1
|
Gopakumar G, Coppo MJC, Diaz-Méndez A, Hartley CA, Devlin JM. Clinical assessment and transcriptome analysis of host immune responses in a vaccination-challenge study using a glycoprotein G deletion mutant vaccine strain of infectious laryngotracheitis virus. Front Immunol 2025; 15:1458218. [PMID: 39926602 PMCID: PMC11802539 DOI: 10.3389/fimmu.2024.1458218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/17/2024] [Indexed: 02/11/2025] Open
Abstract
A glycoprotein-G-deleted live-attenuated vaccine strain of the infectious laryngotracheitis virus (ILTV), ΔgG-ILTV, is safe and efficacious against ILTV challenge. In the current study, the transcriptome of peripheral blood mononuclear cells (PBMCs) of the ΔgG-ILTV-vaccinated group of specific-pathogen-free chickens were compared to those of the nonvaccinated group at 7 days post-vaccination. Tracheal transcriptomes after challenge with virulent ILTV were compared between groups of the non-vaccinated-challenged and the vaccinated-challenged as well as the non-vaccinated-challenged and the uninfected chickens at 4 to 5 days post-challenge. The clinical outcomes after challenge between these groups were also evaluated. Significant differences were observed in the tracheal transcriptome of the non-vaccinated-challenged birds compared to the other two groups. Enriched gene ontologies and pathways that indicated heightened immune responses and impairments to ciliary and neuronal functions, cell junction components, and potential damages to cartilaginous and extracellular components in the trachea of the non-vaccinated-challenged birds were consistent with their severe tracheal pathology compared to the other two groups. On the contrary, the absence of any difference in the tracheal transcriptome between the vaccinated-challenged and the uninfected birds were reflected by the preservation of tracheal mucosal integrity in both groups and mild infiltration of leukocytes in the vaccinated-challenged birds. The results from this study demonstrated that vaccination with ΔgG-ILTV prevented the changes in tracheal transcriptome induced during ILTV challenge, resulting in clinical protection. Additionally, these results also provide insights into the molecular mechanisms underlying the tracheal pathology induced by ILTV infection.
Collapse
Affiliation(s)
- Gayathri Gopakumar
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Mauricio J. C. Coppo
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
- Escuela de Medicina Veterinaria, Universidad Andrés Bello, Concepción, Chile
| | - Andrés Diaz-Méndez
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Carol A. Hartley
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Joanne M. Devlin
- Asia-Pacific Centre for Animal Health, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
2
|
Iannuzo N, Welfley H, Li NC, Johnson MDL, Rojas-Quintero J, Polverino F, Guerra S, Li X, Cusanovich DA, Langlais PR, Ledford JG. CC16 drives VLA-2-dependent SPLUNC1 expression. Front Immunol 2023; 14:1277582. [PMID: 38053993 PMCID: PMC10694244 DOI: 10.3389/fimmu.2023.1277582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
Rationale CC16 (Club Cell Secretory Protein) is a protein produced by club cells and other non-ciliated epithelial cells within the lungs. CC16 has been shown to protect against the development of obstructive lung diseases and attenuate pulmonary pathogen burden. Despite recent advances in understanding CC16 effects in circulation, the biological mechanisms of CC16 in pulmonary epithelial responses have not been elucidated. Objectives We sought to determine if CC16 deficiency impairs epithelial-driven host responses and identify novel receptors expressed within the pulmonary epithelium through which CC16 imparts activity. Methods We utilized mass spectrometry and quantitative proteomics to investigate how CC16 deficiency impacts apically secreted pulmonary epithelial proteins. Mouse tracheal epithelial cells (MTECS), human nasal epithelial cells (HNECs) and mice were studied in naïve conditions and after Mp challenge. Measurements and main results We identified 8 antimicrobial proteins significantly decreased by CC16-/- MTECS, 6 of which were validated by mRNA expression in Severe Asthma Research Program (SARP) cohorts. Short Palate Lung and Nasal Epithelial Clone 1 (SPLUNC1) was the most differentially expressed protein (66-fold) and was the focus of this study. Using a combination of MTECs and HNECs, we found that CC16 enhances pulmonary epithelial-driven SPLUNC1 expression via signaling through the receptor complex Very Late Antigen-2 (VLA-2) and that rCC16 given to mice enhances pulmonary SPLUNC1 production and decreases Mycoplasma pneumoniae (Mp) burden. Likewise, rSPLUNC1 results in decreased Mp burden in mice lacking CC16 mice. The VLA-2 integrin binding site within rCC16 is necessary for induction of SPLUNC1 and the reduction in Mp burden. Conclusion Our findings demonstrate a novel role for CC16 in epithelial-driven host defense by up-regulating antimicrobials and define a novel epithelial receptor for CC16, VLA-2, through which signaling is necessary for enhanced SPLUNC1 production.
Collapse
Affiliation(s)
- Natalie Iannuzo
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Holly Welfley
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | | | | | | | | | - Stefano Guerra
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona, Tucson, AZ, United States
| | - Xingnan Li
- Department of Medicine, Division of Genetics, Genomics, and Precision Medicine, University of Arizona, Tucson, AZ, United States
| | - Darren A. Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, United States
| | - Julie G. Ledford
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| |
Collapse
|
3
|
Barclay AM, Ninaber DK, van Veen S, Hiemstra PS, Ottenhoff THM, van der Does AM, Joosten SA. Airway epithelial cells mount an early response to mycobacterial infection. Front Cell Infect Microbiol 2023; 13:1253037. [PMID: 37822359 PMCID: PMC10562574 DOI: 10.3389/fcimb.2023.1253037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/31/2023] [Indexed: 10/13/2023] Open
Abstract
Lung epithelial cells represent the first line of host defence against foreign inhaled components, including respiratory pathogens. Their responses to these exposures may direct subsequent immune activation to these pathogens. The epithelial response to mycobacterial infections is not well characterized and may provide clues to why some mycobacterial infections are cleared, while others are persistent and pathogenic. We have utilized an air-liquid interface model of human primary bronchial epithelial cells (ALI-PBEC) to investigate the epithelial response to infection with a variety of mycobacteria: Mycobacterium tuberculosis (Mtb), M. bovis (BCG), M. avium, and M. smegmatis. Airway epithelial cells were found to be infected by all four species, albeit at low frequencies. The proportion of infected epithelial cells was lowest for Mtb and highest for M. avium. Differential gene expression analysis revealed a common epithelial host response to mycobacteria, including upregulation of BIRC3, S100A8 and DEFB4, and downregulation of BPIFB1 at 48 h post infection. Apical secretions contained predominantly pro-inflammatory cytokines, while basal secretions contained tissue growth factors and chemokines. Finally, we show that neutrophils were attracted to both apical and basal secretions of infected ALI-PBEC. Neutrophils were attracted in high numbers to apical secretions from PBEC infected with all mycobacteria, with the exception of secretions from M. avium-infected ALI-PBEC. Taken together, our results show that airway epithelial cells are differentially infected by mycobacteria, and react rapidly by upregulation of antimicrobials, and increased secretion of inflammatory cytokines and chemokines which directly attract neutrophils. Thus, the airway epithelium may be an important immunological component in controlling and regulating mycobacterial infections.
Collapse
Affiliation(s)
- Amy M. Barclay
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Dennis K. Ninaber
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Anne M. van der Does
- Department of Pulmonology, Leiden University Medical Center, Leiden, Netherlands
| | - Simone A. Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
4
|
Oyang L, Ouyang L, Yang L, Lin J, Xia L, Tan S, Wu N, Han Y, Yang Y, Li J, Chen X, Tang Y, Su M, Luo X, Li J, Xiong W, Zeng Z, Liao Q, Zhou Y. LPLUNC1 reduces glycolysis in nasopharyngeal carcinoma cells through the PHB1-p53/c-Myc axis. Cancer Sci 2023; 114:870-884. [PMID: 36382614 PMCID: PMC9986081 DOI: 10.1111/cas.15662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer cells prefer glycolysis to support their proliferation. Our previous studies have shown that the long palate, lung, and nasal epithelial cell clone 1 (LPLUNC1) can upregulate prohibitin 1 (PHB1) expression to inhibit the proliferation of nasopharyngeal carcinoma (NPC) cells. Given that PHB1 is an important regulator of cell energy metabolism, we explored whether and how LPLUNC1 regulated glucose glycolysis in NPC cells. LPLUNC1 or PHB1 overexpression decreased glycolysis and increased oxidative phosphorylation (OXPHOS)-related protein expression in NPC cells, promoting phosphorylated PHB1 nuclear translocation through 14-3-3σ. LPLUNC1 overexpression also increased p53 but decreased c-Myc expression in NPC cells, which were crucial for the decrease in glycolysis and increase in OXPHOS-related protein expression induced by LPLUNC1 overexpression. Finally, we found that treatment with all-trans retinoic acid (ATRA) reduced the viability and clonogenicity of NPC cells, decreased glycolysis, and increased OXPHOS-related protein expression by enhancing LPLUNC1 expression in NPC cells. Therefore, the LPLUNC1-PHB1-p53/c-Myc axis decreased glycolysis in NPC cells, and ATRA upregulated LPLUNC1 expression, ATRA maybe a promising drug for the treatment of NPC.
Collapse
Affiliation(s)
- Linda Oyang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lei Ouyang
- Department of Head and Neck Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lixia Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Longzheng Xia
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shiming Tan
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiqing Yang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jian Li
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,University of South China, Changsha, Hunan, China
| | - Xiaohui Chen
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,University of South China, Changsha, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Su
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xia Luo
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jinyun Li
- Department of Head and Neck Surgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health and the Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Translational Radiation Oncology, Changsha, Hunan, China
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Translational Radiation Oncology, Changsha, Hunan, China
| |
Collapse
|
5
|
He L, Zhou S, Li W, Wang Q, Qi Z, Zhou P, Wang Z, Chen J, Li Y, Lin Z. BPIFA2 as a Novel Early Biomarker to Identify Fatal Radiation Injury After Radiation Exposure. Dose Response 2022; 20:15593258221086478. [PMID: 35431693 PMCID: PMC9006374 DOI: 10.1177/15593258221086478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Background Current dosimeters cannot cope with the two tasks of medical rescue in the early stage of nuclear accident, the accurate determination of radiation exposure and the identification of patients with fatal radiation injury. As radiation can cause alterations in serum components, it is feasible to develop biomarkers for radiation injury from serum. This study aims to investigate whether serum BPIFA2 could be used as a potential biomarker of predicting fatal radiation injury in the early stage after nuclear accident. Methods A rabbit anti-mouse BPIFA2 polyclonal antibody was prepared to detect the expression of BPIFA2. C57BL/6J female mice were exposed to total body radiation (TBI) at different dose and Partial body radiation (PBI) at lethal dose to detect the dynamic changes of BPIFA2 in serum at different time points after irradiation by Western blot assay. Results BPIFA2 in mice serum were significantly increased at 1–12 h post-irradiation at .5–10 Gy, and increased again significantly at 3 d after 10 Gy irradiation with associated with mortality closely. It also increased rapidly after PBI and was closely related to injury degree, regardless whether the salivary glands were irradiated. Conclusions The increase of serum BPIFA2 is a novel early biomarker not only for identifying radiation exposure, but also for fatal radiation injury playing a vital role in rational use of medical resources, and greater efficiency of medical treatment to minimize casualties.
