1
|
McKenzie CW, Wilcox RM, Isaiah OJ, Kareta MS, Lee L. Airway ciliary microenvironment responses in mice with primary ciliary dyskinesia and central pair apparatus defects. Sci Rep 2024; 14:28437. [PMID: 39558053 PMCID: PMC11574124 DOI: 10.1038/s41598-024-79877-z] [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: 09/20/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024] Open
Abstract
Dysfunction of motile cilia can impair mucociliary clearance in the airway and result in primary ciliary dyskinesia (PCD). We previously showed that mutations in central pair apparatus (CPA) genes perturb ciliary motility and result in PCD in mouse models. However, little is known about how epithelial cell types in the ciliary microenvironment of the upper airway respond to defects in ciliary motility and mucociliary clearance. Here, we have used single-cell RNA sequencing to investigate responses in tracheal epithelial cells from mice with mutations in CPA genes Cfap221/ Pcdp1, Cfap54, and Spef2. Expected cell types were identified, along with an unidentified cell type not expressing markers of typical airway cells. Deuterosomal cells were found to exist in two states that differ largely in expression of genes involved in differentiation into ciliated cells. Functional enrichment analysis of differentially expressed genes (DEGs) revealed important cellular functions and molecular pathways for each cell type that are altered in mutant mice. Overlapping DEGs shed light on general responses to cilia dysfunction, while unique DEGs indicate that some responses may be specific to the individual mutation and ciliary defect.
Collapse
Affiliation(s)
- Casey W McKenzie
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th St. N., Sioux Falls, Sioux Falls, SD, 57104, USA
| | - Reesa M Wilcox
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th St. N., Sioux Falls, Sioux Falls, SD, 57104, USA
| | - Oduduabasi J Isaiah
- Functional Genomics and Bioinformatics Core, Sanford Research, 2301 E. 60th St. N., Sioux Falls, SD, 57104, USA
| | - Michael S Kareta
- Genetics and Genomics Group, Sanford Research, 2301 E. 60th St. N., Sioux Falls, SD, 57104, USA
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD, 57105, USA
| | - Lance Lee
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 E. 60th St. N., Sioux Falls, Sioux Falls, SD, 57104, USA.
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, 1400 W. 22nd Street, Sioux Falls, SD, 57105, USA.
| |
Collapse
|
2
|
Di YP, Kuhn JM, Mangoni ML. Lung antimicrobial proteins and peptides: from host defense to therapeutic strategies. Physiol Rev 2024; 104:1643-1677. [PMID: 39052018 PMCID: PMC11495187 DOI: 10.1152/physrev.00039.2023] [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: 12/25/2023] [Revised: 06/11/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
Representing severe morbidity and mortality globally, respiratory infections associated with chronic respiratory diseases, including complicated pneumonia, asthma, interstitial lung disease, and chronic obstructive pulmonary disease, are a major public health concern. Lung health and the prevention of pulmonary disease rely on the mechanisms of airway surface fluid secretion, mucociliary clearance, and adequate immune response to eradicate inhaled pathogens and particulate matter from the environment. The antimicrobial proteins and peptides contribute to maintaining an antimicrobial milieu in human lungs to eliminate pathogens and prevent them from causing pulmonary diseases. The predominant antimicrobial molecules of the lung environment include human α- and β-defensins and cathelicidins, among numerous other host defense molecules with antimicrobial and antibiofilm activity such as PLUNC (palate, lung, and nasal epithelium clone) family proteins, elafin, collectins, lactoferrin, lysozymes, mucins, secretory leukocyte proteinase inhibitor, surfactant proteins SP-A and SP-D, and RNases. It has been demonstrated that changes in antimicrobial molecule expression levels are associated with regulating inflammation, potentiating exacerbations, pathological changes, and modifications in chronic lung disease severity. Antimicrobial molecules also display roles in both anticancer and tumorigenic effects. Lung antimicrobial proteins and peptides are promising alternative therapeutics for treating and preventing multidrug-resistant bacterial infections and anticancer therapies.
Collapse
Affiliation(s)
- Yuanpu Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jenna Marie Kuhn
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Maria Luisa Mangoni
- Department of Biochemical Sciences, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
3
|
Iannuzo N, Welfley H, Li NC, Johnson MDL, Rojas-Quintero J, Polverino F, Guerra S, Li X, Cusanovich DA, Langlais PR, Ledford JG. CC16 drives VLA-2-dependent SPLUNC1 expression. Front Immunol 2023; 14:1277582. [PMID: 38053993 PMCID: PMC10694244 DOI: 10.3389/fimmu.2023.1277582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
Rationale CC16 (Club Cell Secretory Protein) is a protein produced by club cells and other non-ciliated epithelial cells within the lungs. CC16 has been shown to protect against the development of obstructive lung diseases and attenuate pulmonary pathogen burden. Despite recent advances in understanding CC16 effects in circulation, the biological mechanisms of CC16 in pulmonary epithelial responses have not been elucidated. Objectives We sought to determine if CC16 deficiency impairs epithelial-driven host responses and identify novel receptors expressed within the pulmonary epithelium through which CC16 imparts activity. Methods We utilized mass spectrometry and quantitative proteomics to investigate how CC16 deficiency impacts apically secreted pulmonary epithelial proteins. Mouse tracheal epithelial cells (MTECS), human nasal epithelial cells (HNECs) and mice were studied in naïve conditions and after Mp challenge. Measurements and main results We identified 8 antimicrobial proteins significantly decreased by CC16-/- MTECS, 6 of which were validated by mRNA expression in Severe Asthma Research Program (SARP) cohorts. Short Palate Lung and Nasal Epithelial Clone 1 (SPLUNC1) was the most differentially expressed protein (66-fold) and was the focus of this study. Using a combination of MTECs and HNECs, we found that CC16 enhances pulmonary epithelial-driven SPLUNC1 expression via signaling through the receptor complex Very Late Antigen-2 (VLA-2) and that rCC16 given to mice enhances pulmonary SPLUNC1 production and decreases Mycoplasma pneumoniae (Mp) burden. Likewise, rSPLUNC1 results in decreased Mp burden in mice lacking CC16 mice. The VLA-2 integrin binding site within rCC16 is necessary for induction of SPLUNC1 and the reduction in Mp burden. Conclusion Our findings demonstrate a novel role for CC16 in epithelial-driven host defense by up-regulating antimicrobials and define a novel epithelial receptor for CC16, VLA-2, through which signaling is necessary for enhanced SPLUNC1 production.
Collapse
Affiliation(s)
- Natalie Iannuzo
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Holly Welfley
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | | | | | | | | | - Stefano Guerra
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona, Tucson, AZ, United States
| | - Xingnan Li
- Department of Medicine, Division of Genetics, Genomics, and Precision Medicine, University of Arizona, Tucson, AZ, United States
| | - Darren A. Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, United States
| | - Julie G. Ledford
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| |
Collapse
|
4
|
Flores-Torres AS, Samarasinghe AE. Impact of Therapeutics on Unified Immunity During Allergic Asthma and Respiratory Infections. FRONTIERS IN ALLERGY 2022; 3:852067. [PMID: 35386652 PMCID: PMC8974821 DOI: 10.3389/falgy.2022.852067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/21/2022] [Indexed: 11/04/2022] Open
Abstract
Asthma is a common chronic respiratory disease that affects millions of people worldwide. Patients with allergic asthma, the most prevalent asthma endotype, are widely considered to possess a defective immune response against some respiratory infectious agents, including viruses, bacteria and fungi. Furthermore, respiratory pathogens are associated with asthma development and exacerbations. However, growing data suggest that the immune milieu in allergic asthma may be beneficial during certain respiratory infections. Immunomodulatory asthma treatments, although beneficial, should then be carefully prescribed to avoid misuse and overuse as they can also alter the host microbiome. In this review, we summarize and discuss recent evidence of the correlations between allergic asthma and the most significant respiratory infectious agents that have a role in asthma pathogenesis. We also discuss the implications of current asthma therapeutics beyond symptom prevention.
Collapse
Affiliation(s)
- Armando S. Flores-Torres
- Division of Pulmonology, Allergy-Immunology, and Sleep, Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States
| | - Amali E. Samarasinghe
- Division of Pulmonology, Allergy-Immunology, and Sleep, Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN, United States
| |
Collapse
|
5
|
Jaiswal AK, Yadav J, Makhija S, Sandey M, Suryawanshi A, Mitra AK, Mishra A. Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) level determines steroid-resistant airway inflammation in aging. Am J Physiol Lung Cell Mol Physiol 2022; 322:L102-L115. [PMID: 34851736 PMCID: PMC8759962 DOI: 10.1152/ajplung.00315.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 01/25/2023] Open
Abstract
Asthma and its heterogeneity change with age. Increased airspace neutrophil numbers contribute to severe steroid-resistant asthma exacerbation in the elderly, which correlates with the changes seen in adults with asthma. However, whether that resembles the same disease mechanism and pathophysiology in aged and adults is poorly understood. Here, we sought to address the underlying molecular mechanism of steroid-resistant airway inflammation development and response to corticosteroid (Dex) therapy in aged mice. To study the changes in inflammatory mechanism, we used a clinically relevant treatment model of house-dust mite (HDM)-induced allergic asthma and investigated lung adaptive immune response in adult (20-22 wk old) and aged (80-82 wk old) mice. Our result indicates an age-dependent increase in airway hyperresponsiveness (AHR), mixed granulomatous airway inflammation comprising eosinophils and neutrophils, and Th1/Th17 immune response with progressive decrease in frequencies and numbers of HDM-bearing dendritic cells (DC) accumulation in the draining lymph node (DLn) of aged mice as compared with adult mice. RNA-Seq experiments of the aged lung revealed short palate, lung, and nasal epithelial clone 1 (SPLUNC1) as one of the steroid-responsive genes, which progressively declined with age and further by HDM-induced inflammation. Moreover, we found increased glycolytic reprogramming, maturation/activation of DCs, the proliferation of OT-II cells, and Th2 cytokine secretion with recombinant SPLUNC1 (rSPLUNC1) treatment. Our results indicate a novel immunomodulatory role of SPLUNC1 regulating metabolic adaptation/maturation of DC. An age-dependent decline in the SPLUNC1 level may be involved in developing steroid-resistant airway inflammation and asthma heterogeneity.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Jyoti Yadav
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Sangeet Makhija
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Maninder Sandey
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | - Amarjit Mishra
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| |
Collapse
|
6
|
Guo Y, Gao F, Wang X, Pan Z, Wang Q, Xu S, Pan S, Li L, Zhao D, Qian J. Spontaneous formation of neutrophil extracellular traps is associated with autophagy. Sci Rep 2021; 11:24005. [PMID: 34907287 PMCID: PMC8671464 DOI: 10.1038/s41598-021-03520-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/02/2021] [Indexed: 12/17/2022] Open
Abstract
Neutrophils release neutrophil extracellular traps (NETs), via NETosis, as a defense mechanism against pathogens. Neutrophils can release NETs spontaneously; however, the mechanisms underlying spontaneous NETosis remain unclear. Neutrophils isolated from healthy donors were tested for NET formation and autophagy at 1, 6, 12, and 24 h after incubation. Autophagy response was evaluated in response to various autophagy inducers and inhibitors. The relationship between autophagy and NETosis was detected in vivo using an ovalbumin-induced mouse model of asthma. We found that the increase in the proportion of spontaneous NETosis was time-dependent. The number of autophagy-positive cells also increased over time and LC3B protein played an integral role in NET formation. Trehalose (an inducer of mTOR-independent autophagy) treatment significantly increased NET formation, whereas rapamycin (an mTOR-dependent autophagy inducer) did not increase NET release by neutrophils. Compared with the control group, 3-methyladenine (an autophagy sequestration inhibitor) and hydroxychloroquine sulfate (autophagosome-lysosome fusion inhibitor) treatments significantly reduced the percentage of NET-positive cells. In vivo studies on ovalbumin-induced asthma lung sections revealed NETs and LC3B and citH3 proteins were found to co-localize with DNA. Our findings suggest that autophagy plays a crucial role in aging-related spontaneous NETosis.
