and assembling a chemical and physical barrier against inhaled pathogens. A deficient amount of surfactant or its functional inactivation is directly linked to a wide range of lung pathologies, including the neonatal respiratory distress syndrome. This paper reviews the main biophysical concepts of surfactant activity and its inactivation mechanisms, and describes the past, present and future roles of surfactant replacement therapy, focusing on the exogenous surfactant preparations marketed worldwide and new formulations under development. The closing section describes the pulmonary surfactant in the context of drug delivery. Thanks to its peculiar composition, biocompatibility, and alveolar spreading capability, the surfactant may work not only as a shuttle to the branched anatomy of the lung for other drugs but also as a modulator for their release, opening to innovative therapeutic avenues for the treatment of several respiratory diseases.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Sgarbi
- Preclinical R&D, Chiesi Farmaceutici, Parma. Italy
| | | | - Xabi Murgia
- Department of Biotechnology, GAIKER Technology Centre, Zamudio. Spain
| | | | | | - Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, Antoine Béclère Medical Center, APHP, South Paris University Hospitals, Paris, France; Physiopathology and Therapeutic Innovation Unit-U999, South Paris-Saclay University, Paris. France
| | - Virgilio Carnielli
- Division of Neonatology, G Salesi Women and Children's Hospital, Polytechnical University of Marche, Ancona. Italy
| | | |
Collapse
|
18
|
Simulated Breathing: Application of Molecular Dynamics Simulations to Pulmonary Lung Surfactant. Symmetry (Basel) 2021. [DOI: 10.3390/sym13071259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In this review, we delve into the topic of the pulmonary surfactant (PS) system, which is present in the respiratory system. The total composition of the PS has been presented and explored, from the types of cells involved in its synthesis and secretion, down to the specific building blocks used, such as the various lipid and protein components. The lipid and protein composition varies across species and between individuals, but ultimately produces a PS monolayer with the same role. As such, the composition has been investigated for the ways in which it imposes function and confers peculiar biophysical characteristics to the system as a whole. Moreover, a couple of theories/models that are associated with the functions of PS have been addressed. Finally, molecular dynamic (MD) simulations of pulmonary surfactant have been emphasized to not only showcase various group’s findings, but also to demonstrate the validity and importance that MD simulations can have in future research exploring the PS monolayer system.
Collapse
|
19
|
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
|
20
|
Legendre M, Butt A, Borie R, Debray MP, Bouvry D, Filhol-Blin E, Desroziers T, Nau V, Copin B, Dastot-Le Moal F, Héry M, Duquesnoy P, Allou N, Bergeron A, Bermudez J, Cazes A, Chene AL, Cottin V, Crestani B, Dalphin JC, Dombret C, Doray B, Dupin C, Giraud V, Gondouin A, Gouya L, Israël-Biet D, Kannengiesser C, Le Borgne A, Leroy S, Longchampt E, Lorillon G, Nunes H, Picard C, Reynaud-Gaubert M, Traclet J, de Vuyst P, Coulomb L'Hermine A, Clement A, Amselem S, Nathan N. Functional assessment and phenotypic heterogeneity of SFTPA1 and SFTPA2 mutations in interstitial lung diseases and lung cancer. Eur Respir J 2020; 56:13993003.02806-2020. [PMID: 32855221 DOI: 10.1183/13993003.02806-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Interstitial lung diseases (ILDs) can be caused by mutations in the SFTPA1 and SFTPA2 genes, which encode the surfactant protein (SP) complex SP-A. Only 11 SFTPA1 or SFTPA2 mutations have so far been reported worldwide, of which five have been functionally assessed. In the framework of ILD molecular diagnosis, we identified 14 independent patients with pathogenic SFTPA1 or SFTPA2 mutations. The present study aimed to functionally assess the 11 different mutations identified and to accurately describe the disease phenotype of the patients and their affected relatives. METHODS The consequences of the 11 SFTPA1 or SFTPA2 mutations were analysed both in vitro, by studying the production and secretion of the corresponding mutated proteins and ex vivo, by analysing SP-A expression in lung tissue samples. The associated disease phenotypes were documented. RESULTS For the 11 identified mutations, protein production was preserved but secretion was abolished. The expression pattern of lung SP-A available in six patients was altered and the family history reported ILD and/or lung adenocarcinoma in 13 out of 14 families (93%). Among the 28 SFTPA1 or SFTPA2 mutation carriers, the mean age at ILD onset was 45 years (range 0.6-65 years) and 48% underwent lung transplantation (mean age 51 years). Seven carriers were asymptomatic. DISCUSSION This study, which expands the molecular and clinical spectrum of SP-A disorders, shows that pathogenic SFTPA1 or SFTPA2 mutations share similar consequences for SP-A secretion in cell models and in lung tissue immunostaining, whereas they are associated with a highly variable phenotypic expression of disease, ranging from severe forms requiring lung transplantation to incomplete penetrance.
Collapse
Affiliation(s)
- Marie Legendre
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France.,Dept of Genetics, Armand Trousseau Hospital, Sorbonne University, Assistance Publique Hôpitaux de Paris (APHP), Paris, France.,Both authors contributed equally
| | - Afifaa Butt
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France.,Both authors contributed equally
| | - Raphaël Borie
- Pulmonology Dept A, Bichat Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | - Marie-Pierre Debray
- Radiology Dept, Bichat Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | - Diane Bouvry
- Pulmonology Dept, EA 2363, Avicenne Hospital, Assistance Publique Hôpitaux de Paris (APHP), Paris 13 University, COMUE Sorbonne Paris Cité, Bobigny, France
| | - Emilie Filhol-Blin
- Dept of Genetics, Armand Trousseau Hospital, Sorbonne University, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Tifenn Desroziers
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France
| | - Valérie Nau
- Dept of Genetics, Armand Trousseau Hospital, Sorbonne University, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Bruno Copin
- Dept of Genetics, Armand Trousseau Hospital, Sorbonne University, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Florence Dastot-Le Moal
- Dept of Genetics, Armand Trousseau Hospital, Sorbonne University, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Mélanie Héry
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France
| | - Philippe Duquesnoy
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France
| | - Nathalie Allou
- Pulmonology Dept, Felix Guyon Hospital, Saint Denis de La Reunion, France
| | - Anne Bergeron
- Pulmonology Dept, Saint Louis Hospital, Université de Paris, Paris, France
| | - Julien Bermudez
- Pulmonology Dept and Lung Transplant Team, North Hospital - Assistance Publique Hôpitaux de Marseille (APHM), Marseille - MEPHI, IHU Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Aurélie Cazes
- Pathology Dept, Bichat Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | | | - Vincent Cottin
- Pulmonology Dept and Coordinating Reference Center for Rare Pulmonary Diseases OrphaLung, Hospices Civils de Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Bruno Crestani
- Radiology Dept, Bichat Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | - Jean-Charles Dalphin
- Pulmonology Dept, UMR-CNRS Chrono-Environnement 6249, CNRS and CHU, Besançon, France
| | - Christine Dombret
- Radiology Dept, Bichat Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | - Bérénice Doray
- Genetic Dept, Felix Guyon Hospital, Saint Denis de La Reunion, France
| | - Clairelyne Dupin
- Pulmonology Dept, Saint Louis Hospital, Université de Paris, Paris, France
| | - Violaine Giraud
- Pulmonology Dept, Ambroise Paré Hospital, Assistance Publique Hôpitaux de Paris (APHP), Boulogne Billancourt, France
| | - Anne Gondouin
- Pulmonology Dept, UMR-CNRS Chrono-Environnement 6249, CNRS and CHU, Besançon, France
| | - Laurent Gouya
- Pulmonology Dept, Saint Louis Hospital, Université de Paris, Paris, France
| | - Dominique Israël-Biet