Collapse
Affiliation(s)
- Lexin He
- College of Life Sciences, North China University of Science and Technology, Tangshan, China
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Shixiang Zhou
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Weihong Li
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Qi Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhenhua Qi
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Pingkun Zhou
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhidong Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Jing Chen
- College of Life Sciences, North China University of Science and Technology, Tangshan, China
| | - Yaqiong Li
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhongwu Lin
- Science Research Management Department of the Academy of Military Sciences, Beijing, China
| |
Collapse
|
6
|
Jaiswal AK, Yadav J, Makhija S, Sandey M, Suryawanshi A, Mitra AK, Mishra A. Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) level determines steroid-resistant airway inflammation in aging. Am J Physiol Lung Cell Mol Physiol 2022; 322:L102-L115. [PMID: 34851736 PMCID: PMC8759962 DOI: 10.1152/ajplung.00315.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 01/25/2023] Open
Abstract
Asthma and its heterogeneity change with age. Increased airspace neutrophil numbers contribute to severe steroid-resistant asthma exacerbation in the elderly, which correlates with the changes seen in adults with asthma. However, whether that resembles the same disease mechanism and pathophysiology in aged and adults is poorly understood. Here, we sought to address the underlying molecular mechanism of steroid-resistant airway inflammation development and response to corticosteroid (Dex) therapy in aged mice. To study the changes in inflammatory mechanism, we used a clinically relevant treatment model of house-dust mite (HDM)-induced allergic asthma and investigated lung adaptive immune response in adult (20-22 wk old) and aged (80-82 wk old) mice. Our result indicates an age-dependent increase in airway hyperresponsiveness (AHR), mixed granulomatous airway inflammation comprising eosinophils and neutrophils, and Th1/Th17 immune response with progressive decrease in frequencies and numbers of HDM-bearing dendritic cells (DC) accumulation in the draining lymph node (DLn) of aged mice as compared with adult mice. RNA-Seq experiments of the aged lung revealed short palate, lung, and nasal epithelial clone 1 (SPLUNC1) as one of the steroid-responsive genes, which progressively declined with age and further by HDM-induced inflammation. Moreover, we found increased glycolytic reprogramming, maturation/activation of DCs, the proliferation of OT-II cells, and Th2 cytokine secretion with recombinant SPLUNC1 (rSPLUNC1) treatment. Our results indicate a novel immunomodulatory role of SPLUNC1 regulating metabolic adaptation/maturation of DC. An age-dependent decline in the SPLUNC1 level may be involved in developing steroid-resistant airway inflammation and asthma heterogeneity.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Jyoti Yadav
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Sangeet Makhija
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Maninder Sandey
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics, Harrison School of Pharmacy, Auburn University, Auburn, Alabama
| | - Amarjit Mishra
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| |
Collapse
|
7
|
Differential Expression of Mitosis and Cell Cycle Regulatory Genes during Recovery from an Acute Respiratory Virus Infection. Pathogens 2021; 10:pathogens10121625. [PMID: 34959580 PMCID: PMC8708581 DOI: 10.3390/pathogens10121625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/27/2022] Open
Abstract
Acute respiratory virus infections can have profound and long-term effects on lung function that persist even after the acute responses have fully resolved. In this study, we examined gene expression by RNA sequencing in the lung tissue of wild-type BALB/c mice that were recovering from a sublethal infection with the pneumonia virus of mice (PVM), a natural rodent pathogen of the same virus family and genus as the human respiratory syncytial virus. We compared these responses to gene expression in PVM-infected mice treated with Lactobacillus plantarum, an immunobiotic agent that limits inflammation and averts the negative clinical sequelae typically observed in response to acute infection with this pathogen. Our findings revealed prominent differential expression of inflammation-associated genes as well as numerous genes and gene families implicated in mitosis and cell-cycle regulation, including cyclins, cyclin-dependent kinases, cell division cycle genes, E2F transcription factors, kinesins, centromere proteins, and aurora kinases, among others. Of particular note was the differential expression of the cell division cycle gene Cdc20b, which was previously identified as critical for the ex vivo differentiation of multi-ciliated cells. Collectively, these findings provided us with substantial insight into post-viral repair processes and broadened our understanding of the mechanisms underlying Lactobacillus-mediated protection.
Collapse
|
8
|
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: 0.5] [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
|
9
|
Kim M, Park J, Bouhaddou M, Kim K, Rojc A, Modak M, Soucheray M, McGregor MJ, O'Leary P, Wolf D, Stevenson E, Foo TK, Mitchell D, Herrington KA, Muñoz DP, Tutuncuoglu B, Chen KH, Zheng F, Kreisberg JF, Diolaiti ME, Gordan JD, Coppé JP, Swaney DL, Xia B, van 't Veer L, Ashworth A, Ideker T, Krogan NJ. A protein interaction landscape of breast cancer. Science 2021; 374:eabf3066. [PMID: 34591612 PMCID: PMC9040556 DOI: 10.1126/science.abf3066] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Minkyu Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Jisoo Park
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Mehdi Bouhaddou
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kyumin Kim
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Ajda Rojc
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Maya Modak
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Margaret Soucheray
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Michael J McGregor
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Patrick O'Leary
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Denise Wolf
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Erica Stevenson
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Tzeh Keong Foo
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Dominique Mitchell
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Kari A Herrington
- Department of Biochemistry and Biophysics, Center for Advanced Light Microscopy, University of California, San Francisco, CA, USA
| | - Denise P Muñoz
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Beril Tutuncuoglu
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Kuei-Ho Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Fan Zheng
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Jason F Kreisberg
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA
| | - Morgan E Diolaiti
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - John D Gordan
- Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.,Division of Hematology/Oncology, University of California, San Francisco, CA, USA
| | - Jean-Philippe Coppé
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Danielle L Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| | - Bing Xia
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Laura van 't Veer
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Alan Ashworth
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Trey Ideker
- The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, CA, USA.,Department of Bioengineering, University of California, San Diego, CA, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA.,The J. David Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA.,Quantitative Biosciences Institute, University of California, San Francisco, CA, USA.,The Cancer Cell Map Initiative, San Francisco and La Jolla, CA, USA
| |
Collapse
|
10
|
Khanal S, Webster M, Niu N, Zielonka J, Nunez M, Chupp G, Slade MD, Cohn L, Sauler M, Gomez JL, Tarran R, Sharma L, Dela Cruz CS, Egan M, Laguna T, Britto CJ. SPLUNC1: a novel marker of cystic fibrosis exacerbations. Eur Respir J 2021; 58:13993003.00507-2020. [PMID: 33958427 DOI: 10.1183/13993003.00507-2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/29/2021] [Indexed: 11/05/2022]
Abstract
Acute pulmonary Exacerbations (AE) are episodes of clinical worsening in cystic fibrosis (CF), often precipitated by infection. Timely detection is critical to minimise morbidity and lung function declines associated with acute inflammation during AE. Based on our previous observations that airway protein Short Palate Lung Nasal epithelium Clone 1 (SPLUNC1) is regulated by inflammatory signals, we investigated the use of SPLUNC1 fluctuations to diagnose and predict AE in CF.We enrolled CF participants from two independent cohorts to measure AE markers of inflammation in sputum and recorded clinical outcomes for a 1-year follow-up period.SPLUNC1 levels were high in healthy controls (n=9, 10.7 μg mL-1), and significantly decreased in CF participants without AE (n=30, 5.7 μg mL-1, p=0.016). SPLUNC1 levels were 71.9% lower during AE (n=14, 1.6 μg mL-1, p=0.0034) regardless of age, sex, CF-causing mutation, or microbiology findings. Cytokines Il-1β and TNFα were also increased in AE, whereas lung function did not consistently decrease. Stable CF participants with lower SPLUNC1 levels were much more likely to have an AE at 60 days (HR: 11.49, Standard Error: 0.83, p=0.0033). Low-SPLUNC1 stable participants remained at higher AE risk even one year after sputum collection (HR: 3.21, Standard Error: 0.47, p=0.0125). SPLUNC1 was downregulated by inflammatory cytokines and proteases increased in sputum during AE.In acute CF care, low SPLUNC1 levels could support a decision to increase airway clearance or to initiate pharmacological interventions. In asymptomatic, stable patients, low SPLUNC1 levels could inform changes in clinical management to improve long-term disease control and clinical outcomes in CF.
Collapse
Affiliation(s)
- Sara Khanal
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Megan Webster
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Naiqian Niu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jana Zielonka
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Myra Nunez
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Martin D Slade
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lauren Cohn
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marie Egan
- Division of Pediatric Pulmonology, Allergy, Immunology, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Theresa Laguna
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
11
|
Yamamoto S, Okamura K, Fujii R, Kawano T, Ueda K, Yajima Y, Shiba K. Specimen-specific drift of densities defines distinct subclasses of extracellular vesicles from human whole saliva. PLoS One 2021; 16:e0249526. [PMID: 33831057 PMCID: PMC8032098 DOI: 10.1371/journal.pone.0249526] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/21/2021] [Indexed: 12/26/2022] Open
Abstract
Extracellular vesicles (EVs) in body fluids constitute heterogenous populations, which mirror their diverse parental cells as well as distinct EV-generation pathways. Various methodologies have been proposed to differentiate EVs in order to deepen the current understanding of EV biology. Equilibrium density-gradient centrifugation has often been used to separate EVs based on their buoyant densities; however, the standard conditions used for the method do not necessarily allow all EVs to move to their equilibrium density positions, which complicates the categorization of EVs. Here, by prolonging ultracentrifugation time to 96 h and fractionating EVs both by floating up or spinning down directions, we allowed 111 EV-associated protein markers from the whole saliva of three healthy volunteers to attain equilibrium. Interestingly, the determined buoyant densities of the markers drifted in a specimen-specific manner, and drift patterns differentiated EVs into at least two subclasses. One class carried classical exosomal markers, such as CD63 and CD81, and the other was characterized by the molecules involved in membrane remodeling or vesicle trafficking. Distinct patterns of density drift may represent the differences in generation pathways of EVs.
