201
|
The Possible Pathogenesis of Idiopathic Pulmonary Fibrosis considering MUC5B. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9712464. [PMID: 31309122 PMCID: PMC6594326 DOI: 10.1155/2019/9712464] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/18/2019] [Accepted: 05/08/2019] [Indexed: 12/27/2022]
Abstract
Background Overexpression of the MUC5B protein is associated with idiopathic pulmonary fibrosis (IPF), but little information is available regarding the pathogenic effects and regulatory mechanisms of overexpressed MUC5B in IPF. Main Body The overexpression of MUC5B in terminal bronchi and honeycomb cysts produces mucosal host defensive dysfunction in the distal airway which may play an important role in the development of IPF. This review addresses the possible association of overexpression of MUC5B, with MUC5B promoter polymorphism, MUC5B gene epigenetic changes, effects of some transcriptional factors, and inflammatory mediators in IPF. In addition, the associated signaling pathways which may influence the expression of MUC5B are also discussed. Conclusion This work has important implications for further exploration of the mechanisms of overexpression of MUC5B in IPF, and future personalized treatment.
Collapse
|
202
|
Affiliation(s)
- Richard C Boucher
- From the Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill
| |
Collapse
|
203
|
Wang Y, Wu Q, Muskhelishvili L, Davis K, Bryant M, Cao X. Assessing the respiratory toxicity of dihydroxyacetone using an in vitro human airway epithelial tissue model. Toxicol In Vitro 2019; 59:78-86. [PMID: 30959092 DOI: 10.1016/j.tiv.2019.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/15/2022]
Abstract
Dihydroxyacetone (DHA) is an approved color additive used in sunless tanning lotions. Recently, there has been an increased use of DHA in sunless tanning booths in a manner that could result in its inhalation during application. In the present study, we have evaluated the potential for DHA causing toxicity via inhalation using a human air-liquid-interface (ALI) in vitro airway epithelial tissue model. ALI airway models have a close structural and functional resemblance to the in vivo airway epithelium, and thus data generated in these models may have relevance for predicting human responses. To simulate in vivo exposure conditions, we employed a method for liquid aerosol generation that mimics the physical form of inhaled chemicals and used doses of DHA and an exposure frequency reflecting human respiratory exposures during tanning sessions. Compared to the vehicle control, cilia beating frequency (CBF) and MUC5AC secretion were significantly decreased after each exposure. However, time-course studies indicated that both CBF and MUC5AC secretion returned to normal levels within 3 days after the treatment. Matrix metalloproteinase (MMP) release, on the other hand, was decreased 24 h after the first exposure and its level returned to baseline after 5 exposures. No significant morphological changes occurred in the DHA-treated cultures after 5 weekly exposures. Our findings indicate that DHA, at concentrations likely to be experienced by humans, has transient toxic effects on human airway ALI cultures.
Collapse
Affiliation(s)
- Yiying Wang
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, United States of America
| | - Qiangen Wu
- Division of Biochemistry Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, United States of America
| | | | - Kelly Davis
- Toxicologic Pathology Associates, Jefferson, AR 72079, United States of America
| | - Matthew Bryant
- Division of Biochemistry Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, United States of America
| | - Xuefei Cao
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, United States of America.
| |
Collapse
|
204
|
Koga Y, Tsurumaki H, Aoki-Saito H, Sato M, Yatomi M, Takehara K, Hisada T. Roles of Cyclic AMP Response Element Binding Activation in the ERK1/2 and p38 MAPK Signalling Pathway in Central Nervous System, Cardiovascular System, Osteoclast Differentiation and Mucin and Cytokine Production. Int J Mol Sci 2019; 20:ijms20061346. [PMID: 30884895 PMCID: PMC6470985 DOI: 10.3390/ijms20061346] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 11/26/2022] Open
Abstract
There are many downstream targets of mitogen-activated protein kinase (MAPK) signalling that are involved in neuronal development, cellular differentiation, cell migration, cancer, cardiovascular dysfunction and inflammation via their functions in promoting apoptosis and cell motility and regulating various cytokines. It has been reported that cyclic AMP response element-binding protein (CREB) is phosphorylated and activated by cyclic AMP signalling and calcium/calmodulin kinase. Recent evidence also points to CREB phosphorylation by the MAPK signalling pathway. However, the specific roles of CREB phosphorylation in MAPK signalling have not yet been reviewed in detail. Here, we describe the recent advances in the study of this MAPK-CREB signalling axis in human diseases. Overall, the crosstalk between extracellular signal-related kinase (ERK) 1/2 and p38 MAPK signalling has been shown to regulate various physiological functions, including central nervous system, cardiac fibrosis, alcoholic cardiac fibrosis, osteoclast differentiation, mucin production in the airway, vascular smooth muscle cell migration, steroidogenesis and asthmatic inflammation. In this review, we focus on ERK1/2 and/or p38 MAPK-dependent CREB activation associated with various diseases to provide insights for basic and clinical researchers.
Collapse
Affiliation(s)
- Yasuhiko Koga
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Hiroaki Tsurumaki
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Haruka Aoki-Saito
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Makiko Sato
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Masakiyo Yatomi
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Kazutaka Takehara
- Department of Allergy and Respiratory Medicine, Gunma University Graduate School of Medicine, 3-39-15 sho-wa machi Maebashi, Gunma 371-8511, Japan.
| | - Takeshi Hisada
- Gunma University Graduate School of Health Sciences, 3-39-22 sho-wa machi Maebashi, Gunma 371-8514, Japan.
| |
Collapse
|
205
|
Abstract
In the past decade, the field of the cellular microbiology of group A Streptococcus (S. pyogenes) infection has made tremendous advances and touched upon several important aspects of pathogenesis, including receptor biology, invasive and evasive phenomena, inflammasome activation, strain-specific autophagic bacterial killing, and virulence factor-mediated programmed cell death. The noteworthy aspect of S. pyogenes-mediated cell signaling is the recognition of the role of M protein in a variety of signaling events, starting with the targeting of specific receptors on the cell surface and on through the induction and evasion of NETosis, inflammasome, and autophagy/xenophagy to pyroptosis and apoptosis. Variations in reports on S. pyogenes-mediated signaling events highlight the complex mechanism of pathogenesis and underscore the importance of the host cell and S. pyogenes strain specificity, as well as in vitro/in vivo experimental parameters. The severity of S. pyogenes infection is, therefore, dependent on the virulence gene expression repertoire in the host environment and on host-specific dynamic signaling events in response to infection. Commonly known as an extracellular pathogen, S. pyogenes finds host macrophages as safe havens wherein it survives and even multiplies. The fact that endothelial cells are inherently deficient in autophagic machinery compared to epithelial cells and macrophages underscores the invasive nature of S. pyogenes and its ability to cause severe systemic diseases. S. pyogenes is still one of the top 10 causes of infectious mortality. Understanding the orchestration of dynamic host signaling networks will provide a better understanding of the increasingly complex mechanism of S. pyogenes diseases and novel ways of therapeutically intervening to thwart severe and often fatal infections.
Collapse
|
206
|
Piazzon MC, Mladineo I, Naya-Català F, Dirks RP, Jong-Raadsen S, Vrbatović A, Hrabar J, Pérez-Sánchez J, Sitjà-Bobadilla A. Acting locally - affecting globally: RNA sequencing of gilthead sea bream with a mild Sparicotyle chrysophrii infection reveals effects on apoptosis, immune and hypoxia related genes. BMC Genomics 2019; 20:200. [PMID: 30866816 PMCID: PMC6416957 DOI: 10.1186/s12864-019-5581-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/03/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Monogenean flatworms are the main fish ectoparasites inflicting serious economic losses in aquaculture. The polyopisthocotylean Sparicotyle chrysophrii parasitizes the gills of gilthead sea bream (GSB, Sparus aurata) causing anaemia, lamellae fusion and sloughing of epithelial cells, with the consequent hypoxia, emaciation, lethargy and mortality. Currently no preventive or curative measures against this disease exist and therefore information on the host-parasite interaction is crucial to find mitigation solutions for sparicotylosis. The knowledge about gene regulation in monogenean-host models mostly comes from freshwater monopysthocotyleans and almost nothing is known about polyopisthocotyleans. The current study aims to decipher the host response at local (gills) and systemic (spleen, liver) levels in farmed GSB with a mild natural S. chrysophrii infection by transcriptomic analysis. RESULTS Using Illumina RNA sequencing and transcriptomic analysis, a total of 2581 differentially expressed transcripts were identified in infected fish when compared to uninfected controls. Gill tissues in contact with the parasite (P gills) displayed regulation of fewer genes (700) than gill portions not in contact with the parasite (NP gills) (1235), most likely due to a local silencing effect of the parasite. The systemic reaction in the spleen was much higher than that at the parasite attachment site (local) (1240), and higher than in liver (334). NP gills displayed a strong enrichment of genes mainly related to immune response and apoptosis. Processes such as apoptosis, inflammation and cell proliferation dominated gills, whereas inhibition of apoptosis, autophagy, platelet activation, signalling and aggregation, and inflammasome were observed in spleen. Proteasome markers were increased in all tissues, whereas hypoxia-related genes were down-regulated in gills and spleen. CONCLUSIONS Contrasting forces seem to be acting at local and systemic levels. The splenic down-regulation could be part of a hypometabolic response, to counteract the hypoxia induced by the parasite damage to the gills and to concentrate the energy on defence and repair responses. Alternatively, it can be also interpreted as the often observed action of helminths to modify host immunity in its own interest. These results provide the first toolkit for future studies towards understanding and management of this parasitosis.
