1
|
Di YP, Mou H. Airway Serous Cells: A Comparative Study of Spatial Distribution and Abundance among Species. JOURNAL OF RESPIRATORY BIOLOGY AND TRANSLATIONAL MEDICINE 2024; 1:10013. [PMID: 39220634 PMCID: PMC11361305 DOI: 10.35534/jrbtm.2024.10013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The conducting airways of the respiratory system play a crucial role in filtering, humidifying, and directing air into the lungs. Among the specialized cell types within these airways, airway serous cells are notable for their secretion of watery, protein-rich fluids and enzymes, which contribute to maintaining airway surface liquid homeostasis and defending against pathogens. However, the distribution and abundance of serous cells across different species in the conducting airways remain poorly understood. In this study, we addressed this gap by investigating the spatial distribution of the airway serous cell-specific marker BPI fold containing family A member 1 (BPIFA1) in humans, pigs, and mice. Our findings demonstrate significant variations in the distribution and abundance of serous cells among these species, potentially reflecting their different respiratory anatomy and evolutionary adaptations to diverse environmental challenges and respiratory demands. In humans and pigs, airway serous cells are predominantly found in the submucosal glands of the trachea and segmental bronchi, frequently overlapping with lysozyme-positive secretory cells. In contrast, rodents like mice exhibit a distinct pattern where serous cells are scarce in submucosal glands. Instead, rodent serous cells are primarily located at the epithelial surface from the trachea to the main bronchi, where many co-express the Club cell-specific protein SCGB1A1. The abundance of serous cells diminishes progressively in the intrapulmonary airways. Given that rodent models are widely utilized in respiratory research, understanding anatomical and cellular differences in airway serous cells is critical for interpreting experimental outcomes and translating findings to human respiratory diseases and therapeutic strategies. This comparative analysis enhances our understanding of airway biology across species and informs the selection and interpretation of animal models in respiratory studies.
Collapse
Affiliation(s)
- Yuanpu Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
2
|
Zhang Y, Lin T, Leung HM, Zhang C, Wilson-Mifsud B, Feldman MB, Puel A, Lanternier F, Couderc LJ, Danion F, Catherinot E, Salvator H, Tcherkian C, Givel C, Xu J, Tearney GJ, Vyas JM, Li H, Hurley BP, Mou H. STAT3 mutation-associated airway epithelial defects in Job syndrome. J Allergy Clin Immunol 2023; 152:538-550. [PMID: 36638921 PMCID: PMC10330947 DOI: 10.1016/j.jaci.2022.12.821] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/30/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Job syndrome is a disease of autosomal dominant hyper-IgE syndrome (AD-HIES). Patients harboring STAT3 mutation are particularly prone to airway remodeling and airway infections. OBJECTIVES Airway epithelial cells play a central role as the first line of defense against pathogenic infection and express high levels of STAT3. This study thus interrogates how AD-HIES STAT3 mutations impact the physiological functions of airway epithelial cells. METHODS This study created human airway basal cells expressing 4 common AD-HIES STAT3 mutants (R382W, V463del, V637M, and Y657S). In addition, primary airway epithelial cells were isolated from a patient with Job syndrome who was harboring a STAT3-S560del mutation and from mice harboring a STAT3-V463del mutation. Cell proliferation, differentiation, barrier function, bacterial elimination, and innate immune responses to pathogenic infection were quantitatively analyzed. RESULTS STAT3 mutations reduce STAT3 protein phosphorylation, nuclear translocation, transcription activity, and protein stability in airway basal cells. As a consequence, STAT3-mutated airway basal cells give rise to airway epithelial cells with abnormal cellular composition and loss of coordinated mucociliary clearance. Notably, AD-HIES STAT3 airway epithelial cells are defective in bacterial killing and fail to initiate vigorous proinflammatory responses and neutrophil transepithelial migration in response to an experimental model of Pseudomonas aeruginosa infection. CONCLUSIONS AD-HIES STAT3 mutations confer numerous abnormalities to airway epithelial cells in cell differentiation and host innate immunity, emphasizing their involvement in the pathogenesis of lung complications in Job syndrome. Therefore, therapies must address the epithelial defects as well as the previously noted immune cell defects to alleviate chronic infections in patients with Job syndrome.
Collapse
Affiliation(s)
- Yihan Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Mass; Departments of Pediatrics, Harvard Medical School, Boston, Mass; Division of Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Mass
| | - Tian Lin
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Mass; Departments of Pediatrics, Harvard Medical School, Boston, Mass; Division of Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Mass
| | - Hui Min Leung
- Wellman Center for Photomedicine, Massachusetts General Hospital, and the Departments of Pediatrics, Harvard Medical School, Boston, Mass; Department of Pathology, Massachusetts General Hospital, Boston, Mass
| | - Cheng Zhang
- Center for Individualized Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minn
| | - Brittany Wilson-Mifsud
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Mass; Departments of Pediatrics, Harvard Medical School, Boston, Mass; Division of Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Mass
| | - Michael B Feldman
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherché (INSERM) U1163, Paris, France; Departments of Medicine, Harvard Medical School, Boston, Mass
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherché (INSERM) U1163, Paris, France; University of Paris, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, NY
| | - Fanny Lanternier
- Institut Pasteur, Université Paris Cité, Centre National de Référence des Mycoses Invasives et Antifongiques, Centre National de la Recherche Scientifique, Unite Mixté de Recherche (UMR) 2000, Paris, France; Service de Maladies Infectieuses, Hôpital Necker, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Louis-Jean Couderc
- Respiratory Diseases Department, Foch Hospital, Suresnes, France; Laboratoire Virologie et Immunologie Moléculaires Suresnes, UMR 0892 Paris-Saclay University, Paris, France
| | - Francois Danion
- Department of Infectious Diseases, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France; Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, Université de Strasbourg, Strasbourg, France
| | | | - Hélène Salvator
- Respiratory Diseases Department, Foch Hospital, Suresnes, France; Laboratoire Virologie et Immunologie Moléculaires Suresnes, UMR 0892 Paris-Saclay University, Paris, France
| | - Colas Tcherkian
- Respiratory Diseases Department, Foch Hospital, Suresnes, France
| | - Claire Givel
- Respiratory Diseases Department, Foch Hospital, Suresnes, France; Laboratoire Virologie et Immunologie Moléculaires Suresnes, UMR 0892 Paris-Saclay University, Paris, France
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, Mich
| | - Guillermo J Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, and the Departments of Pediatrics, Harvard Medical School, Boston, Mass; Department of Pathology, Massachusetts General Hospital, Boston, Mass
| | - Jatin M Vyas
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Mass; Departments of Medicine, Harvard Medical School, Boston, Mass
| | - Hu Li
- Center for Individualized Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minn
| | - Bryan P Hurley
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Mass; Departments of Pediatrics, Harvard Medical School, Boston, Mass; Division of Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Mass
| | - Hongmei Mou
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Mass; Departments of Pediatrics, Harvard Medical School, Boston, Mass; Division of Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, Mass.
| |
Collapse
|
3
|
Immunogenic senescence sensitizes lung cancer to LUNX-targeting therapy. Cancer Immunol Immunother 2021; 71:1403-1417. [PMID: 34674012 PMCID: PMC9123058 DOI: 10.1007/s00262-021-03077-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/30/2021] [Indexed: 11/02/2022]
Abstract
The higher immunogenicity of tumors usually predicts favorable therapeutic responses. Tumor antigens dominate the immunogenic character within tumors. We investigated if there was a targetable tumor antigen during immunogenic chemotherapy within lung cancer. Chemotherapy-induced immunogenic senescence was demonstrated using a multi-marker, three-step workflow, and RNA-sequencing data. The ability of anti-lung-specific X protein (LUNX) antibody to suppress the survival of senescent lung cancer cells was evaluated in vitro and in vivo using real-time cytotoxicity analysis and xenograft mouse models, respectively. The induction of cellular senescence by immunogenic chemotherapy boosted cell-surface shuttling of LUNX and enhanced the immunogenic features of senescent tumor cells, which sensitized lung cancer cells to anti-LUNX antibody-mediated therapy and contributed to tumor suppression. The immunogenic senescence-mediated anti-tumor response was triggered by the direct action of antibody on tumor cells, strengthened by natural-killer cells through an antibody-dependent cell-mediated cytotoxicity response, and ultimately, led to tumor control. Our findings suggest that LUNX is a lung cancer targetable-immunogenic antigen. The proportion of lung cancers responding to LUNX-targeting therapy could be expanded substantially by immunogenic chemotherapy that induces senescence-associated translocation of LUNX to the plasma membrane.