Collapse
Affiliation(s)
- Satoshi Yamamoto
- Division of Protein Engineering, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Oral and Maxillofacial Implantology, Tokyo Dental College, Tokyo, Japan
| | - Kohji Okamura
- Department of Systems BioMedicine, National Center for Child Health and Development, Tokyo, Japan
| | - Risa Fujii
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takamasa Kawano
- Division of Protein Engineering, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Oral Oncology, Oral and Maxillofacial Surgery, Tokyo Dental College, Chiba, Japan
| | - Koji Ueda
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yasutomo Yajima
- Department of Oral and Maxillofacial Implantology, Tokyo Dental College, Tokyo, Japan
| | - Kiyotaka Shiba
- Division of Protein Engineering, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- * E-mail:
| |
Collapse
|
12
|
Innocentini LMAR, Silva AA, Carvalho MA, Coletta RD, Corrêa MEP, Bingle L, Bingle CD, Vargas PA, Lopes MA. Salivary BPIFA proteins are altered in patients undergoing hematopoietic cell transplantation. Oral Dis 2021; 28:1279-1288. [PMID: 33682222 DOI: 10.1111/odi.13832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/29/2020] [Accepted: 03/04/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the expression of BPIFA proteins in the saliva and salivary glands of hematopoietic cell transplant (HCT) patients. MATERIAL AND METHODS This longitudinal study included patients who had undergone autologous HCT (auto-HCT) and allogeneic HCT (allo-HCT), and unstimulated saliva was collected at three time points, with a fourth collection at oral chronic graft-versus-host disease (cGVHD) onset. BPIFA expression was analysed by Western blotting in saliva and immunostaining in the minor salivary glands of cGVHD patients. RESULTS Auto-HCT patients showed increased levels of BPIFA1 (p = .021) and BPIFA2 at D+7 (p = .040), whereas allo-HCT group demonstrated decreased expression of BPIFA2 at D+8 (p = .002) and at D+80 (p = .001) and a significant association between BPIFA2 low levels and hyposalivation was observed (p = .02). BPIFA2 was significantly lower in the cGVHD patients when compared to baseline (p = .04). CONCLUSIONS The results of this study show distinct pattern of expression of BPIF proteins in both auto-HCT and allo-HCT recipients with decreased levels of BPIFA2 during hyposalivation and cGVHD. Further studies are necessary to elucidate these proteins mechanisms and their clinical implications in these groups of patients.
Collapse
Affiliation(s)
- Lara Maria Alencar Ramos Innocentini
- Dentistry and Stomatology Division, Ophthalmology, Otolaryngology and Head and Neck Surgery Department, Clinical Hospital of Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, Brazil
| | - Andreia Aparecida Silva
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Marco Antonio Carvalho
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | | | - Lynne Bingle
- Department of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, UK
| | - Colin D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, Sheffield, UK
| | - Pablo Agustin Vargas
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| | - Márcio Ajudarte Lopes
- Department of Oral Diagnosis, School of Dentistry of Piracicaba, University of Campinas (FOP/UNICAMP), Piracicaba, São Paulo, Brazil
| |
Collapse
|
13
|
Abstract
The lungs are constantly exposed to the external environment and are therefore vulnerable to insults that can cause infection and injury. Maintaining the integrity and barrier function of the lung epithelium requires complex interactions of multiple cell lineages. Elucidating the cellular players and their regulation mechanisms provides fundamental information to deepen understanding about the responses and contributions of lung stem cells. This Review focuses on advances in our understanding of mammalian alveolar epithelial stem cell subpopulations and discusses insights about the regeneration-specific cell status of alveolar epithelial stem cells. We also consider how these advances can inform our understanding of post-injury lung repair processes and lung diseases.
Collapse
Affiliation(s)
- Huijuan Wu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Nan Tang
- National Institute of Biological Sciences, Beijing 102206, China .,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
14
|
Tomazic PV, Darnhofer B, Birner-Gruenberger R. Nasal mucus proteome and its involvement in allergic rhinitis. Expert Rev Proteomics 2020; 17:191-199. [PMID: 32266843 PMCID: PMC7261402 DOI: 10.1080/14789450.2020.1748502] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Nasal mucus is the first line defense barrier against various pathogens including allergens. Proteins in nasal mucus maybe used as biomarkers for diagnosis or future therapeutic strategies. Proteomics opens the possibility to investigate whole human proteomes. Areas Covered: We aimed to analyze the existing literature on nasal mucus and nasal secretions proteomic approaches especially in allergic rhinitis. A PubMed/Medline search was conducted entering the following keywords and combinations: “nasal mucus”, “nasal lavage fluid,” nasal secretions,” “nasal swabs,” “allergic rhinitis,” ”proteins,” and “proteomics.” Expert opinion: The majority of studies focus on single proteins or protein groups mainly using ELISA techniques. Four studies met the criteria using mass spectrometry in the analysis of nasal mucus proteomes in rhinologic diseases. In these studies, 7, 35, 267, and 430 proteins were identified, respectively. These four studies are discussed in this review and put in relation to seven other proteomic studies that focus on nasal lavage fluid and nasal secretions obtained by swabs or filter paper. To put it in a nutshell, proteomics facilitates the investigation of the nasal secretome and its role in healthy and diseased state and as potential biomarkers for new diagnostic or therapeutic approaches.
Collapse
Affiliation(s)
| | - Barbara Darnhofer
- Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, The Omics Center Graz, Graz, Austria
| | - Ruth Birner-Gruenberger
- Diagnostic and Research Institute of Pathology, Diagnostic and Research Center of Molecular Medicine, Medical University of Graz, Graz, Austria.,BioTechMed-Graz, The Omics Center Graz, Graz, Austria.,Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| |
Collapse
|
15
|
Saferali A, Tang AC, Strug LJ, Quon BS, Zlosnik J, Sandford AJ, Turvey SE. Immunomodulatory function of the cystic fibrosis modifier gene BPIFA1. PLoS One 2020; 15:e0227067. [PMID: 31931521 PMCID: PMC6957340 DOI: 10.1371/journal.pone.0227067] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cystic fibrosis (CF) is characterized by a progressive decline in lung function due to airway obstruction, infection, and inflammation. CF patients are particularly susceptible to respiratory infection by a variety of pathogens, and the inflammatory response in CF is dysregulated and prolonged. BPI fold containing family A, member 1 (BPIFA1) and BPIFB1 are proteins expressed in the upper airways that may have innate immune activity. We previously identified polymorphisms in the BPIFA1/BPIFB1 region associated with CF lung disease severity. METHODS We evaluated whether the BPIFA1/BPIFB1 associations with lung disease severity replicated in individuals with CF participating in the International CF Gene Modifier Consortium (n = 6,365). Furthermore, we investigated mechanisms by which the BPIFA1 and BPIFB1 proteins may modify lung disease in CF. RESULTS The association of the G allele of rs1078761 with reduced lung function was replicated in an independent cohort of CF patients (p = 0.001, n = 2,921) and in a meta-analysis of the full consortium (p = 2.39x10-5, n = 6,365). Furthermore, we found that rs1078761G which is associated with reduced lung function was also associated with reduced BPIFA1, but not BPIFB1, protein levels in saliva from CF patients. Functional assays indicated that BPIFA1 and BPIFB1 do not have an anti-bacterial role against P. aeruginosa but may have an immunomodulatory function in CF airway epithelial cells. Gene expression profiling using RNAseq identified Rho GTPase signaling pathways to be altered in CF airway epithelial cells in response to treatment with recombinant BPIFA1 and BPIFB1 proteins. CONCLUSIONS BPIFA1 and BPIFB1 have immunomodulatory activity and genetic variation associated with low levels of these proteins may increase CF lung disease severity.
Collapse
Affiliation(s)
- Aabida Saferali
- Centre for Heart Lung Innovation, University of British Columbia and St Paul’s Hospital, Vancouver, British Columbia, Canada
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Anthony C. Tang
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Lisa J. Strug
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Bradley S. Quon
- Centre for Heart Lung Innovation, University of British Columbia and St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - James Zlosnik
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
| | - Andrew J. Sandford
- Centre for Heart Lung Innovation, University of British Columbia and St Paul’s Hospital, Vancouver, British Columbia, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, University of British Columbia and BC Children’s Hospital, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
16
|
Burbelo PD, Ferré EMN, Chaturvedi A, Chiorini JA, Alevizos I, Lionakis MS, Warner BM. Profiling Autoantibodies against Salivary Proteins in Sicca Conditions. J Dent Res 2019; 98:772-778. [PMID: 31095438 DOI: 10.1177/0022034519850564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Salivary gland dysfunction occurs in several autoimmune and immune-related conditions, including Sjögren syndrome (SS); immune checkpoint inhibitor-induced sicca (ICIS) that develops in some cancer patients and is characterized by severe, sudden-onset dry mouth; and autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). Although subjects with these conditions present with oral dryness and often exhibit inflammatory infiltration of the salivary gland, little is known about the B-cell humoral responses directed against salivary gland protein targets. In this study, autoantibodies were evaluated against Ro52, Ro60, and La, as well as against a panel of 22 proteins derived from the salivary proteome. The tested cohort included healthy volunteers and subjects with SS, ICIS, and APECED without and with sicca. As expected, a high percentage of autoantibody seropositivity was detected against Ro52, Ro60, and La in SS, but only a few ICIS patients were seropositive for these autoantigens. A few APECED subjects also harbored autoantibodies to Ro52 and La, but only Ro60 autoantibodies were weakly associated with a small subset of APECED patients with sicca. Additional testing of the salivary panel failed to detect seropositive autoantibodies against any of the salivary-enriched proteins in the SS and ICIS subjects. However, APECED subjects selectively demonstrated seropositivity against BPI fold containing family A member 1 (BPIFA1), BPI fold containing family A member 2 (BPIFA2)/parotid salivary protein (PSP), and lactoperoxidase, 3 salivary-enriched proteins. Moreover, high levels of serum autoantibodies against BPIFA1 and BPIFA2/PSP occurred in 30% and 67% of the APECED patients with sicca symptoms, respectively, and were associated with an earlier age onset of oral dryness (P = 0.001). These findings highlight the complexity of humoral responses in different sicca diseases and provide new insights and biomarkers for APECED-associated sicca (ClinicalTrials.gov: NCT00001196; NCT00001390; NCT01425892; NCT01386437).