Collapse
Affiliation(s)
- Yun Guo
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Fei Gao
- Department of Intensive Care Unit, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Xin Wang
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Zhenzhen Pan
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Qian Wang
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Shiyao Xu
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Shanshan Pan
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Ling Li
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China
| | - Deyu Zhao
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China.
| | - Jun Qian
- Department of Respiratory Medicine, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
7
|
Li Z, Du Z, Li J, Sun Y. Comparative analysis on lung transcriptome of Mycoplasma ovipneumoniae (Mo) - infected Bashbay sheep and argali hybrid sheep. BMC Vet Res 2021; 17:327. [PMID: 34645427 PMCID: PMC8511284 DOI: 10.1186/s12917-021-03040-3] [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: 10/15/2020] [Accepted: 10/01/2021] [Indexed: 12/04/2022] Open
Abstract
Background Bashbay sheep (Bbs) has a certain degree of resistance to Mycoplasma ovipneumoniae (Mo), however, Argali hybrid sheep (Ahs) is susceptible to Mo. To understand the molecular mechanisms underlying the difference of the susceptibility for Mo infection, RNA-sequencing technology was used to compare the transcriptomic response of the lung tissue of Mo-infected Bbs and Ahs. Results Six Bbs and six Ahs were divided into experimental group and control group respectively, all of them were experimentally infected with Mo by intratracheal injection. For collecting lung tissue samples, three Bbs and three Ahs were sacrificed on day 4 post-infection, and the others were sacrificed on day 14 post-infection. Total RNA extracted from lung tissue were used for transcriptome analyses based on high-throughput sequencing technique and bioinformatics. The results showed that 212 (146 up-regulated, 66 down-regulated) DEGs were found when comparing transcriptomic data of Bbs and Ahs at 4th dpi, besides, 311 (158 up-regulated, 153 down-regulated) DEGs were found at 14th dpi. After GO analysis, three main GO items protein glycosylation, immune response and positive regulation of gene expression were found related to Mo infection. In addition, there were 20 DEGs enriched in these above items, such as SPLUC1 (BPIFA1), P2X7R, DQA, HO-1 and SP-A (SFTPA-1). Conclusions These selected 20 DEGs associated with Mo infection laid the foundation for further study on the underlying molecular mechanism involved in high level of resistance to Mo expressed by Bbs, meanwhile, provided deeper understandings about the development of pathogenicity and host-pathogen interactions. Supplementary Information The online version contains supplementary material available at 10.1186/s12917-021-03040-3.
Collapse
Affiliation(s)
- Zengqiang Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, 832003, China
| | - Zhihui Du
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, 832003, China
| | - Jie Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, 832003, China
| | - Yanming Sun
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang, 832003, China.
| |
Collapse
|
8
|
Khanal S, Webster M, Niu N, Zielonka J, Nunez M, Chupp G, Slade MD, Cohn L, Sauler M, Gomez JL, Tarran R, Sharma L, Dela Cruz CS, Egan M, Laguna T, Britto CJ. SPLUNC1: a novel marker of cystic fibrosis exacerbations. Eur Respir J 2021; 58:13993003.00507-2020. [PMID: 33958427 DOI: 10.1183/13993003.00507-2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/29/2021] [Indexed: 11/05/2022]
Abstract
Acute pulmonary Exacerbations (AE) are episodes of clinical worsening in cystic fibrosis (CF), often precipitated by infection. Timely detection is critical to minimise morbidity and lung function declines associated with acute inflammation during AE. Based on our previous observations that airway protein Short Palate Lung Nasal epithelium Clone 1 (SPLUNC1) is regulated by inflammatory signals, we investigated the use of SPLUNC1 fluctuations to diagnose and predict AE in CF.We enrolled CF participants from two independent cohorts to measure AE markers of inflammation in sputum and recorded clinical outcomes for a 1-year follow-up period.SPLUNC1 levels were high in healthy controls (n=9, 10.7 μg mL-1), and significantly decreased in CF participants without AE (n=30, 5.7 μg mL-1, p=0.016). SPLUNC1 levels were 71.9% lower during AE (n=14, 1.6 μg mL-1, p=0.0034) regardless of age, sex, CF-causing mutation, or microbiology findings. Cytokines Il-1β and TNFα were also increased in AE, whereas lung function did not consistently decrease. Stable CF participants with lower SPLUNC1 levels were much more likely to have an AE at 60 days (HR: 11.49, Standard Error: 0.83, p=0.0033). Low-SPLUNC1 stable participants remained at higher AE risk even one year after sputum collection (HR: 3.21, Standard Error: 0.47, p=0.0125). SPLUNC1 was downregulated by inflammatory cytokines and proteases increased in sputum during AE.In acute CF care, low SPLUNC1 levels could support a decision to increase airway clearance or to initiate pharmacological interventions. In asymptomatic, stable patients, low SPLUNC1 levels could inform changes in clinical management to improve long-term disease control and clinical outcomes in CF.
Collapse
Affiliation(s)
- Sara Khanal
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Megan Webster
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Naiqian Niu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jana Zielonka
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Myra Nunez
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Martin D Slade
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lauren Cohn
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marie Egan
- Division of Pediatric Pulmonology, Allergy, Immunology, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Theresa Laguna
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Liu Q, Wang Z, Zhang W. The Multifunctional Roles of Short Palate, Lung, and Nasal Epithelium Clone 1 in Regulating Airway Surface Liquid and Participating in Airway Host Defense. J Interferon Cytokine Res 2021; 41:139-148. [PMID: 33885339 DOI: 10.1089/jir.2020.0141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) is a kind of secretory protein, and gets expressed abundantly in normal respiratory epithelium of humans. As a natural immune molecule, SPLUNC1 is proved to be involved in inflammatory response and airway host defense. This review focuses on summarizing and discussing the role of SPLUNC1 in regulating airway surface liquid (ASL) and participating in airway host defense. PubMed and MEDLINE were used for searching and identifying the data in this review. The domain of bactericidal/permeability-increasing protein in SPLUNC1 and the α-helix, α4, are essential for SPLUNC1 to exert biological activities. As a natural innate immune molecule, SPLUNC1 plays a significant role in inflammatory response and airway host defense. Its special expression patterns are not only observed in physiological conditions, but also in some respiratory diseases. The mechanisms of SPLUNC1 in airway host defense include modulating ASL volume, acting as a surfactant protein, inhibiting biofilm formation, as well as regulating ASL compositions, such as LL-37, mucins, Neutrophil elastase, and inflammatory cytokines. Besides, potential correlations are found among these different mechanisms, especially among different ASL compositions, which should be further explored in more systematical frameworks. In this review, we summarize the structural characteristics and expression patterns of SPLUNC1 briefly, and mainly discuss the mechanisms of SPLUNC1 exerted in host defense, aiming to provide a theoretical basis and a novel target for future studies and clinical treatments.
Collapse
Affiliation(s)
- Qingluan Liu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhicheng Wang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Kalininskiy A, Kittel J, Nacca NE, Misra RS, Croft DP, McGraw MD. E-cigarette exposures, respiratory tract infections, and impaired innate immunity: a narrative review. PEDIATRIC MEDICINE (HONG KONG, CHINA) 2021; 4:5. [PMID: 34095814 PMCID: PMC8177080 DOI: 10.21037/pm-20-97] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Electronic cigarettes (e-cigarettes) are commonly used devices by adolescents and young adults. Since their introduction, the popularity of e-cigarettes has increased significantly with close to twenty percent of United States high school students reporting current use in 2020. As the number of e-cigarette users has increased, so have reports of vaping related health complications. Overall, respiratory tract infections remain one of the top ten leading causes of death in the US for every age group. Specific to the pediatric population, lower respiratory tract infections are the leading cause for hospitalization. This review highlights the current evidence behind e-cigarette exposure and its association with impaired innate immune function and the risk of lower respiratory tract infections. To date, various preclinical models have evaluated the direct effects of e-cigarette exposure on the innate immune system. More specifically, e-cigarette exposure impairs certain cell types of the innate immune system including the airway epithelium, lung macrophage and neutrophils. Identified effects of e-cigarette exposure common to the lung's innate immunity include abnormal mucus composition, reduced epithelial barrier function, impaired phagocytosis and elevated systemic markers of inflammation. These identified impairments in the lung's innate immunity have been shown to increase adhesion of certain bacteria and fungi as well as to increase virulence of common respiratory pathogens such as influenza virus, Staphylococcus aureus or Streptococcus pneumoniae. Information summarized in this review will provide guidance to healthcare providers, policy advocates and researchers for making informed decisions regarding the associated respiratory health risks of e-cigarette use in pediatric and young adults.
Collapse
Affiliation(s)
- Aleks Kalininskiy
- Department of Medicine, Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester NY, USA
| | - Julie Kittel
- Department of Public Health, University of Rochester Medical Center, Rochester NY, USA
| | - Nicholas E. Nacca
- Department of Emergency Medicine, University of Rochester Medical Center, Rochester NY, USA
| | - Ravi S. Misra
- Department of Pediatrics, Pulmonology, University of Rochester Medical Center, Rochester NY, USA
| | - Daniel P. Croft
- Department of Medicine, Pulmonary Diseases and Critical Care, University of Rochester Medical Center, Rochester NY, USA
| | - Matthew D. McGraw
- Department of Pediatrics, Pulmonology, University of Rochester Medical Center, Rochester NY, USA
| |
Collapse
|
11
|
Li J, Liu H, Zhao N, Wang J, Yang Y, Sun Y. Therapeutic effects of recombinant SPLUNC1 on Mycoplasma ovipneumoniae-infected Argali hybrid sheep. Res Vet Sci 2020; 133:174-179. [PMID: 32992128 DOI: 10.1016/j.rvsc.2020.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/31/2020] [Accepted: 09/10/2020] [Indexed: 11/26/2022]
Abstract
Clinical therapeutic and immunoregulatory effects of recombinant SPLUNC1 protein (rSPLUNC1) were evaluated in Mycoplasma ovipneumoniae (Mo)-infected Argali hybrid sheep (AHS). Group A contained six Bashibai sheep (BS) and groups B-D contained six AHS each. All sheep were manually infected with Mo. Five days post-infection, rSPLUNC1 from BS and AHS was injected intratracheally into group C and D animals; physiological saline was administered to groups A and B. Serum IL-5, IL-6, and IL-9 were quantified by ELISA. After sacrificing the sheep, lung tissues were extracted for pathological examination. The qPCR was used to quantify Mo load in the lungs and evaluate therapeutic efficacy. Serum IL-5, IL-6, and IL-9 concentrations increased during early infection stages in all groups but were significantly lower in groups A, C, and D than in group B on days 14 and 21. On day 21, IL-5 concentrations were lower in group A than in groups C and D. IL-6 concentration in groups A, C, and D was significantly lower than that in group B, and that in groups C and D was significantly lower than that in group A. Mean mycoplasma pneumonia histopathology scores were significantly lower in groups C and D than in group B, and Mo load in group C and D lung tissue decreased significantly compared to that in group B. Intratracheal injection of rSPLUNC1 into Mo-infected sheep decreased the cytokine levels and alleviated clinical symptoms with no mortality. rSPLUNC1 had significant therapeutic effects on Mo-infected AHS and can regulate pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Jie Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Haiyan Liu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Ning Zhao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Jixue Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yi Yang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yanming Sun
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| |
Collapse
|
12
|
Zhang R, Trower J, Wu T. Degradation of bacterial permeability family member A1 (BPIFA1) by house dust mite (HDM) cysteine protease Der p 1 abrogates immune modulator function. Int J Biol Macromol 2020; 164:4022-4031. [PMID: 32890564 PMCID: PMC7467078 DOI: 10.1016/j.ijbiomac.2020.08.214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/19/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023]
Abstract
Bacterial permeability family member A1 (BPIFA1) is one of the most abundant proteins present in normal airway surface liquid (ASL). It is known to be diminished in asthmatic patients' sputum, which causes airway hyperresponsiveness (AHR). What is currently unclear is how environmental factors, such as allergens' impact on BPIFA1's abundance and functions in the context of allergic asthma. House dust mite (HDM) is a predominant domestic source of aeroallergens. The group of proteases found in HDM is thought to cleave multiple cellular protective mechanisms, and therefore foster the development of allergic asthma. Here, we show that BPIFA1 is cleaved by HDM proteases in a time-, dose-, and temperature-dependent manner. We have also shown the main component in HDM that is responsible for BPIFA1's degradation is Der p1. Fragmented BPIFA1 failed to bind E. coli lipopolysaccharide (LPS), and hence elevated TNFα and IL-6 secretion in human whole blood. BPIFA1 degradation is also observed in vivo in bronchoalveolar fluid (BALF) of mice which are intranasally instilled with HDM. These data suggest that proteases associated with environmental allergens such as HDM cleave BPIFA1 and therefore impair its immune modulator function.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Ningxia Medical University, Yinchuan, PR China
| | - Jessika Trower
- Department of Pharmaceutical Sciences, 302 East Lawson Street, North Carolina Central University, Durham, NC 27707, USA
| | - Tongde Wu
- Department of Pharmaceutical Sciences, 302 East Lawson Street, North Carolina Central University, Durham, NC 27707, USA; Biomanufacturing Research Institute & Technology Enterprise (BRITE), 302 East Lawson Street, North Carolina Central University, Durham, NC 27707, USA.