- Pulmonology Dept, Georges Pompidou European Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | - Caroline Kannengiesser
- Genetic Dept, Bichat Hospital, Assistance Publique Hôpitaux de Paris (APHP), Université de Paris, Paris, France
| | | | - Sylvie Leroy
- Pulmonology Dept, Pasteur Hospital, Nice, France
| | | | - Gwenaël Lorillon
- Pulmonology Dept, Saint Louis Hospital, Université de Paris, Paris, France
| | - Hilario Nunes
- Pulmonology Dept, EA 2363, Avicenne Hospital, Assistance Publique Hôpitaux de Paris (APHP), Paris 13 University, COMUE Sorbonne Paris Cité, Bobigny, France
| | | | - Martine Reynaud-Gaubert
- Pulmonology Dept and Lung Transplant Team, North Hospital - Assistance Publique Hôpitaux de Marseille (APHM), Marseille - MEPHI, IHU Méditerranée Infection, Aix-Marseille University, Marseille, France
| | - Julie Traclet
- Pulmonology Dept and Coordinating Reference Center for Rare Pulmonary Diseases OrphaLung, Hospices Civils de Lyon, Claude Bernard University Lyon 1, Lyon, France
| | - Paul de Vuyst
- Pulmonology Dept, Erasme Hospital, Brussels, Belgium
| | | | - Annick Clement
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France.,Pediatric Pulmonology Dept and Reference Center for Rare Lung Diseases RespiRare, Armand Trousseau Hospital, Paris, France
| | - Serge Amselem
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France.,Dept of Genetics, Armand Trousseau Hospital, Sorbonne University, Assistance Publique Hôpitaux de Paris (APHP), Paris, France.,Both authors contributed equally
| | - Nadia Nathan
- Sorbonne Université, Inserm Childhood Genetic Disorders, Armand Trousseau Hospital, Paris, France.,Pediatric Pulmonology Dept and Reference Center for Rare Lung Diseases RespiRare, Armand Trousseau Hospital, Paris, France.,Both authors contributed equally
| |
Collapse
|
21
|
Thorenoor N, Phelps DS, Floros J. Differential Sex-Dependent Regulation of the Alveolar Macrophage miRNome of SP-A2 and co-ex (SP-A1/SP-A2) and Sex Differences Attenuation after 18 h of Ozone Exposure. Antioxidants (Basel) 2020; 9:antiox9121190. [PMID: 33260937 PMCID: PMC7768498 DOI: 10.3390/antiox9121190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Human SP-A1 and SP-A2, encoded by SFTPA1 and SFTPA2, and their genetic variants differentially impact alveolar macrophage (AM) functions and regulation, including the miRNome. We investigated whether miRNome differences previously observed between AM from SP-A2 and SP-A1/SP-A2 mice are due to continued qualitative differences or a delayed response of mice carrying a single gene. Methods: Human transgenic (hTG) mice, carrying SP-A2 or both SP-A genes, and SP-A-KO mice were exposed to filtered air (FA) or ozone (O3). AM miRNA levels, target gene expression, and pathways determined 18 h after O3 exposure. RESULTS: We found (a) differences in miRNome due to sex, SP-A genotype, and exposure; (b) miRNome of both sexes was largely downregulated by O3, and co-ex had fewer changed (≥2-fold) miRNAs than either group; (c) the number and direction of the expression of genes with significant changes in males and females in co-ex are almost the opposite of those in SP-A2; (d) the same pathways were found in the studied groups; and (e) O3 exposure attenuated sex differences with a higher number of genotype-dependent and genotype-independent miRNAs common in both sexes after O3 exposure. Conclusion: Qualitative differences between SP-A2 and co-ex persist 18 h post-O3, and O3 attenuates sex differences.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
- Department of Biochemistry & Molecular Biology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - David S. Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
- Department of Obstetrics & Gynecology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
- Correspondence:
| |
Collapse
|
22
|
Da Silva E, Autilio C, Hougaard KS, Baun A, Cruz A, Perez-Gil J, Sørli JB. Molecular and biophysical basis for the disruption of lung surfactant function by chemicals. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183499. [PMID: 33137304 DOI: 10.1016/j.bbamem.2020.183499] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/29/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
With the intention to move away from animal testing for the toxicological evaluation of chemicals comes the need to develop new approach methodologies which are mechanism-anchored and target relevant key events leading to an adverse outcome. To date, no validated alternative methods are available for studying the acute inhalation toxicity potential of airborne chemicals but the constrained drop surfactometer measuring the surface tension of a drop of lung surfactant presents as a promising candidate. Indeed, the correlation of the increase in minimum surface tension of lung surfactant in vitro with changes in the breathing patterns of mice after inhalation of test compounds has been shown in multiple studies. However, the causal factors leading to lung surfactant inactivation remain speculative. This paper combines molecular and biophysical methods (constrained drop and captive bubble surfactometers, Langmuir-Blodgett balance, epifluorescence microscopy, cryogenic transmission electron microscopy, and differential scanning calorimetry) applied to purified porcine lung surfactant and dipalmitoylphosphatidylcholine interfacial films to gain insights into the disruption of lung surfactant function by three chemicals known to show acute inhalation toxicity (trimethoxyoctylsilane, methyl 3-oxo-2-pentylcyclopentaneacetate, and diisopentyl ether). The results of this study suggest that the test chemicals intercalate between the phospholipids at the air-liquid interface, reduce the stability of the films, and decrease the cohesivity of interface-associated multilayered structures thereby perturbing the lung surfactant surface activity. These findings contribute to a better understanding of chemically-induced lung surfactant function disruption.
Collapse
Affiliation(s)
- Emilie Da Silva
- Department of Environmental Engineering, Technical University of Denmark, Kgs. Lyngby, Denmark; National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Chiara Autilio
- Department of Biochemistry and Molecular Biology, Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| | | | - Anders Baun
- Department of Environmental Engineering, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Antonio Cruz
- Department of Biochemistry and Molecular Biology, Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| | - Jesus Perez-Gil
- Department of Biochemistry and Molecular Biology, Research Institute "Hospital 12 de Octubre (imas12)", Complutense University, Madrid, Spain
| | | |
Collapse
|
23
|
Tekos F, Skaperda Z, Goutzourelas N, Phelps DS, Floros J, Kouretas D. The Importance of Redox Status in the Frame of Lifestyle Approaches and the Genetics of the Lung Innate Immune Molecules, SP-A1 and SP-A2, on Differential Outcomes of COVID-19 Infection. Antioxidants (Basel) 2020; 9:antiox9090784. [PMID: 32854247 PMCID: PMC7554878 DOI: 10.3390/antiox9090784] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
The pandemic of COVID-19 is of great concern to the scientific community. This mainly affects the elderly and people with underlying diseases. People with obesity are more likely to experience unpleasant disease symptoms and increased mortality. The severe oxidative environment that occurs in obesity due to chronic inflammation permits viral activation of further inflammation leading to severe lung disease. Lifestyle affects the levels of inflammation and oxidative stress. It has been shown that a careful diet rich in antioxidants, regular exercise, and fasting regimens, each and/or together, can reduce the levels of inflammation and oxidative stress and strengthen the immune system as they lead to weight loss and activate cellular antioxidant mechanisms and reduce oxidative damage. Thus, a lifestyle change based on the three pillars: antioxidants, exercise, and fasting could act as a proactive preventative measure against the adverse effects of COVID-19 by maintaining redox balance and well-functioning immunity. Moreover, because of the observed diversity in the expression of COVID-19 inflammation, the role of genetics of innate immune molecules, surfactant protein A (SP-A)1 and SP-A2, and their differential impact on the local lung microenvironment and host defense is reviewed as genetics may play a major role in the diverse expression of the disease.