Collapse
|
4
|
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: 0.5] [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
|
5
|
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: 14] [Impact Index Per Article: 2.3] [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
|
6
|
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: 6] [Impact Index Per Article: 1.0] [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
|
7
|
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: 12] [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
|
8
|
Erickson NA, Dietert K, Enders J, Glauben R, Nouailles G, Gruber AD, Mundhenk L. Soluble mucus component CLCA1 modulates expression of leukotactic cytokines and BPIFA1 in murine alveolar macrophages but not in bone marrow-derived macrophages. Histochem Cell Biol 2018; 149:619-633. [PMID: 29610986 PMCID: PMC5999134 DOI: 10.1007/s00418-018-1664-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2018] [Indexed: 11/18/2022]
Abstract
The secreted airway mucus cell protein chloride channel regulator, calcium-activated 1, CLCA1, plays a role in inflammatory respiratory diseases via as yet unidentified pathways. For example, deficiency of CLCA1 in a mouse model of acute pneumonia resulted in reduced cytokine expression with less leukocyte recruitment and the human CLCA1 was shown to be capable of activating macrophages in vitro. Translation of experimental data between human and mouse models has proven problematic due to several CLCA species-specific differences. We therefore characterized activation of macrophages by CLCA1 in detail in solely murine ex vivo and in vitro models. Only alveolar but not bone marrow-derived macrophages freshly isolated from C57BL6/J mice increased their expression levels of several pro-inflammatory and leukotactic cytokines upon CLCA1 stimulation. Among the most strongly regulated genes, we identified the host-protective and immunomodulatory airway mucus component BPIFA1, previously unknown to be expressed by airway macrophages. Furthermore, evidence from an in vivo Staphylococcus aureus pneumonia mouse model suggests that CLCA1 may also modify BPIFA1 expression in airway epithelial cells. Our data underscore and specify the role of mouse CLCA1 in inflammatory airway disease to activate airway macrophages. In addition to its ability to upregulate cytokine expression which explains previous observations in the Clca1-deficient S. aureus pneumonia mouse model, modulation of BPIFA1 expression expands the role of CLCA1 in airway disease to involvement in more complex downstream pathways, possibly including liquid homeostasis, airway protection, and antimicrobial defense.
Collapse
Affiliation(s)
- Nancy A Erickson
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Kristina Dietert
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Jana Enders
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Rainer Glauben
- Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Geraldine Nouailles
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany
| | - Lars Mundhenk
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Strasse 15, 14163, Berlin, Germany.
| |
Collapse
|
9
|
An innate defense peptide BPIFA1/SPLUNC1 restricts influenza A virus infection. Mucosal Immunol 2018; 11:71-81. [PMID: 28513596 DOI: 10.1038/mi.2017.45] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/17/2017] [Indexed: 02/04/2023]
Abstract
The airway epithelium secretes proteins that function in innate defense against infection. Bactericidal/permeability-increasing fold-containing family member A1 (BPIFA1) is secreted into airways and has a protective role during bacterial infections, but it is not known whether it also has an antiviral role. To determine a role in host defense against influenza A virus (IAV) infection and to find the underlying defense mechanism, we developed transgenic mouse models that are deficient in BPIFA1 and used these, in combination with in vitro three-dimensional mouse tracheal epithelial cell (mTEC) cultures, to investigate its antiviral properties. We show that BPIFA1 has a significant role in mucosal defense against IAV infection. BPIFA1 secretion was highly modulated after IAV infection. Mice deficient in BPIFA1 lost more weight after infection, supported a higher viral load and virus reached the peripheral lung earlier, indicative of a defect in the control of infection. Further analysis using mTEC cultures showed that BPIFA1-deficient cells bound more virus particles, displayed increased nuclear import of IAV ribonucleoprotein complexes, and supported higher levels of viral replication. Our results identify a critical role of BPIFA1 in the initial phase of infection by inhibiting the binding and entry of IAV into airway epithelial cells.
Collapse
|
10
|
De Smet EG, Seys LJM, Verhamme FM, Vanaudenaerde BM, Brusselle GG, Bingle CD, Bracke KR. Association of innate defense proteins BPIFA1 and BPIFB1 with disease severity in COPD. Int J Chron Obstruct Pulmon Dis 2017; 13:11-27. [PMID: 29296079 PMCID: PMC5741069 DOI: 10.2147/copd.s144136] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by an abnormal inflammatory response in the lungs caused by the inhalation of noxious particles and gases. The airway epithelium has a protective function against these harmful agents by maintaining a physical barrier and by secreting defensive proteins, such as bactericidal/permeability-increasing fold-containing (BPIF) proteins, BPIFA1 and BPIFB1. However, inconsistent data regarding BPIFA1 expression in smokers and COPD patients have been reported to date. Therefore, we investigated the expression of BPIFA1 and BPIFB1 in a large cohort of never-smokers and smokers with and without COPD, both on the messenger RNA (mRNA) level in lung tissue and on the protein level in airway epithelium. Furthermore, we examined the correlation between BPIFA1 and BPIFB1 levels, goblet cell hyperplasia, and lung function measurements. BPIFA1 and BPIFB1 mRNA expressions were significantly increased in stage III-IV COPD patients compared with stage II COPD patients and subjects without COPD. In addition, protein levels in COPD patients were significantly increased in comparison with subjects without COPD. BPIFA1 and BPIFB1 levels were inversely correlated with measurements of airflow limitation and positively correlated with goblet cell hyperplasia. In addition, by the use of immunofluorescence double staining, we demonstrated the expression of BPIFB1 in goblet cells. In conclusion, we show that BPIFA1 and BPIFB1 levels are elevated in COPD patients and correlate with disease severity.
Collapse
Affiliation(s)
- Elise G De Smet
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Leen JM Seys
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Fien M Verhamme
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bart M Vanaudenaerde
- Laboratory for Respiratory Diseases, Department of Clinical and Experimental Medicine, KU Leuven, Leuven, Belgium
| | - Guy G Brusselle
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Colin D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, UK
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
11
|
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
|
12
|
Wu T, Huang J, Moore PJ, Little MS, Walton WG, Fellner RC, Alexis NE, Peter Di Y, Redinbo MR, Tilley SL, Tarran R. Identification of BPIFA1/SPLUNC1 as an epithelium-derived smooth muscle relaxing factor. Nat Commun 2017; 8:14118. [PMID: 28165446 PMCID: PMC5303822 DOI: 10.1038/ncomms14118] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 11/30/2016] [Indexed: 01/02/2023] Open
Abstract
Asthma is a chronic airway disease characterized by inflammation, mucus hypersecretion and abnormal airway smooth muscle (ASM) contraction. Bacterial permeability family member A1, BPIFA1, is a secreted innate defence protein. Here we show that BPIFA1 levels are reduced in sputum samples from asthmatic patients and that BPIFA1 is secreted basolaterally from healthy, but not asthmatic human bronchial epithelial cultures (HBECs), where it suppresses ASM contractility by binding to and inhibiting the Ca2+ influx channel Orai1. We have localized this effect to a specific, C-terminal α-helical region of BPIFA1. Furthermore, tracheas from Bpifa1-/- mice are hypercontractile, and this phenotype is reversed by the addition of recombinant BPIFA1. Our data suggest that BPIFA1 deficiency in asthmatic airways promotes Orai1 hyperactivity, increased ASM contraction and airway hyperresponsiveness. Strategies that target Orai1 or the BPIFA1 deficiency in asthma may lead to novel therapies to treat this disease.
Collapse
Affiliation(s)
- Tongde Wu
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Julianne Huang
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA.,Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Patrick J Moore
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Michael S Little
- Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - William G Walton
- Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Robert C Fellner
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, US EPA Human Studies Facility, 104 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Y Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, 331 Bridgeside Point Building, Pittsburgh, Pennsylvania 15260, USA
| | - Matthew R Redinbo
- Department of Chemistry, Genome Science Building, 250 Bell Tower Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Stephen L Tilley
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA.,Center for Environmental Medicine, Asthma, and Lung Biology, US EPA Human Studies Facility, 104 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| | - Robert Tarran
- Cystic Fibrosis Center/Marsico Lung Institute, Marsico Hall, 125 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA.,Department of Cell Biology &Physiology, 5200 Medical Biomolecular Research Building, 111 Mason Farm Road, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7248, USA
| |
Collapse
|
13
|
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]
|
14
|
Joo NS, Krouse ME, Choi JY, Cho HJ, Wine JJ. Inhibition of airway surface fluid absorption by cholinergic stimulation. Sci Rep 2016; 6:20735. [PMID: 26846701 PMCID: PMC4742893 DOI: 10.1038/srep20735] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 01/07/2016] [Indexed: 12/28/2022] Open
Abstract
In upper airways airway surface liquid (ASL) depth and clearance rates are both increased by fluid secretion. Secretion is opposed by fluid absorption, mainly via the epithelial sodium channel, ENaC. In static systems, increased fluid depth activates ENaC and decreased depth inhibits it, suggesting that secretion indirectly activates ENaC to reduce ASL depth. We propose an alternate mechanism in which cholinergic input, which causes copious airway gland secretion, also inhibits ENaC-mediated absorption. The conjoint action accelerates clearance, and the increased transport of mucus out of the airways restores ASL depth while cleansing the airways. We were intrigued by early reports of cholinergic inhibition of absorption by airways in some species. To reinvestigate this phenomenon, we studied inward short-circuit currents (Isc) in tracheal mucosa from human, sheep, pig, ferret, and rabbit and in two types of cultured cells. Basal Isc was inhibited 20–70% by the ENaC inhibitor, benzamil. Long-lasting inhibition of ENaC-dependent Isc was also produced by basolateral carbachol in all preparations except rabbit and the H441 cell line. Atropine inhibition produced a slow recovery or prevented inhibition if added before carbachol. The mechanism for inhibition was not determined and is most likely multi-factorial. However, its physiological significance is expected to be increased mucus clearance rates in cholinergically stimulated airways.