| |
Collapse
|
13
|
Comprehensive RNA-Seq profiling of the lung transcriptome of Bashbay sheep in response to experimental Mycoplasma ovipneumoniae infection. PLoS One 2020; 15:e0214497. [PMID: 32639963 PMCID: PMC7343132 DOI: 10.1371/journal.pone.0214497] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 03/14/2019] [Indexed: 01/01/2023] Open
Abstract
The Bashbay sheep (Ovis aries), an indigenous breed of Xinjiang, China, has many excellent characteristics. It is resistant to Mycoplasma ovipneumoniae infection, the causative agent of mycoplasma ovipneumonia, a chronic respiratory disease that is harmful to the sheep industry. To date, knowledge regarding the mechanisms responsible for M. ovipneumoniae pathogenesis in scant. Herein, we report the results of transcriptome profiling of lung tissues from Bashbay sheep experimentally infected with an M. ovipneumoniae strain at 4 and 14 days post-infection, in comparison to mock-infected animals (0 d). Transcriptome profiling was performed by deep RNA sequencing, using the Illumina platform. The analysis of differentially expressed genes was performed to determine concomitant gene-specific temporal patterns of mRNA expression in the lungs after M. ovipneumoniae infection. We found 1048 differentially expressed genes (575 up-regulated, 473 down-regulated) when comparing transcriptomic data at 4 and 0 days post-infection, and 2823 (1362 up-regulated, 1461 down-regulated) when comparing 14 versus 0 days post-infection. Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that the differentially expressed genes at 4 and 14 versus 0 days post-infection were enriched in 245 and 287 pathways, respectively, and the Toll-like receptor (TLR) signaling pathway was considered most closely related to MO infection (p < 0.01). Two pathways (LAMP-TLR2/TLR6-MyD88-MKK6-AP1-IL1B and LAMP-TLR8MyD88-IRF5-RANTES) were identified based on the TLR signaling pathway from differentially expressed genes related M. ovipneumoniae infection. Gene Ontology analysis showed that differentially expressed genes in different groups were enriched for 1580 and 4561 terms, where those most closely related to M. ovipneumoniae infection are positive regulators of inflammatory responses (p < 0.01). These results could aid in understanding how M. ovipneumoniae infection progresses in the lungs and may provide useful information regarding key regulatory pathways.
Collapse
|
14
|
Li Z, Du Z, Sun Y, Wang J, Liu H, Yang Y, Zhao N. Comprehensive RNA-Seq profiling of the lung transcriptome of Argali hybrid sheep in response to experimental Mycoplasma ovipneumoniae infection. Res Vet Sci 2020; 132:57-68. [PMID: 32505020 DOI: 10.1016/j.rvsc.2020.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 05/17/2020] [Accepted: 05/17/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND An experiment was conducted to reveal why the Argali hybrid sheep are susceptible to Mycoplasma ovipneumoniae infection, the causative agent of mycoplasma ovipneumonia, a chronic respiratory disease that is harmful to the sheep industry. RESULTS After nine Argali hybrid sheep, divided into three groups, were experimentally infected with an M. ovipneumoniae strain at 0, 4 and 14 days, transcriptome profiling of lung tissues was performed by deep RNA sequencing, using the Illumina platform. Analysis of differentially expressed genes was performed to determine concomitant gene-specific temporal patterns of mRNA expression in the lungs after M. ovipneumoniae infection. 156 differentially expressed genes (44 up-regulated, 112 down-regulated) were found when comparing transcriptomic data at 4 and 0 days post-infection, and 367 (35 up-regulated, 332 down-regulated) when comparing 14 versus 0 days post-infection. Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that the differentially expressed genes at 4 and 14 versus 0 days post-infection were enriched in 109 and 150 pathways, respectively, and the Primary immunodeficiency pathway was considered most closely related to MO infection (p < .01). Hyper-IgM syndrome was identified based on the B-cell Immunodeficiency signaling pathway from differentially expressed genes related to M. ovipneumoniae infection. Gene Ontology analysis showed that differentially expressed genes in different groups were enriched for 497 and 928 terms, where those most closely related to M. ovipneumoniae infection are ciliated motor damage (p < .01). CONCLUSIONS The situation that ciliary movement is significantly inhibited and B cells in immunodeficiency are possibly the most important reason why Argali hybrid sheep are susceptible to MO.
Collapse
Affiliation(s)
- Zengqiang Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Zhihui Du
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yanming Sun
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Jixue Wang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Haiyan Liu
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Yi Yang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| | - Ning Zhao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China
| |
Collapse
|
15
|
Sharma L, Feng J, Britto CJ, Dela Cruz CS. Mechanisms of Epithelial Immunity Evasion by Respiratory Bacterial Pathogens. Front Immunol 2020. [PMID: 32117248 DOI: 10.3389/fimmu.2020.00091/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Bacterial lung infections are major healthcare challenges killing millions of people worldwide and resulting in a huge economic burden. Both basic and clinical research have elucidated host mechanisms that contribute to the bacterial clearance where an indispensable role of immune cells has been established. However, the role of respiratory epithelial cells in bacterial clearance has garnered limited attention due to their weak inflammatory or phagocytic ability compared to immune cells such as macrophages and neutrophils. These studies often underappreciate the fact that epithelial cells are the most abundant cells in the lung, not only serving as building blocks but also providing immune protection throughout the lung. Epithelial cells function either independently to eradicate the pathogen or communicate with immune cells to orchestrate pathogen clearance. The epithelial cells have multiple mechanisms that include mucus production, antimicrobial peptide production, muco-ciliary clearance, and phagocytosis, all of which contribute to their direct antibacterial function. Secretion of cytokines to recruit immune cells and potentiate their antimicrobial activities is a pathway by which the epithelium contributes to bacterial clearance. Successful pathogens outsmart epithelial resistance and find a way to replicate in sufficient numbers to establish infections in the airway or lung epithelial surfaces. In this mini-review, we discuss evidences that establish important roles for epithelial host defense against invading respiratory bacterial pathogens and demonstrate how pathogens outsmart these epithelial immune mechanisms to successfully establish infection. Finally, we discuss briefly how to boost epithelial immunity to improve outcomes in bacterial lung infections.
Collapse
Affiliation(s)
- Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jingjing Feng
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
16
|
Sharma L, Feng J, Britto CJ, Dela Cruz CS. Mechanisms of Epithelial Immunity Evasion by Respiratory Bacterial Pathogens. Front Immunol 2020; 11:91. [PMID: 32117248 PMCID: PMC7027138 DOI: 10.3389/fimmu.2020.00091] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/14/2020] [Indexed: 01/14/2023] Open
Abstract
Bacterial lung infections are major healthcare challenges killing millions of people worldwide and resulting in a huge economic burden. Both basic and clinical research have elucidated host mechanisms that contribute to the bacterial clearance where an indispensable role of immune cells has been established. However, the role of respiratory epithelial cells in bacterial clearance has garnered limited attention due to their weak inflammatory or phagocytic ability compared to immune cells such as macrophages and neutrophils. These studies often underappreciate the fact that epithelial cells are the most abundant cells in the lung, not only serving as building blocks but also providing immune protection throughout the lung. Epithelial cells function either independently to eradicate the pathogen or communicate with immune cells to orchestrate pathogen clearance. The epithelial cells have multiple mechanisms that include mucus production, antimicrobial peptide production, muco-ciliary clearance, and phagocytosis, all of which contribute to their direct antibacterial function. Secretion of cytokines to recruit immune cells and potentiate their antimicrobial activities is a pathway by which the epithelium contributes to bacterial clearance. Successful pathogens outsmart epithelial resistance and find a way to replicate in sufficient numbers to establish infections in the airway or lung epithelial surfaces. In this mini-review, we discuss evidences that establish important roles for epithelial host defense against invading respiratory bacterial pathogens and demonstrate how pathogens outsmart these epithelial immune mechanisms to successfully establish infection. Finally, we discuss briefly how to boost epithelial immunity to improve outcomes in bacterial lung infections.
Collapse
Affiliation(s)
- Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Jingjing Feng
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States.,Department of Respiratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
17
|
Saferali A, Tang AC, Strug LJ, Quon BS, Zlosnik J, Sandford AJ, Turvey SE. Immunomodulatory function of the cystic fibrosis modifier gene BPIFA1. PLoS One 2020; 15:e0227067. [PMID: 31931521 PMCID: PMC6957340 DOI: 10.1371/journal.pone.0227067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is characterized by a progressive decline in lung function due to airway obstruction, infection, and inflammation. CF patients are particularly susceptible to respiratory infection by a variety of pathogens, and the inflammatory response in CF is dysregulated and prolonged. BPI fold containing family A, member 1 (BPIFA1) and BPIFB1 are proteins expressed in the upper airways that may have innate immune activity. We previously identified polymorphisms in the BPIFA1/BPIFB1 region associated with CF lung disease severity. METHODS We evaluated whether the BPIFA1/BPIFB1 associations with lung disease severity replicated in individuals with CF participating in the International CF Gene Modifier Consortium (n = 6,365). Furthermore, we investigated mechanisms by which the BPIFA1 and BPIFB1 proteins may modify lung disease in CF. RESULTS The association of the G allele of rs1078761 with reduced lung function was replicated in an independent cohort of CF patients (p = 0.001, n = 2,921) and in a meta-analysis of the full consortium (p = 2.39x10-5, n = 6,365). Furthermore, we found that rs1078761G which is associated with reduced lung function was also associated with reduced BPIFA1, but not BPIFB1, protein levels in saliva from CF patients. Functional assays indicated that BPIFA1 and BPIFB1 do not have an anti-bacterial role against P. aeruginosa but may have an immunomodulatory function in CF airway epithelial cells. Gene expression profiling using RNAseq identified Rho GTPase signaling pathways to be altered in CF airway epithelial cells in response to treatment with recombinant BPIFA1 and BPIFB1 proteins. CONCLUSIONS BPIFA1 and BPIFB1 have immunomodulatory activity and genetic variation associated with low levels of these proteins may increase CF lung disease severity.