Collapse
Affiliation(s)
- Fotios Tekos
- Department of Biochemistry-Biotechnology, University of Thessaly, 41500 Larissa, Greece; (F.T.); (Z.S.); (N.G.)
| | - Zoi Skaperda
- Department of Biochemistry-Biotechnology, University of Thessaly, 41500 Larissa, Greece; (F.T.); (Z.S.); (N.G.)
| | - Nikolaos Goutzourelas
- Department of Biochemistry-Biotechnology, University of Thessaly, 41500 Larissa, Greece; (F.T.); (Z.S.); (N.G.)
| | - David S. Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) and Departments of Pediatrics, Hershey, PA 17033, USA; (D.S.P.); (J.F.)
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) and Departments of Pediatrics, Hershey, PA 17033, USA; (D.S.P.); (J.F.)
- Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Demetrios Kouretas
- Department of Biochemistry-Biotechnology, University of Thessaly, 41500 Larissa, Greece; (F.T.); (Z.S.); (N.G.)
- Correspondence: ; Tel.: +30-2410-565-277; Fax: +30-2410-565-290
| |
Collapse
|
24
|
Thorenoor N, S. Phelps D, Kala P, Ravi R, Floros Phelps A, M. Umstead T, Zhang X, Floros J. Impact of Surfactant Protein-A Variants on Survival in Aged Mice in Response to Klebsiella pneumoniae Infection and Ozone: Serendipity in Action. Microorganisms 2020; 8:microorganisms8091276. [PMID: 32825654 PMCID: PMC7570056 DOI: 10.3390/microorganisms8091276] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 01/10/2023] Open
Abstract
Innate immune molecules, SP-A1 (6A2, 6A4) and SP-A2 (1A0, 1A3), differentially affect young mouse survival after infection. Here, we investigated the impact of SP-A variants on the survival of aged mice. hTG mice carried a different SP-A1 or SP-A2 variant and SP-A-KO were either infected with Klebsiella pneumoniae or exposed to filtered air (FA) or ozone (O3) prior to infection, and their survival monitored over 14 days. In response to infection alone, no gene- or sex-specific (except for 6A2) differences were observed; variant-specific survival was observed (1A0 > 6A4). In response to O3, gene-, sex-, and variant-specific survival was observed with SP-A2 variants showing better survival in males than females, and 1A0 females > 1A3 females. A serendipitous, and perhaps clinically important observation was made; mice exposed to FA prior to infection exhibited significantly better survival than infected alone mice. 1A0 provided an overall better survival in males and/or females indicating a differential role for SP-A genetics. Improved ventilation, as provided by FA, resulted in a survival of significant magnitude in aged mice and perhaps to a lesser extent in young mice. This may have clinical application especially within the context of the current pandemic.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence: (N.T.); (J.F.)
| | - David S. Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
| | - Padma Kala
- Independent Consultant, Upper Saddle River, NJ 07458, USA;
| | - Radhika Ravi
- Division of Anesthesia, Department of Surgery, Veterans Affairs New Jersey Health Care System, 385 Tremont Avenue, East Orange, NJ 07018, USA;
| | | | - Todd M. Umstead
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
| | - Xuesheng Zhang
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.S.P.); (T.M.U.); (X.Z.)
- Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence: (N.T.); (J.F.)
| |
Collapse
|
25
|
Thorenoor N, Kawasawa YI, Gandhi CK, Floros J. Sex-Specific Regulation of Gene Expression Networks by Surfactant Protein A (SP-A) Variants in Alveolar Macrophages in Response to Klebsiella pneumoniae. Front Immunol 2020; 11:1290. [PMID: 32670284 PMCID: PMC7326812 DOI: 10.3389/fimmu.2020.01290] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/21/2020] [Indexed: 01/01/2023] Open
Abstract
Surfactant protein A (SP-A) in addition to its surfactant-related functions interacts with alveolar macrophages (AM), the guardian cells of innate immunity in the lungs, and regulates many of its functions under basal condition and in response to various pressures, such as infection and oxidative stress. The human SP-A locus consists of two functional genes, SFTPA1 and SFTPA2, and one pseudogene. The functional genes encode human SP-A1 and SP-A2 proteins, respectively, and each has been identified with several genetic variants. SP-A variants differ in their ability to regulate lung function mechanics and survival in response to bacterial infection. Here, we investigated the effect of hSP-A variants on the AM gene expression profile in response to Klebsiella pneumoniae infection. We used four humanized transgenic (hTG) mice that each carried SP-A1 (6A2, 6A4) or SP-A2 (1A0, 1A3), and KO. AM gene expression profiling was performed after 6 h post-infection. We found: (a) significant sex differences in the expression of AM genes; (b) in response to infection, 858 (KO), 196 (6A2), 494 (6A4), 276 (1A0), and 397 (1A3) genes were identified (P < 0.05) and some of these were differentially expressed with ≥2 fold, specific to either males or females; (c) significant SP-A1 and SP-A2 variant-specific differences in AM gene expression; (d) via Ingenuity Pathway Analysis (IPA), key pathways and molecules were identified that had direct interaction with TP53, TNF, and cell cycle signaling nodes; (e) of the three pathways (TNF, TP-53, and cell cycle signaling nodes) studied here, all variants except SP-A2 (1A3) female, showed significance for at least 2 of these pathways, and KO male showed significance for all three pathways; (f) validation of key molecules exhibited variant-specific significant differences in the expression between sexes and a similarity in gene expression profile was observed between KO and SP-A1. These results reveal for the first time a large number of biologically relevant functional pathways influenced in a sex-specific manner by SP-A variants in response to infection. These data may assist in studying molecular mechanisms of SP-A-mediated AM gene regulation and potentially identify novel therapeutic targets for K. pneumoniae infection.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Biochemistry & Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Pharmacology & Biochemistry & Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
26
|
Lipid-Protein and Protein-Protein Interactions in the Pulmonary Surfactant System and Their Role in Lung Homeostasis. Int J Mol Sci 2020; 21:ijms21103708. [PMID: 32466119 PMCID: PMC7279303 DOI: 10.3390/ijms21103708] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 05/22/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary surfactant is a lipid/protein complex synthesized by the alveolar epithelium and secreted into the airspaces, where it coats and protects the large respiratory air–liquid interface. Surfactant, assembled as a complex network of membranous structures, integrates elements in charge of reducing surface tension to a minimum along the breathing cycle, thus maintaining a large surface open to gas exchange and also protecting the lung and the body from the entrance of a myriad of potentially pathogenic entities. Different molecules in the surfactant establish a multivalent crosstalk with the epithelium, the immune system and the lung microbiota, constituting a crucial platform to sustain homeostasis, under health and disease. This review summarizes some of the most important molecules and interactions within lung surfactant and how multiple lipid–protein and protein–protein interactions contribute to the proper maintenance of an operative respiratory surface.