Collapse
Affiliation(s)
- M Carla Piazzon
- Fish Pathology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Ribera de Cabanes, Castellón, Spain.
| | | | - Fernando Naya-Català
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Ribera de Cabanes, Castellón, Spain.,Biotechvana, Parc Cientific, Universitat de Valencia, Valencia, Spain
| | - Ron P Dirks
- Future Genomics Technology, Leiden, The Netherlands
| | | | | | - Jerko Hrabar
- Institute of Oceanography and Fisheries, Split, Croatia
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Ribera de Cabanes, Castellón, Spain
| | - Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Institute of Aquaculture Torre de la Sal (IATS-CSIC), Ribera de Cabanes, Castellón, Spain
| |
Collapse
|
207
|
Ogino K, Nagaoka K, Ito T, Takemoto K, Okuda T, Nakayama SF, Ogino N, Seki Y, Hamada H, Takashiba S, Fujikura Y. Involvement of PM2.5-bound protein and metals in PM2.5-induced allergic airway inflammation in mice. Inhal Toxicol 2019; 30:498-508. [PMID: 30849251 DOI: 10.1080/08958378.2018.1561769] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The aim of this study was to investigate the protein and trace element components of PM2.5 and their contribution to the allergic airway inflammation in BALB/c mice. METHODS PM2.5, treated at high temperature and with a strong acid to hydrolyze any protein content and remove trace elements, was administered to BALB/c mice. Allergic airway inflammation was compared between the three groups (saline, pure PM2.5 and treated PM2.5) by evaluating airway hyperresponsiveness (AHR), bronchoalveolar lavage fluid (BALF) cells, serum IgE, the mRNA of various cytokine (IL-4, IL-5, IL-13, eotaxin-1 and CXCL3), mucus protein mRNA (MUC5ac and MUC5b) and the filtration of inflammatory cells in the lung. RESULTS The treatment of PM2.5 with a strong acid at a high temperature attenuated AHR, eosinophil percentage in BALF, mRNA levels of IL-13 and CXCL3 and peribronchial inflammation. On the contrary, the percentage of neutrophils in BALF, mRNA expression of MIP2α, EGFR, Nrf2, and TLR4 and 4-OH-2-nonenal levels in the lung was increased. Moreover, the treatment of the PM2.5 reduced PM2.5-bound proteins as well as the percentages of the trace elements in PM2.5 in the order Zn > Cu > Pb > P > S > Mn > Fe > Ca > Ni, whereas the percentage of C, Si and Cl increased. CONCLUSIONS PM2.5 collected by of the cyclone system induced allergic airway inflammation in mice. PM2.5-bound proteins and acid-soluble metals may be involved in the pathogenesis of PM2.5-induced allergic airway inflammation.
Collapse
Affiliation(s)
- Keiki Ogino
- a Department of Public Health , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science , Okayama , Japan
| | - Kenjiro Nagaoka
- a Department of Public Health , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science , Okayama , Japan
| | - Tatsuo Ito
- a Department of Public Health , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science , Okayama , Japan
| | - Kei Takemoto
- a Department of Public Health , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science , Okayama , Japan
| | - Tomoaki Okuda
- b Department of Applied Chemistry, Faculty of Science and Technology , Keio University , Yokohama , Japan
| | - Shoji F Nakayama
- c Center for Environmental Health Sciences, Integrated Health Risk Assessment Section , Tsukuba , Japan
| | - Noriyoshi Ogino
- a Department of Public Health , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science , Okayama , Japan.,d Third Department of Internal Medicine School of Medicine , University of Occupational and Environmental Health , Kitakyushu , Japan
| | - Yuka Seki
- a Department of Public Health , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science , Okayama , Japan
| | - Hiroki Hamada
- e Department of Life Science, Faculty of Science, Laboratory of Food Preventive Medicine , Okayama University of Science , Okayama , Japan
| | - Shogo Takashiba
- f Department of Patho-Physiology, Division of Periodontal Science , Okayama University Graduate School of Medicine , Okayama , Japan
| | - Yoshihisa Fujikura
- g Department of Anatomy, Biology and Medicine, Division of Morphological Analysis, Faculty of Medicine , Oita University , Oita , Japan
| |
Collapse
|
208
|
Erickson-DiRenzo E, Leydon C, Thibeault SL. Methodology for the establishment of primary porcine vocal fold epithelial cell cultures. Laryngoscope 2019; 129:E355-E364. [PMID: 30848488 DOI: 10.1002/lary.27909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/04/2019] [Accepted: 02/12/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE A current lack of methods for epithelial cell culture significantly hinders our understanding of the role of the epithelial and mucus barriers in vocal fold health and disease. Our first objective was to establish reproducible techniques for the isolation and culture of primary porcine vocal fold epithelial cells. Our second objective was to evaluate the functional significance of cell cultures using an in vitro exposure to an inflammatory cytokine. METHODS Epithelial cells were isolated from porcine vocal folds and expanded in culture. Characterization of cultures was completed by immunostaining with markers for pan-cytokeratin (epithelial cells), vimentin (stromal cells), von Willebrand factor (endothelial cell), and MUC1 and MUC4 (mucin) glycoproteins. Established epithelial cell cultures were then exposed to the inflammatory cytokine tumor necrosis factor alpha (TNF-α) for 24-hours, and transcript expression of MUC1 and MUC4 was evaluated. RESULTS Reproducible, porcine vocal fold epithelial cell cultures, demonstrating cobblestone appearance characteristic of the typical morphology of epithelial cell cultures were created. Cells showed positive staining for pan-cytokeratin with limited expression of vimentin and von Willebrand factor. Epithelial cells also expressed MUC1 and MUC4. TNF-α significantly increased transcript expression of MUC4. CONCLUSION Here, we present the first report of successful culture of primary porcine vocal fold epithelial cells. Cultures will provide researchers with a valuable new in vitro tool to investigate vocal fold epithelium and mucus as well as the effects of common challenges, including inflammatory cytokines, on these barriers. LEVEL OF EVIDENCE NA Laryngoscope, 129:E355-E364, 2019.
Collapse
Affiliation(s)
- Elizabeth Erickson-DiRenzo
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, University of Wisconsin-Madison, Madison, Wisconsin, U.S.A
| | - Ciara Leydon
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, University of Wisconsin-Madison, Madison, Wisconsin, U.S.A
| | - Susan L Thibeault
- Department of Surgery, Division of Otolaryngology-Head & Neck Surgery, University of Wisconsin-Madison, Madison, Wisconsin, U.S.A
| |
Collapse
|
209
|
Modulation of Mucin ( MUC2, MUC5AC and MUC5B) mRNA Expression and Protein Production and Secretion in Caco-2/HT29-MTX Co-Cultures Following Exposure to Individual and Combined Aflatoxin M1 and Ochratoxin A. Toxins (Basel) 2019; 11:toxins11020132. [PMID: 30813459 PMCID: PMC6409803 DOI: 10.3390/toxins11020132] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Aflatoxin M1 (AFM1) and ochratoxin A (OTA), which widely coexist in milk, may pose a serious threat to human health. Mucin is a major component of the intestinal mucus layer, which plays an important role in maintaining intestinal mucosal homeostasis. However, the effect of mycotoxins AFM1 and OTA on intestinal mucin production is still not clear. This study aimed to investigate individual and interactive effects of mycotoxins AFM1 and OTA on the intestinal barrier and the mRNA expression of intestinal mucin (MUC2, MUC5AC and MUC5B) and on protein production in Caco-2/HT29-MTX cultures after 48 h of exposure. Our results show that individual mycotoxins and their mixtures significantly reduced intestinal cell viability and transepithelial electrical resistance (TEER) values, as well as significantly altered intestinal mucin mRNA expression and protein abundance. Moreover, OTA showed toxicity similar to AFM1 in cell viability and TEER value at the same concentration. When the two mycotoxins acted in combination, the synergistic effects observed in the assessment of cell viability and protein abundance in all mono- and co-cultures. In general, this study provides evidence that AFM1 and OTA can damage the intestine, and it contributes to optimized maximum permissible limits of mycotoxins in milk.
Collapse
|
210
|
Rojas DA, Iturra PA, Méndez A, Ponce CA, Bustamante R, Gallo M, Bórquez P, Vargas SL. Increase in secreted airway mucins and partial Muc5b STAT6/FoxA2 regulation during Pneumocystis primary infection. Sci Rep 2019; 9:2078. [PMID: 30765827 PMCID: PMC6376022 DOI: 10.1038/s41598-019-39079-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/19/2018] [Indexed: 12/19/2022] Open
Abstract
Airway mucus responses to subclinical infections may explain variations in progression of chronic lung diseases and differences in clinical expression of respiratory infections across individuals. Pneumocystis associates to more severe Chronic Obstructive Pulmonary Disease (COPD), asthma, respiratory distress of premature newborns, and is a consistent subclinical infection between 2 and 5 months of age when hospitalizations for respiratory cause and infant mortality are higher. This atypical fungus associates to increased mucin 5AC (MUC5AC), a central effector of Th2-type allergic inflammation, in infant lungs. However, mucus progression, expression of MUC5B essential for airway defense, and potential for pharmacologic modulation of mucus during Pneumocystis infection remain unknown. We measured MUC5B and Pneumocystis in infant lungs, and progression of mucin levels and effect of inhibition of the STAT6/FoxA2 mucus pathway using Kaempferol, a JAK/STAT6 inhibitor, in immunocompetent rats during Pneumocystis primary infection. Pneumocystis associated to increased MUC5B in infant lungs. Muc5b increased earlier and more abundantly than Muc5ac during experimental primary infection suggesting an acute defensive response against Pneumocystis as described against bacteria, while increased Muc5ac levels supports an ongoing allergic, Th2 lymphocyte-type response during primary Pneumocystis infection. Kaempferol partly reversed Muc5b stimulation suggesting limited potential for pharmacological modulation via the STAT6-FoxA2 pathway.