Collapse
Affiliation(s)
- P D Burbelo
- 1 Dental Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - E M N Ferré
- 2 Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - A Chaturvedi
- 1 Dental Clinical Research Core, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - J A Chiorini
- 3 Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - I Alevizos
- 4 Sjogren's Clinic, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - M S Lionakis
- 2 Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - B M Warner
- 3 Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.,4 Sjogren's Clinic, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
17
|
Fu XG, Huang Z, Zhou SJ, Yang J, Peng YJ, Cao LY, Guo H, Wu GH, Lin YH, Huang BY. Novel heterozygous BPIFC variant in a Chinese pedigree with hereditary trichilemmal cysts. Mol Genet Genomic Med 2019; 7:e697. [PMID: 31033252 PMCID: PMC6565563 DOI: 10.1002/mgg3.697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/15/2019] [Accepted: 04/08/2019] [Indexed: 12/30/2022] Open
Abstract
Background Trichilemmal cysts (TCs) are common intradermal or subcutaneous cysts, which are commonly sporadic and rarely autosomal dominantly inherited. However, little is known about the disease‐determining genes in families with TCs exhibiting Mendelian inheritance. Objective The aim of this study was to identify the causative gene in a family with TCs. Methods Whole‐exome sequencing was performed on a TCs family to identify the candidate gene. Sanger sequencing was conducted to validate the candidate variants and familial segregation. Results We identified the heterozygous variant c.3G>C (p.Met1?) within the BPIFC gene. Sanger sequencing confirmed the cosegregation of this variant with the TCs phenotype in the family by demonstrating the presence of the heterozygous variant in all the 12 affected and absence in all the seven unaffected individuals. This variant was found to be absent in dbSNP141, 1,000 Genomes database and 500 ethnicity matched controls. Conclusion Our results imply that BPIFC is a causative gene in this Chinese family with hereditary TCs. Further studies should be performed to validate the role of BPIFC in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Xian-Guo Fu
- Department of Central Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China.,Department of Clinical Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Zhao Huang
- Department of Pathology, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Su-Juan Zhou
- Department of Pathology, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Jing Yang
- Department of Central Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Yun-Juan Peng
- Department of Clinical Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Luo-Yuan Cao
- Department of Central Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Hua Guo
- Department of Pathology, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Guang-Hui Wu
- Department of Neurosurgery, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Ying-Hua Lin
- Department of Central Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| | - Bao-Ying Huang
- Department of Central Laboratory, Ningde Municipal Hospital, Fujian Medical University, Ningde, Fujian, China
| |
Collapse
|
18
|
Marcinko MC, Darrow AL, Tuia AJ, Shohet RV. Sex influences susceptibility to methamphetamine cardiomyopathy in mice. Physiol Rep 2019; 7:e14036. [PMID: 30891941 PMCID: PMC6424857 DOI: 10.14814/phy2.14036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/22/2022] Open
Abstract
In this study, we created a mouse model of methamphetamine cardiomyopathy that reproduces the chronic, progressive dosing commonly encountered in addicted subjects. We gradually increased the quantity of methamphetamine given to C57Bl/6 mice from 5 to 40 mg/kg over 2 or 5 months during two study periods. At the fifth month, heart weight was increased, echocardiograms showed a dilated cardiomyopathy and survival was lower in males, with less effect in females. Interestingly, these findings correspond to previous observations in human patients, suggesting greater male susceptibility to the effects of methamphetamine on the heart. Transcriptional analysis showed changes in genes dysregulated in previous methamphetamine neurological studies as well as many that likely play a role in cardiac response to this toxic stress. We expect that a deeper understanding of the molecular biology of methamphetamine exposure in the heart will provide insights into the mechanism of cardiomyopathy in addicts and potential routes to more effective treatment.
Collapse
MESH Headings
- Amphetamine-Related Disorders/complications
- Amphetamine-Related Disorders/genetics
- Amphetamine-Related Disorders/metabolism
- Animals
- Cardiomyopathy, Dilated/etiology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/physiopathology
- Disease Models, Animal
- Female
- Male
- Methamphetamine
- Mice, Inbred C57BL
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Risk Assessment
- Risk Factors
- Sex Factors
- Time Factors
- Transcription, Genetic
Collapse
Affiliation(s)
- Marie C. Marcinko
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| | - April L. Darrow
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| | - Aaron J. Tuia
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| | - Ralph V. Shohet
- Department of MedicineUniversity of Hawaii John A. Burns School of MedicineHonoluluHawaii
| |
Collapse
|
19
|
The Role of BPIFA1 in Upper Airway Microbial Infections and Correlated Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2021890. [PMID: 30255091 PMCID: PMC6140130 DOI: 10.1155/2018/2021890] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/04/2018] [Accepted: 08/07/2018] [Indexed: 12/16/2022]
Abstract
The mucosa is part of the first line of immune defense against pathogen exposure in humans and prevents viral and bacterial infection of the soft palate, lungs, uvula, and nasal cavity that comprise the ear-nose-throat (ENT) region. Bactericidal/permeability-increasing fold containing family A, member 1 (BPIFA1) is a secretory protein found in human upper aerodigestive tract mucosa. This innate material is secreted in mucosal fluid or found in submucosal tissue in the human soft palate, lung, uvula, and nasal cavity. BPIFA1 is a critical component of the innate immune response that prevents upper airway diseases. This review will provide a brief introduction of the roles of BPIFA1 in the upper airway (with a focus on the nasal cavity, sinus, and middle ear), specifically its history, identification, distribution in various human tissues, function, and diagnostic value in various upper airway infectious diseases.
Collapse
|
20
|
Nikolić MZ, Sun D, Rawlins EL. Human lung development: recent progress and new challenges. Development 2018; 145:145/16/dev163485. [PMID: 30111617 PMCID: PMC6124546 DOI: 10.1242/dev.163485] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent studies have revealed biologically significant differences between human and mouse lung development, and have reported new in vitro systems that allow experimental manipulation of human lung models. At the same time, emerging clinical data suggest that the origins of some adult lung diseases are found in embryonic development and childhood. The convergence of these research themes has fuelled a resurgence of interest in human lung developmental biology. In this Review, we discuss our current understanding of human lung development, which has been profoundly influenced by studies in mice and, more recently, by experiments using in vitro human lung developmental models and RNA sequencing of human foetal lung tissue. Together, these approaches are helping to shed light on the mechanisms underlying human lung development and disease, and may help pave the way for new therapies. Summary: This Review describes how recent technological advances have shed light on the mechanisms underlying human lung development and disease, and outlines the future challenges in this field.
Collapse
Affiliation(s)
- Marko Z Nikolić
- Wellcome Trust/CRUK Gurdon Institute, Wellcome Trust/MRC Stem Cell Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK.,University of Cambridge School of Clinical Medicine, Department of Medicine, Cambridge CB2 0QQ, UK
| | - Dawei Sun
- Wellcome Trust/CRUK Gurdon Institute, Wellcome Trust/MRC Stem Cell Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK
| | - Emma L Rawlins
- Wellcome Trust/CRUK Gurdon Institute, Wellcome Trust/MRC Stem Cell Institute, Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK
| |
Collapse
|
21
|
Tata A, Kobayashi Y, Chow RD, Tran J, Desai A, Massri AJ, McCord TJ, Gunn MD, Tata PR. Myoepithelial Cells of Submucosal Glands Can Function as Reserve Stem Cells to Regenerate Airways after Injury. Cell Stem Cell 2018; 22:668-683.e6. [PMID: 29656943 DOI: 10.1016/j.stem.2018.03.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/01/2018] [Accepted: 03/21/2018] [Indexed: 01/12/2023]
Abstract
Cells demonstrate plasticity following injury, but the extent of this phenomenon and the cellular mechanisms involved remain underexplored. Using single-cell RNA sequencing (scRNA-seq) and lineage tracing, we uncover that myoepithelial cells (MECs) of the submucosal glands (SMGs) proliferate and migrate to repopulate the airway surface epithelium (SE) in multiple injury models. Specifically, SMG-derived cells display multipotency and contribute to basal and luminal cell types of the SMGs and SE. Ex vivo expanded MECs have the potential to repopulate and differentiate into SE cells when grafted onto denuded airway scaffolds. Significantly, we find that SMG-like cells appear on the SE of both extra- and intra-lobular airways of large animal lungs following severe injury. We find that the transcription factor SOX9 is necessary for MEC plasticity in airway regeneration. Because SMGs are abundant and present deep within airways, they may serve as a reserve cell source for enhancing human airway regeneration.
Collapse
Affiliation(s)
- Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ryan D Chow
- Department of Genetics, Systems Biology Institute, Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jasmine Tran
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Avani Desai
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Abdull J Massri
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Timothy J McCord
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael Dee Gunn
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
22
|
Mulay A, Hood DW, Williams D, Russell C, Brown SDM, Bingle L, Cheeseman M, Bingle CD. Loss of the homeostatic protein BPIFA1, leads to exacerbation of otitis media severity in the Junbo mouse model. Sci Rep 2018; 8:3128. [PMID: 29449589 PMCID: PMC5814562 DOI: 10.1038/s41598-018-21166-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/30/2018] [Indexed: 02/02/2023] Open
Abstract
Otitis Media (OM) is characterized by epithelial abnormalities and defects in innate immunity in the middle ear (ME). Although, BPIFA1, a member of the BPI fold containing family of putative innate defence proteins is abundantly expressed by the ME epithelium and SNPs in Bpifa1 have been associated with OM susceptibility, its role in the ME is not well characterized. We investigated the role of BPIFA1 in protection of the ME and the development of OM using murine models. Loss of Bpifa1 did not lead to OM development. However, deletion of Bpifa1 in Evi1Jbo/+ mice, a model of chronic OM, caused significant exacerbation of OM severity, thickening of the ME mucosa and increased collagen deposition, without a significant increase in pro-inflammatory gene expression. Our data suggests that BPIFA1 is involved in maintaining homeostasis within the ME under steady state conditions and its loss in the presence of inflammation, exacerbates epithelial remodelling leading to more severe OM.