Collapse
|
27
|
Gandhi CK, Chen C, Wu R, Yang L, Thorenoor N, Thomas NJ, DiAngelo SL, Spear D, Keim G, Yehya N, Floros J. Association of SNP-SNP Interactions of Surfactant Protein Genes with Pediatric Acute Respiratory Failure. J Clin Med 2020; 9:jcm9041183. [PMID: 32326132 PMCID: PMC7231046 DOI: 10.3390/jcm9041183] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 12/19/2022] Open
Abstract
The hallmarks of pediatric acute respiratory failure (ARF) are dysregulated inflammation and surfactant dysfunction. The objective is to study association of surfactant protein (SP) genes’ single nucleotide polymorphisms (SNPs) with ARF and its morbidity: pulmonary dysfunction at discharge (PDAD), employing a single-, two-, and three-SNP interaction model. We enrolled 468 newborn controls and 248 children aged ≤ 24 months with ARF; 86 developed PDAD. Using quantitative genetic principles, we tested the association of SP genes SNPs with ARF and PDAD. We observed a dominant effect of rs4715 of the SFTPC on ARF risk. In a three-SNP model, we found (a) 34 significant interactions among SNPs of SFTPA1, SFTPA2, and SFTPC associated with ARF (p = 0.000000002–0.05); 15 and 19 of those interactions were associated with increased and decreased risk for ARF, respectively; (b) intergenic SNP–SNP interactions of both hydrophobic and hydrophilic SP genes associated with PDAD (p = 0.00002–0.03). The majority of intra- and intergenic interactions associated with ARF involve the SFTPA2 SNPs, whereas most of the intra- and intergenic interactions associated with PDAD are of SFTPA1 SNPs. We also observed a dominant effect of haplotypes GG of SFTPA1 associated with increased and AA of SFTPC associated with decreased ARF risk (p = 0.02). To the best of our knowledge, this is the first study showing an association of complex interactions of SP genes with ARF and PDAD. Our data indicate that SP genes polymorphisms may contribute to ARF pathogenesis and subsequent PDAD and/or may serve as markers for disease susceptibility in healthy children.
Collapse
Affiliation(s)
- Chintan K. Gandhi
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Chixiang Chen
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Rongling Wu
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Lili Yang
- School of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Nithyananda Thorenoor
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Neal J. Thomas
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
- Department of Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Susan L. DiAngelo
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Debbie Spear
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
| | - Garrett Keim
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Nadir Yehya
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joanna Floros
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (C.K.G.)
- Department of Obstetrics & Gynecology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence:
| |
Collapse
|
28
|
Wang X, Zhang Y, Mei H, An C, Liu C, Zhang Y, Zhang Y, Xin C. Study on the Relationship Between Respiratory Distress Syndrome and SP-A1 (rs1059057) Gene Polymorphism in Mongolian Very Premature Infants. Front Pediatr 2020; 8:81. [PMID: 32257981 PMCID: PMC7090089 DOI: 10.3389/fped.2020.00081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 02/18/2020] [Indexed: 11/23/2022] Open
Abstract
Aim: To study the relationship between rs1059057 polymorphism of pulmonary surfactant protein A1 (SP-A1) and respiratory distress syndrome (RDS) in Mongolian very premature infants. Methods: Applying the strategy of case-control study, 120 Mongolian RDS very premature infants (58 males and 62 females) in the western part of Inner Mongolia were selected as the case group, and 120 subjects of non-RDS very premature infants (56 males and 64 females) with the same nationality, same sex and similar gestational age were used as the control group. The single nucleotide polymorphism (SNP) site rs1059057 of SP-A1 was genotyped using polymerase chain reaction-single strand conformational polymorphism (PCR-SSCP). Results: Two genotypes, A/G and A/A, were detected at the SP-A1 rs1059057 locus in the western part of Inner Mongolia. In the case group, the frequencies of two genotypes were 53 and 47%, and the frequencies of A allele and G allele were 73 and 27%, respectively. In the control group, the frequencies of the two genotypes were 42 and 58%, and the frequencies of A allele and G allele were 79 and 21%, respectively. There was no significant difference in the genotype frequency of SP-A1 (rs1059057) locus between the case group and the control group (X 2 = 3.275, P > 0.05), and no significant difference in allele frequency between the case group and the control group (X 2 = 2.255, P > 0.05). Conclusion: The genotypes and allele frequencies of SP-A1 (rs1059057) locus were not associated with the incidence of RDS in Mongolian very premature infants in western Inner Mongolia.
Collapse
Affiliation(s)
- Xiaoli Wang
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yuheng Zhang
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hua Mei
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Caiyan An
- Clinical Medical Research Center of the Affiliated Hospital, Inner Mongolia Medical University, Hohhot, China
| | - Chunzhi Liu
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yayu Zhang
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yanbo Zhang
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Chun Xin
- Division of Neonatology, Department of Pediatric, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
29
|
In Vitro Functional and Structural Characterization of A Synthetic Clinical Pulmonary Surfactant with Enhanced Resistance to Inhibition. Sci Rep 2020; 10:1385. [PMID: 31992800 PMCID: PMC6987218 DOI: 10.1038/s41598-020-58248-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/13/2020] [Indexed: 11/25/2022] Open
Abstract
CHF5633 is a novel synthetic clinical pulmonary surfactant preparation composed by two phospholipid species, dipalmitoyl phosphatidylcholine (DPPC) and palmitoyloleoyl phosphatidylglycerol (POPG), and synthetic analogues of the hydrophobic surfactant proteins SP-B and SP-C. In this study, the interfacial properties of CHF5633 in the absence and in the presence of inhibitory serum proteins have been assessed in comparison with a native surfactant purified from porcine lungs and with poractant alpha, a widely used clinical surfactant preparation. The study of the spreading properties of CHF5633 in a Wilhelmy balance, its ability to adsorb and accumulate at air-liquid interfaces as revealed by a multiwell fluorescence assay, and its dynamic behavior under breathing-like compression-expansion cycling in a Captive Bubble Surfactometer (CBS), all revealed that CHF5633 exhibits a good behavior to reduce and sustain surface tensions to values below 5 mN/m. CHF5633 shows somehow slower initial interfacial adsorption than native surfactant or poractant alpha, but a better resistance to inhibition by serum proteins than the animal-derived clinical surfactant, comparable to that of the full native surfactant complex. Interfacial CHF5633 films formed in a Langmuir-Blodgett balance coupled with epifluorescence microscopy revealed similar propensity to segregate condensed lipid domains under compression than films made by native porcine surfactant or poractant alpha. This ability of CHF5633 to segregate condensed lipid phases can be related with a marked thermotropic transition from ordered to disordered membrane phases as exhibited by differential scanning calorimetry (DSC) of CHF5633 suspensions, occurring at similar temperatures but with higher associated enthalpy than that shown by poractant alpha. The good interfacial behavior of CHF5633 tested under physiologically meaningful conditions in vitro and its higher resistance to inactivation by serum proteins, together with its standardized and well-defined composition, makes it a particularly useful therapeutic preparation to be applied in situations associated with lung inflammation and edema, alone or in combined strategies to exploit surfactant-facilitated drug delivery.
Collapse
|
30
|
Arroyo R, Khan MA, Echaide M, Pérez-Gil J, Palaniyar N. SP-D attenuates LPS-induced formation of human neutrophil extracellular traps (NETs), protecting pulmonary surfactant inactivation by NETs. Commun Biol 2019; 2:470. [PMID: 31872075 PMCID: PMC6915734 DOI: 10.1038/s42003-019-0662-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/30/2019] [Indexed: 02/08/2023] Open
Abstract
An exacerbated amount of neutrophil extracellular traps (NETs) can cause dysfunction of systems during inflammation. However, host proteins and factors that suppress NET formation (NETosis) are not clearly identified. Here we show that an innate immune collectin, pulmonary surfactant protein-D (SP-D), attenuates lipopolysaccharide (LPS)-mediated NETosis in human neutrophils by binding to LPS. SP-D deficiency in mice (Sftpd-/-) leads to excess NET formation in the lungs during LPS-mediated inflammation. In the absence of SP-D, NETs inhibit the surface-active properties of lung surfactant, essential to prevent the collapse of alveoli, the air breathing structures of the lungs. SP-D reverses NET-mediated inhibition of surfactant and restores the biophysical properties of surfactant. To the best of our knowledge, this study establishes for the first time that (i) SP-D suppresses LPS-mediated NETosis, (ii) NETs inhibit pulmonary surfactant function in the absence of SP-D, and (iii) SP-D can restore NET-mediated inhibition of the surfactant system.