Collapse
Affiliation(s)
- Aabida Saferali
- Centre for Heart Lung Innovation, University of British Columbia and St Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anthony C. Tang
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Lisa J. Strug
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Bradley S. Quon
- Centre for Heart Lung Innovation, University of British Columbia and St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - James Zlosnik
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Andrew J. Sandford
- Centre for Heart Lung Innovation, University of British Columbia and St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
18
|
Kim DK, Wi YC, Shin SJ, Kim KR, Kim DW, Cho SH. Diverse phenotypes and endotypes of fungus balls caused by mixed bacterial colonization in chronic rhinosinusitis. Int Forum Allergy Rhinol 2019; 9:1360-1366. [PMID: 31403760 DOI: 10.1002/alr.22410] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND The pathogenic roles of fungus and bacteria in chronic rhinosinusitis (CRS) remain unclear. Recently, we described the bacterial ball, which is distinct from the fungus ball, as an unusual phenotype of bacterial infection. In this study, we investigated the clinical, histopathologic, and immunologic characteristics of sinonasal microorganic materials, including fungus ball and bacterial ball. METHODS In this study, we enrolled 80 CRS patients with sinonasal microorganic materials and 10 control subjects who underwent skull base surgery or endoscopic dacryocystorhinostomy and had no signs or symptoms of nasal inflammation. All specimens were stained with hematoxylin-eosin, Gomori-methenamine-silver, and Gram stain to identify fungal organisms and Gram-positive/negative bacterial colonies. The expression of tumor necrosis factor (TNF)-α; interleukin (IL)-1β; S100A7; S100A8/A9; and short, palate, lung, and nasal epithelial clone 1 (SPLUNC1) were evaluated by enzyme-linked immunosorbent assay using sinus lavage fluid. RESULTS We histologically classified sinonasal microorganic materials into the following 4 groups: fungus ball (n = 45); bacterial ball (n = 6); mixed ball (formed by a mixture of fungus and bacteria, n = 27); and double ball (formed by separate fungal and bacterial balls, n = 2). Compared with the fungus ball, the mixed ball was more frequently detected in immunocompromised patients (p < 0.0001). In addition, TNF-α expression was significantly higher in fungus and mixed balls than in control, whereas the mixed ball showed higher expression of IL-1β compared with the fungus ball. Moreover, the expression of S100A7 and S100A8/A9 protein in the mixed ball was significantly decreased when compared with the fungus ball, whereas there was no significant difference in SPLUNC1 expression between fungus and mixed balls. CONCLUSION Our findings suggest that fungal and bacterial interactions are diverse in CRS. Specifically, the mixed ball is prevalent in CRS with an immunocompromised state and it may decrease epithelial barrier function.
Collapse
Affiliation(s)
- Dong-Kyu Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Young Chan Wi
- Department of Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Su-Jin Shin
- Department of Pathology, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Kyung Rae Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Hanyang University College of Medicine, Seoul, Republic of Korea
| | - Dae Woo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seok Hyun Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Hanyang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
Jones-Nelson O, Hilliard JJ, DiGiandomenico A, Warrener P, Alfaro A, Cheng L, Stover CK, Cohen TS, Sellman BR. The Neutrophilic Response to Pseudomonas Damages the Airway Barrier, Promoting Infection by Klebsiella pneumoniae. Am J Respir Cell Mol Biol 2019; 59:745-756. [PMID: 30109945 DOI: 10.1165/rcmb.2018-0107oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pseudomonas aeruginosa and Klebsiella pneumoniae are two common gram-negative pathogens that are associated with bacterial pneumonia and can often be isolated from the same patient. We used a mixed-pathogen pneumonia infection model in which mice were infected with sublethal concentrations of P. aeruginosa and K. pneumoniae, resulting in significant lethality, outgrowth of both bacteria in the lung, and systemic dissemination of K. pneumoniae. Inflammation, induced by P. aeruginosa activation of Toll-like receptor 5, results in prolonged neutrophil recruitment to the lung and increased levels of neutrophil elastase in the airway, resulting in lung damage and epithelial barrier dysfunction. Live P. aeruginosa was not required to potentiate K. pneumoniae infection, and flagellin alone was sufficient to induce lethality when delivered along with Klebsiella. Prophylaxis with an anti-Toll-like receptor 5 antibody or Sivelestat, a neutrophil elastase inhibitor, reduced neutrophil influx, inflammation, and mortality. Furthermore, pathogen-specific monoclonal antibodies targeting P. aeruginosa or K. pneumoniae prevented the outgrowth of both bacteria and reduced host inflammation and lethality. These findings suggest that coinfection with P. aeruginosa may enable the outgrowth and dissemination of K. pneumoniae, and that a pathogen- or host-specific prophylactic approach targeting P. aeruginosa may prevent or limit the severity of such infections by reducing neutrophil-induced lung damage.
Collapse
Affiliation(s)
| | | | | | | | - Alex Alfaro
- 2 Department of Laboratory Animal Research, and
| | - Lily Cheng
- 3 Department of Translational Science, MedImmune, LLC, Gaithersburg, Maryland
| | | | | | | |
Collapse
|
20
|
Interleukin 1 Receptor-Like 1 (IL1RL1) Promotes Airway Bacterial and Viral Infection and Inflammation. Infect Immun 2019; 87:IAI.00340-19. [PMID: 31061143 DOI: 10.1128/iai.00340-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 01/14/2023] Open
Abstract
Interleukin 1 receptor-like 1 (IL1RL1), also known as suppression of tumorigenicity 2 (ST2), is the receptor for interleukin 33 (IL-33) and has been increasingly studied in type 2 inflammation. An increase in airway IL-33/ST2 signaling in asthma has been associated with eosinophilic inflammation, but little is known about the role of ST2 in neutrophilic inflammation. Airway Mycoplasma pneumoniae and human rhinovirus (HRV) infections are linked to neutrophilic inflammation during acute exacerbations of asthma. However, whether ST2 contributes to M. pneumoniae- and HRV-mediated airway inflammation is poorly understood. The current study sought to determine the functions of ST2 during airway M. pneumoniae or HRV infection. In cultured normal human primary airway epithelial cells, ST2 overexpression (OE) increased the production of neutrophilic chemoattractant IL-8 in the absence or presence of M. pneumoniae or HRV1B infection. ST2 OE also enhanced HRV1B-induced IP-10, a chemokine involved in asthma exacerbations. In the M. pneumoniae-infected mouse model, ST2 deficiency, in contrast to sufficiency, significantly reduced the levels of neutrophils following acute (≤24 h) infection, while in the HRV1B-infected mouse model, ST2 deficiency significantly reduced the levels of proinflammatory cytokines KC, IP-10, and IL-33 in bronchoalveolar lavage (BAL) fluid. Overall, ST2 overexpression in human epithelial cells and ST2 sufficiency in mice increased the M. pneumoniae and HRV loads in cell supernatants and BAL fluid. After pathogen infection, ST2-deficient mice showed a higher level of the host defense protein lactotransferrin in BAL fluid. Our data suggest that ST2 promotes proinflammatory responses (e.g., neutrophils) to airway bacterial and viral infection and that blocking ST2 signaling may broadly attenuate airway infection and inflammation.
Collapse
|
21
|
Abstract
The airways are under continuous assault from aerosolized bacteria and oral flora. The bacteria present in the airways and gastrointestinal tract of neonates promote immune maturation and protect against asthma pathogenesis. Later bacterial infections and perturbations to the microbiome can contribute to asthma pathogenesis, persistence, and severity.
Collapse
Affiliation(s)
- Michael Insel
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of Arizona Health Sciences, University of Arizona College of Medicine - Tucson, 1501 North Campbell Avenue, PO Box 245017, Tucson, AZ 85724, USA
| | - Monica Kraft
- Department of Medicine, College of Medicine Tucson, Asthma and Airway Disease Research Center, University of Arizona Health Sciences, University of Arizona College of Medicine - Tucson, 1501 North Campbell Avenue, PO Box 245017, Tucson, AZ 85724, USA.
| |
Collapse
|
22
|
Schaefer N, Li X, Seibold MA, Jarjour NN, Denlinger LC, Castro M, Coverstone AM, Teague WG, Boomer J, Bleecker ER, Meyers DA, Moore WC, Hawkins GA, Fahy J, Phillips BR, Mauger DT, Dakhama A, Gellatly S, Pavelka N, Berman R, Di YP, Wenzel SE, Chu HW. The effect of BPIFA1/SPLUNC1 genetic variation on its expression and function in asthmatic airway epithelium. JCI Insight 2019; 4:127237. [PMID: 30996135 DOI: 10.1172/jci.insight.127237] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/14/2019] [Indexed: 11/17/2022] Open
Abstract
Bacterial permeability family member A1 (BPIFA1), also known as short palate, lung, and nasal epithelium clone 1 (SPLUNC1), is a protein involved in the antiinflammatory response. The goal of this study was to determine whether BPIFA1 expression in asthmatic airways is regulated by genetic variations, altering epithelial responses to type 2 cytokines (e.g., IL-13). Nasal epithelial cells from patients with mild to severe asthma were collected from the National Heart, Lung, and Blood Institute Severe Asthma Research Program centers, genotyped for rs750064, and measured for BPIFA1. To determine the function of rs750064, cells were cultured at air-liquid interface and treated with IL-13 with or without recombinant human BPIFA1 (rhBPIFA1). Noncultured nasal cells with the rs750064 CC genotype had significantly less BPIFA1 mRNA expression than the CT and TT genotypes. Cultured CC versus CT and TT cells without stimulation maintained less BPIFA1 expression. With IL-13 treatment, CC genotype cells secreted more eotaxin-3 than CT and TT genotype cells. Also, rhBPIFA1 reduced IL-13-mediated eotaxin-3. BPIFA1 mRNA levels negatively correlated with serum IgE and fractional exhaled nitric oxide. Baseline FEV1% levels were lower in the asthma patients with the CC genotype (n = 1,016). Our data suggest that less BPIFA1 in asthma patients with the CC allele may predispose them to greater eosinophilic inflammation, which could be attenuated by rhBPIFA1 protein therapy.
Collapse
Affiliation(s)
| | - Xingnan Li
- University of Arizona, Tucson, Arizona, USA
| | | | | | | | - Mario Castro
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | - Jonathan Boomer
- Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | - Wendy C Moore
- Wake Forest University, Winston-Salem, North Carolina, USA
| | | | - John Fahy
- UCSF, San Francisco, California, USA
| | | | - David T Mauger
- Pennsylvania State University, Centre County, Pennsylvania, USA
| | | | | | | | | | - Y Peter Di
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
23
|
Antibacterial Properties and Efficacy of a Novel SPLUNC1-Derived Antimicrobial Peptide, α4-Short, in a Murine Model of Respiratory Infection. mBio 2019; 10:mBio.00226-19. [PMID: 30967458 PMCID: PMC6456746 DOI: 10.1128/mbio.00226-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The rise of superbugs underscores the urgent need for novel antimicrobial agents. Antimicrobial peptides (AMPs) have the ability to kill superbugs regardless of resistance to traditional antibiotics. However, AMPs often display a lack of efficacy in vivo. Sequence optimization and engineering are promising but may result in increased host toxicity. We report here the optimization of a novel AMP (α4-short) derived from the multifunctional respiratory protein SPLUNC1. The AMP α4-short demonstrated broad-spectrum activity against superbugs as well as in vivo efficacy in the P. aeruginosa pneumonia model. Further exploration for clinical development is warranted. Multidrug resistance (MDR) by bacterial pathogens constitutes a global health crisis, and resistance to treatment displayed by biofilm-associated infections (e.g., cystic fibrosis, surgical sites, and medical implants) only exacerbates a problem that is already difficult to overcome. Antimicrobial peptides (AMPs) are a promising class of therapeutics that may be useful in the battle against antibiotic resistance, although certain limitations have hindered their clinical development. The goal of this study was to examine the therapeutic potential of novel AMPs derived from the multifunctional respiratory host defense protein SPLUNC1. Using standard growth inhibition and antibiofilm assays, we demonstrated that a novel structurally optimized AMP, α4-short, was highly effective against the most common group of MDR bacteria while showing broad-spectrum bactericidal and antibiofilm activities. With negligible hemolysis and toxicity to white blood cells, the new peptide also demonstrated in vivo efficacy when delivered directly into the airway in a murine model of Pseudomonas aeruginosa-induced respiratory infection. The data warrant further exploration of SPLUNC1-derived AMPs with optimized structures to assess the potential application to difficult-to-cure biofilm-associated infections.
Collapse
|
24
|
Marcinko MC, Darrow AL, Tuia AJ, Shohet RV. Sex influences susceptibility to methamphetamine cardiomyopathy in mice. Physiol Rep 2019; 7:e14036. [PMID: 30891941 PMCID: PMC6424857 DOI: 10.14814/phy2.14036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/22/2022] Open
Abstract
In this study, we created a mouse model of methamphetamine cardiomyopathy that reproduces the chronic, progressive dosing commonly encountered in addicted subjects. We gradually increased the quantity of methamphetamine given to C57Bl/6 mice from 5 to 40 mg/kg over 2 or 5 months during two study periods. At the fifth month, heart weight was increased, echocardiograms showed a dilated cardiomyopathy and survival was lower in males, with less effect in females. Interestingly, these findings correspond to previous observations in human patients, suggesting greater male susceptibility to the effects of methamphetamine on the heart. Transcriptional analysis showed changes in genes dysregulated in previous methamphetamine neurological studies as well as many that likely play a role in cardiac response to this toxic stress. We expect that a deeper understanding of the molecular biology of methamphetamine exposure in the heart will provide insights into the mechanism of cardiomyopathy in addicts and potential routes to more effective treatment.