Collapse
Affiliation(s)
- Apoorva Mulay
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Derek W Hood
- MRC Mammalian Genetics Unit, MRC Harwell Institute, Didcot, UK
| | - Debbie Williams
- MRC Mammalian Genetics Unit, MRC Harwell Institute, Didcot, UK
| | - Catherine Russell
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Steve D M Brown
- MRC Mammalian Genetics Unit, MRC Harwell Institute, Didcot, UK
| | - Lynne Bingle
- Oral and Maxillofacial Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Michael Cheeseman
- Roslin Institute, University of Edinburgh, Edinburgh, UK.,Division of Pathology, University of Edinburgh, Edinburgh, UK
| | - Colin D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK. .,Florey Institute for Host Pathogen Interactions, University of Sheffield, Sheffield, UK.
| |
Collapse
|
23
|
Mundhenk L, Erickson NA, Klymiuk N, Gruber AD. Interspecies diversity of chloride channel regulators, calcium-activated 3 genes. PLoS One 2018; 13:e0191512. [PMID: 29346439 PMCID: PMC5773202 DOI: 10.1371/journal.pone.0191512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/06/2018] [Indexed: 12/15/2022] Open
Abstract
Members of the chloride channel regulators, calcium-activated (CLCA) family, have been implicated in diverse biomedical conditions, including chronic inflammatory airway diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis, the activation of macrophages, and the growth and metastatic spread of tumor cells. Several observations, however, could not be repeated across species boundaries and increasing evidence suggests that select CLCA genes are particularly prone to dynamic species-specific evolvements. Here, we systematically characterized structural and expressional differences of the CLCA3 gene across mammalian species, revealing a spectrum of gene duplications, e.g., in mice and cows, and of gene silencing via diverse chromosomal modifications in pigs and many primates, including humans. In contrast, expression of a canonical CLCA3 protein from a single functional gene seems to be evolutionarily retained in carnivores, rabbits, guinea pigs, and horses. As an accepted asthma model, we chose the cat to establish the tissue and cellular expression pattern of the CLCA3 protein which was primarily found in mucin-producing cells of the respiratory tract and in stratified epithelia of the esophagus. Our results suggest that, among developmental differences in other CLCA genes, the CLCA3 gene possesses a particularly high dynamic evolutionary diversity with pivotal consequences for humans and other primates that seem to lack a CLCA3 protein. Our data also help to explain previous contradictory results on CLCA3 obtained from different species and warrant caution in extrapolating data from animal models in conditions where CLCA3 may be involved.
Collapse
Affiliation(s)
- Lars Mundhenk
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- * E-mail:
| | - Nancy A. Erickson
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Nikolai Klymiuk
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilians-Universität, Oberschleissheim, Germany
| | - Achim D. Gruber
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
24
|
Bpifcl modulates kiss2 expression under the influence of 11-ketotestosterone in female zebrafish. Sci Rep 2017; 7:7926. [PMID: 28801581 PMCID: PMC5554142 DOI: 10.1038/s41598-017-08248-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 07/10/2017] [Indexed: 11/21/2022] Open
Abstract
The bactericidal/permeability-increasing (BPI) fold-containing (BPIF) superfamily of genes expressed in the brain are purportedly involved in modulating brain function in response to stress, such as inflammation. Kisspeptin, encoded by kiss, is affected by inflammation in the brain; therefore, BPIF family genes might be involved in the modulation of kisspeptin in the brain. In this study, we investigated the expression of BPIF family C, like (bpifcl) in zebrafish brain and its involvement in kiss2 regulation. The identified, full-length sequence of a bpifcl isoform expressed in the zebrafish brain contained the BPI fold shared by BPIF family members. bpifcl mRNA expression in female zebrafish brains was significantly higher than that in males. Exposure of female zebrafish to 11-ketotestosterone decreased bpifcl and kiss2 mRNA expression. bpifcl knockdown by bpifcl-specific small interfering RNA administration to female zebrafish brain decreased kiss2 mRNA expression. bpifcl expression was widely distributed in the brain, including in the dorsal zone of the periventricular hypothalamus (Hd). Furthermore, bpifcl was also expressed in KISS2 neurons in the Hd. These results suggest that the Bpifcl modulates kiss2 mRNA expression under the influence of testosterone in the Hd of female zebrafish.
Collapse
|
25
|
Nashida T, Shimomura-Kuroki J, Mizuhashi F, Haga-Tsujimura M, Yoshimura K, Hayashi-Sakai S. Presence of BPIFB1 in saliva from non-obese diabetic mice. Odontology 2017; 106:117-124. [PMID: 28748269 DOI: 10.1007/s10266-017-0312-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/22/2017] [Indexed: 01/09/2023]
Abstract
We previously showed that mRNA expression of BPIFB1 (Bpifb1), an antibacterial protein in the palate, lung, and nasal epithelium clone protein family, was increased in parotid acinar cells in non-obese diabetic (NOD, NOD/ShiJcl) mice, which is an animal model for Sjögren's syndrome. However, we did not previously assess the protein levels. In this report, we confirmed the expression of BPIFB1 protein in the parotid glands of NOD mice. Immunoblotting of subcellular fractions revealed that BPIBB1 was localised in secretory granules in parotid glands from NOD mice, and was almost not in parotid glands from the control mice. BPIFB1 had N-linked glycan that reacted with Aleuria aurantia lectin, which caused two types of spots with a slightly different pI and molecular weight. The expression of BPIFB1 protein was also demonstrated by immunohistochemistry. BPIFB1 was detected in the saliva from NOD mice but not in the saliva from the control mice, indicating individual constitution. BPIFB1 in saliva may be applied to other research as a diagnostic marker.
Collapse
Affiliation(s)
- Tomoko Nashida
- Department of Biochemistry, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan.
| | - Junko Shimomura-Kuroki
- Department of Pediatric Dentistry, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Fumi Mizuhashi
- Department of Removable Prosthodontics, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Maiko Haga-Tsujimura
- Department of Histology, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Ken Yoshimura
- Department of Anatomy, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Sachiko Hayashi-Sakai
- Division of Oral and Maxillofacial Radiology, Niigata University Graduate School of Medical and Dental Science, 2-5274 Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| |
Collapse
|
26
|
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
|
27
|
Hall SC, Hassis ME, Williams KE, Albertolle ME, Prakobphol A, Dykstra AB, Laurance M, Ona K, Niles RK, Prasad N, Gormley M, Shiboski C, Criswell LA, Witkowska HE, Fisher SJ. Alterations in the Salivary Proteome and N-Glycome of Sjögren's Syndrome Patients. J Proteome Res 2017; 16:1693-1705. [PMID: 28282148 PMCID: PMC9668345 DOI: 10.1021/acs.jproteome.6b01051] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We used isobaric mass tagging (iTRAQ) and lectin affinity capture mass spectrometry (MS)-based workflows for global analyses of parotid saliva (PS) and whole saliva (WS) samples obtained from patients diagnosed with primary Sjögren's Syndrome (pSS) who were enrolled in the Sjögren's International Collaborative Clinical Alliance (SICCA) as compared with two control groups. The iTRAQ analyses revealed up- and down-regulation of numerous proteins that could be involved in the disease process (e.g., histones) or attempts to mitigate the ensuing damage (e.g., bactericidal/permeability increasing fold containing family (BPIF) members). An immunoblot approach applied to independent sample sets confirmed the pSS associated up-regulation of β2-microglobulin (in PS) and down-regulation of carbonic anhydrase VI (in WS) and BPIFB2 (in PS). Beyond the proteome, we profiled the N-glycosites of pSS and control samples. They were enriched for glycopeptides using lectins Aleuria aurantia and wheat germ agglutinin, which recognize fucose and sialic acid/N-acetyl glucosamine, respectively. MS analyses showed that pSS is associated with increased N-glycosylation of numerous salivary glycoproteins in PS and WS. The observed alterations of the salivary proteome and N-glycome could be used as pSS biomarkers enabling easier and earlier detection of this syndrome while lending potential new insights into the disease process.
Collapse
Affiliation(s)
- Steven C. Hall
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Maria E. Hassis
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Katherine E. Williams
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Matthew E. Albertolle
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Akraporn Prakobphol
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Andrew B. Dykstra
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Megan Laurance
- Library and Center for Knowledge Management, University of California, San Francisco, San Francisco, California 94143, United States
| | - Katherine Ona
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Richard K. Niles
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Namrata Prasad
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Matthew Gormley
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Caroline Shiboski
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Lindsey A. Criswell
- Department of Orofacial Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Russel/Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, San Francisco, California 94143, United States
| | - H. Ewa Witkowska
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| | - Susan J. Fisher
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, United States
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
28
|
Assessment of Pasteurella multocida A Lipopolysaccharide, as an Adhesin in an In Vitro Model of Rabbit Respiratory Epithelium. Vet Med Int 2017; 2017:8967618. [PMID: 28251016 PMCID: PMC5303596 DOI: 10.1155/2017/8967618] [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: 09/25/2016] [Revised: 12/06/2016] [Accepted: 12/29/2016] [Indexed: 11/18/2022] Open
Abstract
The role of the P. multocida lipopolysaccharide (LPS) as a putative adhesin during the early stages of infection with this bacterium in the respiratory epithelium of rabbits was investigated. By light microscopy and double enzyme labeling of nasal septa tissues, the amount of bacteria attached to the respiratory epithelium and the amount of LPS present in goblet cells at different experimental times were estimated. Transmission electron microscopy (TEM) and LPS labeling with colloidal gold particles were also used to determine the exact location of LPS in the cells. Septa that were challenged with LPS of P. multocida and 30 minutes later with P. multocida showed more adherent bacteria and more severe lesions than the other treatments. Free LPS was observed in the lumen of the nasal septum, forming bilamellar structures and adhering to the cilia, microvilli, cytoplasmic membrane, and cytoplasm of epithelial ciliated and goblet cells. The above findings suggest that P. multocida LPS plays an important role in the process of bacterial adhesion and that it has the ability of being internalized into host cells.