Collapse
Affiliation(s)
- Diego A Rojas
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Pablo A Iturra
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Andrea Méndez
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Carolina A Ponce
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Rebeca Bustamante
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile
| | - Miriam Gallo
- Servicio Médico Legal de Santiago, Av. La Paz 1012, Independencia, Santiago, 8380454, Chile
| | - Pamela Bórquez
- Servicio Médico Legal de Santiago, Av. La Paz 1012, Independencia, Santiago, 8380454, Chile
| | - Sergio L Vargas
- Biomedical Sciences Institute, University of Chile School of Medicine, Independencia 1027, Independencia, Santiago, 8380453, Chile.
| |
Collapse
|
211
|
Interleukin-Mediated Pendrin Transcriptional Regulation in Airway and Esophageal Epithelia. Int J Mol Sci 2019; 20:ijms20030731. [PMID: 30744098 PMCID: PMC6386862 DOI: 10.3390/ijms20030731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
Pendrin (SLC26A4), a Cl−/anion exchanger, is expressed at high levels in kidney, thyroid, and inner ear epithelia, where it has an essential role in bicarbonate secretion/chloride reabsorption, iodide accumulation, and endolymph ion balance, respectively. Pendrin is expressed at lower levels in other tissues, such as airways and esophageal epithelia, where it is transcriptionally regulated by the inflammatory cytokines interleukin (IL)-4 and IL-13 through a signal transducer and activator of transcription 6 (STAT6)-mediated pathway. In the airway epithelium, increased pendrin expression during inflammatory diseases leads to imbalances in airway surface liquid thickness and mucin release, while, in the esophageal epithelium, dysregulated pendrin expression is supposed to impact the intracellular pH regulation system. In this review, we discuss some of the recent findings on interleukin-mediated transcriptional regulation of pendrin and how this dysregulation impacts airway and esophagus epithelial homeostasis during inflammatory diseases.
Collapse
|
212
|
Bishwal SC, Das MK, Badireddy VK, Dabral D, Das A, Mahapatra AR, Sahu S, Malakar D, Singh II, Mazumdar H, Patgiri SJ, Deka T, Kapfo W, Liegise K, Kupa RU, Debnath S, Bhowmik R, Debnath R, Behera RK, Pillai MG, Deuri P, Nath R, Khalo KP, Sing WA, Pandit B, Das A, Bhattacharya S, Behera D, Saikia L, Khamo V, Nanda RK. Sputum Proteomics Reveals a Shift in Vitamin D-binding Protein and Antimicrobial Protein Axis in Tuberculosis Patients. Sci Rep 2019; 9:1036. [PMID: 30705350 PMCID: PMC6355791 DOI: 10.1038/s41598-018-37662-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/13/2018] [Indexed: 01/11/2023] Open
Abstract
Existing understanding of molecular composition of sputum and its role in tuberculosis patients is variously limited to its diagnostic potential. We sought to identify infection induced sputum proteome alteration in active/non tuberculosis patients (A/NTB) and their role in altered lung patho-physiology. Out of the study population (n = 118), sputum proteins isolated from discovery set samples (n = 20) was used for an 8-plex isobaric tag for relative and absolute concentration analysis. A minimum set of protein with at least log2(ATB/NTB) >±1.0 in ATB was selected as biosignature and validated in 32 samples. Predictive accuracy was calculated from area under the receiver operating characteristic curve (AUC of ROC) using a confirmatory set (n = 50) by Western blot analysis. Mass spectrometry analysis identified a set of 192 sputum proteins, out of which a signature of β-integrin, vitamin D binding protein:DBP, uteroglobin, profilin and cathelicidin antimicrobial peptide was sufficient to differentiate ATB from NTB. AUC of ROC of the biosignature was calculated to 0.75. A shift in DBP-antimicrobial peptide (AMP) axis in the lungs of tuberculosis patients is observed. The identified sputum protein signature is a promising panel to differentiate ATB from NTB groups and suggest a deregulated DBP-AMP axis in lungs of tuberculosis patients.
Collapse
Affiliation(s)
- Subasa C Bishwal
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,School of Life Sciences, Sambalpur University, JyotiVihar, Sambalpur, India
| | - Mrinal K Das
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Vinod K Badireddy
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Deepti Dabral
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Aleena Das
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Alok R Mahapatra
- School of Life Sciences, Sambalpur University, JyotiVihar, Sambalpur, India
| | - Sukanya Sahu
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - I Ibungo Singh
- Department of Respiratory Medicine, Regional Institute of Medical Sciences, Imphal, India
| | - Himanghsu Mazumdar
- Department of Microbiology, Assam Medical College and Hospital, Dibrugarh, India
| | - Saurav J Patgiri
- Department of Microbiology, Assam Medical College and Hospital, Dibrugarh, India
| | - Trinayan Deka
- Department of Microbiology, Assam Medical College and Hospital, Dibrugarh, India
| | - Wetetsho Kapfo
- Healthcare Laboratory and Research Centre, Naga Hospital Authority Kohima, Nagaland, India
| | - Kevideme Liegise
- Healthcare Laboratory and Research Centre, Naga Hospital Authority Kohima, Nagaland, India
| | - Rukuwe-U Kupa
- Healthcare Laboratory and Research Centre, Naga Hospital Authority Kohima, Nagaland, India
| | - Sanjita Debnath
- Department of Respiratory Medicine, Agartala Government Medical College, Agartala, India
| | - Rajesh Bhowmik
- Department of Respiratory Medicine, Agartala Government Medical College, Agartala, India
| | - Rahul Debnath
- Department of Respiratory Medicine, Agartala Government Medical College, Agartala, India
| | - Rajendra K Behera
- School of Life Sciences, Sambalpur University, JyotiVihar, Sambalpur, India
| | | | - Pranjal Deuri
- Department of Microbiology, Assam Medical College and Hospital, Dibrugarh, India
| | - Reema Nath
- Department of Microbiology, Assam Medical College and Hospital, Dibrugarh, India
| | - K Pewezo Khalo
- Department of Microbiology, Naga Hospital Authority Kohima, Nagaland, India
| | - W Asoka Sing
- Department of Respiratory Medicine, Regional Institute of Medical Sciences, Imphal, India
| | - Bhaswati Pandit
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | - Anjan Das
- Department of Respiratory Medicine, Agartala Government Medical College, Agartala, India
| | | | - Digambar Behera
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Lahari Saikia
- Department of Microbiology, Assam Medical College and Hospital, Dibrugarh, India
| | - Vinotsole Khamo
- Healthcare Laboratory and Research Centre, Naga Hospital Authority Kohima, Nagaland, India
| | - Ranjan K Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.
| |
Collapse
|
213
|
McAuley JL, Gilbertson BP, Trifkovic S, Brown LE, McKimm-Breschkin JL. Influenza Virus Neuraminidase Structure and Functions. Front Microbiol 2019; 10:39. [PMID: 30761095 PMCID: PMC6362415 DOI: 10.3389/fmicb.2019.00039] [Citation(s) in RCA: 267] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/10/2019] [Indexed: 12/31/2022] Open
Abstract
With the constant threat of emergence of a novel influenza virus pandemic, there must be continued evaluation of the molecular mechanisms that contribute to virulence. Although the influenza A virus surface glycoprotein neuraminidase (NA) has been studied mainly in the context of its role in viral release from cells, accumulating evidence suggests it plays an important, multifunctional role in virus infection and fitness. This review investigates the various structural features of NA, linking these with functional outcomes in viral replication. The contribution of evolving NA activity to viral attachment, entry and release of virions from infected cells, and maintenance of functional balance with the viral hemagglutinin are also discussed. Greater insight into the role of this important antiviral drug target is warranted.
Collapse
Affiliation(s)
- Julie L McAuley
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Brad P Gilbertson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sanja Trifkovic
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.,Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Lorena E Brown
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jennifer L McKimm-Breschkin
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
214
|
Krishn SR, Ganguly K, Kaur S, Batra SK. Ramifications of secreted mucin MUC5AC in malignant journey: a holistic view. Carcinogenesis 2019; 39:633-651. [PMID: 29415129 DOI: 10.1093/carcin/bgy019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Heavily glycosylated secreted mucin MUC5AC, by the virtue of its cysteine-rich repeats, can form inter- and intramolecular disulfide linkages resulting in complex polymers, which in turn craft the framework of the polymeric mucus gel on epithelial cell surfaces. MUC5AC is a molecule with versatile functional implications including barrier functions to epithelial cells, host-pathogen interaction, immune cell attraction to sites of premalignant or malignant lesions and tumor progression in a context-dependent manner. Differential expression, glycosylation and localization of MUC5AC have been associated with a plethora of benign and malignant pathologies. In this era of robust technologies, overexpression strategies and genetically engineered mouse models, MUC5AC is emerging as a potential diagnostic, prognostic and therapeutic target for various malignancies. Considering the clinical relevance of MUC5AC, this review holistically encompasses its genomic organization, domain structure, glycosylation patterns, regulation, functional and molecular connotation from benign to malignant pathologies. Furthermore, we have here explored the incipient and significant experimental tools that are being developed to study this structurally complex and evolutionary conserved gel-forming mucin.