Collapse
Affiliation(s)
- Nam Soo Joo
- The Cystic Fibrosis Research Laboratory, Stanford University, Stanford, CA 94305-2130, USA
| | - Mauri E Krouse
- The Cystic Fibrosis Research Laboratory, Stanford University, Stanford, CA 94305-2130, USA
| | - Jae Young Choi
- The Cystic Fibrosis Research Laboratory, Stanford University, Stanford, CA 94305-2130, USA.,Department of Otorhinolaryngology, Yonsei University, Seoul, Korea
| | - Hyung-Ju Cho
- The Cystic Fibrosis Research Laboratory, Stanford University, Stanford, CA 94305-2130, USA.,Department of Otorhinolaryngology, Yonsei University, Seoul, Korea
| | - Jeffrey J Wine
- The Cystic Fibrosis Research Laboratory, Stanford University, Stanford, CA 94305-2130, USA
| |
Collapse
|
15
|
Interleukin-13 Inhibits Lipopolysaccharide-Induced BPIFA1 Expression in Nasal Epithelial Cells. PLoS One 2015; 10:e0143484. [PMID: 26646664 PMCID: PMC4672888 DOI: 10.1371/journal.pone.0143484] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 11/05/2015] [Indexed: 02/01/2023] Open
Abstract
Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein is expressed in human nasopharyngeal and respiratory epithelium and has demonstrated antimicrobial activity. SPLUNC1 is now referred to as bactericidal/permeability-increasing fold containing family A, member 1 (BPIFA1). Reduced BPIFA1 expression is associated with bacterial colonization in patients with chronic rhinosinusitis with nasal polyps (CRSwNP). Interleukin 13 (IL-13), predominately secreted by T helper 2 (TH2) cells, has been found to contribute to airway allergies and suppress BPIFA1 expression in nasal epithelial cells. However, the molecular mechanism of IL-13 perturbation of bacterial infection and BPIFA1 expression in host airways remains unclear. In this study, we found that lipopolysaccharide (LPS)-induced BPIFA1 expression in nasal epithelial cells was mediated through the JNK/c-Jun signaling pathway and AP-1 activation. We further demonstrated that IL-13 downregulated the LPS-induced activation of phosphorylated JNK and c-Jun, followed by attenuation of BPIFA1 expression. Moreover, the immunohistochemical analysis showed that IL-13 prominently suppressed BPIFA1 expression in eosinophilic CRSwNP patients with bacterial infection. Taken together, these results suggest that IL-13 plays a critical role in attenuation of bacteria-induced BPIFA1 expression that may result in eosinophilic CRSwNP.
Collapse
|
16
|
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 2015; 100:363-9. [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] [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
|
17
|
Ghosh A, Boucher RC, Tarran R. Airway hydration and COPD. Cell Mol Life Sci 2015; 72:3637-52. [PMID: 26068443 PMCID: PMC4567929 DOI: 10.1007/s00018-015-1946-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 02/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the prevalent causes of worldwide mortality and encompasses two major clinical phenotypes, i.e., chronic bronchitis (CB) and emphysema. The most common cause of COPD is chronic tobacco inhalation. Research focused on the chronic bronchitic phenotype of COPD has identified several pathological processes that drive disease initiation and progression. For example, the lung's mucociliary clearance (MCC) system performs the critical task of clearing inhaled pathogens and toxic materials from the lung. MCC efficiency is dependent on: (1) the ability of apical plasma membrane ion channels such as the cystic fibrosis transmembrane conductance regulator (CFTR) and the epithelial Na(+) channel (ENaC) to maintain airway hydration; (2) ciliary beating; and (3) appropriate rates of mucin secretion. Each of these components is impaired in CB and likely contributes to the mucus stasis/accumulation seen in CB patients. This review highlights the cellular components responsible for maintaining MCC and how this process is disrupted following tobacco exposure and with CB. We shall also discuss existing therapeutic strategies for the treatment of chronic bronchitis and how components of the MCC can be used as biomarkers for the evaluation of tobacco or tobacco-like-product exposure.
Collapse
Affiliation(s)
- Arunava Ghosh
- Cystic Fibrosis Center/Marsico Lung Institute and the Department of Cell Biology and Physiology, The University of North Carolina, 7102 Marsico Hall, Chapel Hill, NC, 27599-7248, USA
| | - R C Boucher
- Cystic Fibrosis Center/Marsico Lung Institute and the Department of Cell Biology and Physiology, The University of North Carolina, 7102 Marsico Hall, Chapel Hill, NC, 27599-7248, USA
| | - Robert Tarran
- Cystic Fibrosis Center/Marsico Lung Institute and the Department of Cell Biology and Physiology, The University of North Carolina, 7102 Marsico Hall, Chapel Hill, NC, 27599-7248, USA.
| |
Collapse
|
18
|
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: 46] [Impact Index Per Article: 5.1] [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
|
19
|
Leeming GH, Kipar A, Hughes DJ, Bingle L, Bennett E, Moyo NA, Tripp RA, Bigley AL, Bingle CD, Sample JT, Stewart JP. Gammaherpesvirus infection modulates the temporal and spatial expression of SCGB1A1 (CCSP) and BPIFA1 (SPLUNC1) in the respiratory tract. J Transl Med 2015; 95:610-24. [PMID: 25531566 PMCID: PMC4450743 DOI: 10.1038/labinvest.2014.162] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/23/2014] [Accepted: 11/11/2014] [Indexed: 11/09/2022] Open
Abstract
Murine γ-herpesvirus 68 (MHV-68) infection of Mus musculus-derived strains of mice is an established model of γ-herpesvirus infection. We have previously developed an alternative system using a natural host, the wood mouse (Apodemus sylvaticus), and shown that the MHV-68 M3 chemokine-binding protein contributes significantly to MHV-68 pathogenesis. Here we demonstrate in A. sylvaticus using high-density micro-arrays that M3 influences the expression of genes involved in the host response including Scgb1a1 and Bpifa1 that encode potential innate defense proteins secreted into the respiratory tract. Further analysis of MHV-68-infected animals showed that the levels of both protein and RNA for SCGB1A1 and BPIFA1 were decreased at day 7 post infection (p.i.) but increased at day 14 p.i. as compared with M3-deficient and mock-infected animals. The modulation of expression was most pronounced in bronchioles but was also present in the bronchi and trachea. Double staining using RNA in situ hybridization and immunohistology demonstrated that much of the BPIFA1 expression occurs in club cells along with SCGB1A1 and that BPIFA1 is stored within granules in these cells. The increase in SCGB1A1 and BPIFA1 expression at day 14 p.i. was associated with the differentiation of club cells into mucus-secreting cells. Our data highlight the role of club cells and the potential of SCGB1A1 and BPIFA1 as innate defense mediators during respiratory virus infection.
Collapse
Affiliation(s)
- Gail H Leeming
- Department of Infection Biology, University of Liverpool, Liverpool, UK,Department of Veterinary Pathology, School of Veterinary Science, University of Liverpool, Liverpool, UK
| | - Anja Kipar
- Department of Infection Biology, University of Liverpool, Liverpool, UK,Department of Veterinary Pathology, School of Veterinary Science, University of Liverpool, Liverpool, UK,Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - David J Hughes
- Department of Infection Biology, University of Liverpool, Liverpool, UK
| | - Lynne Bingle
- Academic Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Elaine Bennett
- Department of Infection Biology, University of Liverpool, Liverpool, UK
| | - Nathifa A Moyo
- Department of Infection Biology, University of Liverpool, Liverpool, UK
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | - Alison L Bigley
- Investigative and Translational Pathology, AstraZeneca, R&D Innovative Medicines, Global Safety Assessment, Macclesfield, UK
| | - Colin D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Jeffery T Sample
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - James P Stewart
- Department of Infection Biology, University of Liverpool, Liverpool, UK,Department of Infection Biology, University of Liverpool, Liverpool Science Park IC2, 146 Brownlow Hill, Liverpool L3 5RF, UK. E-mail:
| |
Collapse
|
20
|
Lee B, Robinson KM, McHugh KJ, Scheller EV, Mandalapu S, Chen C, Di YP, Clay ME, Enelow RI, Dubin PJ, Alcorn JF. Influenza-induced type I interferon enhances susceptibility to gram-negative and gram-positive bacterial pneumonia in mice. Am J Physiol Lung Cell Mol Physiol 2015; 309:L158-67. [PMID: 26001778 DOI: 10.1152/ajplung.00338.2014] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/16/2015] [Indexed: 11/22/2022] Open
Abstract
Suppression of type 17 immunity by type I interferon (IFN) during influenza A infection has been shown to enhance susceptibility to secondary bacterial pneumonia. Although this mechanism has been described in coinfection with gram-positive bacteria, it is unclear whether similar mechanisms may impair lung defense against gram-negative infections. Furthermore, precise delineation of the duration of type I IFN-associated susceptibility to bacterial infection remains underexplored. Therefore, we investigated the effects of preceding influenza A virus infection on subsequent challenge with the gram-negative bacteria Escherichia coli or Pseudomonas aeruginosa and the temporal association between IFN expression with susceptibility to Staphylococcus aureus challenge in a mouse model of influenza and bacterial coinfection. Here we demonstrate that preceding influenza A virus led to increased lung E. coli and P. aeruginosa bacterial burden, which was associated with suppression of type 17 immunity and attenuation of antimicrobial peptide expression. Enhanced susceptibility to S. aureus coinfection ceased at day 14 of influenza infection, when influenza-associated type I IFN levels had returned to baseline levels, further suggesting a key role for type I IFN in coinfection pathogenesis. These findings further implicate type I IFN-associated suppression of type 17 immunity and antimicrobial peptide production as a conserved mechanism for enhanced susceptibility to both gram-positive and gram-negative bacterial coinfection during influenza infection.