Collapse
Affiliation(s)
- Raquel Arroyo
- 1Department of Biochemistry, Faculty of Biology, Complutense University, 28040 Madrid, Spain
- Research Institute "Hospital 12 de Octubre (imas12)", 28041 Madrid, Spain
- 3Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
| | - Meraj Alam Khan
- 3Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- 4Department of Laboratory Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| | - Mercedes Echaide
- 1Department of Biochemistry, Faculty of Biology, Complutense University, 28040 Madrid, Spain
- Research Institute "Hospital 12 de Octubre (imas12)", 28041 Madrid, Spain
| | - Jesús Pérez-Gil
- 1Department of Biochemistry, Faculty of Biology, Complutense University, 28040 Madrid, Spain
- Research Institute "Hospital 12 de Octubre (imas12)", 28041 Madrid, Spain
| | - Nades Palaniyar
- 3Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 1X8 Canada
- 4Department of Laboratory Medicine and Pathobiology, and Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON Canada
| |
Collapse
|
31
|
Nalian A, Umstead TM, Yang CH, Silveyra P, Thomas NJ, Floros J, McCormack FX, Chroneos ZC. Structural and Functional Determinants of Rodent and Human Surfactant Protein A: A Synthesis of Binding and Computational Data. Front Immunol 2019; 10:2613. [PMID: 31781112 PMCID: PMC6856657 DOI: 10.3389/fimmu.2019.02613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/21/2019] [Indexed: 11/23/2022] Open
Abstract
Surfactant protein A (SP-A) provides surfactant stability, first line host defense, and lung homeostasis by binding surfactant phospholipids, pathogens, alveolar macrophages (AMs), and epithelial cells. Non-primates express one SP-A protein whereas humans express two: SP-A1 and SP-A2 with core intra- and inter-species differences in the collagen-like domain. Here, we used macrophages and solid phase binding assays to discern structural correlates of rat (r) and human (h) SP-A function. Binding assays using recombinant rSP-A expressed in insect cells showed that lack of proline hydroxylation, truncations of amino-terminal oligomerization domains, and site-directed serine (S) or alanine (A) mutagenesis of cysteine 6 (C6S), glutamate 195 (E195A), and glutamate 171 (E171A) in the carbohydrate recognition domain (CRD) all impaired SP-A binding. Replacement of arginine 197 with alanine found in hSP-A (R197A), however, restored the binding of hydroxyproline-deficient rSP-A to the SP-A receptor SP-R210 similar to native rat and human SP-A. In silico calculation of Ca++ coordination bond length and solvent accessibility surface area revealed that the “humanized” R197A substitution alters topology and solvent accessibility of the Ca++ coordination residues of the CRD domain. Binding assays in mouse AMs that were exposed to either endogenous SP-A or hSP-A1 (6A2) and hSP-A2 (1A0) isoforms in vivo revealed that mouse SP-A is a functional hybrid of hSP-A1 and hSP-A2 in regulating SP-A receptor occupancy and binding affinity. Binding assays using neonatal and adult human AMs indicates that the interaction of SP-A1 and SP-A2 with AMs is developmentally regulated. Furthermore, our data indicate that the auxiliary ion coordination loop encompassing the conserved E171 residue may comprise a conserved site of interaction with macrophages, and SP-R210 specifically, that merits further investigation to discern conserved and divergent SP-A functions between species. In summary, our findings support the notion that complex structural adaptation of SP-A regulate conserved and species specific AM functions in vertebrates.
Collapse
Affiliation(s)
- Armen Nalian
- Department of Biology, Stephen F. Austin State University, Nacogdoches, TX, United States.,The Center of Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Todd M Umstead
- Department of Pediatrics, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States
| | - Ching-Hui Yang
- The Center of Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Patricia Silveyra
- Department of Pediatrics, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States
| | - Neal J Thomas
- Department of Pediatrics, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Department of Public Health Sciences, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Center of Host Defense and Inflammatory Disease Research, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States
| | - Francis X McCormack
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Zissis C Chroneos
- The Center of Biomedical Research, University of Texas Health Science Center at Tyler, Tyler, TX, United States.,Department of Pediatrics, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Pulmonary Immunology and Physiology Laboratory, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States.,Department of Microbiology and Immunology, Pennsylvania State University College of Medicine and PennState Health Children's Hospital, Hershey, PA, United States
| |
Collapse
|
32
|
Thorenoor N, Kawasawa YI, Gandhi CK, Zhang X, Floros J. Differential Impact of Co-expressed SP-A1/SP-A2 Protein on AM miRNome; Sex Differences. Front Immunol 2019; 10:1960. [PMID: 31475015 PMCID: PMC6707024 DOI: 10.3389/fimmu.2019.01960] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/02/2019] [Indexed: 12/04/2022] Open
Abstract
In humans there are two surfactant protein A (SP-A) functional genes SFTPA1 and SFTPA2 encoding innate immune molecules, SP-A1 and SP-A2, respectively, with numerous genetic variants each. SP-A interacts and regulates many of the functions of alveolar macrophages (AM). It is shown that SP-A variants differ in their ability to regulate the AM miRNome in response to oxidative stress (OxS). Because humans have both SP-A gene products, we were interested to determine the combined effect of co-expressed SP-A1/SP-A2 (co-ex) in response to ozone (O3) induced OxS on AM miRNome. Human transgenic (hTG) mice, carrying both SP-A1/SP-A2 (6A2/1A0, co-ex) and SP-A- KO were utilized. The hTG and KO mice were exposed to filtered air (FA) or O3 and miRNA levels were measured after AM isolation with or without normalization to KO. We found: (i) The AM miRNome of co-ex males and females in response to OxS to be largely downregulated after normalization to KO, but after Bonferroni multiple comparison analysis only in females the AM miRNome remained significantly different compared to control (FA); (ii) The targets of the significantly changed miRNAs were downregulated in females and upregulated in males; (iii) Several of the validated mRNA targets were involved in pro-inflammatory response, anti-apoptosis, cell cycle, cellular growth and proliferation; (iv) The AM of SP-A2 male, shown, previously to have major effect on the male AM miRNome in response to OxS, shared similarities with the co-ex, namely in pathways involved in the pro-inflammatory response and anti-apoptosis but also exhibited differences with the cell-cycle, growth, and proliferation pathway being involved in co-ex and ROS homeostasis in SP-A2 male. We speculate that the presence of both gene products vs. single gene products differentially impact the AM responses in males and females in response to OxS.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Departments of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Chintan K Gandhi
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xuesheng Zhang
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
33
|
Noutsios GT, Thorenoor N, Zhang X, Phelps DS, Umstead TM, Durrani F, Floros J. Major Effect of Oxidative Stress on the Male, but Not Female, SP-A1 Type II Cell miRNome. Front Immunol 2019; 10:1514. [PMID: 31354704 PMCID: PMC6635478 DOI: 10.3389/fimmu.2019.01514] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/17/2019] [Indexed: 11/15/2022] Open
Abstract
Pulmonary surfactant protein A (SP-A) plays an important role in surfactant metabolism and lung innate immunity. In humans there are two proteins, SP-A1 and SP-A2, encoded by SFTPA1 and SFTPA2, respectively, which are produced by the alveolar type II cells (T2C). We sought to investigate the differential influence of SP-A1 and SP-A2 in T2C miRNome under oxidative stress (OxS). SP-A knock out (KO) and hTG male and female mice expressing SP-A1 or SP-A2 as well as gonadectomized (Gx) mice were exposed to O3-induced oxidative stress (OxS) or filtered air (FA). Expression of miRNAs and mRNAs was measured in the T2C of experimental animals. (a) In SP-A1 males after normalizing to KO males, significant changes were observed in the miRNome in terms of sex-OxS effects, with 24 miRNAs being differentially expressed under OxS. (b) The mRNA targets of the dysregulated miRNAs included Ago2, Ddx20, Plcg2, Irs1, Elf2, Jak2, Map2k4, Bcl2, Ccnd1, and Vhl. We validated the expression levels of these transcripts, and observed that the mRNA levels of all of these targets were unaffected in SP-A1 T2C but six of these were significantly upregulated in the KO (except Bcl2 that was downregulated). (c) Gondadectomy had a major effect on the expression of miRNAs and in three of the mRNA targets (Irs1, Bcl2, and Vhl). Ccnd1 was upregulated in KO regardless of Gx. (d) The targets of the significantly changed miRNAs are involved in several pathways including MAPK signaling pathway, cell cycle, anti-apoptosis, and other. In conclusion, in response to OxS, SP-A1 and male hormones appear to have a major effect in the T2C miRNome.