Collapse
|
29
|
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: 40] [Impact Index Per Article: 5.0] [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
|
30
|
Alves DBM, Bingle L, Bingle CD, Lourenço SV, Silva AA, Pereira DL, Vargas PA. BPI-fold (BPIF) containing/plunc protein expression in human fetal major and minor salivary glands. Braz Oral Res 2017; 31:e6. [PMID: 28099576 DOI: 10.1590/1807-3107bor-2017.vol31.0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/23/2016] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to determine expression, not previously described, of PLUNC (palate, lung, and nasal epithelium clone) (BPI-fold containing) proteins in major and minor salivary glands from very early fetal tissue to the end of the second trimester and thus gain further insight into the function of these proteins. Early fetal heads, and major and minor salivary glands were collected retrospectively and glands were classified according to morphodifferentiation stage. Expression of BPI-fold containing proteins was localized through immunohistochemistry. BPIFA2, the major BPI-fold containing protein in adult salivary glands, was detected only in the laryngeal pharynx; the lack of staining in salivary glands suggested salivary expression is either very late in development or is only in adult tissues. Early expression of BPIFA1 was seen in the trachea and nasal cavity with salivary gland expression only seen in late morphodifferentiation stages. BPIFB1 was seen in early neural tissue and at later stages in submandibular and sublingual glands. BPIFA1 is significantly expressed in early fetal oral tissue but BPIFB1 has extremely limited expression and the major salivary BPIF protein (BPIFA2) is not produced in fetal development. Further studies, with more sensitive techniques, will confirm the expression pattern and enable a better understanding of embryonic BPIF protein function.
Collapse
Affiliation(s)
- Daniel Berretta Moreira Alves
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, SP, Brazil
| | - Lynne Bingle
- University of Sheffield, School of Clinical Dentistry, Academic Unit of Oral and Maxillofacial Pathology, Sheffield, UK
| | - Colin David Bingle
- University of Sheffield, Medical School, Royal Hallamshire Hospital, Academic Unit of Respiratory Medicine, Sheffield, UK
| | - Silvia Vanessa Lourenço
- Universidade de São Paulo - USP, School of Dentistry, Department of General Pathology, São Paulo-SP, Brazil
| | - Andréia Aparecida Silva
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, SP, Brazil
| | - Débora Lima Pereira
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, SP, Brazil
| | - Pablo Agustin Vargas
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Oral Diagnosis, Piracicaba, SP, Brazil
| |
Collapse
|
31
|
Chu CY, Qiu X, Wang L, Bhattacharya S, Lofthus G, Corbett A, Holden-Wiltse J, Grier A, Tesini B, Gill SR, Falsey AR, Caserta MT, Walsh EE, Mariani TJ. The Healthy Infant Nasal Transcriptome: A Benchmark Study. Sci Rep 2016; 6:33994. [PMID: 27658638 PMCID: PMC5034274 DOI: 10.1038/srep33994] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/06/2016] [Indexed: 01/21/2023] Open
Abstract
Responses by resident cells are likely to play a key role in determining the severity of respiratory disease. However, sampling of the airways poses a significant challenge, particularly in infants and children. Here, we report a reliable method for obtaining nasal epithelial cell RNA from infants for genome-wide transcriptomic analysis, and describe baseline expression characteristics in an asymptomatic cohort. Nasal epithelial cells were collected by brushing of the inferior turbinates, and gene expression was interrogated by RNA-seq analysis. Reliable recovery of RNA occurred in the absence of adverse events. We observed high expression of epithelial cell markers and similarity to the transcriptome for intrapulmonary airway epithelial cells. We identified genes displaying low and high expression variability, both inherently, and in response to environmental exposures. The greatest gene expression differences in this asymptomatic cohort were associated with the presence of known pathogenic viruses and/or bacteria. Robust bacteria-associated gene expression patterns were significantly associated with the presence of Moraxella. In summary, we have developed a reliable method for interrogating the infant airway transcriptome by sampling the nasal epithelium. Our data demonstrates both the fidelity and feasibility of our methodology, and describes normal gene expression and variation within a healthy infant cohort.
Collapse
Affiliation(s)
- Chin-Yi Chu
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester NY, USA
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester NY, USA
| | - Lu Wang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester NY, USA
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester NY, USA
| | - Gerry Lofthus
- Department of Medicine, University of Rochester Medical Center, Rochester NY, USA
| | - Anthony Corbett
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester NY, USA
| | - Jeanne Holden-Wiltse
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester NY, USA
| | - Alex Grier
- Department of Microbiology and Immunology and University of Rochester Medical Center, Rochester NY, USA
| | - Brenda Tesini
- Division of Pediatric Infectious Diseases, University of Rochester Medical Center, Rochester NY, USA
| | - Steven R. Gill
- Department of Microbiology and Immunology and University of Rochester Medical Center, Rochester NY, USA
| | - Ann R. Falsey
- Department of Medicine, Rochester General Hospital University of Rochester Medical Center, Rochester NY, USA
| | - Mary T. Caserta
- Division of Pediatric Infectious Diseases, University of Rochester Medical Center, Rochester NY, USA
| | - Edward E. Walsh
- Department of Medicine, Rochester General Hospital University of Rochester Medical Center, Rochester NY, USA
| | - Thomas J. Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester NY, USA
| |
Collapse
|
32
|
Differential short palate, lung, and nasal epithelial clone 1 suppression in eosinophilic and noneosinophilic chronic rhinosinusitis with nasal polyps: implications for pathogenesis and treatment. Curr Opin Allergy Clin Immunol 2016; 16:31-8. [PMID: 26658012 DOI: 10.1097/aci.0000000000000228] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Short palate, lung, and nasal epithelial clone 1 (SPLUNC1) is an epithelium-secreted protein that is involved in innate immunity. A protective role for SPLUNC1 in lower respiratory inflammation and chronic rhinosinusitis (CRS) has recently been recognized. RECENT FINDINGS An impaired epithelial immune barrier has been proposed to play a critical role in the pathogenesis of CRS. Recent research has demonstrated that SPLUNC1 is profoundly reduced in polyp tissues of CRS with nasal polyps (CRSwNP) compared with control tissues. Studies investigating the differential expression of SPLUNC1 in eosinophilic and noneosinophilic CRSwNP have been published. Nasal SPLUNC1 expression was inhibited by Th2 cytokines (IL-4 and IL-13) but was stimulated by toll-like receptor (TLR) agonists and glucocorticoids. Decreased SPLUNC1 expression in the sinus mucosa is associated with positive Pseudomonas aeruginosa bacterial colonization and poor surgical outcomes in CRS patients. SUMMARY These studies identify the role of SPLUNC1 in sinonasal innate immunity and the pathogenesis of CRS. Defective expression of SPLUNC1 in CRSwNP patients may lead to insufficient maintenance of the epithelial barrier function and enhanced bacterial colonization. The use of SPLUNC1 as a therapeutic target for CRSwNP remains to be determined.
Collapse
|
33
|
Arba M, Iavarone F, Vincenzoni F, Manconi B, Vento G, Tirone C, Cabras T, Castagnola M, Messana I, Sanna MT. Proteomic characterization of the acid-insoluble fraction of whole saliva from preterm human newborns. J Proteomics 2016; 146:48-57. [DOI: 10.1016/j.jprot.2016.06.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/31/2016] [Accepted: 06/15/2016] [Indexed: 11/25/2022]
|
34
|
Walton WG, Ahmad S, Little MR, Kim CS, Tyrrell J, Lin Q, Di YP, Tarran R, Redinbo MR. Structural Features Essential to the Antimicrobial Functions of Human SPLUNC1. Biochemistry 2016; 55:2979-91. [PMID: 27145151 PMCID: PMC4887393 DOI: 10.1021/acs.biochem.6b00271] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
SPLUNC1 is an abundantly secreted innate immune protein in the mammalian respiratory tract that exerts bacteriostatic and antibiofilm effects, binds to lipopolysaccharide (LPS), and acts as a fluid-spreading surfactant. Here, we unravel the structural elements essential for the surfactant and antimicrobial functions of human SPLUNC1 (short palate lung nasal epithelial clone 1). A unique α-helix (α4) that extends from the body of SPLUNC1 is required for the bacteriostatic, surfactant, and LPS binding activities of this protein. Indeed, we find that mutation of just four leucine residues within this helical motif to alanine is sufficient to significantly inhibit the fluid spreading abilities of SPLUNC1, as well as its bacteriostatic actions against Gram-negative pathogens Burkholderia cenocepacia and Pseudomonas aeruginosa. Conformational flexibility in the body of SPLUNC1 is also involved in the bacteriostatic, surfactant, and LPS binding functions of the protein as revealed by disulfide mutants introduced into SPLUNC1. In addition, SPLUNC1 exerts antibiofilm effects against Gram-negative bacteria, although α4 is not involved in this activity. Interestingly, though, the introduction of surface electrostatic mutations away from α4 based on the unique dolphin SPLUNC1 sequence, and confirmed by crystal structure, is shown to impart antibiofilm activity against Staphylococcus aureus, the first SPLUNC1-dependent effect against a Gram-positive bacterium reported to date. Together, these data pinpoint SPLUNC1 structural motifs required for the antimicrobial and surfactant actions of this protective human protein.
Collapse
Affiliation(s)
- William G. Walton
- Departments of Chemistry, Biochemistry and Microbiology, 4350 Genome Sciences Building, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Saira Ahmad
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Michael R. Little
- Departments of Chemistry, Biochemistry and Microbiology, 4350 Genome Sciences Building, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| | - Christine S.K. Kim
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Jean Tyrrell
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Qiao Lin
- Department of Environmental and Occupational Health, 331 Bridgeside Point Building, University of Pittsburgh, Pittsburgh, PA 15260
| | - Y. Peter Di
- Department of Environmental and Occupational Health, 331 Bridgeside Point Building, University of Pittsburgh, Pittsburgh, PA 15260
| | - Robert Tarran
- Marsico Lung Institute, Cystic Fibrosis/Pulmonary Research and Treatment Center, 7102 Marsico Hall, University of North Carolina, Chapel Hill, NC 27599-7248, USA
| | - Matthew R. Redinbo
- Departments of Chemistry, Biochemistry and Microbiology, 4350 Genome Sciences Building, University of North Carolina, Chapel Hill, NC 27599-3290, USA
| |
Collapse
|
35
|
Nashida T, Yoshimura K, Yoshie S, Mizuhashi F, Shimomura-Kuroki J. Upregulation of Bpifb1 expression in the parotid glands of non-obese diabetic mice. Oral Dis 2016; 22:46-52. [PMID: 26769076 DOI: 10.1111/odi.12377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/14/2015] [Accepted: 09/30/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To define the increased mRNA expression of Bpifb1, a member of the bactericidal/permeability-increasing protein family, in parotid acinar cells from non-obese diabetic (NOD) mice, an animal model for Sjögren's syndrome. MATERIALS AND METHODS Parotid acinar cells were prepared from female NOD (NOD/ShiJcl) mice with or without diabetes, as well as from control (C57BL/6JJcl) mice. Total RNA and homogenate were prepared from the parotid acinar cells. Embryonic cDNA from a Mouse MTC(™) Panel I kit was used. The expression of Bpifb1 was determined by cDNA microarray analysis, RT-PCR, real-time PCR, northern blotting and in situ hybridization. RESULTS The expression of Bpifb1 mRNA was high in parotid acinar cells from diabetic and non-diabetic NOD mice at 5-50 weeks of age. Acinar cells in the C57BL/6 mice had a low expression of Bpifb1 mRNA at an age >8 weeks, but had a relatively high expression in the foetus and infantile stages. CONCLUSIONS Bpifb1 mRNA is upregulated in parotid acinar cells in NOD mice, but its expression is not related to the onset of diabetes. These findings suggest that high expression levels of Bpifb1 might predict disease traits before the onset of autoimmunity.