Collapse
Affiliation(s)
- Benjamin Lee
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Keven M Robinson
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania; Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Kevin J McHugh
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Erich V Scheller
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Sivanarayana Mandalapu
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Chen Chen
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Y Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Michelle E Clay
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Richard I Enelow
- Department of Medicine, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Patricia J Dubin
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - John F Alcorn
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania;
| |
Collapse
|
21
|
Gao J, Ohlmeier S, Nieminen P, Toljamo T, Tiitinen S, Kanerva T, Bingle L, Araujo B, Rönty M, Höyhtyä M, Bingle CD, Mazur W, Pulkkinen V. Elevated sputum BPIFB1 levels in smokers with chronic obstructive pulmonary disease: a longitudinal study. Am J Physiol Lung Cell Mol Physiol 2015; 309:L17-26. [PMID: 25979078 DOI: 10.1152/ajplung.00082.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/12/2015] [Indexed: 01/22/2023] Open
Abstract
A previous study involving a proteomic screen of induced sputum from smokers and patients with chronic obstructive pulmonary disease (COPD) demonstrated elevated levels of bactericidal/permeability-increasing fold-containing protein B1 (BPIFB1). The aim of the present study was to further evaluate the association of sputum BPIFB1 levels with smoking and longitudinal changes in lung function in smokers with COPD. Sputum BPIFB1 was characterized by two-dimensional gel electrophoresis and mass spectrometry. The expression of BPIFB1 in COPD was investigated by immunoblotting and immunohistochemistry using sputum and lung tissue samples. BPIFB1 levels were also assessed in induced sputum from nonsmokers (n = 31), smokers (n = 169), and patients with COPD (n = 52) via an ELISA-based method. The longitudinal changes in lung function during the 4-year follow-up period were compared with the baseline sputum BPIFB1 levels. In lung tissue samples, BPIFB1 was localized to regions of goblet cell metaplasia. Secreted and glycosylated BPIFB1 was significantly elevated in the sputum of patients with COPD compared with that of smokers and nonsmokers. Sputum BPIFB1 levels correlated with pack-years and lung function as measured by forced expiratory volume in 1 s (FEV1) % predicted and FEV1/FVC (forced vital capacity) at baseline and after the 4-year follow-up in all participants. The changes in lung function over 4 years were significantly associated with BPIFB1 levels in current smokers with COPD. In conclusion, higher sputum concentrations of BPIFB1 were associated with changes of lung function over time, especially in current smokers with COPD. BPIFB1 may be involved in the pathogenesis of smoking-related lung diseases.
Collapse
Affiliation(s)
- J Gao
- HUCH Heart and Lung Center, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - S Ohlmeier
- Proteomics Core Facility, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - P Nieminen
- Medical Informatics and Statistics Group, University of Oulu, Oulu, Finland
| | - T Toljamo
- Department of Pulmonary Medicine, Lapland Central Hospital, Rovaniemi, Finland
| | | | - T Kanerva
- HUCH Heart and Lung Center, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - L Bingle
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK; and
| | - B Araujo
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK; and
| | - M Rönty
- HUSLAB, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - M Höyhtyä
- Medix Biochemica, Kauniainen, Finland
| | - C D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK; and
| | - W Mazur
- HUCH Heart and Lung Center, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - V Pulkkinen
- HUCH Heart and Lung Center, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland;
| |
Collapse
|
22
|
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: 1.6] [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
|
23
|
Zheng X, Cheng M, Fu B, Fan X, Wang Q, Yu X, Sun R, Tian Z, Wei H. Targeting LUNX inhibits non-small cell lung cancer growth and metastasis. Cancer Res 2015; 75:1080-90. [PMID: 25600649 DOI: 10.1158/0008-5472.can-14-1831] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
There remains a great need for effective therapies for lung cancer, the majority of which are non-small cell lung cancers (NSCLC). Here, we report the identification of a novel candidate therapeutic target, LUNX, as a molecule overexpressed in primary NSCLC and lymph node metastases that is associated with reduced postoperative survival. Functional studies demonstrated that LUNX overexpression promoted lung cancer cell migration and proliferation by interactions with the chaperone protein 14-3-3. Conversely, LUNX silencing disrupted primary tumor growth, local invasion, and metastatic colonization. The finding that LUNX was expressed on cell membranes prompted us to generate and characterize LUNX antibodies as a candidate therapeutic. Anti-LUNX could downregulate LUNX and reduce lung cancer cell proliferation and migration in vitro. Administered in vivo to mice bearing lung cancer xenografts, anti-LUNX could slow tumor growth and metastasis and improve mouse survival. Together, our work provides a preclinical proof of concept for LUNX as a novel candidate target for immunotherapy in lung cancer.
Collapse
Affiliation(s)
- Xiaohu Zheng
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Min Cheng
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Anhui Province Hospital Affiliated Anhui Medical University, Hefei, Anhui, China
| | - Binqing Fu
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaolei Fan
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Qing Wang
- Anhui Chest Hospital, Hefei, Anhui, China
| | - Xiaoqing Yu
- The First People's Hospital of Hefei, Hefei, Anhui, China
| | - Rui Sun
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.
| | - Haiming Wei
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
24
|
Zhang W, Zeng Z, Wei F, Chen P, Schmitt DC, Fan S, Guo X, Liang F, Shi L, Liu Z, Zhang Z, Xiang B, Zhou M, Huang D, Tang K, Li X, Xiong W, Tan M, Li G, Li X. SPLUNC1 is associated with nasopharyngeal carcinoma prognosis and plays an important role in all-trans-retinoic acid-induced growth inhibition and differentiation in nasopharyngeal cancer cells. FEBS J 2014; 281:4815-29. [PMID: 25161098 DOI: 10.1111/febs.13020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 06/19/2014] [Accepted: 08/22/2014] [Indexed: 12/13/2022]
Abstract
Human SPLUNC1 can suppress nasopharyngeal carcinoma (NPC) tumor formation; however, the correlation between SPLUNC1expression and NPC patient prognosis has not been reported. In the present study, we used a large-scale sample of 1015 tissue cores to detect SPLUNC1 expression and its association with patient prognosis. SPLUNC1 expression was reduced in NPC samples compared to nontumor nasopharyngeal epithelium tissues. Positive expression of SPLUNC1 in NPC predicted a better prognosis (disease-free survival, P = 0.034; overall survival, P = 0.048). Cox's proportional hazards model revealed that SPLUNC1 could be a significant prognostic factor affecting disease-free survival (P = 0.027). A cDNA micro-array analyzed by significant analysis of micro-array (SAM) and ingenuity pathway analysis (IPA) revealed that an indirect interaction existed between SPLUNC1 and retinoic acid (RA) in the cancer regulatory network. To further investigate the molecular mechanisms involved, we utilized several bioinformatics tools and identified 12 retinoid X receptors heterodimer binding sites in the promoter region of the SPLUNC1 gene. The transcriptional activity of the SPLUNC1 promoter was up-regulated significantly by all-trans-retinoic acid (ATRA). SPLUNC1 and retinoic acid receptor expression were induced significantly by ATRA, and removal of ATRA led to a progressive loss of SPLUNC1 and retinoic acid receptor expression. ATRA inhibited proliferation and induced the differentiation of NPC cells. Interestingly, over-expression of SPLUNC1 sensitized NPC cells to ATRA, whereas knockdown of SPLUNC1 in HNE1 cells increased cell viability. Under SPLUNC1 knockdown conditions, differentiation was reversed by ATRA treatment. We concluded that SPLUNC1 could potentially predict prognosis for NPC patients and play an important role in ATRA-induced growth inhibition and differentiation in NPC cells.
Collapse
Affiliation(s)
- Wenling Zhang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis of Ministry of Health and Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
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: 3.0] [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
|
26
|
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: 2.6] [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
|
27
|
Liu Y, Di ME, Chu HW, Liu X, Wang L, Wenzel S, Di YP. Increased susceptibility to pulmonary Pseudomonas infection in Splunc1 knockout mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:4259-68. [PMID: 24048904 DOI: 10.4049/jimmunol.1202340] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The airway epithelium is the first line of host defense against pathogens. The short palate, lung, and nasal epithelium clone (SPLUNC)1 protein is secreted in respiratory tracts and is a member of the bacterial/permeability increasing (BPI) fold-containing protein family, which shares structural similarities with BPI-like proteins. On the basis of its homology with BPIs and restricted expression of SPLUNC1 in serous cells of submucosal glands and surface epithelial cells of the upper respiratory tract, SPLUNC1 is thought to possess antimicrobial activity in host defense. SPLUNC1 is also reported to have surfactant properties, which may contribute to anti-biofilm defenses. The objective of this study was to determine the in vivo functions of SPLUNC1 following Pseudomonas aeruginosa infection and to elucidate the underlying mechanism by using a knockout (KO) mouse model with a genetic ablation of Splunc1. Splunc1 KO mice showed accelerated mortality and increased susceptibility to P. aeruginosa infection with significantly decreased survival rates, increased bacterial burdens, exaggerated tissue injuries, and elevated proinflammatory cytokine levels as compared with those of their wild-type littermates. Increased neutrophil infiltration in Splunc1 KO mice was accompanied by elevated chemokine levels, including Cxcl1, Cxcl2, and Ccl20. Furthermore, the expression of several epithelial secretory proteins and antimicrobial molecules was considerably suppressed in the lungs of Splunc1 KO mice. The deficiency of Splunc1 in mouse airway epithelium also results in increased biofilm formation of P. aeruginosa. Taken together, our results support that the ablation of Splunc1 in mouse airways affects the mucociliary clearance, resulting in decreased innate immune response during Pseudomonas-induced respiratory infection.