Collapse
Affiliation(s)
- George T Noutsios
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Xuesheng Zhang
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - David S Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Todd M Umstead
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Faryal Durrani
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, Pennsylvania State University, Hershey, PA, United States.,Department of Obstetrics and Gynecology, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
34
|
Wang G, Umstead TM, Hu S, Mikerov AN, Phelps DS, Floros J. Differential Effects of Human SP-A1 and SP-A2 on the BAL Proteome and Signaling Pathways in Response to Klebsiella pneumoniae and Ozone Exposure. Front Immunol 2019; 10:561. [PMID: 30972061 PMCID: PMC6443908 DOI: 10.3389/fimmu.2019.00561] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/04/2019] [Indexed: 12/29/2022] Open
Abstract
Surfactant protein A (SP-A) plays critical roles in host defense, regulation of inflammation and surfactant metabolism in the lung. The human SP-A locus consists of two functional genes, SFTPA1 and SFTPA2 encoding surfactant proteins SP-A1 and SP-A2, respectively. Structural and functional differences exist between SP-A1 and SP-A2 in vitro and in vivo. Ozone is a major air pollutant with a negative impact on many biological processes. In this study we used humanized transgenic (hTG) SP-A1 and SP-A2 mice, and SP-A KO mice to study in vivo effects of SP-A1 and SP-A2 on the bronchoalveolar lavage (BAL) proteomic profile and associated signaling pathways in response to ozone or filtered air (FA) exposure and Klebsiella pneumoniae infection. The BAL samples were harvested 24 h after ozone (2 ppm for 3 h) or FA exposure and infection and analyzed by two-dimensional difference gel electrophoresis (2D-DIGE) and MALDI-ToF/ToF. We found: that (1) Ozone exposure, but not infection, is a major factor for increases in total BAL protein content. (2) A total of 36 proteins were identified, accounting for 89.62% of the BAL proteins resolved by the 2D-DIGE system. (3) The number of proteins in which levels were altered more than 25% following infection and FA exposure was: SP-A2 > SP-A1 > KO for male mice, and SP-A2 ≈ SP-A1 > KO for female mice. (4) The number of proteins with more than 25% increase/decrease after ozone exposure and infection was: SP-A2 > SP-A1 ≈ KO, with the majority being increases in male mice and decreases in female mice. (5) Eleven out of the 36 proteins, including annexin A5, glutathione S-transferase A4, SP-A1/SP-A2, and 14-3-3 zeta protein, exhibited significant differences among SP-A genotypes. The acute phase response (APR) that includes the NF-kB signaling pathway plays a critical role, followed by Nrf2-mediated oxidative response, and others. These associated with SP-A genotype, sex, and ozone-induced oxidative stress in response to infection. We concluded that human SP-A2 and SP-A1 exhibit differential genotype-and sex-dependent innate immune responses to microbial pathogens and/or ozone-induced oxidative stress by modulating proteomic patterns and signaling pathways in the lung.
Collapse
Affiliation(s)
- Guirong Wang
- Department of Pediatrics, Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Todd M Umstead
- Department of Pediatrics, Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Sanmei Hu
- Department of Pediatrics, Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Anatoly N Mikerov
- Department of Pediatrics, Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David S Phelps
- Department of Pediatrics, Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
35
|
Thorenoor N, Umstead TM, Zhang X, Phelps DS, Floros J. Survival of Surfactant Protein-A1 and SP-A2 Transgenic Mice After Klebsiella pneumoniae Infection, Exhibits Sex-, Gene-, and Variant Specific Differences; Treatment With Surfactant Protein Improves Survival. Front Immunol 2018; 9:2404. [PMID: 30459763 PMCID: PMC6232836 DOI: 10.3389/fimmu.2018.02404] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/28/2018] [Indexed: 01/23/2023] Open
Abstract
Surfactant protein A (SP-A) is involved in lung innate host defense and surfactant-related functions. The human SFTPA1 and SFTPA2 genes encode SP-A1 and SP-2 proteins, and each gene has been identified with numerous genetic variants. SP-A1 and SP-A2 differentially enhance bacterial phagocytosis. Sex differences have been observed in pulmonary disease and in survival of wild type and SP-A knockout (KO) mice. The impact of human SP-A variants on survival after infection is unknown. In this study, we determined whether SP-A variants differentially affect survival of male and female mice infected with Klebsiella pneumoniae. Transgenic (TG) mice, where each carries a different human (h) SP-A1 (6A2, 6A4), SP-A2 (1A0, 1A3) variant or both variants SP-A1/SP-A2 (6A2/1A0, co-ex), and SP-A- KO, were utilized. The hTG and KO mice were infected intratracheally with K. pneumoniae bacteria, and groups of KO mice were treated with SP-A1 or SP-A2 either prior to and/or at the time of infection and survival for both experimental groups was monitored over 14 days. The binding of purified SP-A1 and SP-A2 proteins to phagocytic and non-phagocytic cells and expression of cell surface proteins in alveolar macrophages (AM) from SP-A1 and SP-A2 mice was examined. We observed gene-, variant-, and sex-specific (except for co-ex) differences with females showing better survival: (a) Gene-specific differences: co-ex = SP-A2 > SP-A1 > KO (both sexes); (b) Variant-specific survival co-ex (6A2/1A0) = 1A0 > 1A3 = 6A2 > 6A4 (both sexes); (c) KO mice treated with SPs (SP-A1 or SP-A2) proteins exhibit significantly (p < 0.05) better survival; (d) SP-A1 and SP-A2 differentially bind to phagocytic, but not to non-phagocytic cells, and AM from SP-A1 and SP-A2 hTG mice exhibit differential expression of cell surface proteins. Our results indicate that sex and SP-A genetics differentially affect survival after infection and that exogenous SP-A1/SP-A2 treatment significantly improves survival. We postulate that the differential SP-A1/SP-A2 binding to the phagocytic cells and the differential expression of cell surface proteins that bind SP-A by AM from SP-A1 and SP-A2 mice play a role in this process. These findings provide insight into the importance of sex and innate immunity genetics in survival following infection.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Todd M Umstead
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xuesheng Zhang
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - David S Phelps
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Joanna Floros
- Center for Host defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, United States.,Department of Obstetrics & Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
36
|
Guagliardo R, Pérez-Gil J, De Smedt S, Raemdonck K. Pulmonary surfactant and drug delivery: Focusing on the role of surfactant proteins. J Control Release 2018; 291:116-126. [PMID: 30321577 DOI: 10.1016/j.jconrel.2018.10.012] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 11/30/2022]
Abstract
Pulmonary surfactant (PS) has been extensively studied because of its primary role in mammalian breathing. The deposition of this surface-active material at the alveolar air-water interface is essential to lower surface tension, thus avoiding alveolar collapse during expiration. In addition, PS is involved in host defense, facilitating the clearance of potentially harmful particulates. PS has a unique composition, including 92% of lipids and 8% of surfactant proteins (SPs) by mass. Although they constitute the minor fraction, SPs to a large extent orchestrate PS-related functions. PS contains four surfactant proteins (SPs) that can be structurally and functionally divided in two groups, i.e. the large hydrophilic SP-A and SP-D and the smaller hydrophobic SP-B and SP-C. The former belong to the family of collectins and are involved in opsonization processes, thus promoting uptake of pathogens and (nano)particles by phagocytic cell types. The latter SPs regulate interfacial surfactant adsorption dynamics, facilitating (phospho)lipid transfer and membrane fusion processes. In the context of pulmonary drug delivery, the exploitation of PS as a carrier to promote drug spreading along the alveolar interface is gaining interest. In addition, recent studies investigated the interaction of PS with drug-loaded nanoparticles (nanomedicines) following pulmonary administration, which strongly influences their biological fate, drug delivery efficiency and toxicological profile. Interestingly, the specific biophysical mode-of-action of the four SPs affect the drug delivery process of nanomedicines both on the extra-and intracellular level, modulating pulmonary distribution, cell targeting and intracellular delivery. This knowledge can be harnessed to exploit SPs for the design of unique and bio-inspired drug delivery strategies.