Collapse
MESH Headings
- Amphetamine-Related Disorders/complications
- Amphetamine-Related Disorders/genetics
- Amphetamine-Related Disorders/metabolism
- Animals
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/physiopathology
- Disease Models, Animal
- Female
- Male
- Methamphetamine
- Mice, Inbred C57BL
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Risk Assessment
- Risk Factors
- Sex Factors
- Time Factors
- Transcription, Genetic
Collapse
Affiliation(s)
- Marie C. Marcinko
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| | - April L. Darrow
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| | - Aaron J. Tuia
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| | - Ralph V. Shohet
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| |
Collapse
|
25
|
Britto CJ, Niu N, Khanal S, Huleihel L, Herazo-Maya JD, Thompson A, Sauler M, Slade MD, Sharma L, Dela Cruz CS, Kaminski N, Cohn LE. BPIFA1 regulates lung neutrophil recruitment and interferon signaling during acute inflammation. Am J Physiol Lung Cell Mol Physiol 2018; 316:L321-L333. [PMID: 30461288 DOI: 10.1152/ajplung.00056.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bpifa1 (BPI fold-containing group A member 1) is an airway host-protective protein with immunomodulatory properties that binds to LPS and is regulated by infectious and inflammatory signals. Differential expression of Bpifa1 has been widely reported in lung disease, yet the biological significance of this observation is unclear. We sought to understand the role of Bpifa1 fluctuations in modulating lung inflammation. We treated wild-type (WT) and Bpifa1-/- mice with intranasal LPS and performed immunological and transcriptomic analyses of lung tissue to determine the immune effects of Bpifa1 deficiency. We show that neutrophil (polymorphonuclear cells, PMNs) lung recruitment and transmigration to the airways in response to LPS is impaired in Bpifa1-/- mice. Transcriptomic analysis revealed a signature of 379 genes that differentiated Bpifa1-/- from WT mice. During acute lung inflammation, the most downregulated genes in Bpifa1-/- mice were Cxcl9 and Cxcl10. Bpifa1-/- mice had lower bronchoalveolar lavage concentrations of C-X-C motif chemokine ligand 10 (Cxcl10) and Cxcl9, interferon-inducible PMN chemokines. This was consistent with lower expression of IFNγ, IFNλ, downstream IFN-stimulated genes, and IFN-regulatory factors, which are important for the innate immune response. Administration of Cxcl10 before LPS treatment restored the inflammatory response in Bpifa1-/- mice. Our results identify a novel role for Bpifa1 in the regulation of Cxcl10-mediated PMN recruitment to the lungs via IFNγ and -λ signaling during acute inflammation.
Collapse
Affiliation(s)
- Clemente J Britto
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Naiqian Niu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Sara Khanal
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Luai Huleihel
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Jose D Herazo-Maya
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Alison Thompson
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Martin D Slade
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut.,Yale University School of Public Health, Department of Environmental Health Sciences , New Haven, Connecticut
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Lauren E Cohn
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
26
|
Yu Z, Deslouches B, Walton WG, Redinbo MR, Di YP. Enhanced biofilm prevention activity of a SPLUNC1-derived antimicrobial peptide against Staphylococcus aureus. PLoS One 2018; 13:e0203621. [PMID: 30216370 PMCID: PMC6138395 DOI: 10.1371/journal.pone.0203621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/23/2018] [Indexed: 11/18/2022] Open
Abstract
SPLUNC1 is a multifunctional protein of the airway with antimicrobial properties. We previously reported that it displayed antibiofilm activities against P. aeruginosa. The goal of this study was to determine whether (1) the antibiofilm property is broad (including S. aureus, another prevalent organism in cystic fibrosis); (2) the α4 region is responsible for such activity; and (3), if so, this motif could be structurally optimized as an antimicrobial peptide with enhanced activities. We used S. aureus biofilm-prevention assays to determine bacterial biomass in the presence of SPLUNC1 and SPLUNC1Δα4 recombinant proteins, or SPLUNC1-derived peptides (α4 and α4M1), using the well-established crystal-violet biofilm detection assay. The SPLUNC1Δα4 showed markedly reduced biofilm prevention compared to the parent protein. Surprisingly, the 30-residue long α4 motif alone demonstrated minimal biofilm prevention activities. However, structural optimization of the α4 motif resulted in a modified peptide (α4M1) with significantly enhanced antibiofilm properties against methicillin–sensitive (MSSA) and–resistant (MRSA) S. aureus, including six different clinical strains of MRSA and the well-known USA300. Hemolytic activity was undetectable at up to 100μM for the peptides. The data warrant further investigation of α4-derived AMPs to explore the potential application of antimicrobial peptides to combat bacterial biofilm-related infections.
Collapse
Affiliation(s)
- Zhongjie Yu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Molecular Genetics, Institute for Translational Medicine, Qingdao University, Qingdao, China
| | - Berthony Deslouches
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - William G. Walton
- Departments of Chemistry, Biochemistry, and Microbiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Matthew R. Redinbo
- Departments of Chemistry, Biochemistry, and Microbiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Y. Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
27
|
The Role of BPIFA1 in Upper Airway Microbial Infections and Correlated Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2021890. [PMID: 30255091 PMCID: PMC6140130 DOI: 10.1155/2018/2021890] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/04/2018] [Accepted: 08/07/2018] [Indexed: 12/16/2022]
Abstract
The mucosa is part of the first line of immune defense against pathogen exposure in humans and prevents viral and bacterial infection of the soft palate, lungs, uvula, and nasal cavity that comprise the ear-nose-throat (ENT) region. Bactericidal/permeability-increasing fold containing family A, member 1 (BPIFA1) is a secretory protein found in human upper aerodigestive tract mucosa. This innate material is secreted in mucosal fluid or found in submucosal tissue in the human soft palate, lung, uvula, and nasal cavity. BPIFA1 is a critical component of the innate immune response that prevents upper airway diseases. This review will provide a brief introduction of the roles of BPIFA1 in the upper airway (with a focus on the nasal cavity, sinus, and middle ear), specifically its history, identification, distribution in various human tissues, function, and diagnostic value in various upper airway infectious diseases.
Collapse
|
28
|
Huang SJ, Xu YM, Lau ATY. Electronic cigarette: A recent update of its toxic effects on humans. J Cell Physiol 2018; 233:4466-4478. [PMID: 29215738 DOI: 10.1002/jcp.26352] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 11/29/2017] [Indexed: 02/05/2023]
Abstract
Electronic cigarettes (e-cigarettes), battery-powered and liquid-vaporizing devices, were invented to replace the conventional cigarette (c-cigarette) smoking for the sake of reducing the adverse effects on multiple organ systems that c-cigarettes have induced. Although some of the identified harmful components in e-cigarettes were alleged to be measured in lower quantity than those in c-cigarettes, researchers unveiled that the toxic effects of e-cigarettes should not be understated. This review is sought for an attempt to throw light on several typical types of e-cigarette components (tobacco-specific nitrosamines, carbonyl compounds, and volatile organic compounds) by revealing their possible impacts on human bodies through different action mechanisms characterized by alteration of specific biomarkers on cellular and molecular levels. In addition, this review is intended to draw the limelight that like c-cigarettes, e-cigarettes could also be accompanied with toxic effects on whole human body, which are especially apparent on respiratory system. From head to foot, from physical aspect to chemical aspect, from genotype to phenotype, potential alterations will take place upon the intake of the liquid aerosol.
Collapse
Affiliation(s)
- Shu-Jie Huang
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Andy T Y Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
29
|
Wang H, Jiang D, Li W, Wang S. Increased expression of BPI fold-containing family A member 1 is associated with metastasis and poor prognosis in human colorectal carcinoma. Oncol Lett 2017; 14:4231-4236. [PMID: 28943932 DOI: 10.3892/ol.2017.6662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/26/2017] [Indexed: 12/13/2022] Open
Abstract
Bactericidal or permeability-increasing protein fold-containing family A member 1 (BPIFA1) has been demonstrated to be involved in inflammatory responses in the upper airway and the progression of non-small cell lung cancer. However, the expression levels of BPIFA1 and its clinical prognostic significance in colorectal carcinoma (CRC) has not yet been elucidated. Reverse transcription-polymerase chain reaction and immunohistochemistry were used to analyze the expression levels of BPIFA1 in CRC and normal mucosal tissues. The associations between BPIFA1 expression levels and clinicopathological characteristics, and its predictive value for prognosis in CRC, were statistically evaluated as appropriate. The expression levels of BPIFA1 were revealed to be upregulated at the transcriptional and translational levels in CRC tissues, compared with in normal mucosal tissues. A high expression level of BPIFA1 is significantly associated with invasion depth (P=0.040), lymph node metastasis (P=0.035) and distant metastasis (P=0.010). Furthermore, Kaplan-Meier analysis indicated that BIPFA1 overexpression is associated with short survival time, and the Cox proportional hazards model of risk analysis indicated that BPIFA1 is an independent prognostic factor for patients with CRC. The results of the present study suggested that BPIFA1 expression is upregulated in CRC tissues, and that an increased expression level of BPIFA1 is associated with tumor invasion, metastasis and poor prognosis, indicating that BPIFA1 may be a potential clinical prognostic predictor and therapeutic target for patients with CRC.
Collapse
Affiliation(s)
- Huanan Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dongmei Jiang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wenlu Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shuang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
30
|
Waites KB, Xiao L, Liu Y, Balish MF, Atkinson TP. Mycoplasma pneumoniae from the Respiratory Tract and Beyond. Clin Microbiol Rev 2017; 30:747-809. [PMID: 28539503 PMCID: PMC5475226 DOI: 10.1128/cmr.00114-16] [Citation(s) in RCA: 470] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Mycoplasma pneumoniae is an important cause of respiratory tract infections in children as well as adults that can range in severity from mild to life-threatening. Over the past several years there has been much new information published concerning infections caused by this organism. New molecular-based tests for M. pneumoniae detection are now commercially available in the United States, and advances in molecular typing systems have enhanced understanding of the epidemiology of infections. More strains have had their entire genome sequences published, providing additional insights into pathogenic mechanisms. Clinically significant acquired macrolide resistance has emerged worldwide and is now complicating treatment. In vitro susceptibility testing methods have been standardized, and several new drugs that may be effective against this organism are undergoing development. This review focuses on the many new developments that have occurred over the past several years that enhance our understanding of this microbe, which is among the smallest bacterial pathogens but one of great clinical importance.
Collapse
Affiliation(s)
- Ken B Waites
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Li Xiao
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yang Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China, and Key Laboratory of Clinical Pharmacology of Antibiotics, Ministry of Health, Shanghai, China
| | | | - T Prescott Atkinson
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
31
|
Jiang D, Matsuda J, Berman R, Schaefer N, Stevenson C, Gross J, Zhang B, Sanchez A, Li L, Chu HW. A novel mouse model of conditional IRAK-M deficiency in myeloid cells: application in lung Pseudomonas aeruginosa infection. Innate Immun 2017; 23:206-215. [PMID: 28120642 DOI: 10.1177/1753425916684202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Myeloid cells such as macrophages are critical to innate defense against infection. IL-1 receptor-associated kinase M (IRAK-M) is a negative regulator of TLR signaling during bacterial infection, but the role of myeloid cell IRAK-M in bacterial infection is unclear. Our goal was to generate a novel conditional knockout mouse model to define the role of myeloid cell IRAK-M during bacterial infection. Myeloid cell-specific IRAK-M knockout mice were generated by crossing IRAK-M floxed mice with LysM-Cre knock-in mice. The resulting LysM-Cre+/IRAK-Mfl/wt and control (LysM-Cre-/IRAK-Mfl/wt) mice were intranasally infected with Pseudomonas aeruginosa (PA). IRAK-M deletion, inflammation, myeloperoxidase (MPO) activity and PA load were measured in leukocytes, bronchoalveolar lavage (BAL) fluid and lungs. PA killing assay with BAL fluid was performed to determine mechanisms of IRAK-M-mediated host defense. IRAK-M mRNA and protein levels in alveolar and lung macrophages were significantly reduced in LysM-Cre+/IRAK-Mfl/wt mice compared with control mice. Following PA infection, LysM-Cre+/IRAK-Mfl/wt mice have enhanced lung neutrophilic inflammation, including MPO activity, but reduced PA load. The increased lung MPO activity in LysM-Cre+/IRAK-Mfl/wt mouse BAL fluid reduced PA load. Generation of IRAK-M conditional knockout mice will enable investigators to determine precisely the function of IRAK-M in myeloid cells and other types of cells during infection and inflammation.