Collapse
Affiliation(s)
- Yanyan Liu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260
| | | | | | | | | | | | | |
Collapse
|
28
|
Britto CJ, Liu Q, Curran DR, Patham B, Dela Cruz CS, Cohn L. Short palate, lung, and nasal epithelial clone-1 is a tightly regulated airway sensor in innate and adaptive immunity. Am J Respir Cell Mol Biol 2013; 48:717-24. [PMID: 23470624 DOI: 10.1165/rcmb.2012-0072oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Short palate, lung, and nasal epithelial clone-1 (SPLUNC1) is a protein abundantly expressed by the respiratory epithelium of the proximal lower respiratory tract, a site of great environmental exposure. Previous studies showed that SPLUNC1 exerts antimicrobial effects, regulates airway surface liquid and mucociliary clearance, and suppresses allergic airway inflammation. We studied SPLUNC1 to gain insights into its role in host defense. In the lower respiratory tract, concentrations of SPLUNC1 are high under basal conditions. In models of pneumonia caused by common respiratory pathogens, and in Th1-induced and Th2-induced airway inflammation, SPLUNC1 secretion is markedly reduced. Pathogen-associated molecular patterns and IFN-γ act directly on airway epithelial cells to inhibit SPLUNC1 mRNA expression. Thus, SPLUNC1 is quickly suppressed during infection, in response to an insult on the epithelial surface. These experiments highlight the finely tuned fluctuations of SPLUNC1 in response to exposures in the respiratory tract, and suggest that the loss of SPLUNC1 is a crucial feature of host defense across air-breathing animal species.
Collapse
Affiliation(s)
- Clemente J Britto
- Section of Pulmonary and Critical Care, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | |
Collapse
|
29
|
Mawson AR. Role of Fat-Soluble Vitamins A and D in the Pathogenesis of Influenza: A New Perspective. ACTA ACUST UNITED AC 2013. [DOI: 10.5402/2013/246737] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Reduced exposure to solar radiation, leading to a deficiency of vitamin D and hence impaired innate immunity, has been suggested as a trigger for influenza viral replication and as an explanation of seasonal influenza. Although this hypothesis accounts for many unexplained facts about the epidemiology of influenza, gaps remain in understanding the pathogenesis and manifestations of the disease. Several observations suggest a role for vitamin A compounds (retinoids) in the disease. This paper presents a new model of the etiopathogenesis of influenza, suggesting that host resistance and susceptibility depend importantly on the ratio of vitamin D to vitamin A activity. Retinoid concentrations within normal physiological limits appear to inhibit influenza pathogenesis whereas higher background concentrations (i.e., very low vitamin D : A ratios) increase the risk of severe complications of the disease. There is also evidence that influenza-induced or preexisting liver disease, diabetes, and obesity worsen the severity of infection, possibly via liver dysfunction and alterations in retinoid metabolism. The model could be tested by determining the presence of retinoids in the secretions of patients with influenza and by studies of retinoid profiles in patients and controls. Potential strategies for prevention and treatment are discussed.
Collapse
Affiliation(s)
- Anthony R. Mawson
- Department of Health Policy and Management, School of Health Sciences, College of Public Service, Jackson State University,
350 West Woodrow Wilson Avenue, Room 229, Jackson, MS 39213, USA
| |
Collapse
|
30
|
Jiang D, Wenzel SE, Wu Q, Bowler RP, Schnell C, Chu HW. Human neutrophil elastase degrades SPLUNC1 and impairs airway epithelial defense against bacteria. PLoS One 2013; 8:e64689. [PMID: 23741370 PMCID: PMC3669426 DOI: 10.1371/journal.pone.0064689] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 04/17/2013] [Indexed: 12/13/2022] Open
Abstract
Background Acute exacerbations of chronic obstructive pulmonary disease (AECOPD) are a significant cause of mortality of COPD patients, and pose a huge burden on healthcare. One of the major causes of AECOPD is airway bacterial (e.g. nontypeable Haemophilus influenzae [NTHi]) infection. However, the mechanisms underlying bacterial infections during AECOPD remain poorly understood. As neutrophilic inflammation including increased release of human neutrophil elastase (HNE) is a salient feature of AECOPD, we hypothesized that HNE impairs airway epithelial defense against NTHi by degrading airway epithelial host defense proteins such as short palate, lung, and nasal epithelium clone 1 (SPLUNC1). Methodology/Main Results Recombinant human SPLUNC1 protein was incubated with HNE to confirm SPLUNC1 degradation by HNE. To determine if HNE-mediated impairment of host defense against NTHi was SPLUNC1-dependent, SPLUNC1 protein was added to HNE-treated primary normal human airway epithelial cells. The in vivo function of SPLUNC1 in NTHi defense was investigated by infecting SPLUNC1 knockout and wild-type mice intranasally with NTHi. We found that: (1) HNE directly increased NTHi load in human airway epithelial cells; (2) HNE degraded human SPLUNC1 protein; (3) Recombinant SPLUNC1 protein reduced NTHi levels in HNE-treated human airway epithelial cells; (4) NTHi levels in lungs of SPLUNC1 knockout mice were increased compared to wild-type mice; and (5) SPLUNC1 was reduced in lungs of COPD patients. Conclusions Our findings suggest that SPLUNC1 degradation by neutrophil elastase may increase airway susceptibility to bacterial infections. SPLUNC1 therapy likely attenuates bacterial infections during AECOPD.
Collapse
Affiliation(s)
- Di Jiang
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Sally E. Wenzel
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Qun Wu
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Russell P. Bowler
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Christina Schnell
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
31
|
LPLUNC1 suppresses IL-6-induced nasopharyngeal carcinoma cell proliferation via inhibiting the Stat3 activation. Oncogene 2013; 33:2098-109. [PMID: 23708661 DOI: 10.1038/onc.2013.161] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 03/02/2013] [Accepted: 03/14/2013] [Indexed: 12/28/2022]
Abstract
Tumor-associated macrophage (TAM)-related chronic inflammation and interleukin-6 (IL-6) contribute to the progression of nasopharyngeal carcinoma (NPC). In this study, we characterized TAMs and IL-6 expression in 212 biopsied NPC and 119 non-tumor nasopharyngeal epithelium (NPE) tissues by tissue array. In comparison with that in the NPE tissues, more TAM infiltrates and a higher density of IL-6 expression were detected in NPC tissues, which were associated with the poor survival of NPC patients. In contrast, little or no LPLUNC1, a regulator of inflammation, expression was detected in NPC tissues, and the levels of LPLUNC1 expression in the NPC were associated negatively with the numbers of TAMs and the levels of IL-6 expression, but positively with the survival of NPC patients. Induction of LPLUNC1 overexpression in NPC cells mitigated lipopolysaccharide (LPS)-induced IL-6, IL-8, tumor necrosis factor-α and IL-1β expression or treatment of THP-1 macrophages with LPLUNC1 inhibited spontaneous and LPS-induced IL-6 expression in vitro. IL-6-promoted NPC cell proliferation in a dose- and time-dependent manner, accompanied by increasing cyclin D1 and Bcl-2 expression and the Stat3 activation, but inhibiting Bax and p21 expression. Induction of LPLUNC1 overexpression inhibited NPC cell proliferation, induced NPC cell arrest, promoted NPC cell apoptosis even after IL-6 stimulation and inhibited the growth of implanted NPC tumors in vivo, which were associated with decreasing cyclin D1 and Bcl-2 expression and the Janus kinase 2 (JAK2)/Stat3 activation, but enhancing Bax and p21 expression. These results suggest that LPLUNC1 can inhibit inflammation and NPC growth by downregulating the Stat3 pathway.