Collapse
Affiliation(s)
- Shiv Ram Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
215
|
Kim HG, Choi TY, Bae CH, Choi YS, Na HG, Song SY, Kim YD. Effect of Resolvin D1 and E1 on Mucin Expression in Human Airway Epithelial Cells. KOREAN JOURNAL OF OTORHINOLARYNGOLOGY-HEAD AND NECK SURGERY 2019; 62:28-35. [DOI: 10.3342/kjorl-hns.2018.00164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/09/2018] [Indexed: 07/25/2023]
|
216
|
Fischer AJ, Pino-Argumedo MI, Hilkin BM, Shanrock CR, Gansemer ND, Chaly AL, Zarei K, Allen PD, Ostedgaard LS, Hoffman EA, Stoltz DA, Welsh MJ, Abou Alaiwa MH. Mucus strands from submucosal glands initiate mucociliary transport of large particles. JCI Insight 2019; 4:124863. [PMID: 30626743 DOI: 10.1172/jci.insight.124863] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/29/2018] [Indexed: 01/21/2023] Open
Abstract
Mucus produced by submucosal glands is a key component of respiratory mucociliary transport (MCT). When it emerges from submucosal gland ducts, mucus forms long strands on the airway surface. However, the function of those strands is uncertain. To test the hypothesis that mucus strands facilitate transport of large particles, we studied newborn pigs. In ex vivo experiments, interconnected mucus strands moved over the airway surface, attached to immobile spheres, and initiated their movement by pulling them. Stimulating submucosal gland secretion with methacholine increased the percentage of spheres that moved and shortened the delay until mucus strands began moving spheres. To disrupt mucus strands, we applied reducing agents tris-(2-carboxyethyl)phosphine and dithiothreitol. They decreased the fraction of moving spheres and delayed initiation of movement for spheres that did move. We obtained similar in vivo results with CT-based tracking of microdisks in spontaneously breathing pigs. Methacholine increased the percentage of microdisks moving and reduced the delay until they were propelled up airways. Aerosolized tris-(2-carboxyethyl)phosphine prevented those effects. Once particles started moving, reducing agents did not alter their speed either ex vivo or in vivo. These findings indicate that submucosal glands produce mucus in the form of strands and that the strands initiate movement of large particles, facilitating their removal from airways.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Keyan Zarei
- Department of Internal Medicine and Pappajohn Biomedical Institute.,Department of Biomedical Engineering
| | | | | | - Eric A Hoffman
- Department of Biomedical Engineering.,Department of Radiology, and
| | - David A Stoltz
- Department of Internal Medicine and Pappajohn Biomedical Institute.,Department of Biomedical Engineering.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Michael J Welsh
- Department of Internal Medicine and Pappajohn Biomedical Institute.,Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.,Howard Hughes Medical Institute, University of Iowa, Iowa City, Iowa USA
| | - Mahmoud H Abou Alaiwa
- Department of Internal Medicine and Pappajohn Biomedical Institute.,Department of Biomedical Engineering
| |
Collapse
|
217
|
Xu S, Hui Y, Shu J, Qian J, Li L. Characterization of the human mucin 5AC promoter and its regulation by the histone acetyltransferase P300. Int J Mol Med 2019; 43:1263-1270. [PMID: 30628655 PMCID: PMC6365035 DOI: 10.3892/ijmm.2019.4054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 12/31/2018] [Indexed: 12/14/2022] Open
Abstract
Histone acetylation is important in the modification of gene transcription in asthma and is regulated by histone acetyltransferases (HATs). P300 (P300 HAT) is an enzyme that is able to acetylate a wide variety of proteins. The modification of core histones can further regulate gene transcription, cell proliferation and other cell processes. Airway mucus hypersecretion is one of the most serious pathophysiological symptoms of chronic airway inflammatory diseases, and the human mucin 5AC (MUC5AC) gene has been reported to be a major component of respiratory secretions related to asthma and chronic obstructive pulmonary disease. In the present study, the 5′ sequence of the human MUC5AC gene with a 1,348-bp DNA sequence was amplified from human A549 cells genomic DNA by polymerase chain reaction (PCR), and the product of the PCR was sequenced. By promoter deletion analysis, five promoter segments with different lengths were amplified by PCR. The products were identified by DNA sequencing and the six promoter segments were inserted into pGL3-enhancer vectors. The core promoter area was identified with a series of 5′ deletion promoter plasmids using luciferase reporter assays. MUC5AC promoter activity, and the mRNA and protein expression levels of MUC5AC were observed in P300 wild-type, P300 mutant, P300 small interfering RNA and P300 control groups. The results showed that the core promoter area of MUC5AC was located within the −935/+48 region and that P300 reduced the expression of MUC5AC in A549 cells.
Collapse
Affiliation(s)
- Shiyao Xu
- Division of Pediatric Pulmonology, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Yu Hui
- Division of Pediatric Pulmonology, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jin Shu
- Department of Pediatrics, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Jun Qian
- Division of Pediatric Pulmonology, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Ling Li
- Division of Pediatric Pulmonology, Wuxi Children's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|
218
|
Unuma K, Aki T, Yamashita A, Yoshikawa A, Uemura K. Hydrogen sulfide donor NaHS causes bronchitis with enhanced respiratory secretion in rats. J Toxicol Sci 2019; 44:107-112. [DOI: 10.2131/jts.44.107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Ayaka Yamashita
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
- Yamaguchi University School of Medicine
| | - Ayaka Yoshikawa
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| |
Collapse
|
219
|
Muc5b overexpression causes mucociliary dysfunction and enhances lung fibrosis in mice. Nat Commun 2018; 9:5363. [PMID: 30560893 PMCID: PMC6299094 DOI: 10.1038/s41467-018-07768-9] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 11/14/2018] [Indexed: 01/16/2023] Open
Abstract
The gain-of-function MUC5B promoter variant rs35705950 is the dominant risk factor for developing idiopathic pulmonary fibrosis (IPF). Here we show in humans that MUC5B, a mucin thought to be restricted to conducting airways, is co-expressed with surfactant protein C (SFTPC) in type 2 alveolar epithelia and in epithelial cells lining honeycomb cysts, indicating that cell types involved in lung fibrosis in distal airspace express MUC5B. In mice, we demonstrate that Muc5b concentration in bronchoalveolar epithelia is related to impaired mucociliary clearance (MCC) and to the extent and persistence of bleomycin-induced lung fibrosis. We also establish the ability of the mucolytic agent P-2119 to restore MCC and to suppress bleomycin-induced lung fibrosis in the setting of Muc5b overexpression. Our findings suggest that mucociliary dysfunction might play a causative role in bleomycin-induced pulmonary fibrosis in mice overexpressing Muc5b, and that MUC5B in distal airspaces is a potential therapeutic target in humans with IPF.
Collapse
|
220
|
Momtazi G, Lambrecht BN, Naranjo JR, Schock BC. Regulators of A20 (TNFAIP3): new drug-able targets in inflammation. Am J Physiol Lung Cell Mol Physiol 2018; 316:L456-L469. [PMID: 30543305 DOI: 10.1152/ajplung.00335.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Persistent activation of the transcription factor Nuclear factor-κB (NF-κB) is central to the pathogenesis of many inflammatory disorders, including those of the lung such as cystic fibrosis (CF), asthma, and chronic obstructive pulmonary disease (COPD). Despite recent advances in treatment, management of the inflammatory component of these diseases still remains suboptimal. A20 is an endogenous negative regulator of NF-κB signaling, which has been widely described in several autoimmune and inflammatory disorders and more recently in terms of chronic lung disorders. However, the underlying mechanism for the apparent lack of A20 in CF, COPD, and asthma has not been investigated. Transcriptional regulation of A20 is complex and requires coordination of different transcription factors. In this review we examine the existing body of research evidence on the regulation of A20, concentrating on pulmonary inflammation. Special focus is given to the repressor downstream regulatory element antagonist modulator (DREAM) and its nuclear and cytosolic action to regulate inflammation. We provide evidence that would suggest the A20-DREAM axis to be an important player in (airway) inflammatory responses and point to DREAM as a potential future therapeutic target for the modification of phenotypic changes in airway inflammatory disorders. A schematic summary describing the role of DREAM in inflammation with a focus on chronic lung diseases as well as the possible consequences of altered DREAM expression on immune responses is provided.
Collapse
Affiliation(s)
- G Momtazi
- Centre for Experimental Medicine, Queen's University of Belfast , Belfast , United Kingdom
| | - B N Lambrecht
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - J R Naranjo
- Spanish Network for Biomedical Research in Neurodegenerative Diseases (Centro Investigación Biomédica en Red Enfermedades Neurodegenerativas), Instituto de Salud Carlos III, Madrid, Spain.,National Biotechnology Center, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - B C Schock
- Centre for Experimental Medicine, Queen's University of Belfast , Belfast , United Kingdom
| |
Collapse
|
221
|
Adnane M, Meade KG, O'Farrelly C. Cervico-vaginal mucus (CVM) - an accessible source of immunologically informative biomolecules. Vet Res Commun 2018; 42:255-263. [PMID: 30117040 PMCID: PMC6244541 DOI: 10.1007/s11259-018-9734-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/02/2018] [Indexed: 01/05/2023]
Abstract
Cervico-vaginal mucus (CVM), the product of epithelial cells lining the uterus, cervix and vagina, is secreted to facilitate uterine lubrication and microbial clearance. Predominantly composed of water and mucins, CVM also contains high levels of immuno-active proteins such as immunoglobulin A (IgA), lactoferrin and lysozyme which protect against infection by blocking adhesion and mediating microbial killing. The repertoire of cytokines, chemokines and antimicrobial peptides is predominantly generated by the secretions of endometrial epithelial cells into the uterine lumen and concentrated in the CVM. The quantity and relative proportions of these inflammatory biomarkers are affected by diverse factors including the estrus cycle and health status of the animal and therefore potentially provide important diagnostic and prognostic indicators. We propose that measuring molecular signatures in bovine CVM could be a useful approach to identifying and monitoring genital tract pathologies in beef and dairy cows.
Collapse
Affiliation(s)
- Mounir Adnane
- School of Biochemistry and Immunology & School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
- Institute of Veterinary Sciences, Tiaret, Algeria
| | - Kieran G Meade
- Animal & Bioscience Research Department, Animal & Grassland Research and Innovation Centre, Teagasc, Grange, Co. Meath, Ireland
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology & School of Medicine, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
222
|
Cantero-Recasens G, Butnaru CM, Brouwers N, Mitrovic S, Valverde MA, Malhotra V. Sodium channel TRPM4 and sodium/calcium exchangers (NCX) cooperate in the control of Ca 2+-induced mucin secretion from goblet cells. J Biol Chem 2018; 294:816-826. [PMID: 30482841 DOI: 10.1074/jbc.ra117.000848] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/21/2018] [Indexed: 01/08/2023] Open
Abstract
Regulated mucin secretion is essential for the formation of the mucus layer that protects the underlying epithelial cells from foreign particles. Alterations in the quantity or quality of secreted mucins are therefore detrimental to airway and colon physiology. Based on various biochemical assays in several human cell lines, we report here that Na+/Ca2+ exchanger 2 (NCX2) works in conjunction with transient receptor potential cation channel subfamily M member 4 (TRPM4), and perhaps TRPM5, Na+ channels to control Ca2+-mediated secretion of both mucin 2 (MUC2) and MUC5AC from HT29-18N2 colonic cancer cells. Differentiated normal bronchial epithelial (NHBE) cells and tracheal cells from patients with cystic fibrosis (CFT1-LC3) expressed only TRPM4 and all three isoforms of NCXs. Blocking the activity of TRPM4 or NCX proteins abrogated MUC5AC secretion from NHBE and CFT1-LC3 cells. Altogether, our findings reveal that NCX and TRPM4/TRPM5 are both required for mucin secretion. We therefore propose that these two proteins could be potential pharmacological targets to control mucus-related pathologies such as cystic fibrosis.