Collapse
Affiliation(s)
- Roberta Guagliardo
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Jesús Pérez-Gil
- Departamento de Bioquimica y Biologia Molecular, Facultad de Biologia, Research Institute Hospital 12 Octubre, Universidad Complutense, José Antonio Novais 2, 28040 Madrid, Spain.
| | - Stefaan De Smedt
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Laboratory of General Biochemistry and Physical Pharmacy, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
37
|
Lin Z, Thorenoor N, Wu R, DiAngelo SL, Ye M, Thomas NJ, Liao X, Lin TR, Warren S, Floros J. Genetic Association of Pulmonary Surfactant Protein Genes, SFTPA1, SFTPA2, SFTPB, SFTPC, and SFTPD With Cystic Fibrosis. Front Immunol 2018; 9:2256. [PMID: 30333828 PMCID: PMC6175982 DOI: 10.3389/fimmu.2018.02256] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/11/2018] [Indexed: 01/03/2023] Open
Abstract
Surfactant proteins (SP) are involved in surfactant function and innate immunity in the human lung. Both lung function and innate immunity are altered in CF, and altered SP levels and genetic association are observed in Cystic Fibrosis (CF). We hypothesized that single nucleotide polymorphisms (SNPs) within the SP genes associate with CF or severity subgroups, either through single SNP or via SNP-SNP interactions between two SNPs of a given gene (intragenic) and/or between two genes (intergenic). We genotyped a total of 17 SP SNPs from 72 case-trio pedigree (SFTPA1 (5), SFTPA2 (4), SFTPB (4), SFTPC (2), and SFTPD (2)), and identified SP SNP associations by applying quantitative genetic principles. The results showed (a) Two SNPs, SFTPB rs7316 (p = 0.0083) and SFTPC rs1124 (p = 0.0154), each associated with CF. (b) Three intragenic SNP-SNP interactions, SFTPB (rs2077079, rs3024798), and SFTPA1 (rs1136451, rs1059057 and rs4253527), associated with CF. (c) A total of 34 intergenic SNP-SNP interactions among the 4 SP genes to be associated with CF. (d) No SNP-SNP interaction was observed between SFTPA1 or SFTPA2 and SFTPD. (e) Equal number of SNP-SNP interactions were observed between SFTPB and SFTPA1/SFTPA2 (n = 7) and SP-B and SFTPD (n = 7). (f) SFTPC exhibited significant SNP-SNP interactions with SFTPA1/SFTPA2 (n = 11), SFTPB (n = 4) and SFTPD (n = 3). (g) A single SFTPB SNP was associated with mild CF after Bonferroni correction, and several intergenic interactions that are associated (p < 0.01) with either mild or moderate/severe CF were observed. These collectively indicate that complex SNP-SNP interactions of the SP genes may contribute to the pulmonary disease in CF patients. We speculate that SPs may serve as modifiers for the varied progression of pulmonary disease in CF and/or its severity.
Collapse
Affiliation(s)
- Zhenwu Lin
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Nithyananda Thorenoor
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Rongling Wu
- Public Health Science, College of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Susan L. DiAngelo
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Meixia Ye
- Public Health Science, College of Medicine, Pennsylvania State University, Hershey, PA, United States
- Center for Computational Biology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing, China
| | - Neal J. Thomas
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Xiaojie Liao
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Tony R. Lin
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Stuart Warren
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Pennsylvania State University, Hershey, PA, United States
- Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
38
|
Bornelöv S, Seroussi E, Yosefi S, Benjamini S, Miyara S, Ruzal M, Grabherr M, Rafati N, Molin AM, Pendavis K, Burgess SC, Andersson L, Friedman-Einat M. Comparative omics and feeding manipulations in chicken indicate a shift of the endocrine role of visceral fat towards reproduction. BMC Genomics 2018; 19:295. [PMID: 29695257 PMCID: PMC5922311 DOI: 10.1186/s12864-018-4675-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 04/15/2018] [Indexed: 02/18/2023] Open
Abstract
Background The mammalian adipose tissue plays a central role in energy-balance control, whereas the avian visceral fat hardly expresses leptin, the key adipokine in mammals. Therefore, to assess the endocrine role of adipose tissue in birds, we compared the transcriptome and proteome between two metabolically different types of chickens, broilers and layers, bred towards efficient meat and egg production, respectively. Results Broilers and layer hens, grown up to sexual maturation under free-feeding conditions, differed 4.0-fold in weight and 1.6-fold in ovarian-follicle counts, yet the relative accumulation of visceral fat was comparable. RNA-seq and mass-spectrometry (MS) analyses of visceral fat revealed differentially expressed genes between broilers and layers, 1106 at the mRNA level (FDR ≤ 0.05), and 203 at the protein level (P ≤ 0.05). In broilers, Ingenuity Pathway Analysis revealed activation of the PTEN-pathway, and in layers increased response to external signals. The expression pattern of genes encoding fat-secreted proteins in broilers and layers was characterized in the RNA-seq and MS data, as well as by qPCR on visceral fat under free feeding and 24 h-feed deprivation. This characterization was expanded using available RNA-seq data of tissues from red junglefowl, and of visceral fat from broilers of different types. These comparisons revealed expression of new adipokines and secreted proteins (LCAT, LECT2, SERPINE2, SFTP1, ZP1, ZP3, APOV1, VTG1 and VTG2) at the mRNA and/or protein levels, with dynamic gene expression patterns in the selected chicken lines (except for ZP1; FDR/P ≤ 0.05) and feed deprivation (NAMPT, SFTPA1 and ZP3) (P ≤ 0.05). In contrast, some of the most prominent adipokines in mammals, leptin, TNF, IFNG, and IL6 were expressed at a low level (FPKM/RPKM< 1) and did not show differential mRNA expression neither between broiler and layer lines nor between fed vs. feed-deprived chickens. Conclusions Our study revealed that RNA and protein expression in visceral fat changes with selective breeding, suggesting endocrine roles of visceral fat in the selected phenotypes. In comparison to gene expression in visceral fat of mammals, our findings points to a more direct cross talk of the chicken visceral fat with the reproductive system and lower involvement in the regulation of appetite, inflammation and insulin resistance. Electronic supplementary material The online version of this article (10.1186/s12864-018-4675-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Susanne Bornelöv
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23, Uppsala, Sweden.,Present Address: Wellcome Trust Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Eyal Seroussi
- Agricultural Research Organization, Volcani Center, Rishon LeZion, Israel
| | - Sara Yosefi
- Agricultural Research Organization, Volcani Center, Rishon LeZion, Israel
| | - Sharon Benjamini
- Agricultural Research Organization, Volcani Center, Rishon LeZion, Israel.,Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, 76100, Rehovot, Israel
| | - Shoval Miyara
- Agricultural Research Organization, Volcani Center, Rishon LeZion, Israel
| | - Mark Ruzal
- Agricultural Research Organization, Volcani Center, Rishon LeZion, Israel
| | - Manfred Grabherr
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23, Uppsala, Sweden.,Bioinformatics Infrastructure for Life Sciences, Uppsala University, Uppsala, Sweden
| | - Nima Rafati