Collapse
|
32
|
Tsou YA, Peng MT, Wu YF, Lai CH, Lin CD, Tai CJ, Tsai MH, Chen CM, Chen HC. Decreased PLUNC expression in nasal polyps is associated with multibacterial colonization in chronic rhinosinusitis patients. Eur Arch Otorhinolaryngol 2013; 271:299-304. [PMID: 23644997 DOI: 10.1007/s00405-013-2535-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 04/25/2013] [Indexed: 10/26/2022]
Abstract
PLUNC (palate, lung, and nasal epithelium clone) is an epithelium-secreted protein that plays a crucial role in the host's defense against bacterial infection. The function of PLUNC in the sinus remains poorly understood. To examine whether the expression levels of PLUNC could serve as a predictive outcome biomarker for patients with CRSwNP and bacterial colonization, we investigated the association of PLUNC expression levels with bacterial colonization in the sinuses. A total of 174 patients who underwent sinus surgery for chronic rhinosinusitis with nasal polyps (CRSwNP) were enrolled in this study. The tissue samples obtained from patients were examined using preoperative sinus computed tomography (CT) scans, postoperative bacterial cultures, and nasal polyp examinations. PLUNC mRNA and protein expression were quantified using RT-PCR and immunohistochemistry. We identified that decreased PLUNC expression is associated with multibacterial colonization (P = 0.0001), specifically those mediated by Staphyloccocus aureus (P = 0.037) and Pseudomonas aeruginosa (P = 0.002). The patients who required repeated sinus surgeries for recurrent or persistent sinusitis also presented much lower PLUNC expression than those who did not require repeated sinus surgery (P = 0.001). However, gender, age, and CT scores were not associated with PLUNC expression. These results suggest that reduced PLUNC expression is associated with bacterial colonization as well as treatment outcome in CRSwNP patients. Investigation of the association between PLUNC expressions and chronic rhinosinusitis may lead to the development of a novel biomarker for treatment outcome in CRSwNP patients.
Collapse
Affiliation(s)
- Yung-An Tsou
- Department of Otolaryngology-Head and Neck Surgery, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Liu Y, Bartlett JA, Di ME, Bomberger JM, Chan YR, Gakhar L, Mallampalli RK, McCray PB, Di YP. SPLUNC1/BPIFA1 contributes to pulmonary host defense against Klebsiella pneumoniae respiratory infection. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1519-31. [PMID: 23499554 DOI: 10.1016/j.ajpath.2013.01.050] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 01/10/2013] [Accepted: 01/14/2013] [Indexed: 12/12/2022]
Abstract
Epithelial host defense proteins comprise a critical component of the pulmonary innate immune response to infection. The short palate, lung, nasal epithelium clone (PLUNC) 1 (SPLUNC1) protein is a member of the bactericidal/permeability-increasing (BPI) fold-containing (BPIF) protein family, sharing structural similarities with BPI-like proteins. SPLUNC1 is a 25 kDa secretory protein that is expressed in nasal, oropharyngeal, and lung epithelia, and has been implicated in airway host defense against Pseudomonas aeruginosa and other organisms. SPLUNC1 is reported to have surfactant properties, which may contribute to anti-biofilm defenses. The objective of this study was to assess the importance of SPLUNC1 surfactant activity in airway epithelial secretions and to explore its biological relevance in the context of a bacterial infection model. Using cultured airway epithelia, we confirmed that SPLUNC1 is critically important for maintenance of low surface tension in airway fluids. Furthermore, we demonstrated that recombinant SPLUNC1 (rSPLUNC1) significantly inhibited Klebsiella pneumoniae biofilm formation on airway epithelia. We subsequently found that Splunc1(-/-) mice were significantly more susceptible to infection with K. pneumoniae, confirming the likely in vivo relevance of this anti-biofilm effect. Our data indicate that SPLUNC1 is a crucial component of mucosal innate immune defense against pulmonary infection by a relevant airway pathogen, and provide further support for the novel hypothesis that SPLUNC1 protein prevents bacterial biofilm formation through its ability to modulate surface tension of airway fluids.
Collapse
Affiliation(s)
- Yang Liu
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sayeed S, Nistico L, St Croix C, Di YP. Multifunctional role of human SPLUNC1 in Pseudomonas aeruginosa infection. Infect Immun 2013; 81:285-91. [PMID: 23132494 PMCID: PMC3536124 DOI: 10.1128/iai.00500-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 10/24/2012] [Indexed: 12/14/2022] Open
Abstract
The human short PLUNC1 (SPLUNC1) protein has been identified as a component of the pulmonary antimicrobial response based on its structural similarity to the bactericidal/permeability-increasing (BPI) protein. Using a genetically modified mouse model, we recently verified the antimicrobial activity of SPLUNC1 against Pseudomonas aeruginosa in vivo. To further define the mechanism of epithelial SPLUNC1-mediated antibacterial action, we carried out studies to determine how SPLUNC1 protects the host from acute respiratory infections. P. aeruginosa treated with recombinant human SPLUNC1 protein showed decreased growth in vitro. This antibacterial activity was due to growth inhibition, as a consequence of a SPLUNC1-induced increase in bacterial cell permeability. Removal of SPLUNC1 allowed the recovery of P. aeruginosa and suggested no permanent cell injury or direct killing of bacteria. Further investigation showed coating of bacterial cells by SPLUNC1. We suggest that this "bacterial cell coating" is necessary for the bacteriostatic function of SPLUNC1. Additionally, we demonstrated a novel role for SPLUNC1 as a chemoattractant that facilitated migration of macrophages and neutrophils. Taking the findings together, we propose synergistic roles for human SPLUNC1 as an antibacterial agent with bacteriostatic and chemotactic activities.
Collapse
Affiliation(s)
- Sameera Sayeed
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Laura Nistico
- Allegheny Singer Research Institute, Allegheny General Hospital, Pittsburgh, Pennsylvania, USA
| | - Claudette St Croix
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Y. Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
35
|
GUO XF, CHEN P, LI XY, LI XL, LI GY. The Structure and Function of SPLUNC1:Novel Class of Innate Immune Protective Molecules*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2011.00436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Abstract
Airway epithelial healing is defined as restoration of health or soundness; to cure. Our research indicates that two types of progenitor cells participate in this process: the tissue-specific stem cell (TSC) and the facultative basal progenitor (FBP). The TSC restores the epithelium to its normal structure and function. Thus, the TSC regenerates the epithelium. In contrast, the FBP-derived epithelium is characterized by regions of cellular hyperplasia and hypoplasia. Since the FBP-derived epithelium deviates from normal, we term the FBP-mediated process repair. Our work indicates that the TSC responds to signals from other epithelial cells, including the FBP. These signals instruct the TSC to proliferate or to select one of several differentiation pathways. We interpret these data in the context of Stephen Padget's "seed and soil" paradigm. Therein, Padget explained that metastasis of a tumor, the seed, to a specific site, the soil, was determined by the growth and differentiation requirements of the tumor cell. By extending the seed and soil paradigm to airway epithelial healing, we suggest that proliferation and differentiation of the TSC, the seed, is determined by its interactions with other cell types, the soil. Based on this concept, we provide a set of suggestions for development of cell-based therapies that are directed toward chronic airways disease.
Collapse
|
37
|
Di YP, Zhao J, Harper R. Cigarette smoke induces MUC5AC protein expression through the activation of Sp1. J Biol Chem 2012; 287:27948-58. [PMID: 22700966 DOI: 10.1074/jbc.m111.334375] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cigarette smoke (CS) exposure is associated with increased mucus production and chronic obstructive pulmonary disease (COPD). MUC5AC is the major inducible mucus gene in the airway. The purpose of this investigation was to elucidate the mechanisms of CS-induced activation of MUC5AC gene transcription. We observed that the region -3724/-3224 of the MUC5AC promoter is critical for CS-induced gene transcriptional activity and that this region contains two Sp1 binding sites. Using a lung-relevant model, we observed that CS increased nuclear Sp1 protein expression. Consequently, CS exposure resulted in enhanced Sp1-DNA binding activity and Sp1 trans-activation. Co-transfection of the MUC5AC-luc reporter with Sp1 expression plasmids resulted in significantly increased MUC5AC-luc activity, whereas co-treatment with mithramycin A, a Sp1 inhibitor, abolished CS-induced MUC5AC promoter activity. Using mobility shift assay and chromatin immunoprecipitation, we demonstrated that two Sp1 binding sites in the MUC5AC promoter are functional and responsive to CS exposure. A mutation of either Sp1 binding site in the MUC5AC promoter significantly decreased CS-induced promoter activity. Together, these data indicate that CS induces MUC5AC gene transcription predominantly through increased Sp1 nuclear protein levels and increased Sp1 binding to its promoter region.
Collapse
Affiliation(s)
- Y Peter Di
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA.
| | | | | |
Collapse
|
38
|
Abstract
PLUNC (palate, lung and nasal epithelium clone) protein is an abundant secretory product of epithelia throughout the mammalian conducting airways. Despite its homology with the innate immune defence molecules BPI (bactericidal/permeability-increasing protein) and LBP (lipopolysaccharide-binding protein), it has been difficult to define the functions of PLUNC. Based on its marked hydrophobicity and expression pattern, we hypothesized that PLUNC is an airway surfactant. We found that purified recombinant human PLUNC exhibited potent surfactant activity by several different measures, and experiments with airway epithelial cell lines and primary cultures indicate that native PLUNC makes a significant contribution to the overall surface tension in airway epithelial secretions. Interestingly, we also found that physiologically relevant concentrations of PLUNC-inhibited Pseudomonas aeruginosa biofilm formation in vitro without acting directly as a bactericide. This finding suggests that PLUNC protein may inhibit biofilm formation by airway pathogens, perhaps through its dispersant properties. Our data, along with reports from other groups on activity against some airway pathogens, expand on an emerging picture of PLUNC as a multifunctional protein, which plays a novel role in airway defences at the air/liquid interface.