Collapse
Affiliation(s)
- Gerard Cantero-Recasens
- From the Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Cristian M Butnaru
- From the Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Nathalie Brouwers
- From the Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Sandra Mitrovic
- the University Hospital of Basel, Clinical Chemistry, Petersgraben 4, 4031 Basel, Switzerland
| | - Miguel A Valverde
- the Laboratory of Molecular Physiology, Faculty of Health and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Catalonia, Spain, and
| | - Vivek Malhotra
- From the Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain, .,the Universitat Pompeu Fabra (UPF), Barcelona, Spain.,the Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluis Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
223
|
Panganiban RA, Sun M, Dahlin A, Park HR, Kan M, Himes BE, Mitchel JA, Iribarren C, Jorgenson E, Randell SH, Israel E, Tantisira K, Shore S, Park JA, Weiss ST, Wu AC, Lu Q. A functional splice variant associated with decreased asthma risk abolishes the ability of gasdermin B to induce epithelial cell pyroptosis. J Allergy Clin Immunol 2018; 142:1469-1478.e2. [PMID: 29330013 PMCID: PMC6037620 DOI: 10.1016/j.jaci.2017.11.040] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 11/12/2017] [Accepted: 11/22/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Genetic variants in the chromosomal region 17q21 are consistently associated with asthma. However, mechanistic studies have not yet linked any of the associated variants to a function that could influence asthma, and as a result, the identity of the asthma gene(s) remains elusive. OBJECTIVES We sought to identify and characterize functional variants in the 17q21 locus. METHODS We used the Exome Aggregation Consortium browser to identify coding (amino acid-changing) variants in the 17q21 locus. We obtained asthma association measures for these variants in both the Genetic Epidemiology Research in Adult Health and Aging (GERA) cohort (16,274 cases and 38,269 matched controls) and the EVE Consortium study (5,303 asthma cases and 12,560 individuals). Gene expression and protein localization were determined by quantitative RT-PCR and fluorescence immunostaining, respectively. Molecular and cellular studies were performed to determine the functional effects of coding variants. RESULTS Two coding variants (rs2305480 and rs11078928) of the gasdermin B (GSDMB) gene in the 17q21 locus were associated with lower asthma risk in both GERA (odds ratio, 0.92; P = 1.01 × 10-6) and EVE (odds ratio, 0.85; joint PEVE = 1.31 × 10-13). In GERA, rs11078928 had a minor allele frequency (MAF) of 0.45 in unaffected (nonasthmatic) controls and 0.43 in asthma cases. For European Americans in EVE, the MAF of rs2305480 was 0.45 for controls and 0.39 for cases; for all EVE subjects, the MAF was 0.32 for controls and 0.27 for cases. GSDMB is highly expressed in differentiated airway epithelial cells, including the ciliated cells. We found that, when the GSDMB protein is cleaved by inflammatory caspase-1 to release its N-terminal fragment, potent pyroptotic cell death is induced. The splice variant rs11078928 deletes the entire exon 6, which encodes 13 amino acids in the critical N-terminus, and abolishes the pyroptotic activity of the GSDMB protein. CONCLUSIONS Our study identified a functional asthma variant in the GSDMB gene of the 17q21 locus and implicates GSDMB-mediated epithelial cell pyroptosis in pathogenesis.
Collapse
Affiliation(s)
- Ronald A Panganiban
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Maoyun Sun
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Amber Dahlin
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Hae-Ryung Park
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Mengyuan Kan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Blanca E Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jennifer A Mitchel
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, Calif
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, Calif
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, University of North Carolina, Chapel Hill, NC
| | - Elliot Israel
- Asthma Research Center, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Kelan Tantisira
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Stephanie Shore
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Jin-Ah Park
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Ann Chen Wu
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass; Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School, Boston, Mass
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Mass.
| |
Collapse
|
224
|
Winkelmann VE, Thompson KE, Neuland K, Jaramillo AM, Fois G, Schmidt H, Wittekindt OH, Han W, Tuvim MJ, Dickey BF, Dietl P, Frick M. Inflammation-induced upregulation of P2X 4 expression augments mucin secretion in airway epithelia. Am J Physiol Lung Cell Mol Physiol 2018; 316:L58-L70. [PMID: 30358443 DOI: 10.1152/ajplung.00157.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mucus clearance provides an essential innate defense mechanism to keep the airways and lungs free of particles and pathogens. Baseline and stimulated mucin secretion from secretory airway epithelial cells need to be tightly regulated to prevent mucus hypersecretion and mucus plugging of the airways. It is well established that extracellular ATP is a potent stimulus for regulated mucus secretion. Previous studies revealed that ATP acts via metabotropic P2Y2 purinoreceptors on goblet cells. Extracellular ATP, however, is also a potent agonist for ionotropic P2X purinoreceptors. Expression of several P2X isoforms has been reported in airways, but cell type-specific expression and the function thereof remained elusive. With this study, we now provide evidence that P2X4 is the predominant P2X isoform expressed in secretory airway epithelial cells. After IL-13 treatment of either human primary tracheal epithelial cells or mice, P2X4 expression is upregulated in vitro and in vivo under conditions of chronic inflammation, mucous metaplasia, and hyperplasia. Upregulation of P2X4 is strongest in MUC5AC-positive goblet cells. Moreover, activation of P2X4 by extracellular ATP augments intracellular Ca2+ signals and mucin secretion, whereas Ca2+ signals and mucin secretion are dampened by inhibition of P2X4 receptors. These data provide new insights into the purinergic regulation of mucin secretion and add to the emerging picture that P2X receptors modulate exocytosis of large secretory organelles and secretion of macromolecular vesicle cargo.
Collapse
Affiliation(s)
| | - Kristin E Thompson
- Centre de Recherche Saint-Antoine, INSERM, Université Pierre et Marie Curie-Université Paris 06, Sorbonne Universités, Paris , France
| | - Kathrin Neuland
- Institute of General Physiology, Ulm University , Ulm , Germany
| | - Ana M Jaramillo
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Giorgio Fois
- Institute of General Physiology, Ulm University , Ulm , Germany
| | - Hanna Schmidt
- Institute of General Physiology, Ulm University , Ulm , Germany
| | | | - Wei Han
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Michael J Tuvim
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Burton F Dickey
- Department of Pulmonary Medicine, MD Anderson Cancer Center , Houston, Texas
| | - Paul Dietl
- Institute of General Physiology, Ulm University , Ulm , Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University , Ulm , Germany
| |
Collapse
|
225
|
Kim MH, Bae CH, Choi YS, Na HG, Song SY, Kim YD. Endoplasmic Reticulum Stress Induces MUC5AC and MUC5B Expression in Human Nasal Airway Epithelial Cells. Clin Exp Otorhinolaryngol 2018; 12:181-189. [PMID: 30336657 PMCID: PMC6453786 DOI: 10.21053/ceo.2018.00493] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/09/2018] [Indexed: 01/08/2023] Open
Abstract
Objectives Endoplasmic reticulum (ER) stress is known to be associated with inflammatory airway diseases, and three major transmembrane receptors: double-stranded RNA-activated protein kinase-like ER kinase, inositol requiring enzyme 1, and activating transcription factor 6 (ATF6) play important roles in ER stress-related proinflammatory signaling. However, the effects of ER stress and these three major signaling pathways on the regulation of the production of airway mucins in human nasal airway epithelial cells have not been elucidated. Methods In primary human nasal epithelial cells, the effect of tunicamycin (an ER stress inducer) and 4-phenylbutyric acid (4-PBA, ER stress inhibitor) on the expression of MUC5AC and MUC5B was investigated by reverse transcriptasepolymerase chain reaction, real-time polymerase chain reaction, enzyme immunoassay, and immunoblot analysis. Small interfering RNA (siRNA) transfection was used to identify the mechanisms involved. Results Tunicamycin increased the expressions of MUC5AC and MUC5B and the mRNA expressions of ER stress-related signaling molecules, including spliced X-box binding protein 1 (XBP-1), transcription factor CCAAT-enhancer-binding protein homologous protein (CHOP), and ATF6. In addition, 4-PBA attenuated the tunicamycin-induced expressions of MUC5AC and MUC5B and the mRNA expressions of ER stress-related signaling molecules. Furthermore, siRNA knockdowns of XBP-1, CHOP, and ATF6 blocked the tunicamycin-induced mRNA expressions and glycoprotein productions of MUC5AC and MUC5B. Conclusion. These results demonstrate that ER stress plays an important role in the regulation of MUC5AC and MUC5B via the activations of XBP-1, CHOP, and ATF6 in human nasal airway epithelial cells.