Collapse
Affiliation(s)
- T Nashida
- Departments of Biochemistry, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - K Yoshimura
- Departments of Anatomy, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - S Yoshie
- Departments of Histology, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - F Mizuhashi
- Departments of Removable Prosthodontics, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| | - J Shimomura-Kuroki
- Departments of Pediatric Dentistry, The Nippon Dental University School of Life Dentistry at Niigata, Chuo-ku, Niigata, Japan
| |
Collapse
|
36
|
Britto CJ, Cohn L. Bactericidal/Permeability-increasing protein fold-containing family member A1 in airway host protection and respiratory disease. Am J Respir Cell Mol Biol 2015; 52:525-34. [PMID: 25265466 DOI: 10.1165/rcmb.2014-0297rt] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bactericidal/permeability-increasing protein fold-containing family member A1 (BPIFA1), formerly known as SPLUNC1, is one of the most abundant proteins in respiratory secretions and has been identified with increasing frequency in studies of pulmonary disease. Its expression is largely restricted to the respiratory tract, being highly concentrated in the upper airways and proximal trachea. BPIFA1 is highly responsive to airborne pathogens, allergens, and irritants. BPIFA1 actively participates in host protection through antimicrobial, surfactant, airway surface liquid regulation, and immunomodulatory properties. Its expression is modulated in multiple lung diseases, including cystic fibrosis, chronic obstructive pulmonary disease, respiratory malignancies, and idiopathic pulmonary fibrosis. However, the role of BPIFA1 in pulmonary pathogenesis remains to be elucidated. This review highlights the versatile properties of BPIFA1 in antimicrobial protection and its roles as a sensor of environmental exposure and regulator of immune cell function. A greater understanding of the contribution of BPIFA1 to disease pathogenesis and activity may clarify if BPIFA1 is a biomarker and potential drug target in pulmonary disease.
Collapse
Affiliation(s)
- Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
37
|
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: 24] [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
|
38
|
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: 2.0] [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
|
39
|
Salivary BPIFA1 (SPLUNC1) and BPIFA2 (SPLUNC2 A) are modified by head and neck cancer radiotherapy. Oral Surg Oral Med Oral Pathol Oral Radiol 2015; 119:48-58. [DOI: 10.1016/j.oooo.2014.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 09/16/2014] [Accepted: 09/22/2014] [Indexed: 12/11/2022]
|
40
|
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: 1.5] [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
|
41
|
Ning F, Wang C, Berry KZ, Kandasamy P, Liu H, Murphy RC, Voelker DR, Nho CW, Pan CH, Dai S, Niu L, Chu HW, Zhang G. Structural characterization of the pulmonary innate immune protein SPLUNC1 and identification of lipid ligands. FASEB J 2014; 28:5349-60. [PMID: 25223608 DOI: 10.1096/fj.14-259291] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The short palate, lung and nasal epithelial clone 1 (SPLUNC1) protein is a member of the palate, lung, and nasal epithelium clone (PLUNC) family, also known as bactericidal/permeability-increasing (BPI) fold-containing protein, family A, member 1 (BPIFA1). SPLUNC1 is an abundant protein in human airways, but its function remains poorly understood. The lipid ligands of SPLUNC1 as well as other PLUNC family members are largely unknown, although some reports provide evidence that lipopolysaccharide (LPS) could be a lipid ligand. Unlike previous hypotheses, we found significant structural differences between SPLUNC1 and BPI. Recombinant SPLUNC1 produced in HEK 293 cells harbored several molecular species of sphingomyelin and phosphatidylcholine as its ligands. Significantly, in vitro lipid-binding studies failed to demonstrate interactions between SPLUNC1 and LPS, lipoteichoic acid, or polymyxin B. Instead, one of the major and most important pulmonary surfactant phospholipids, dipalmitoylphosphatidylcholine (DPPC), bound to SPLUNC1 with high affinity and specificity. We found that SPLUNC1 could be the first protein receptor for DPPC. These discoveries provide insight into the specific determinants governing the interaction between SPLUNC1 and lipids and also shed light on novel functions that SPLUNC1 and other PLUNC family members perform in host defense.
Collapse
Affiliation(s)
- Fangkun Ning
- School of Life Sciences, University of Science and Technology of China, Hefei, China; Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Chao Wang
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Karin Zemski Berry
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Haolin Liu
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA
| | - Dennis R Voelker
- Department of Medicine, National Jewish Health, Denver, Colorado, USA; and
| | - Chu Won Nho
- Functional Food Center, Korea Institute of Science and Technology, GangNeung, Korea
| | - Choel-Ho Pan
- Functional Food Center, Korea Institute of Science and Technology, GangNeung, Korea
| | - Shaodong Dai
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| | - Liwen Niu
- School of Life Sciences, University of Science and Technology of China, Hefei, China;
| | - Hong-Wei Chu
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA; Department of Medicine, National Jewish Health, Denver, Colorado, USA; and
| | - Gongyi Zhang
- Integrated Department of Immunology, National Jewish Health and University of Colorado Denver, Denver, Colorado, USA
| |
Collapse
|
42
|
Tsou YA, Lin CC, Tai CJ, Tsai MH, Tsai TC, Chen CM. Chronic Rhinosinusitis and the Risk of Nasopharyngeal Cancer in a Taiwanese Health Study. Am J Rhinol Allergy 2014; 28:168-72. [PMID: 25197911 DOI: 10.2500/ajra.2014.28.4083] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Although epidemiological and laboratory studies report that chronic inflammatory conditions contribute to the pathogenesis of cancer, it remains controversial whether chronic rhinosinusitis (CRS) results in nasopharyngeal cancer (NPC). Methods Retrospective cohort study was performed from the National Health Insurance (NHI) Taiwan database. This study prospectively examined whether CRS or nasal polyposis is associated with NPC risk in the NHI, a population-based cohort of 231,490 Taiwan Chinese individuals with a mean age of 32 years, recruited between 2000 and 2006. We collected information from the Longitudinal Health Insurance Database. Each subject completed an interview including questions about medical conditions, and the NPC occurrence and survival statuses were determined by linkage to population-based NHI registries in Taiwan. In addition, each NPC and CRS subject had completed an interview on medical condition to confirm their diagnosis. Results After adjustment for age, sex, hypertension, diabetes mellitus, allergic rhinitis, otitis media, coronary artery disease, pharyngitis, and tonsillitis, individuals with rhinosinusitis were found to have a 3.55-fold increased risk of developing NPC compared with individuals without rhinosinusitis (hazard ratio = 3.55; 95% CI = 2.22–5.69). The same results were also observed when the study subjects were analyzed without comorbidities. Conclusion Adult patients with rhinosinusitis should be followed up with regard to the nasopharynx for at least 3 years, particularly repeat sinusitis patients.
Collapse
Affiliation(s)
- Yung-An Tsou
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Che-Chen Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Chin-Jaan Tai
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Hsui Tsai
- Department of Otolaryngology Head and Neck Surgery, School of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Tung-Chou Tsai
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
43
|
Tsou YA, Huang HJ, Lin WWY, Chen CYC. Investigation of anti-infection mechanism of lactoferricin and splunc-1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2014; 2014:907028. [PMID: 24876880 PMCID: PMC4021689 DOI: 10.1155/2014/907028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/08/2014] [Accepted: 02/08/2014] [Indexed: 12/11/2022]
Abstract
The innate immune system is the first line in the defense system and prevents the body from further bacteria, virus, or fungal infections. Most of the innate immune system is relevant to mucosa immunity. Lactotransferrin is secreted from the human mammal breast duct epithelial tissue and strengthens infant immunity to defense with regard to outward pathogens. Splunc-1 is also an innate material secreted from the soft palate, lung, nasal cavity epithelium, and mucosa. It helps with mucosa defense against bacterial, virus, and even fungus. LPS is the main etiology of Gram-negative bacilla infection source. And studies of lactoferricin and slpunc-1 both can combine with LPS and subsequently cause insults to the mucosa. Although, we know that both of them partake in an important role in innate immunity, we do not know the effects when they work together. In this study, we just overview silicon stimulation to examine the combination of Lactoferricin and Splunc-1 and the effect with regard to LPS.
Collapse
Affiliation(s)
- Yung An Tsou
- Otolaryngology Head and Neck Surgery, China Medical University Hospital, Taichung 40402, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Hung-Jin Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Wesley Wen Yang Lin
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
| | - Calvin Yu-Chian Chen
- School of Medicine, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
44
|
Irander K, Borres MP, Ghafouri B. The effects of physical exercise and smoking habits on the expression of SPLUNC1 in nasal lavage fluids from allergic rhinitis subjects. Int J Pediatr Otorhinolaryngol 2014; 78:618-22. [PMID: 24512783 DOI: 10.1016/j.ijporl.2014.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/11/2014] [Accepted: 01/14/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Palate lung nasal epithelial clone (PLUNC) is a family of proteins, which are proposed to participate in the innate immune defense against infections in the upper aero-digestive tract. The aim of this study was to investigate the expression of SPLUNC1 in allergic rhinitis subjects with considerations taken to the mucosal function and smoking habits. METHODS The participants, recruited from a cohort followed from infancy, were re-examined at the age of 18 years regarding allergy development. Based on medical histories and skin prick tests the participants were classified into groups with persistent allergic rhinitis (n=18), intermittent allergic rhinitis (n = 8) and healthy controls (n = 13). Seven subjects (3, 2 and 2 in each group, respectively) reported smoking habits. The SPLUNC1 levels in nasal lavage fluids were analyzed by Western blot. Changes in the volume of the proper nasal cavity before and after physical exercise (Vol2(increase)) were analyzed by acoustic rhinometry. RESULTS Compared to the control group the SPLUNC1 level was significantly lower in the persistent allergy group (3.8 ± 3.4 OD vs. 1.3 ± 1.5 OD; p = 0.02), but not in the intermittent allergy group without current exposure to allergens (3.6 ± 4.7 OD). No differences were found in Vol2(increase) between any of the allergy groups and controls. In smokers Vol2(increase) was significantly reduced (p < 0.01) and the SPLUNC1 levels were lower compared to non-smokers. A significant correlation was found between SPLUNC1 and Vol2(increase) (p < 0.01; r = 0.53) in non-smokers. CONCLUSIONS Current allergen exposure has an impact on SPLUNC1 expression in nasal lavage fluid, why allergy ought to be considered in study populations where analyses of SPLUNC1 levels are included in the reports. The normal nasal decongestion after exercise was not affected by allergy in contrast to smoking habits. The correlation between SPLUNC1 levels and Vol2(increase) in non-smokers may indicate involvement of SPLUNC1in the regulation of the normal function of the nasal mucosa. Complementary studies are needed to confirm the smoke-related reduction of SPLUNC1 expression and to analyze the possible participation of SPLUNC1 in the nasal mucosa regulation.