Collapse
|
39
|
Functional roles of SPLUNC1 in the innate immune response against Gram-negative bacteria. Biochem Soc Trans 2011; 39:1051-5. [PMID: 21787346 DOI: 10.1042/bst0391051] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PLUNC (palate, lung and nasal epithelium clone)-associated gene originally referred to one gene, but now has been extended to represent a gene family that consists of a number of genes with peptide sequence homologies and predicted structural similarities. PLUNC-like proteins display sequence homology with BPI (bactericidal/permeability-increasing protein), a 456-residue cationic protein produced by precursors of polymorphonuclear leucocytes that have been shown to possess both bactericidal and LPS (lipopolysaccharide)-binding activities. The human PLUNC is also known as LUNX (lung-specific X protein), NASG (nasopharyngeal carcinoma-related protein) and SPURT (secretory protein in upper respiratory tract). The gene originally named PLUNC is now recognized as SPLUNC1. Its gene product SPLUNC1 is a secretory protein that is abundantly expressed in cells of the surface epithelium in the upper respiratory tracts and secretory glands in lung, and in the head and the neck region. The functional role of SPLUNC1 in innate immunity has been suggested but not clearly defined. The present review describes recent findings that support antimicrobial and anti-inflammatory functions of SPLUNC1 in Gram-negative bacteria-induced respiratory infection.
Collapse
|
40
|
Quantitation of SPLUNC1 in saliva with an xMAP particle-based antibody capture and detection immunoassay. Arch Oral Biol 2011; 57:197-204. [PMID: 21925642 DOI: 10.1016/j.archoralbio.2011.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 08/17/2011] [Accepted: 08/21/2011] [Indexed: 11/22/2022]
Abstract
The short palate lung and nasal epithelial clone 1 (SPLUNC1) protein may be differentially expressed in oral infections, oral inflammatory disorders, or oral malignancies and may be involved in innate immune responses in the oral cavity. However, the actual concentration of SPLUNC1 in saliva has not previously been determined. In this study, we determined the concentrations of SPLUNC1 in saliva using a particle-based antibody capture and detection immunoassay. A commercial goat anti-rhSPLUNC1 polyclonal antibody (AF1897) was linked to fluorescent polystyrene microspheres and used as the capture antibody. A commercial mouse IgG2b anti-rhSPLUNC1 monoclonal antibody (MAB1897) was biotinylated and used as the detection antibody. Western blot and 2-dimensional fluorescence difference gel electrophoresis (2-D DIGE) analysis of immunoprecipitated rhSPLUNC1 and SPLUNC1 from saliva were used to show that the capture AF1897 and detection MAB1897 antibodies both recognized SPLUNC1. Protein concentrations in saliva from 20 subjects ranged from 0.9 to 23.9mg/ml; SPLUNC1 concentrations ranged from 34.7ng/ml to 13.8μg/ml; and SPLUNC concentrations normalized per mg of total salivary protein ranged from 4.7ng/ml to 5.3μg/ml. These results show that SPLUNC1 is detected in saliva in a variety of concentrations. This immunoassay may prove to be useful in determining the concentration of SPLUNC1 in saliva for assessing its role in the pathogenesis of oral infections, oral inflammatory disorders, or oral malignancies.
Collapse
|
41
|
Fornander L, Ghafouri B, Kihlström E, Åkerlind B, Schön T, Tagesson C, Lindahl M. Innate immunity proteins and a new truncated form of SPLUNC1 in nasopharyngeal aspirates from infants with respiratory syncytial virus infection. Proteomics Clin Appl 2011; 5:513-22. [DOI: 10.1002/prca.201100016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 06/16/2011] [Accepted: 07/11/2011] [Indexed: 11/07/2022]
|
42
|
Abstract
Although gene expression studies have shown that human PLUNC (palate, lung and nasal epithelium clone) proteins are predominantly expressed in the upper airways, nose and mouth, and proteomic studies have indicated they are secreted into airway and nasal lining fluids and saliva, there is currently little information concerning the localization of human PLUNC proteins. Our studies have focused on the localization of three members of this protein family, namely SPLUNC1 (short PLUNC1), SPLUNC2 and LPLUNC1 (long PLUNC1). Western blotting has indicated that PLUNC proteins are highly glycosylated, whereas immunohistochemical analysis demonstrated distinct patterns of expression. For example, SPLUNC2 is expressed in serous cells of the major salivary glands and in minor mucosal glands, whereas SPLUNC1 is expressed in the mucous cells of these glands. LPLUNC1 is a product of a population of goblet cells in the airway epithelium and nasal passages and expressed in airway submucosal glands and minor glands of the oral and nasal cavities. SPLUNC1 is also found in the epithelium of the upper airways and nasal passages and in airway submucosal glands, but is not co-expressed with LPLUNC1. We suggest that this differential expression may be reflected in the function of individual PLUNC proteins.
Collapse
|
43
|
Lukinskiene L, Liu Y, Reynolds SD, Steele C, Stripp BR, Leikauf GD, Kolls JK, Di YP. Antimicrobial activity of PLUNC protects against Pseudomonas aeruginosa infection. THE JOURNAL OF IMMUNOLOGY 2011; 187:382-90. [PMID: 21632717 DOI: 10.4049/jimmunol.1001769] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Epithelial antimicrobial activity may protect the lung against inhaled pathogens. The bactericidal/permeability-increasing protein family has demonstrated antimicrobial activity in vitro. PLUNC (palate, lung, and nasal epithelium associated) is a 25-kDa secreted protein that shares homology with bactericidal/permeability-increasing proteins and is expressed in nasopharyngeal and respiratory epithelium. The objective of this study was to determine whether PLUNC can limit Pseudomonas aeruginosa infection in mice. Transgenic mice (Scgb1a1-hPLUNC) were generated in which human PLUNC (hPLUNC) was directed to the airway epithelium with the Scgb1a1 promoter. The hPLUNC protein (hPLUNC) was detected in the epithelium throughout the trachea and bronchial airways and in bronchoalveolar lavage fluid. Bronchoalveolar lavage fluid from transgenic mice exhibited higher antibacterial activity than that from wild type littermates in vitro. After in vivo P. aeruginosa challenge, Scgb1a1-hPLUNC transgenic mice displayed enhanced bacterial clearance. This was accompanied by a decrease in neutrophil infiltration and cytokine levels. More importantly, the overexpressed hPLUNC in Scgb1a1-hPLUNC transgenic mouse airway significantly enhanced mouse survival against P. aeruginosa-induced respiratory infection. These data indicate that PLUNC is a novel antibacterial protein that likely plays a critical role in airway epithelium-mediated innate immune response.
Collapse
Affiliation(s)
- Lina Lukinskiene
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lee BY, Cho S, Shin DH, Kim H. Genome-wide association study of copy number variations associated with pulmonary function measures in Korea Associated Resource (KARE) cohorts. Genomics 2010; 97:101-5. [PMID: 21059387 DOI: 10.1016/j.ygeno.2010.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/29/2010] [Accepted: 11/02/2010] [Indexed: 01/13/2023]
Abstract
Copy number variation (CNV) is an attractive emerging approach to study the association with various diseases. We performed a CNV-based genome-wide association study of pulmonary function measures (FEV(1), FVC, and FEV(1)/FVC) in KARE cohorts. Affymetrix Genome-wide Human SNP Array 5.0 was used to measure genome-wide variation and CNV segmentation was performed using Golden Helix SVS 7.0. Single and multivariate regressions were used for the association study using the R statistical package and the Dabatase for Annotation, Visualization and Integrated (DAVID v6.7b) tool for the functional annotation. We identified significantly associated 1260 CNVs with pulmonary function measures of FEV(1) and FVC. Functional gene classification and annotation analysis found 5 highly enriched clusters, the BPI/LBP/Plunc superfamily, myosin, serpin peptidase inhibitor, protein tyrosine phosphatase, and olfactory receptors. According to the functional annotation, gene-based CNVs are likely to be involved in the pathogenesis and inflammatory responsiveness of pulmonary diseases.
Collapse
Affiliation(s)
- Bo-Young Lee
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, San 56-1, Sillim-dong, Gwanak-gu, Seoul 151-742, Republic of Korea.
| | | | | | | |
Collapse
|
45
|
Chu HW, Gally F, Thaikoottathil J, Janssen-Heininger YM, Wu Q, Zhang G, Reisdorph N, Case S, Minor M, Smith S, Jiang D, Michels N, Simon G, Martin RJ. SPLUNC1 regulation in airway epithelial cells: role of Toll-like receptor 2 signaling. Respir Res 2010; 11:155. [PMID: 21054862 PMCID: PMC2992501 DOI: 10.1186/1465-9921-11-155] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 11/05/2010] [Indexed: 12/15/2022] Open
Abstract
Background Respiratory infections including Mycoplasma pneumoniae (Mp) contribute to various chronic lung diseases. We have shown that mouse short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein was able to inhibit Mp growth. Further, airway epithelial cells increased SPLUNC1 expression upon Mp infection. However, the mechanisms underlying SPLUNC1 regulation remain unknown. In the current study, we investigated if SPLUNC1 production following Mp infection is regulated through Toll-like receptor 2 (TLR2) signaling. Methods Airway epithelial cell cultures were utilized to reveal the contribution of TLR2 signaling including NF-κB to SPLUNC1 production upon bacterial infection and TLR2 agonist stimulation. Results Mp and TLR2 agonist Pam3CSK4 increased SPLUNC1 expression in tracheal epithelial cells from wild type, but not TLR2-/- BALB/c mice. RNA interference (short-hairpin RNA) of TLR2 in normal human bronchial epithelial cells under air-liquid interface cultures significantly reduced SPLUNC1 levels in Mp-infected or Pam3CSK4-treated cells. Inhibition and activation of NF-κB pathway decreased and increased SPLUNC1 production in airway epithelial cells, respectively. Conclusions Our data for the first time suggest that airway epithelial TLR2 signaling is pivotal in mycoplasma-induced SPLUNC1 production, thus improving our understanding of the aberrant SPLUNC1 expression in airways of patients suffering from chronic lung diseases with bacterial infections.