Collapse
Affiliation(s)
- Min Han Kim
- Department of Otorhinolaryngology-Head and Neck surgery, College of Medicine, Yeungnam University, Daegu, Korea
| | - Chang Hoon Bae
- Department of Otorhinolaryngology-Head and Neck surgery, College of Medicine, Yeungnam University, Daegu, Korea
| | - Yoon Seok Choi
- Department of Otorhinolaryngology-Head and Neck surgery, College of Medicine, Yeungnam University, Daegu, Korea
| | - Hyung Gyun Na
- Department of Otorhinolaryngology-Head and Neck surgery, College of Medicine, Yeungnam University, Daegu, Korea
| | - Si-Youn Song
- Department of Otorhinolaryngology-Head and Neck surgery, College of Medicine, Yeungnam University, Daegu, Korea
| | - Yong-Dae Kim
- Department of Otorhinolaryngology-Head and Neck surgery, College of Medicine, Yeungnam University, Daegu, Korea.,Regional Center for Respiratory Diseases, Yeungnam University Medical Center, Daegu, Korea
| |
Collapse
|
226
|
Mucin 2 (MUC2) modulates the aggressiveness of breast cancer. Breast Cancer Res Treat 2018; 173:289-299. [PMID: 30317423 DOI: 10.1007/s10549-018-4989-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/29/2018] [Indexed: 01/01/2023]
Abstract
PURPOSE Tumors that secrete large volumes of mucus are chemotherapy resistant, however, mechanisms underlying this resistance are unknown. One protein highly expressed in mucin secreting breast cancers is the secreted mucin, Mucin 2 (MUC2). While MUC2 is expressed in some breast cancers it is absent in normal breast tissue, implicating it in breast cancer. However, the effects of MUC2 on breast cancer are largely unknown. This study examined the role of MUC2 in modulating breast cancer proliferation, response to chemotherapy and metastasis. METHODS Using patient derived xenografts we developed two novel cell lines, called BCK4 and PT12, which express high levels of MUC2. To modulate MUC2 levels, BCK4 and PT12 cells were engineered to express shRNA targeted to MUC2 (shMUC2, low MUC2) or a non-targeting control (shCONT, high MUC2) and proliferation and apoptosis were measured in vitro and in vivo. BCK4 cells with shCONT or shMUC2 were labeled with GFP-luciferase and examined in an experimental metastasis model; disease burden and site specific dissemination were monitored by intravital imaging and fluorescence guided dissection, respectively. RESULTS Proliferation decreased in BCK4 and PT12 shMUC2 cells versus control cells both in vitro and in vivo. Chemotherapy induced minimal apoptosis in control cells expressing high MUC2 but increased apoptosis in shMUC2 cells containing low MUC2. An experimental metastasis model showed disease burden decreased when breast cancer cells contained low versus high MUC2. Treatment with Epidermal Growth Factor (EGF) increased MUC2 expression in BCK4 cells; this induction was abolished by the EGF-receptor inhibitor, Erlotinib. CONCLUSIONS MUC2 plays an important role in mediating proliferation, apoptosis and metastasis of breast cancer cells. MUC2 may be important in guiding treatment and predicting outcomes in breast cancer patients.
Collapse
|
227
|
Choi W, Yang AX, Waltenburg MA, Choe S, Steiner M, Radwan A, Lin J, Maddox CW, Stern AW, Fredrickson RL, Lau GW. FOXA2 depletion leads to mucus hypersecretion in canine airways with respiratory diseases. Cell Microbiol 2018; 21:e12957. [DOI: 10.1111/cmi.12957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Woosuk Choi
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Alina X. Yang
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Michelle A. Waltenburg
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Shawn Choe
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Madeline Steiner
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Ahmed Radwan
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Jingjun Lin
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Carrol W. Maddox
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
- Veterinary Diagnostic Laboratory, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Adam W. Stern
- Veterinary Diagnostic Laboratory, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
- Department of Veterinary Clinical Medicine, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Richard L. Fredrickson
- Veterinary Diagnostic Laboratory, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
- Department of Veterinary Clinical Medicine, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| | - Gee W. Lau
- Department of Pathobiology, College of Veterinary MedicineUniversity of Illinois at Urbana‐Champaign Urbana Illinois USA
| |
Collapse
|
228
|
Mucins trigger dispersal of Pseudomonas aeruginosa biofilms. NPJ Biofilms Microbiomes 2018; 4:23. [PMID: 30323945 PMCID: PMC6180003 DOI: 10.1038/s41522-018-0067-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/20/2018] [Accepted: 09/04/2018] [Indexed: 02/07/2023] Open
Abstract
Mucus is a biological gel that lines all wet epithelia in the body, including the mouth, lungs, and digestive tract, and has evolved to protect the body from pathogenic infection. However, microbial pathogenesis is often studied in mucus-free environments that lack the geometric constraints and microbial interactions in physiological three-dimensional mucus gels. We developed fluid-flow and static test systems based on purified mucin polymers, the major gel-forming constituents of the mucus barrier, to understand how the mucus barrier influences bacterial virulence, particularly the integrity of Pseudomonas aeruginosa biofilms, which can become resistant to immune clearance and antimicrobial agents. We found that mucins separate the cells in P. aeruginosa biofilms and disperse them into suspension. Other viscous polymer solutions did not match the biofilm disruption caused by mucins, suggesting that mucin-specific properties mediate the phenomenon. Cellular dispersion depended on functional flagella, indicating a role for swimming motility. Taken together, our observations support a model in which host mucins are key players in the regulation of microbial virulence. These mucins should be considered in studies of mucosal pathogenesis and during the development of novel strategies to treat biofilms. Biofilms are an important survival strategy for pathogenic bacteria including Pseudomonas aeruginosa and whilst mucins play a role the regulation of microbial virulence, microbial pathogenesis on mucosal tissues is often studied in mucin-free contexts. Here, Katharina Ribbeck and colleagues at the Massachusetts Institute of Technology used native purified mucin polymers and examined their effects on the integrity of Pseudomonas aeruginosa biofilms. The mucins dissolved the biofilms by separating the bacteria, which was not observed in other viscous alternative substances examined, but this did rely on functional bacterial motility. Here the authors provide evidence that mucins are involved in suppressing bacterial virulence and should be included in systems used to assess bacterial pathogenesis on mucosal tissues.
Collapse
|
229
|
Piqué N, De Servi B. Rhinosectan ® spray (containing xyloglucan) on the ciliary function of the nasal respiratory epithelium; results of an in vitro study. Allergy Asthma Clin Immunol 2018; 14:41. [PMID: 30337943 PMCID: PMC6174573 DOI: 10.1186/s13223-018-0268-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/23/2018] [Indexed: 12/19/2022] Open
Abstract
Background To assess the effects of Rhinosectan® spray, a medical device containing xyloglucan, on nasal ciliary function (in MucilAir™Nasal cells). Methods MucilAir™Nasal, a three-dimensional organotypic airway tissue model (with different cell types), was treated with Rhinosectan® (30 µl) or with a control (saline solution). The effects of Rhinosectan® were evaluated at 15 and 60 min post-exposure by: measurement of the cilia beating frequency (Hz), mucin detection (Enzyme-Linked Lectin Assay—ELLA), mucociliary clearance (µm/s) and phagocytosis assay (fluorescence). Results Exposure of MucilAir™ to Rhinosectan® did not alter the cilia beating frequency at 15 and 60 min post-exposure (diluted and undiluted). Exposure to Rhinosectan® (undiluted) during 60 min increased mucociliary clearance (93.3 ± 2.1 µm/s vs. 80.9 ± 1.8 µm/s; p < 0.01) and phagocytic activity (1.89-fold increase) in comparison with saline solution. Moreover, a significant decrease in mucin concentration was observed after 15 min of exposure (171.4 ng/ml vs. 306.5 ng/ml; p < 0.01) and at 60 min post-treatment (242.7 ng/ml vs. 339 ng/ml; p < 0.05). Conclusions The application of Rhinosectan® to nasal epithelial cells does not impair ciliary movement, enhances mucociliary clearance and facilitates phagocytosis while reducing mucin secretion, which are optimal properties for the management of rhinitis and associated conditions.
Collapse
Affiliation(s)
- Núria Piqué
- 1Department of Microbiology and Parasitology, Pharmacy Faculty, Universitat de Barcelona (UB), Diagonal Sud, Facultat de Farmàcia, Edifici A, Av Joan XXIII, 27-31, 08028 Barcelona, Spain.,2Institut de Recerca en Nutrició i Seguretat Alimentària de la UB (INSA-UB), Universitat de Barcelona, Barcelona, Spain
| | | |
Collapse
|
230
|
Hussain SS, George S, Singh S, Jayant R, Hu CA, Sopori M, Chand HS. A Small Molecule BH3-mimetic Suppresses Cigarette Smoke-Induced Mucous Expression in Airway Epithelial Cells. Sci Rep 2018; 8:13796. [PMID: 30218002 PMCID: PMC6138652 DOI: 10.1038/s41598-018-32114-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 09/03/2018] [Indexed: 01/07/2023] Open
Abstract
Cigarette smoke (CS) exposure is one of the primary risk factors associated with the chronic mucous hypersecretion (CMH). The antiapoptotic protein, Bcl-2 sustains hyperplastic mucous cells, and the airway epithelium of ex-smokers with CMH as well as mice exposed to chronic CS showed increased Bcl-2 expression. Therefore, we investigated whether Bcl-2 plays a role in CS-induced mucous expression. Primary airway epithelial cells (AECs) of murine and human origin were treated with CS extract (CSE), and there was a concentration- and time-dependent increase in secretory mucin (MUC5AC), mucous regulator (SPDEF) and Bcl-2 expression. Using differentiated human AECs cultured on air-liquid interface, EGFR and ERK1/2 pathways were interrogated. Bcl-2 activity was blocked using a small molecule BH3 mimetic ABT-263 that disrupts the Bcl-2 interaction with pro-apoptotic proteins. The ABT-263 treatment resulted in the downregulation of CSE-induced mucus expression and disrupted the EGFR-signaling while inducing the apoptosis and the pro-apoptotic protein, Bik expression. This strategy significantly suppressed the mainstream CS-induced mucous phenotype in a 3-D human airway epithelium model. Therefore, the present study suggests that CS induces Bcl-2 expression to help promote mucous cell survival; and small molecule BH3 mimetics targeting Bcl-2 could be useful in suppressing the CS-induced mucous response.