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Anna-Maja Molin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Ken Pendavis
- College of Agriculture and Life Sciences, University of Arizona, Tucson, USA
| | - Shane C Burgess
- College of Agriculture and Life Sciences, University of Arizona, Tucson, USA
| | - Leif Andersson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23, Uppsala, Sweden.,Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, SE-750 07, Uppsala, Sweden.,Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843-4458, USA
| | | |
Collapse
|
39
|
Differential effects of innate immune variants of surfactant protein-A1 (SFTPA1) and SP-A2 (SFTPA2) in airway function after Klebsiella pneumoniae infection and sex differences. Respir Res 2018; 19:23. [PMID: 29394894 PMCID: PMC5797374 DOI: 10.1186/s12931-018-0723-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/16/2018] [Indexed: 01/15/2023] Open
Abstract
Background Surfactant Protein-A (SP-A) is a major protein component of surfactant and plays a role in surfactant-related functions and innate immunity. Human SP-A consists of two functional genes, SFTPA1 and SFTPA2, encoding SP-A1 and SP-A2 proteins, respectively and each is identified with numerous genetic variants. These differentially enhance bacterial phagocytosis, with SP-A2 variants being more effective than SP-A1. Methods Lung functions of humanized transgenic (hTG) mice that carry different SP-A1 and SP-A2 variants or both variants SP-A1/SP-A2 (6A2/1A0, co-ex), as well as SP-A knockout (KO), were studied. The animals were connected to a flexiVent system to obtain forced oscillation technique (FOT) measurements and the data were analyzed using various models. Lung function was assessed after infection (baseline) and following inhaled methacholine concentrations (0–50 mg/mL). Results Here, we investigated the role of SP-A variants on airway function after Klebsiella pneumoniae (Kp) infection (baseline) and following inhaled methacholine. We found that: 1) in the absence of methacholine no significant differences were observed between SP-A1 and SP-A2 variants and/or SP-A knockout (KO) except for sex differences in most of the parameters studied. 2) In response to methacholine, i) sex differences were observed that were reverse of those observed in the absence of methacholine; ii) SP-A2 (1A3) gene variant in males exhibited increased total and central airway resistance (Rrs and Rn) versus all other variants; iii) In females, SP-A2 (1A3) and SP-A1 (6A2) variants had similar increases in total and central airway resistance (Rrs and Rn) versus all other variants; iv) Allele-specific differences were observed, a) with SP-A2 (1A3) exhibiting significantly higher lung functions versus SP-A2 (1A0) in both sexes, except for Crs, and b) SP-A1 (6A2, 6A4) had more diverse changes in lung function in both sexes. Conclusion We conclude that, in response to infection and methacholine, SP-A variants differentially affect lung function and exhibit sex-specific differences consistent with previously reported findings of functional differences of SP-A variants. Thus, the observed changes in respiratory function mechanics provide insight into the role and importance of genetic variation of innate immune molecules, such as SP-A, on mechanical consequences of lung function after infection and inhaled substances.
Collapse
|
40
|
Echaide M, Autilio C, Arroyo R, Perez-Gil J. Restoring pulmonary surfactant membranes and films at the respiratory surface. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1725-1739. [PMID: 28341439 DOI: 10.1016/j.bbamem.2017.03.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/14/2017] [Accepted: 03/19/2017] [Indexed: 02/08/2023]
Abstract
Pulmonary surfactant is a complex of lipids and proteins assembled and secreted by the alveolar epithelium into the thin layer of fluid coating the respiratory surface of lungs. There, surfactant forms interfacial films at the air-water interface, reducing dramatically surface tension and thus stabilizing the air-exposed interface to prevent alveolar collapse along respiratory mechanics. The absence or deficiency of surfactant produces severe lung pathologies. This review describes some of the most important surfactant-related pathologies, which are a cause of high morbidity and mortality in neonates and adults. The review also updates current therapeutic approaches pursuing restoration of surfactant operative films in diseased lungs, mainly through supplementation with exogenous clinical surfactant preparations. This article is part of a Special Issue entitled: Membrane Lipid Therapy: Drugs Targeting Biomembranes edited by Pablo V. Escribá.
Collapse
Affiliation(s)
- Mercedes Echaide
- Dept. Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, Madrid, Spain
| | - Chiara Autilio
- Dept. Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, Madrid, Spain
| | - Raquel Arroyo
- Dept. Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, Madrid, Spain
| | - Jesus Perez-Gil
- Dept. Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, Madrid, Spain.
| |
Collapse
|
41
|
Surfactant protein A: A key player in lung homeostasis. Int J Biochem Cell Biol 2016; 81:151-155. [PMID: 27836807 DOI: 10.1016/j.biocel.2016.11.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/27/2016] [Accepted: 11/07/2016] [Indexed: 11/21/2022]
Abstract
The respiratory tract is continually exposed to various insults that are a permanent threat to the maintenance of lung homeostasis. Repair of the parenchyma structure, particularly of the alveolar epithelium, requires complex cellular strategies. Among the molecular components that play an important role in these processes are the surfactant proteins (SPs), particularly SP-A. The present review examines current evidence regarding the role of SP-A in lung host defence mechanisms through its implication in innate/adaptive immunity of the lung and epithelium integrity and repair. New information on SP-A deficiency in various forms of pulmonary diseases could help define therapeutic strategies aimed at restoring functional SP-A within the alveolar structure.
Collapse
|
42
|
Olmeda B, Martínez-Calle M, Pérez-Gil J. Pulmonary surfactant metabolism in the alveolar airspace: Biogenesis, extracellular conversions, recycling. Ann Anat 2016; 209:78-92. [PMID: 27773772 DOI: 10.1016/j.aanat.2016.09.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/22/2016] [Accepted: 09/25/2016] [Indexed: 01/03/2023]
Abstract
Pulmonary surfactant is a lipid-protein complex that lines and stabilizes the respiratory interface in the alveoli, allowing for gas exchange during the breathing cycle. At the same time, surfactant constitutes the first line of lung defense against pathogens. This review presents an updated view on the processes involved in biogenesis and intracellular processing of newly synthesized and recycled surfactant components, as well as on the extracellular surfactant transformations before and after the formation of the surface active film at the air-water interface. Special attention is paid to the crucial regulation of surfactant homeostasis, because its disruption is associated with several lung pathologies.
Collapse
Affiliation(s)
- Bárbara Olmeda
- Department of Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, 28040 Madrid, Spain
| | - Marta Martínez-Calle
- Department of Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, 28040 Madrid, Spain
| | - Jesus Pérez-Gil
- Department of Biochemistry, Faculty of Biology, and Research Institute "Hospital 12 de Octubre", Complutense University, 28040 Madrid, Spain.
| |
Collapse
|