Collapse
Affiliation(s)
- Hong Wei Chu
- Department of Medicine, National Jewish Health, and the University of Colorado Denver, Denver, CO, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wright PL, Yu J, Di YPP, Homer RJ, Chupp G, Elias JA, Cohn L, Sessa WC. Epithelial reticulon 4B (Nogo-B) is an endogenous regulator of Th2-driven lung inflammation. ACTA ACUST UNITED AC 2010; 207:2595-607. [PMID: 20975041 PMCID: PMC2989775 DOI: 10.1084/jem.20100786] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The reticulon protein Nogo-B is highly expressed in the lungs, and its loss augments lung inflammation in part as a result of decreased expression of the antiinflammatory protein PLUNC. Nogo-B is a member of the reticulon family of proteins (RTN-4B) that is highly expressed in lung tissue; however, its function remains unknown. We show that mice with Th2-driven lung inflammation results in a loss of Nogo expression in airway epithelium and smooth muscle compared with nonallergic mice, a finding which is replicated in severe human asthma. Mice lacking Nogo-A/B (Nogo-KO) display an exaggerated asthma-like phenotype, and epithelial reconstitution of Nogo-B in transgenic mice blunts Th2-mediated lung inflammation. Microarray analysis of lungs from Nogo-KO mice reveals a marked reduction in palate lung and nasal clone (PLUNC) gene expression, and the levels of PLUNC are enhanced in epithelial Nogo-B transgenic mice. Finally, transgenic expression of PLUNC into Nogo-KO mice rescues the enhanced asthmatic-like responsiveness in these KO mice. These data identify Nogo-B as a novel protective gene expressed in lung epithelia, and its expression regulates the levels of the antibacterial antiinflammatory protein PLUNC.
Collapse
Affiliation(s)
- Paulette L Wright
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
The multifunctional host defense peptide SPLUNC1 is critical for homeostasis of the mammalian upper airway. PLoS One 2010; 5:e13224. [PMID: 20949060 PMCID: PMC2951362 DOI: 10.1371/journal.pone.0013224] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 09/14/2010] [Indexed: 12/15/2022] Open
Abstract
Otitis media (OM) is a highly prevalent pediatric disease caused by normal flora of the nasopharynx that ascend the Eustachian tube and enter the middle ear. As OM is a disease of opportunity, it is critical to gain an increased understanding of immune system components that are operational in the upper airway and aid in prevention of this disease. SPLUNC1 is an antimicrobial host defense peptide that is hypothesized to contribute to the health of the airway both through bactericidal and non-bactericidal mechanisms. We used small interfering RNA (siRNA) technology to knock down expression of the chinchilla ortholog of human SPLUNC1 (cSPLUNC1) to begin to determine the role that this protein played in prevention of OM. We showed that knock down of cSPLUNC1 expression did not impact survival of nontypeable Haemophilus influenzae, a predominant causative agent of OM, in the chinchilla middle ear under the conditions tested. In contrast, expression of cSPLUNC1 was essential for maintenance of middle ear pressure and efficient mucociliary clearance, key defense mechanisms of the tubotympanum. Collectively, our data have provided the first in vivo evidence that cSPLUNC1 functions to maintain homeostasis of the upper airway and, thereby, is critical for protection of the middle ear.
Collapse
|
48
|
Yeh TH, Lee SY, Hsu WC. Expression of SPLUNC1 protein in nasal polyp epithelial cells in air-liquid interface culture treated with IL-13. Am J Rhinol Allergy 2010; 24:17-20. [PMID: 20109312 DOI: 10.2500/ajra.2010.24.3381] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) protein is an airway epithelial cell-derived molecule exerting host defense against pathogen. However, the function and regulation of SPLUNC1 in nasal epithelial cells are still unclear. Chronic rhinosinusitis with nasal polyps (CRSwNPs) is a disorder characterized by eosinophilic Th2 inflammation and frequent microbial colonization. The pathogenesis has been postulated as a disturbed mucosal immune response. This study investigates the SPLUNC1 expression of nasal polyp epithelial cells in air-liquid interface (ALI) culture and after treating with Th2 inflammatory cytokines IL-13. METHODS Human nasal polyp epithelial cells isolated from patients with CRSwNPs were put in different cell culture models at days 0 and 21 and were assessed for expression of SPLUNC1 by microarray. Cultured cells in ALI plus retinoic acid (ALI + RA) model were then incubated with 0, 1, 10, and 100 ng/mL human recombinant IL-13 for up to 5 days. The expression of SPLUNC1 was assessed by real-time quantitative polymerase chain reaction (RT-Q-PCR), reverse-transcriptase PCR (RT-PCR) and Western blot analysis. RESULTS ALI + RA culture model harvesting ciliary differentiated nasal epithelial cells constitutively expressed high levels of SPLUNC1. In contrast, SPLUNC1 is reduced under classic submerged single layer culture. SPLUNC1 is also dose-responsively down-regulated after incubation with IL-13. CONCLUSION A microenvironmental milieu containing IL-13 may be detrimental to the host innate immunity response, at least in part, through the inhibition of SPLUNC1 production.
Collapse
Affiliation(s)
- Te-Huei Yeh
- Department of Otolaryngology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | | | | |
Collapse
|
49
|
Linderholm AL, Onitsuka J, Xu C, Chiu M, Lee WM, Harper RW. All-trans retinoic acid mediates DUOX2 expression and function in respiratory tract epithelium. Am J Physiol Lung Cell Mol Physiol 2010; 299:L215-21. [PMID: 20511343 DOI: 10.1152/ajplung.00015.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
DUOX1 and DUOX2 are members of the NADPH oxidase family that are specifically regulated to produce hydrogen peroxide in epithelia of the thyroid, gastrointestinal tract, and respiratory tract. The determinants of DUOX1 or DUOX2 expression in various tissues have not been established. Using respiratory tract epithelial cells as a model, we investigated changes in DUOX mRNA and protein expression during the first 10 days of differentiation. By comparing a respiratory tract cell line, HBE1, with primary tracheobronchial epithelial (TBE) cells, we determined that DUOX2 was significantly expressed only in cell conditions that included all-trans retinoic acid (ATRA). In HBE1 cells, DUOX2 mRNA increased 6-fold after ATRA treatment. Similarly, ATRA induced a 19-fold increase in DUOX2 mRNA expression in primary TBE cells with parallel increases in DUOX protein and DUOX-mediated H(2)O(2) production as well. In addition, DUOX2 induction by rhinovirus required the presence of ATRA. ATRA had no effect on DUOX1 expression for all the conditions studied. Our data indicate that for respiratory epithelial cells, ATRA is important in the regulation of DUOX2 expression, function, and rhinovirus-mediated DUOX2 inducibility.
Collapse
|
50
|
Human LPLUNC1 is a secreted product of goblet cells and minor glands of the respiratory and upper aerodigestive tracts. Histochem Cell Biol 2010; 133:505-15. [PMID: 20237794 PMCID: PMC2852594 DOI: 10.1007/s00418-010-0683-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2010] [Indexed: 11/13/2022]
Abstract
Long PLUNC1 (LPLUNC1, C20orf114) is a member of a family of poorly described proteins (PLUNCS) expressed in the upper respiratory tract and oral cavity, which may function in host defence. Although it is one of the most highly expressed genes in the upper airways and has been identified in sputum and nasal secretions by proteomic studies, localisation of LPLUNC1 protein has not yet been described. We developed affinity purified antibodies and localised the protein in tissues of the human respiratory tract, oro- and nasopharynx. We have complemented these studies with analysis of LPLUNC1 expression in primary human lung cell cultures and used Western blotting to study the protein in cell culture secretions and in BAL. LPLUNC1 is a product of a population of goblet cells in the airway epithelium and nasal passages and is also present in airway submucosal glands and minor glands of the oral and nasal cavities. The protein is not expressed in peripheral lung epithelial cells. LPLUNC1 is present in bronchoalveolar lavage fluid as two glycosylated isoforms and primary airway epithelial cells produce identical proteins as they undergo mucociliary differentiation. Our results suggest that LPLUNC1 is an abundant, secreted product of goblet cells and minor mucosal glands of the respiratory tract and oral cavity and suggest that the protein functions in the complex milieu that protects the mucosal surfaces in these locations.
Collapse
|