Collapse
Affiliation(s)
- K Irander
- Allergy Center, ENT Section, University Hospital, Linköping, Sweden
| | - M P Borres
- Department of Women's and Children's Health, Uppsala University, Sweden; Thermo Fisher Scientific, Uppsala, Sweden
| | - B Ghafouri
- Department of Medical and Health Sciences, Division of Community Medicine Rehabilitation Medicine, Faculty of Health Sciences, Linköping University, and Pain and Rehabilitation Centre, County Council of Östergötland, Linköping, Sweden; Occupational and Environmental Medicine, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, and Centre of Occupational and Environmental Medicine, County Council of Östergötland, Linköping, Sweden.
| |
Collapse
|
45
|
Holmes A, Rodrigues E, van der Wielen P, Lyons K, Haigh B, Wheeler T, Dawes P, Cannon R. Adherence ofCandida albicansto silicone is promoted by the human salivary protein SPLUNC2/PSP/BPIFA2. Mol Oral Microbiol 2014; 29:90-8. [DOI: 10.1111/omi.12048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/16/2014] [Indexed: 11/28/2022]
Affiliation(s)
- A.R. Holmes
- Sir John Walsh Research Institute; School of Dentistry; University of Otago; Dunedin New Zealand
| | - E. Rodrigues
- Sir John Walsh Research Institute; School of Dentistry; University of Otago; Dunedin New Zealand
| | - P. van der Wielen
- Sir John Walsh Research Institute; School of Dentistry; University of Otago; Dunedin New Zealand
| | - K.M. Lyons
- Sir John Walsh Research Institute; School of Dentistry; University of Otago; Dunedin New Zealand
| | - B.J. Haigh
- AgResearch Ltd; Ruakura Research Centre; Hamilton New Zealand
| | - T.T. Wheeler
- AgResearch Ltd; Ruakura Research Centre; Hamilton New Zealand
| | - P.J.D. Dawes
- Department of Surgical Sciences; Dunedin School of Medicine; University of Otago; Dunedin New Zealand
| | - R.D. Cannon
- Sir John Walsh Research Institute; School of Dentistry; University of Otago; Dunedin New Zealand
| |
Collapse
|
46
|
Di YP, Tkach AV, Yanamala N, Stanley S, Gao S, Shurin MR, Kisin ER, Kagan VE, Shvedova A. Dual acute proinflammatory and antifibrotic pulmonary effects of short palate, lung, and nasal epithelium clone-1 after exposure to carbon nanotubes. Am J Respir Cell Mol Biol 2013; 49:759-67. [PMID: 23721177 DOI: 10.1165/rcmb.2012-0435oc] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Carbon nanotubes (CNTs; allotropes of carbon with a cylindrical nanostructure) have emerged as one of the most commonly used types of nanomaterials, with numerous applications in industry and biomedicine. However, the inhalation of CNTs has been shown to elicit pulmonary toxicity, accompanied by a robust inflammatory response with an early-onset fibrotic phase. Epithelial host-defense proteins represent an important component of the pulmonary innate immune response to foreign inhalants such as particles and bacteria. The short palate, lung, and nasal epithelium clone-1 (SPLUNC1) protein, a member of the bactericidal/permeability-increasing-fold (BPIF)-containing protein family, is a 25-kD secretory protein that is expressed in nasal, oropharyngeal, and lung epithelia, and has been shown to have multiple functions, including antimicrobial and chemotactic activities, as well as surfactant properties. This study sought to assess the importance of SPLUNC1-mediated pulmonary responses in airway epithelial secretions, and to explore the biological relevance of SPLUNC1 to inhaled particles in a single-walled carbon nanotube (SWCNT) model. Using Scgb1a1-hSPLUNC1 transgenic mice, we observed that SPLUNC1 significantly modified host inflammatory responses by increasing leukocyte recruitment and enhancing phagocytic activity. Furthermore, we found that transgenic mice were more susceptible to SWCNT exposure at the acute phase, but showed resistance against lung fibrogenesis through pathological changes in the long term. The binding of SPLUNC1 also attenuated SWCNT-induced TNF-α secretion by RAW 264.7 macrophages. Taken together, our data indicate that SPLUNC1 is an important component of mucosal innate immune defense against pulmonary inhaled particles.
Collapse
Affiliation(s)
- Y Peter Di
- 1 Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hobbs CA, Da Tan C, Tarran R. Does epithelial sodium channel hyperactivity contribute to cystic fibrosis lung disease? J Physiol 2013; 591:4377-87. [PMID: 23878362 DOI: 10.1113/jphysiol.2012.240861] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Airway epithelia absorb Na+ through the epithelial Na+ channel (ENaC) and secrete Cl- through the cystic fibrosis transmembrane conductance regulator (CFTR) anion channel. This balance maintains sufficient airway surface liquid hydration to permit efficient mucus clearance, which is needed to maintain sterility of the lung. Cystic fibrosis (CF) is a common autosomal recessive inherited disease caused by mutations in the CFTR gene that lead to the reduction or elimination of the CFTR protein. CF is a multi-organ disease that affects epithelia lining the intestines, lungs, pancreas, sweat ducts and vas deferens, among others. CF lungs are characterized by viscous, dehydrated mucus, persistent neutrophilia and chronic infections. ENaC is negatively regulated by CFTR and, in patients with CF, the absence of CFTR results in a double hit of reduced Cl-/HCO3- and H2O secretion as well as ENaC hyperactivity and increased Na+ and H2O absorption. Together, these effects are hypothesized to trigger mucus dehydration, resulting in a failure to clear mucus. Rehydrating CF mucus has become a recent clinical focus and yields important end-points for clinical trials. However, while ENaC hyperactivity in CF airways has been detected in vivo and in vitro, recent data have brought the role of ENaC in CF lung disease pathogenesis into question. This review will focus on our current understanding of the contribution of ENaC to CF pathogenesis.
Collapse
Affiliation(s)
- Carey A Hobbs
- R. Tarran: 7125 Thurston Bowles Building, UNC, Chapel Hill, NC 27599-7248, USA.
| | | | | |
Collapse
|
48
|
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: 96] [Impact Index Per Article: 8.0] [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
|
49
|
Musa M, Wilson K, Sun L, Mulay A, Bingle L, Marriott HM, LeClair EE, Bingle CD. Differential localisation of BPIFA1 (SPLUNC1) and BPIFB1 (LPLUNC1) in the nasal and oral cavities of mice. Cell Tissue Res 2012; 350:455-64. [PMID: 22986921 PMCID: PMC3505551 DOI: 10.1007/s00441-012-1490-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 08/16/2012] [Indexed: 01/14/2023]
Abstract
Despite being initially identified in mice, little is known about the sites of production of members of the BPI fold (BPIF) containing (PLUNC) family of putative innate defence proteins in this species. These proteins have largely been considered to be specificaly expressed in the respiratory tract, and we have recently shown that they exhibit differential expression in the epithelium of the proximal airways. In this study, we have used species-specific antibodies to systematically localize two members of this protein family; BPIFA1 (PLUNC/SPLUNC1) and BPIFB1 (LPLUNC1) in adult mice. In general, these proteins exhibit distinct and only partially overlapping localization. BPIFA1 is highly expressed in the respiratory epithelium and Bowman’s glands of the nasal passages, whereas BPIFB1 is present in small subset of goblet cells in the nasal passage and pharynx. BPIFB1 is also present in the serous glands in the proximal tongue where is co-localised with the salivary gland specific family member, BPIFA2E (parotid secretory protein) and also in glands of the soft palate. Both proteins exhibit limited expression outside of these regions. These results are consistent with the localization of the proteins seen in man. Knowledge of the complex expression patterns of BPIF proteins in these regions will allow the use of tractable mouse models of disease to dissect their function.
Collapse
Affiliation(s)
- Maslinda Musa
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, University of Sheffield, UK
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Although the biology the PLUNC (recently renamed BPI fold, BPIF) family of secreted proteins is poorly understood, multiple array based studies have suggested that some are differentially expressed in lung diseases. We have examined the expression of BPIFB1 (LPLUNC1), the prototypic two-domain containing family member, in lungs from CF patients and in mouse models of CF lung disease. BPIFB1 was localized in CF lung samples along with BPIFA1, MUC5AC, CD68 and NE and directly compared to histologically normal lung tissues and that of bacterial pneumonia. We generated novel antibodies to mouse BPIF proteins to conduct similar studies on ENaC transgenic (ENaC-Tg) mice, a model for CF-like lung disease. Small airways in CF demonstrated marked epithelial staining of BPIFB1 in goblet cells but staining was absent from alveolar regions. BPIFA1 and BPIFB1 were not co-localised in the diseased lungs. In ENaC-Tg mice there was strong staining of both proteins in the airways and luminal contents. This was most marked for BPIFB1 and was noted within 2 weeks of birth. The two proteins were present in distinct cells within epithelium. BPIFB1 was readily detected in BAL from ENaC-Tg mice but was absent from wild-type mice. Alterations in the expression of BPIF proteins is associated with CF lung disease in humans and mice. It is unclear if this elevation of protein production, which results from phenotypic alteration of the cells within the diseased epithelium, plays a role in the pathogenesis of the disease.
Collapse
|