Collapse
Affiliation(s)
- Di Jiang
- 1 Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Jennifer Matsuda
- 2 Office of Academic Affairs and Department of Biomedical Research, National Jewish Health, Denver, CO, USA
| | - Reena Berman
- 1 Department of Medicine, National Jewish Health, Denver, CO, USA
| | | | - Connor Stevenson
- 1 Department of Medicine, National Jewish Health, Denver, CO, USA
| | - James Gross
- 2 Office of Academic Affairs and Department of Biomedical Research, National Jewish Health, Denver, CO, USA
| | - Bicheng Zhang
- 2 Office of Academic Affairs and Department of Biomedical Research, National Jewish Health, Denver, CO, USA
| | - Amelia Sanchez
- 1 Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Liwu Li
- 3 Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Hong Wei Chu
- 1 Department of Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|
32
|
Shang YP, Lin L, Li CC. [Streptococcus pneumoniae induces SPLUNC1 and the regulatory effects of resveratrol]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2017; 19:111-116. [PMID: 28100333 PMCID: PMC7390127 DOI: 10.7499/j.issn.1008-8830.2017.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 11/24/2016] [Indexed: 06/06/2023]
Abstract
OBJECTIVE To investigate the host-defense role of short palate, lung, and nasal epithelium clone 1 (SPLUNC1) in Streptococcus pneumoniae (SP) infection and the effect of resveratrol (Res) on SPLUNC1 expression, and to provide new thoughts for the treatment of diseases caused by SP infection. METHODS According to the multiplicity of infection (MOI), BEAS-2B cells with SP infection were divided into control group, MOI20 SP group, and MOI50 SP group. According to the different concentrations of Res, the BEAS-2B cells with MOI20 SP infection pretreated by Res were divided into 12.5Res+SP group, 25Res+SP group, and 50Res+SP group (the final concentrations of Res were 12.5, 25, and 50 μmol/L, respectively). Cell Counting Kit-8 was used to measure cell activity and determine the optimal concentration and action time of SP and Res. In the formal experiment, the cells were divided into control group, Res group, SP group, and Res+SP group. Real-time PCR and ELISA were used to measure the mRNA and protein expression of SPLUNC1. RESULTS Over the time of SP infection, cell activity tended to decrease. Compared with the control group and the MOI20 SP group, the MOI50 SP group had a reduction in cell activity. Compared with the MOI20 SP group, the 25Res+SP group had increased cell activity and the 50Res+SP group had reduced cell activity (P<0.05). MOI20 SP bacterial suspension and 25 μmol/L Res were used for the formal experiment. Over the time of SP infection, the mRNA expression of SPLUNC1 in BEAS-2B cells firstly increased and then decreased in the SP group and the Res+SP group (P<0.05). Compared with the SP group, the Res+SP group had significant increases in the mRNA and protein expression of SPLUNC1 at all time points (P<0.05). Compared with the control group, the Res group had no significant changes in the mRNA and protein expression of SPLUNC1 (P>0.05). CONCLUSIONS SP infection can induce SPLUNC1 expression and the host-defense role of SPLUNC1. Res can upregulate SPLUNC1 expression during the development of infection and enhance cell protection in a concentration- and time-dependent manner.
Collapse
Affiliation(s)
- Yan-Ping Shang
- Department of Pediatric Pulmonology, Second Affiliated Hospital/Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | | | | |
Collapse
|
33
|
Yamamoto T, Kida Y, Sakamoto Y, Kuwano K. Mpn491, a secreted nuclease ofMycoplasma pneumoniae, plays a critical role in evading killing by neutrophil extracellular traps. Cell Microbiol 2016; 19. [DOI: 10.1111/cmi.12666] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Takeshi Yamamoto
- Division of Microbiology, Department of Infectious Medicine; Kurume University School of Medicine; Kurume 830-0011 Japan
| | - Yutaka Kida
- Division of Microbiology, Department of Infectious Medicine; Kurume University School of Medicine; Kurume 830-0011 Japan
| | - Yuichi Sakamoto
- Division of Microbiology, Department of Infectious Medicine; Kurume University School of Medicine; Kurume 830-0011 Japan
| | - Koichi Kuwano
- Division of Microbiology, Department of Infectious Medicine; Kurume University School of Medicine; Kurume 830-0011 Japan
| |
Collapse
|
34
|
Short Palate, Lung, and Nasal Epithelial Clone 1 Has Antimicrobial and Antibiofilm Activities against the Burkholderia cepacia Complex. Antimicrob Agents Chemother 2016; 60:6003-12. [PMID: 27458217 DOI: 10.1128/aac.00975-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/17/2016] [Indexed: 02/07/2023] Open
Abstract
The opportunistic bacteria of the Burkholderia cepacia complex (Bcc) are extremely pathogenic to cystic fibrosis (CF) patients, and acquisition of Bcc bacteria is associated with a significant increase in mortality. Treatment of Bcc infections is difficult because the bacteria are multidrug resistant and able to survive in biofilms. Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) is an innate defense protein that is secreted by the upper airways and pharynx. While SPLUNC1 is known to have antimicrobial functions, its effects on Bcc strains are unclear. We therefore tested the hypothesis that SPLUNC1 is able to impair Bcc growth and biofilm formation. We found that SPLUNC1 exerted bacteriostatic effects against several Bcc clinical isolates, including B. cenocepacia strain J2315 (50% inhibitory concentration [IC50] = 0.28 μM), and reduced biofilm formation and attachment (IC50 = 0.11 μM). We then determined which domains of SPLUNC1 are responsible for its antimicrobial activity. Deletions of SPLUNC1's N terminus and α6 helix did not affect its function. However, deletion of the α4 helix attenuated antimicrobial activity, while the corresponding α4 peptide displayed antimicrobial activity. Chronic neutrophilia is a hallmark of CF lung disease, and neutrophil elastase (NE) cleaves SPLUNC1. However, we found that the ability of SPLUNC1 to disrupt biofilm formation was significantly potentiated by NE pretreatment. While the impact of CF on SPLUNC1-Bcc interactions is not currently known, our data suggest that understanding this interaction may have important implications for CF lung disease.
Collapse
|
35
|
Shen W, Chen K, Sun Y, Guo H, Chen D, Cao Y. Cloning and sequence analysis of Wild Argali short palate, lung and nasal epithelium clone 1 cDNA. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 30:736-742. [PMID: 27620892 PMCID: PMC5411834 DOI: 10.5713/ajas.15.0557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 11/02/2015] [Accepted: 09/10/2016] [Indexed: 11/27/2022]
|
36
|
LeMessurier KS, Lin Y, McCullers JA, Samarasinghe AE. Antimicrobial peptides alter early immune response to influenza A virus infection in C57BL/6 mice. Antiviral Res 2016; 133:208-17. [PMID: 27531368 DOI: 10.1016/j.antiviral.2016.08.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/12/2016] [Indexed: 02/07/2023]
Abstract
Influenza is a disease of the respiratory system caused by single stranded RNA viruses with varying genotypes. Immunopathogenesis to influenza viruses differs based on virus strain, dose, and mouse strain used in laboratory models. Although effective mucosal immune defenses are important in early host defense against influenza, information on the kinetics of these immune defense mechanisms during the course of influenza infection is limited. We investigated changes to antimicrobial peptides and primary innate immune cells at early time points after infection and compared these variables between two prominent H1N1 influenza A virus (IAV) strains, A/CA/04/2009 and A/PR/08/1934 in C57BL/6 mice. Alveolar and parenchymal macrophage ratios were altered after IAV infection and pro-inflammatory cytokine production in macrophages was induced after IAV infection. Genes encoding antimicrobial peptides, β-defensin (Defb4), bactericidal-permeability increasing protein (Bpifa1), and cathelicidin antimicrobial peptide (Camp), were differentially regulated after IAV infection and the kinetics of Defb4 expression differed in response to each virus strain. Beta-defensin reduced infectivity of A/CA/04/2009 virus but not A/PR/08/1934. Beta defensins also changed the innate immune cell profile wherein mice pre-treated with β-defensin had increased alveolar macrophages and CD103(+) dendritic cells, and reduced CD11b(+) dendritic cells and neutrophils. In addition to highlighting that immune responses may vary based on influenza virus strain used, our data suggest an important role for antimicrobial peptides in host defense against influenza virus.
Collapse
Affiliation(s)
- Kim S LeMessurier
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Yanyan Lin
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Jonathan A McCullers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Amali E Samarasinghe
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
37
|
Liu H, Zhang X, Wu J, French SW, He Z. New insights on the palate, lung, and nasal epithelium clone (PLUNC) proteins: Based on molecular and functional analysis of its homolog of YH1/SPLUNC1. Exp Mol Pathol 2016; 100:363-369. [PMID: 26654795 DOI: 10.1016/j.yexmp.2015.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 12/02/2015] [Indexed: 12/12/2022]
Abstract
The palate, lung, and nasal epithelium clone (PLUNC) proteins are intricate immune molecules and arisen questions from them are still unresolved. In order to identify the role of PLUNC family proteins, we had analyzed its homolog protein YH1/SPLUNC1, which highly expresses in nontumor nasopharyngeal epithelium while expresses weakly in nasopharyngeal carcinoma (NPC) tissues. It is found that YH1/SPLUNC1 protein expression level was higher in chronic normal nasopharynx inflammatory cells compared to NPC tissue cells. An approach to produce active YH1/SPLUNC1 protein had been established and recombinant YH1/SPLUNC1 protein could bind to all four Gram-positive and four Gram-negative bacteria we tested, and triggered the aggregation of those bacteria. Interestingly, YH1/SPLUNC1 protein has antimicrobial activity, and it can directly kill Escherichia coli and Acinetobacter haemolyticus. The microorganism cell showed morphological changes in cell wall such as cell damage and cytoplasmic leakage after exposure to YH1/SPLUNC1 protein, indicating that YH1/SPLUNC1 directly killed the microorganisms by cell wall permeabilization. All these results indicated that YH1/SPLUNC1 might be an important antimicrobial protein involved in innate immunity defense.