Collapse
Affiliation(s)
- Shah S Hussain
- Department of Immunology & Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL - 33199, USA
| | - Shebin George
- Department of Immunology & Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL - 33199, USA
| | - Shashi Singh
- Lovelace Respiratory Research Institute, Albuquerque, NM - 87108, USA
| | - Rahul Jayant
- Department of Immunology & Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL - 33199, USA
| | - Chien-An Hu
- Department of Biochemistry and Molecular Biology, University of New Mexico, Albuquerque, NM - 87131, USA
| | - Mohan Sopori
- Lovelace Respiratory Research Institute, Albuquerque, NM - 87108, USA
| | - Hitendra S Chand
- Department of Immunology & Nano-Medicine, Institute of NeuroImmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL - 33199, USA.
| |
Collapse
|
231
|
Marcos-López M, Calduch-Giner JA, Mirimin L, MacCarthy E, Rodger HD, O'Connor I, Sitjà-Bobadilla A, Pérez-Sánchez J, Piazzon MC. Gene expression analysis of Atlantic salmon gills reveals mucin 5 and interleukin 4/13 as key molecules during amoebic gill disease. Sci Rep 2018; 8:13689. [PMID: 30209326 PMCID: PMC6135806 DOI: 10.1038/s41598-018-32019-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 08/20/2018] [Indexed: 12/19/2022] Open
Abstract
Amoebic gill disease (AGD) is one of the main diseases affecting Atlantic salmon (Salmo salar L.) mariculture. Hallmarks of AGD are hyperplasia of the lamellar epithelium and increased production of gill mucus. This study investigated the expression of genes involved in mucus secretion, cell cycle regulation, immunity and oxidative stress in gills using a targeted 21-gene PCR array. Gill samples were obtained from experimental and natural Neoparamoeba perurans infections, and sampling points included progressive infection stages and post-freshwater treatment. Up-regulation of genes related to mucin secretion and cell proliferation, and down-regulation of pro-inflammatory and pro-apoptotic genes were associated with AGD severity, while partial restoration of the gill homeostasis was detected post-treatment. Mucins and Th2 cytokines accoun ted for most of the variability observed between groups highlighting their key role in AGD. Two mucins (muc5, muc18) showed differential regulation upon disease. Substantial up-regulation of the secreted muc5 was detected in clinical AGD, and the membrane bound muc18 showed an opposite pattern. Th2 cytokines, il4/13a and il4/13b2, were significantly up-regulated from 2 days post-infection onwards, and changes were lesion-specific. Despite the differences between experimental and natural infections, both yielded comparable results that underline the importance of the studied genes in the respiratory organs of fish, and during AGD progression.
Collapse
Affiliation(s)
- Mar Marcos-López
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland. .,FishVet Group Ireland, Unit 7b Oranmore Business Park, Oranmore, Co, Galway, H91 XP3F, Ireland.
| | - Josep A Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain
| | - Luca Mirimin
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland
| | - Eugene MacCarthy
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland
| | - Hamish D Rodger
- FishVet Group Ireland, Unit 7b Oranmore Business Park, Oranmore, Co, Galway, H91 XP3F, Ireland
| | - Ian O'Connor
- Marine and Freshwater Research Centre, Galway-Mayo Institute of Technology, Dublin Road, Galway, Co., Galway, H91 T8NW, Ireland
| | - Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain
| | - M Carla Piazzon
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal (IATS-CSIC), Castellón, 12595, Spain.
| |
Collapse
|
232
|
Gungl A, Biasin V, Wilhelm J, Olschewski A, Kwapiszewska G, Marsh LM. Fra2 Overexpression in Mice Leads to Non-allergic Asthma Development in an IL-13 Dependent Manner. Front Immunol 2018; 9:2018. [PMID: 30233597 PMCID: PMC6133984 DOI: 10.3389/fimmu.2018.02018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/16/2018] [Indexed: 12/28/2022] Open
Abstract
Background: Asthma is a complex chronic inflammatory disease characterised by airway inflammation, remodelling and hyperresponsiveness (AHR). Members of the AP-1 transcription factor family play important roles in the activation of the immune system and the control of cellular responses; however, their role in the development of asthma has not been well studied. We aimed to investigate the role of the lesser known AP-1 family member, Fra2 in experimental asthma. Methods: Phenotypic characterisation and gene expression profiling was performed on Fra2 (TG) overexpressing and wild-type mice. The efficacy of therapeutic interventions in regulating the Fra2 phenotype was determined. Results: Transcriptional profiling of TG mice revealed a high abundance of regulated genes associated with airway remodelling, inflammation and mucus production. A concomitant increase in peribronchial collagen deposition, smooth muscle thickening and mucus production was observed. TG mice possessed increased inflammatory infiltration in the lung, predominantly consisting of eosinophils and T-cells and elevated expression of Th2 cytokines and eotaxin. Furthermore, TG mice possessed severe AHR in response to increasing doses of methacholine. Glucocorticoid treatment led to a partial improvement of the asthma phenotype, whereas blockade of IL-13 via neutralising antibodies ameliorated AHR and mucus production, but had no effect on collagen deposition. Conclusion: We here describe a novel model for non-allergic asthma that does not require the application of exogenous allergens, which mimics several key features of the disease, such as airway inflammation, remodelling and hyperresponsiveness. Fra2 may represent a key molecule coordinating multiple aspects of asthma pathogenesis.
Collapse
Affiliation(s)
- Anna Gungl
- Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Valentina Biasin
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Jochen Wilhelm
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center, Giessen, Germany.,German Center for Lung Research, Justus-Liebig University, Giessen, Germany
| | - Andrea Olschewski
- Otto Loewi Research Center, Medical University of Graz, Graz, Austria.,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Medical University of Graz, Graz, Austria.,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| |
Collapse
|
233
|
Cho HJ, Kim CH. Oxygen matters: hypoxia as a pathogenic mechanism in rhinosinusitis. BMB Rep 2018; 51:59-64. [PMID: 29366441 PMCID: PMC5836558 DOI: 10.5483/bmbrep.2018.51.2.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Indexed: 12/21/2022] Open
Abstract
The airway epithelium is the first place, where a defense mechanism is initiated against environmental stimuli. Mucociliary transport (MCT), which is the defense mechanism of the airway and the role of airway epithelium as mechanical barriers are essential in innate immunity. To maintain normal physiologic function, normal oxygenation is critical for the production of energy for optimal cellular functions. Several pathologic conditions are associated with a decrease in oxygen tension in airway epithelium and chronic sinusitis is one of the airway diseases, which is associated with the hypoxic condition, a potent inflammatory stimulant. We have observed the overexpression of the hypoxia-inducible factor 1 (HIF-1), an essential factor for oxygen homeostasis, in the epithelium of sinus mucosa in sinusitis patients. In a series of previous reports, we have found hypoxia-induced mucus hyperproduction, especially by MUC5AC hyperproduction, disruption of epithelial barrier function by the production of VEGF, and down-regulation of junctional proteins such as ZO-1 and E-cadherin. Furthermore, hypoxia-induced inflammation by HMGB1 translocation into the cytoplasm results in the release of IL-8 through a ROS-dependent mechanism in upper airway epithelium. In this mini-review, we briefly introduce and summarize current progress in the pathogenesis of sinusitis related to hypoxia. The investigation of hypoxia-related pathophysiology in airway epithelium will suggest new insights on airway inflammatory diseases, such as rhinosinusitis for clinical application and drug development.
Collapse
Affiliation(s)
- Hyung-Ju Cho
- Department of Otorhinolaryngology, and The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Chang-Hoon Kim
- Department of Otorhinolaryngology, and The Airway Mucus Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
234
|
Val S, Poley M, Anna K, Nino G, Brown K, Pérez-Losada M, Gordish-Dressman H, Preciado D. Characterization of mucoid and serous middle ear effusions from patients with chronic otitis media: implication of different biological mechanisms? Pediatr Res 2018; 84:296-305. [PMID: 29915406 PMCID: PMC6185811 DOI: 10.1038/s41390-018-0060-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/02/2018] [Accepted: 05/10/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chronic otitis media with effusion (COME) is characterized by persistent middle ear effusions that are in most cases highly viscous, but some patients present with serous fluid. This study aimed at comprehensively characterizing the macromolecular composition of mucoid vs. serous middle ear effusions (MEEs). METHODS MEEs from patients with COME were analyzed for proteins by mass spectrometry (MS) and western blot techniques, total DNA quantity, bacterial DNA (16S sequencing), and cytokine content. Proteomics datasets were studied in Ingenuity Pathway Analysis (IPA). RESULTS Mucoid samples showed a global tendency of increased pro-inflammatory mediators. Interleukin-1β (IL-1β) and IL-10 were significantly more abundant in serous samples (p < 0.01). Mucoid samples had higher DNA quantity (p = 0.04), more likely to be positive in MUC5B protein (p = 0.008) and higher peptide counts (12,786 vs. 2225), as well as an overall larger number of identified proteins (331 vs. 177), compared to serous. IPA found the mucoid sample dataset to be related to immune cell function and epithelial remodeling, whereas the serous sample dataset showed acute responses and blood-related proteins. Interestingly, serous samples showed more bacterial DNA than mucoid ones, with less bacterial genera variability. CONCLUSION This study demonstrates divergent immune responses in children with COME by effusion quality.
Collapse
Affiliation(s)
- Stéphanie Val
- Sheikh Zayed Center for Pediatric Surgical Innovation, Children's National Health System, Washington, DC, USA.
| | - Marian Poley
- Sheikh Zayed Center for Pediatric Surgical Innovation, Children’s National Health System, Washington DC
| | - Krueger Anna
- Sheikh Zayed Center for Pediatric Surgical Innovation, Children’s National Health System, Washington DC
| | - Gustavo Nino
- Center for Genetic Medicine Research, Children’s National Health System, Washington DC,Division of Pediatric Pulmonology, Children’s National Health System, Washington DC
| | - Kristy Brown
- Center for Genetic Medicine Research, Children’s National Health System, Washington DC
| | - Marcos Pérez-Losada
- Computational Biology Institute, Milken Institute School of Public Health, George Washington University, Ashburn VA,CIBIO-InBIO, Universidade do Porto, Campus Agrário de Vairão, Vairão, Portugal
| | | | - Diego Preciado
- Sheikh Zayed Center for Pediatric Surgical Innovation, Children’s National Health System, Washington DC,Division of Pediatric Otolaryngology, Children’s National Health System, Washington DC
| |
Collapse
|
235
|
Huong TN, Yan Y, Jumat MR, Lui J, Tan BH, Wang DY, Sugrue RJ. A sustained antiviral host response in respiratory syncytial virus infected human nasal epithelium does not prevent progeny virus production. Virology 2018; 521:20-32. [PMID: 29870884 DOI: 10.1016/j.virol.2018.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/09/2018] [Accepted: 05/18/2018] [Indexed: 12/01/2022]
Abstract
Respiratory syncytial virus infection was examined using a human nasal epithelial cell model. Maximum levels of shed-virus were produced at between 3 and 5 days post-infection (dpi), and the infectivity of the shed-virus was stable up to 10 dpi. The highest levels of interferon signalling were recorded at 2dpi, and infection induced a widespread antivirus response in the nasal epithelium, involving both infected cells and non-infected cells. Although these cellular responses were associated with reduced levels of progeny virus production and restricted virus spread, they did not inhibit the infectivity virus that is shed early in infection. In the clinical context these data suggest that although the host cell response in the nasal epithelium may restrict the levels of progeny virus particles produced, the stability of the shed-virus in the nasal mucosa may be an important factor in both disease progression and virus transmission.