Collapse
Affiliation(s)
- Hui Liu
- China-America Cancer Research Institute, Guangdong Medical College, No. 1 New City Ave, Songshan Lake High-Tech. Area, Dongguan 523808, China; Department of Pathology, University of California, Harbor UCLA Medical Center, Torrance, CA 90509, United States
| | - Xiangning Zhang
- China-America Cancer Research Institute, Guangdong Medical College, No. 1 New City Ave, Songshan Lake High-Tech. Area, Dongguan 523808, China
| | - Jingjing Wu
- China-America Cancer Research Institute, Guangdong Medical College, No. 1 New City Ave, Songshan Lake High-Tech. Area, Dongguan 523808, China
| | - Samuel W French
- Department of Pathology, University of California, Harbor UCLA Medical Center, Torrance, CA 90509, United States
| | - Zhiwei He
- China-America Cancer Research Institute, Guangdong Medical College, No. 1 New City Ave, Songshan Lake High-Tech. Area, Dongguan 523808, China.
| |
Collapse
|
38
|
Walton WG, Ahmad S, Little MR, Kim CS, Tyrrell J, Lin Q, Di YP, Tarran R, Redinbo MR. Structural Features Essential to the Antimicrobial Functions of Human SPLUNC1. Biochemistry 2016; 55:2979-91. [PMID: 27145151 PMCID: PMC4887393 DOI: 10.1021/acs.biochem.6b00271] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SPLUNC1 is an abundantly secreted innate immune protein in the mammalian respiratory tract that exerts bacteriostatic and antibiofilm effects, binds to lipopolysaccharide (LPS), and acts as a fluid-spreading surfactant. Here, we unravel the structural elements essential for the surfactant and antimicrobial functions of human SPLUNC1 (short palate lung nasal epithelial clone 1). A unique α-helix (α4) that extends from the body of SPLUNC1 is required for the bacteriostatic, surfactant, and LPS binding activities of this protein. Indeed, we find that mutation of just four leucine residues within this helical motif to alanine is sufficient to significantly inhibit the fluid spreading abilities of SPLUNC1, as well as its bacteriostatic actions against Gram-negative pathogens Burkholderia cenocepacia and Pseudomonas aeruginosa. Conformational flexibility in the body of SPLUNC1 is also involved in the bacteriostatic, surfactant, and LPS binding functions of the protein as revealed by disulfide mutants introduced into SPLUNC1. In addition, SPLUNC1 exerts antibiofilm effects against Gram-negative bacteria, although α4 is not involved in this activity. Interestingly, though, the introduction of surface electrostatic mutations away from α4 based on the unique dolphin SPLUNC1 sequence, and confirmed by crystal structure, is shown to impart antibiofilm activity against Staphylococcus aureus, the first SPLUNC1-dependent effect against a Gram-positive bacterium reported to date. Together, these data pinpoint SPLUNC1 structural motifs required for the antimicrobial and surfactant actions of this protective human protein.
Collapse
Affiliation(s)
- William G. Walton
- Departments of Chemistry, Biochemistry and Microbiology, 4350 Genome Sciences Building, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Saira Ahmad
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Michael R. Little
- Departments of Chemistry, Biochemistry and Microbiology, 4350 Genome Sciences Building, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Christine S.K. Kim
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Jean Tyrrell
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Qiao Lin
- Department of Environmental and Occupational Health, 331 Bridgeside Point Building, University of Pittsburgh, Pittsburgh, PA 15260
| | - Y. Peter Di
- Department of Environmental and Occupational Health, 331 Bridgeside Point Building, University of Pittsburgh, Pittsburgh, PA 15260
| | - Robert Tarran
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Matthew R. Redinbo
- Departments of Chemistry, Biochemistry and Microbiology, 4350 Genome Sciences Building, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| |
Collapse
|
39
|
Saferali A, Obeidat M, Bérubé JC, Lamontagne M, Bossé Y, Laviolette M, Hao K, Nickle DC, Timens W, Sin DD, Postma DS, Strug LJ, Gallins PJ, Paré PD, Bingle CD, Sandford AJ. Polymorphisms associated with expression of BPIFA1/BPIFB1 and lung disease severity in cystic fibrosis. Am J Respir Cell Mol Biol 2016; 53:607-14. [PMID: 25574903 DOI: 10.1165/rcmb.2014-0182oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BPI fold containing family A, member 1 (BPIFA1) and BPIFB1 are putative innate immune molecules expressed in the upper airways. Because of their hypothesized roles in airway defense, these molecules may contribute to lung disease severity in cystic fibrosis (CF). We interrogated BPIFA1/BPIFB1 single-nucleotide polymorphisms in data from an association study of CF modifier genes and found an association of the G allele of rs1078761 with increased lung disease severity (P = 2.71 × 10(-4)). We hypothesized that the G allele of rs1078761 is associated with decreased expression of BPIFA1 and/or BPIFB1. Genome-wide lung gene expression and genotyping data from 1,111 individuals with lung disease, including 51 patients with CF, were tested for associations between genotype and BPIFA1 and BPIFB1 gene expression levels. Findings were validated by quantitative PCR in a subset of 77 individuals. Western blotting was used to measure BPIFA1 and BPIFB1 protein levels in 93 lung and 101 saliva samples. The G allele of rs1078761 was significantly associated with decreased mRNA levels of BPIFA1 (P = 4.08 × 10(-15)) and BPIFB1 (P = 0.0314). These findings were confirmed with quantitative PCR and Western blotting. We conclude that the G allele of rs1078761 may be detrimental to lung function in CF owing to decreased levels of BPIFA1 and BPIFB1.
Collapse
Affiliation(s)
- Aabida Saferali
- 1 Centre for Heart Lung Innovation, UBC and St. Paul's Hospital, Vancouver, British Columbia
| | - Ma'en Obeidat
- 1 Centre for Heart Lung Innovation, UBC and St. Paul's Hospital, Vancouver, British Columbia
| | | | - Maxime Lamontagne
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Yohan Bossé
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec and.,3 Department of Molecular Medicine, Laval University, Québec, Quebec
| | - Michel Laviolette
- 2 Institut Universitaire de Cardiologie et de Pneumologie de Québec and
| | - Ke Hao
- 4 Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - David C Nickle
- 5 Merck Research Laboratories, Boston, Massachusetts.,6 Merck, Rahway, New Jersey.,7 Genetics, Rosetta Inpharmatics, Merck, Seattle, Washington
| | - Wim Timens
- Departments of 8 Pathology and Medical Biology and
| | - Don D Sin
- 1 Centre for Heart Lung Innovation, UBC and St. Paul's Hospital, Vancouver, British Columbia
| | - Dirkje S Postma
- 9 Pulmonary Medicine and Tuberculosis, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lisa J Strug
- 10 Program in Genetics and Genome Biology, The Hospital For Sick Children, and Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Paul J Gallins
- 11 Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Peter D Paré
- 1 Centre for Heart Lung Innovation, UBC and St. Paul's Hospital, Vancouver, British Columbia
| | - Colin D Bingle
- 12 Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, United Kingdom
| | - Andrew J Sandford
- 1 Centre for Heart Lung Innovation, UBC and St. Paul's Hospital, Vancouver, British Columbia
| |
Collapse
|
40
|
Aragao-Santiago L, Hillaireau H, Grabowski N, Mura S, Nascimento TL, Dufort S, Coll JL, Tsapis N, Fattal E. Compared in vivo toxicity in mice of lung delivered biodegradable and non-biodegradable nanoparticles. Nanotoxicology 2015; 10:292-302. [DOI: 10.3109/17435390.2015.1054908] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Letícia Aragao-Santiago
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| | - Hervé Hillaireau
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| | - Nadège Grabowski
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| | - Simona Mura
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| | - Thais L. Nascimento
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| | - Sandrine Dufort
- INSERM U823, Institut Albert Bonniot, Grenoble, France, and
- Université Joseph Fourier, Grenoble, France
| | - Jean-Luc Coll
- INSERM U823, Institut Albert Bonniot, Grenoble, France, and
- Université Joseph Fourier, Grenoble, France
| | - Nicolas Tsapis
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| | - Elias Fattal
- Faculté de Pharmacie, Institut Galien Paris-Sud, LabEx LERMIT, Université Paris-Sud, Chatenay-Malabry Cedex, France,
- CNRS, UMR 8612, Chatenay-Malabry Cedex, France,
| |
Collapse
|
41
|
Therapeutic Effects of α1-Antitrypsin on Psedumonas aeruginosa Infection in ENaC Transgenic Mice. PLoS One 2015; 10:e0141232. [PMID: 26509529 PMCID: PMC4624966 DOI: 10.1371/journal.pone.0141232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease with many airway pathological features, including aberrant epithelial sodium channel (ENaC) function, persistent Pseudomonas aeruginosa (PA) infection and neutrophil-dominant inflammation. PA infection in CF airways is difficult to treat due to antibiotic resistance and other factors. Recently, α1-antitrypsin (A1AT) have been shown to be effective to reduce CF airway PA infection. However, there is a dearth of studies about the mechanisms underlying A1AT's therapeutic effects. The goal of our study is to provide an animal model of A1AT therapy in CF lungs. ENaC transgenic mice with PA infection were used as a CF-like model. Mice were intratracheally treated with PA or saline (control) in a fibrin plug. Two hours after PA infection, aerosolized A1AT were delivered to mouse lungs once daily. At day 1 and day 3 post PA infection, lung inflammation, PA load as well as host defence protein short palate, lung, and nasal epithelium clone 1 (SPLUNC1) were measured. At day 1 post PA infection when A1AT was delivered once to ENaC transgenic mouse lungs, A1AT did not reduce lung inflammation (e.g., neutrophils) and PA load. However, at day 3 post PA infection when ENaC transgenic mice received three repeated A1AT treatments, a significant decrease in airspace inflammation and PA load was observed. Although A1AT prevented the loss of SPLUNC1 in bronchoalveolar lavage fluid of PA-infected wild-type mice, it did not restore SPLUNC1 levels in ENaC transgenic mice. Our current study has provided a valid and quick A1AT therapeutic model in CF-like lungs that may serve as a platform for future mechanistic studies about how A1AT exerts beneficial effects in human CF patients.
Collapse
|
42
|
Britto CJ, Cohn L. Bactericidal/Permeability-increasing protein fold-containing family member A1 in airway host protection and respiratory disease. Am J Respir Cell Mol Biol 2015; 52:525-34. [PMID: 25265466 DOI: 10.1165/rcmb.2014-0297rt] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bactericidal/permeability-increasing protein fold-containing family member A1 (BPIFA1), formerly known as SPLUNC1, is one of the most abundant proteins in respiratory secretions and has been identified with increasing frequency in studies of pulmonary disease. Its expression is largely restricted to the respiratory tract, being highly concentrated in the upper airways and proximal trachea. BPIFA1 is highly responsive to airborne pathogens, allergens, and irritants. BPIFA1 actively participates in host protection through antimicrobial, surfactant, airway surface liquid regulation, and immunomodulatory properties. Its expression is modulated in multiple lung diseases, including cystic fibrosis, chronic obstructive pulmonary disease, respiratory malignancies, and idiopathic pulmonary fibrosis. However, the role of BPIFA1 in pulmonary pathogenesis remains to be elucidated. This review highlights the versatile properties of BPIFA1 in antimicrobial protection and its roles as a sensor of environmental exposure and regulator of immune cell function. A greater understanding of the contribution of BPIFA1 to disease pathogenesis and activity may clarify if BPIFA1 is a biomarker and potential drug target in pulmonary disease.
Collapse
Affiliation(s)
- Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
43
|
Xue JL, Yi L, Yan ZH, Li X, Wang XJ, Wei PJ, Zeng JE, Zhao YL, Zhang HT. SPLUNC1 Is a Significant Marker in Pleural Effusion from Lung Cancer Compared to Tuberculosis. Monoclon Antib Immunodiagn Immunother 2015; 34:206-12. [PMID: 26090599 DOI: 10.1089/mab.2014.0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
SPLUNC1 (Short palate, lung and nasal epithelium clone1) protein is an abundant secretory product of epithelia present throughout the conducting airways. Although its function is still not fully known, most studies have focused on its defensive effect in the infection of human airways and its potential to serve as a molecular marker for lung cancer. In this study, we further evaluated the SPLUNC1 expression in patients with lung disease to explore its role in cancer or tuberculosis at the protein level. We generated a panel of antibodies by using protein from a eukaryotic expression system as the immunogen to mice. It was the panel of SPLUNC1 monoclonal antibodies that allowed us to comparatively determine SPLUNC1 protein in lung cancer and tuberculosis infection by detecting sera and pleural effusion other than airway surface. The results showed that the SPLUNC1 level was not significantly changed either from sera of lung cancer or control. There was a significant increase in pleural effusion from lung cancer when compared to tuberculosis. These results indicate that SPLUNC1 may be a useful marker for tracing lung cancer cells, based on its epithelial origin property in pleural effusion.
Collapse
Affiliation(s)
- Jun L Xue
- 1 Department of Endocrinology, Jingzhou Clinical Medical College, Yangtze University , Jingzhou, People's Republic of China
| | - Ling Yi
- 2 Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute , Beijing, People's Republic of China
| | - Zhou H Yan
- 2 Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute , Beijing, People's Republic of China
| | - Xin Li
- 3 Hebei Chest Hospital , Shijiazhuang, People's Republic of China
| | - Xiao J Wang
- 2 Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute , Beijing, People's Republic of China
| | - Pang J Wei
- 2 Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute , Beijing, People's Republic of China
| | - Jiao E Zeng
- 1 Department of Endocrinology, Jingzhou Clinical Medical College, Yangtze University , Jingzhou, People's Republic of China
| | - Yan L Zhao
- 4 Chinese Center for Disease Control and Prevention , Beijing, People's Republic of China
| | - Hong T Zhang
- 2 Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute , Beijing, People's Republic of China
| |
Collapse
|
44
|
Bartlett JA, Meyerholz DK, Wohlford-Lenane CL, Naumann PW, Salzman NH, McCray PB. Increased susceptibility to otitis media in a Splunc1-deficient mouse model. Dis Model Mech 2015; 8:501-8. [PMID: 25765466 PMCID: PMC4415896 DOI: 10.1242/dmm.019646] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/07/2015] [Indexed: 12/15/2022] Open
Abstract
Otitis media (inflammation of the middle ear) is one of the most common diseases of early childhood. Susceptibility to otitis is influenced by a number of factors, including the actions of innate immune molecules secreted by the epithelia lining the nasopharynx, middle ear and Eustachian tube. The SPLUNC1 (short palate, lung, nasal epithelial clone 1) protein is a highly abundant secretory product of the mammalian nasal, oral and respiratory mucosa that is thought to play a multifunctional role in host defense. In this study we investigated Splunc1 expression in the ear of the mouse, and examined whether this protein contributes to overall host defense in the middle ear and/or Eustachian tube. We found that Splunc1 is highly expressed in both the surface epithelium and in submucosal glands in these regions in wild-type mice. In mice lacking Splunc1, we noted histologically an increased frequency of otitis media, characterized by the accumulation of leukocytes (neutrophils with scattered macrophages), proteinaceous fluid and mucus in the middle ear lumens. Furthermore, many of these mice had extensive remodeling of the middle ear wall, suggesting a chronic course of disease. From these observations, we conclude that loss of Splunc1 predisposes mice to the development of otitis media. The Splunc1−/− mouse model should help investigators to better understand both the biological role of Splunc1 as well as host defense mechanisms in the middle ear. Summary: We document expression of the innate immune factor Splunc1 in the murine middle ear and Eustachian tube, and describe spontaneous development of otitis media in mice lacking functional Splunc1.
Collapse
Affiliation(s)
- Jennifer A Bartlett
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - David K Meyerholz
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | - Paul W Naumann
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nita H Salzman
- Department of Pediatrics, Division of Gastroenterology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Paul B McCray
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
45
|
Electronic cigarette liquid increases inflammation and virus infection in primary human airway epithelial cells. PLoS One 2014; 9:e108342. [PMID: 25244293 PMCID: PMC4171526 DOI: 10.1371/journal.pone.0108342] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 08/27/2014] [Indexed: 11/19/2022] Open
Abstract
Background/Objective The use of electronic cigarettes (e-cigarettes) is rapidly increasing in the United States, especially among young people since e-cigarettes have been perceived as a safer alternative to conventional tobacco cigarettes. However, the scientific evidence regarding the human health effects of e-cigarettes on the lung is extremely limited. The major goal of our current study is to determine if e-cigarette use alters human young subject airway epithelial functions such as inflammatory response and innate immune defense against respiratory viral (i.e., human rhinovirus, HRV) infection. Methodology/Main Results We examined the effects of e-cigarette liquid (e-liquid) on pro-inflammatory cytokine (e.g., IL-6) production, HRV infection and host defense molecules (e.g., short palate, lung, and nasal epithelium clone 1, SPLUNC1) in primary human airway epithelial cells from young healthy non-smokers. Additionally, we examined the role of SPLUNC1 in lung defense against HRV infection using a SPLUNC1 knockout mouse model. We found that nicotine-free e-liquid promoted IL-6 production and HRV infection. Addition of nicotine into e-liquid further amplified the effects of nicotine-free e-liquid. Moreover, SPLUNC1 deficiency in mice significantly increased lung HRV loads. E-liquid inhibited SPLUNC1 expression in primary human airway epithelial cells. These findings strongly suggest the deleterious health effects of e-cigarettes in the airways of young people. Our data will guide future studies to evaluate the impact of e-cigarettes on lung health in human populations, and help inform the public about potential health risks of e-cigarettes.
Collapse
|
46
|
Ning F, Wang C, Berry KZ, Kandasamy P, Liu H, Murphy RC, Voelker DR, Nho CW, Pan CH, Dai S, Niu L, Chu HW, Zhang G. Structural characterization of the pulmonary innate immune protein SPLUNC1 and identification of lipid ligands. FASEB J 2014; 28:5349-60. [PMID: 25223608 DOI: 10.1096/fj.14-259291] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The short palate, lung and nasal epithelial clone 1 (SPLUNC1) protein is a member of the palate, lung, and nasal epithelium clone (PLUNC) family, also known as bactericidal/permeability-increasing (BPI) fold-containing protein, family A, member 1 (BPIFA1). SPLUNC1 is an abundant protein in human airways, but its function remains poorly understood. The lipid ligands of SPLUNC1 as well as other PLUNC family members are largely unknown, although some reports provide evidence that lipopolysaccharide (LPS) could be a lipid ligand. Unlike previous hypotheses, we found significant structural differences between SPLUNC1 and BPI. Recombinant SPLUNC1 produced in HEK 293 cells harbored several molecular species of sphingomyelin and phosphatidylcholine as its ligands. Significantly, in vitro lipid-binding studies failed to demonstrate interactions between SPLUNC1 and LPS, lipoteichoic acid, or polymyxin B. Instead, one of the major and most important pulmonary surfactant phospholipids, dipalmitoylphosphatidylcholine (DPPC), bound to SPLUNC1 with high affinity and specificity. We found that SPLUNC1 could be the first protein receptor for DPPC. These discoveries provide insight into the specific determinants governing the interaction between SPLUNC1 and lipids and also shed light on novel functions that SPLUNC1 and other PLUNC family members perform in host defense.
Collapse
Affiliation(s)
- Fangkun Ning
- School of Life Sciences, University of Science and Technology of China, Hefei, China; Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Chao Wang
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Karin Zemski Berry
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Haolin Liu
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA
| | - Dennis R Voelker
- Department of Medicine, National Jewish Health, Denver, Colorado, USA; and
| | - Chu Won Nho
- Functional Food Center, Korea Institute of Science and Technology, GangNeung, Korea
| | - Choel-Ho Pan
- Functional Food Center, Korea Institute of Science and Technology, GangNeung, Korea
| | - Shaodong Dai
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Liwen Niu
- School of Life Sciences, University of Science and Technology of China, Hefei, China;
| | - Hong-Wei Chu
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA; Department of Medicine, National Jewish Health, Denver, Colorado, USA; and
| | - Gongyi Zhang
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| |
Collapse
|
47
|
Tarran R, Redinbo MR. Mammalian short palate lung and nasal epithelial clone 1 (SPLUNC1) in pH-dependent airway hydration. Int J Biochem Cell Biol 2014; 52:130-5. [PMID: 24631954 PMCID: PMC4048990 DOI: 10.1016/j.biocel.2014.03.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/22/2014] [Accepted: 03/03/2014] [Indexed: 12/12/2022]
Abstract
The epithelia that line the conducting airways are the lung's first point of contact with inhaled pathogens and toxicants. As such, they are known to play an important role in the lung's innate defense system, which includes (i) the production of airway surface liquid (ASL) that helps cleanse the airways through the physical removal of pathogens and toxicants on the mucociliary escalator and (ii) the secretion of anti-microbial proteins into the ASL to kill inhaled pathogens. Interestingly, the recently crystallized short palate lung and nasal epithelial clone 1 (SPLUNC1) protein appears to be a multi-functional protein. That is, it not only acts as an anti-microbial agent, but also modulates ASL homeostasis by acting as an endogenous inhibitor of the epithelial Na(+) channel (ENaC). This review will focus on the latter function of SPLUNC1, and will discuss new structural and physiological data regarding SPLUNC1's failure to function as a regulator of ASL hydration in CF airways.
Collapse
Affiliation(s)
- Robert Tarran
- Cystic Fibrosis/Pulmonary Research and Treatment Center, Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Matthew R Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
48
|
Tsou YA, Huang HJ, Lin WWY, Chen CYC. Investigation of anti-infection mechanism of lactoferricin and splunc-1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2014; 2014:907028. [PMID: 24876880 PMCID: PMC4021689 DOI: 10.1155/2014/907028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/08/2014] [Accepted: 02/08/2014] [Indexed: 12/11/2022]
Abstract
The innate immune system is the first line in the defense system and prevents the body from further bacteria, virus, or fungal infections. Most of the innate immune system is relevant to mucosa immunity. Lactotransferrin is secreted from the human mammal breast duct epithelial tissue and strengthens infant immunity to defense with regard to outward pathogens. Splunc-1 is also an innate material secreted from the soft palate, lung, nasal cavity epithelium, and mucosa. It helps with mucosa defense against bacterial, virus, and even fungus. LPS is the main etiology of Gram-negative bacilla infection source. And studies of lactoferricin and slpunc-1 both can combine with LPS and subsequently cause insults to the mucosa. Although, we know that both of them partake in an important role in innate immunity, we do not know the effects when they work together. In this study, we just overview silicon stimulation to examine the combination of Lactoferricin and Splunc-1 and the effect with regard to LPS.
Collapse
Affiliation(s)
- Yung An Tsou
- Otolaryngology Head and Neck Surgery, China Medical University Hospital, Taichung 40402, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Hung-Jin Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Wesley Wen Yang Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
| | - Calvin Yu-Chian Chen
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
49
|
Wei Y, Xia W, Ye X, Fan Y, Shi J, Wen W, Yang P, Li H. The antimicrobial protein short palate, lung, and nasal epithelium clone 1 (SPLUNC1) is differentially modulated in eosinophilic and noneosinophilic chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2014; 133:420-8. [DOI: 10.1016/j.jaci.2013.09.052] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Revised: 09/09/2013] [Accepted: 09/12/2013] [Indexed: 01/18/2023]
|
50
|
Di YP, Tkach AV, Yanamala N, Stanley S, Gao S, Shurin MR, Kisin ER, Kagan VE, Shvedova A. Dual acute proinflammatory and antifibrotic pulmonary effects of short palate, lung, and nasal epithelium clone-1 after exposure to carbon nanotubes. Am J Respir Cell Mol Biol 2013; 49:759-67. [PMID: 23721177 DOI: 10.1165/rcmb.2012-0435oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Carbon nanotubes (CNTs; allotropes of carbon with a cylindrical nanostructure) have emerged as one of the most commonly used types of nanomaterials, with numerous applications in industry and biomedicine. However, the inhalation of CNTs has been shown to elicit pulmonary toxicity, accompanied by a robust inflammatory response with an early-onset fibrotic phase. Epithelial host-defense proteins represent an important component of the pulmonary innate immune response to foreign inhalants such as particles and bacteria. The short palate, lung, and nasal epithelium clone-1 (SPLUNC1) protein, a member of the bactericidal/permeability-increasing-fold (BPIF)-containing protein family, is a 25-kD secretory protein that is expressed in nasal, oropharyngeal, and lung epithelia, and has been shown to have multiple functions, including antimicrobial and chemotactic activities, as well as surfactant properties. This study sought to assess the importance of SPLUNC1-mediated pulmonary responses in airway epithelial secretions, and to explore the biological relevance of SPLUNC1 to inhaled particles in a single-walled carbon nanotube (SWCNT) model. Using Scgb1a1-hSPLUNC1 transgenic mice, we observed that SPLUNC1 significantly modified host inflammatory responses by increasing leukocyte recruitment and enhancing phagocytic activity. Furthermore, we found that transgenic mice were more susceptible to SWCNT exposure at the acute phase, but showed resistance against lung fibrogenesis through pathological changes in the long term. The binding of SPLUNC1 also attenuated SWCNT-induced TNF-α secretion by RAW 264.7 macrophages. Taken together, our data indicate that SPLUNC1 is an important component of mucosal innate immune defense against pulmonary inhaled particles.
Collapse
Affiliation(s)
- Y Peter Di
- 1 Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|