Collapse
Affiliation(s)
- Tra Nguyen Huong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Yan Yan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Republic of Singapore
| | - Muhammad Raihan Jumat
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore
| | - Jing Lui
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Republic of Singapore
| | - Boon Huan Tan
- Detection and Diagnostics Laboratory, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Republic of Singapore
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 119228, Republic of Singapore
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Republic of Singapore.
| |
Collapse
|
236
|
Birket S. A clean sweep: mucin bundles clear the airway. Eur Respir J 2018; 52:52/2/1801144. [DOI: 10.1183/13993003.01144-2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/02/2018] [Indexed: 11/05/2022]
|
237
|
Lakshmi SP, Reddy AT, Banno A, Reddy RC. Airway Epithelial Cell Peroxisome Proliferator-Activated Receptor γ Regulates Inflammation and Mucin Expression in Allergic Airway Disease. THE JOURNAL OF IMMUNOLOGY 2018; 201:1775-1783. [PMID: 30061200 DOI: 10.4049/jimmunol.1800649] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/06/2018] [Indexed: 11/19/2022]
Abstract
Airway epithelial cells (AECs) orchestrate inflammatory responses to airborne irritants that enter the respiratory system. A viscous mucus layer produced by goblet cells in the airway epithelium also contributes to a physiological defense mechanism through the physical and chemical barriers it provides. Dysregulation or impairment in these functions has been implicated as a cause of the chronic inflammation and tissue remodeling that constitute major pathological features of asthma. In particular, mucus hypersecretion leading to airway obstruction and impaired pulmonary function is associated with morbidity and mortality in asthma patients. Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated transcription factor involved in a variety of cellular processes. Accumulating evidence indicates that PPARγ agonists antagonize exaggerated inflammatory responses, yet PPARγ's precise role in airway remodeling/mucus hypersecretion has yet to be defined. In this study, we created an AEC-specific PPARγ (AEC-PPARγ) deletion to investigate PPARγ's functions in a murine model of allergic airway disease. AEC-PPARγ deficiency exaggerated airway hyperresponsiveness, inflammation, cytokine expression, and tissue remodeling. We also found that PPARγ directly bound to a PPAR response element found in MUC5AC and repressed gene expression. Likewise, PPARγ regulated mucin and inflammatory factors in primary human bronchial epithelial cells. In light of the current standard therapies' limited and inadequate direct effect on airway mucus hypersecretion, our study showing AEC-PPARγ's role as a transcriptional repressor of MUC5AC highlights this receptor's potential as a pharmacological target for asthma.
Collapse
Affiliation(s)
- Sowmya P Lakshmi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Aravind T Reddy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| | - Asoka Banno
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and
| | - Raju C Reddy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213; and .,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240
| |
Collapse
|
238
|
Song H, Jung BK, Cho J, Chai JY. Worm expulsion of Gymnophalloides seoi from C57BL/6 mice: role of metacercarial exosomes in upregulating TLR2 and MUC2 expression in intestinal tissues. Parasitol Res 2018; 117:3309-3314. [PMID: 30019213 DOI: 10.1007/s00436-018-6002-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/04/2018] [Indexed: 11/26/2022]
Abstract
Gymnophalloides seoi worms were rapidly expelled from C57BL/6 mice within days 3-6 post-infection probably due to operation of mucosal innate immunity. To understand better the mucosal immunity related to worm expulsion from the host, we isolated exosomes of G. seoi metacercariae and investigated their role in induction of mRNA and protein expression of several Toll-like receptors and mucin-related factors in vitro. G. seoi-secreted exosomes were collected using differential ultracentrifugation, and cellular internalization of the exosomes into HT-29 intestinal epithelial cells was visualized by confocal microscopy. The expression of TLR2 and MUC2 in HT-29 cells was up-regulated in stimulation with the exosomes. We suggest that G. seoi-secreted exosomes offer a new point of view in the mechanism of worm expulsion from the host through enhancement of TLR2 and MUC2 expression.
Collapse
Affiliation(s)
- Hyemi Song
- Korea Association of Health Promotion, Institute of Parasitic Diseases, Seoul, 07649, South Korea
| | - Bong-Kwang Jung
- Korea Association of Health Promotion, Institute of Parasitic Diseases, Seoul, 07649, South Korea
| | - Jaeeun Cho
- Korea Association of Health Promotion, Institute of Parasitic Diseases, Seoul, 07649, South Korea
| | - Jong-Yil Chai
- Korea Association of Health Promotion, Institute of Parasitic Diseases, Seoul, 07649, South Korea.
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
239
|
Saco TV, Breitzig MT, Lockey RF, Kolliputi N. Epigenetics of Mucus Hypersecretion in Chronic Respiratory Diseases. Am J Respir Cell Mol Biol 2018; 58:299-309. [PMID: 29096066 DOI: 10.1165/rcmb.2017-0072tr] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Asthma, chronic obstructive pulmonary disease, and cystic fibrosis are three chronic pulmonary diseases that affect an estimated 420 million individuals across the globe. A key factor contributing to each of these conditions is mucus hypersecretion. Although management of these diseases is vastly studied, researchers have only begun to scratch the surface of the mechanisms contributing to mucus hypersecretion. Epigenetic regulation of mucus hypersecretion, other than microRNA post-translational modification, is even more scarcely researched. Detailed study of epigenetic mechanisms, such as DNA methylation and histone modification, could not only help to better the understanding of these respiratory conditions but also reveal new treatments for them. Because mucus hypersecretion is such a complex event, there are innumerable genes involved in the process, which are beyond the scope of a single review. Therefore, the purpose of this review is to narrow the focus and summarize specific epigenetic research that has been conducted on a few aspects of mucus hypersecretion in asthma, chronic obstructive pulmonary disease, cystic fibrosis, and some cancers. Specifically, this review emphasizes the contribution of DNA methylation and histone modification of particular genes involved in mucus hypersecretion to identify possible targets for the development of future therapies for these conditions. Elucidating the role of epigenetics in these respiratory diseases may provide a breath of fresh air to millions of affected individuals around the world.
Collapse
Affiliation(s)
- Tara V Saco
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Mason T Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
240
|
Bakshani CR, Morales-Garcia AL, Althaus M, Wilcox MD, Pearson JP, Bythell JC, Burgess JG. Evolutionary conservation of the antimicrobial function of mucus: a first defence against infection. NPJ Biofilms Microbiomes 2018; 4:14. [PMID: 30002868 PMCID: PMC6031612 DOI: 10.1038/s41522-018-0057-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/05/2018] [Accepted: 06/06/2018] [Indexed: 02/07/2023] Open
Abstract
Mucus layers often provide a unique and multi-functional hydrogel interface between the epithelial cells of organisms and their external environment. Mucus has exceptional properties including elasticity, changeable rheology and an ability to self-repair by re-annealing, and is therefore an ideal medium for trapping and immobilising pathogens and serving as a barrier to microbial infection. The ability to produce a functional surface mucosa was an important evolutionary step, which evolved first in the Cnidaria, which includes corals, and the Ctenophora. This allowed the exclusion of non-commensal microbes and the subsequent development of the mucus-lined digestive cavity seen in higher metazoans. The fundamental architecture of the constituent glycoprotein mucins is also evolutionarily conserved. Although an understanding of the biochemical interactions between bacteria and the mucus layer are important to the goal of developing new antimicrobial strategies, they remain relatively poorly understood. This review summarises the physicochemical properties and evolutionary importance of mucus, which make it so successful in the prevention of bacterial infection. In addition, the strategies developed by bacteria to counteract the mucus layer are also explored.
Collapse
Affiliation(s)
- Cassie R Bakshani
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ana L Morales-Garcia
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Mike Althaus
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew D Wilcox
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - Jeffrey P Pearson
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | - John C Bythell
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - J Grant Burgess
- School of Natural and Environmental Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
241
|
Restrepo MI, Sibila O, Anzueto A. Pneumonia in Patients with Chronic Obstructive Pulmonary Disease. Tuberc Respir Dis (Seoul) 2018; 81:187-197. [PMID: 29962118 PMCID: PMC6030662 DOI: 10.4046/trd.2018.0030] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 03/30/2018] [Accepted: 04/01/2018] [Indexed: 11/24/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a frequent comorbid condition associated with increased morbidity and mortality. Pneumonia is the most common infectious disease condition. The purpose of this review is to evaluate the impact of pneumonia in patients with COPD. We will evaluate the epidemiology and factors associated with pneumonia. We are discussing the clinical characteristics of COPD that may favor the development of infections conditions such as pneumonia. Over the last 10 years, there is an increased evidence that COPD patients treated with inhaled corticosteroids are at increased risk to develp pneumonia. We will review the avaialbe information as well as the possible mechanism for this events. We also discuss the impact of influenza and pneumococcal vaccination in the prevention of pneumonia in COPD patients.
Collapse
Affiliation(s)
- Marcos I Restrepo
- South Texas Veterans Health Care System, San Antonio, TX, USA
- Veterans Evidence Based Research Dissemination and Implementation Center (VERDICT) (MR), San Antonio, TX, USA
| | - Oriol Sibila
- Servei de Pneumologia, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Antonio Anzueto
- South Texas Veterans Health Care System, San Antonio, TX, USA
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
242
|
Kim SS, Kim CH, Kim JW, Kung HC, Park TW, Shin YS, Kim JD, Ryu S, Kim WJ, Choi YH, Song KS. Airborne particulate matter increases MUC5AC expression by downregulating Claudin-1 expression in human airway cells. BMB Rep 2018; 50:516-521. [PMID: 28946937 PMCID: PMC5683821 DOI: 10.5483/bmbrep.2017.50.10.100] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Indexed: 12/15/2022] Open
Abstract
CLB2.0, a constituent of PM, induces secretion of multiple cytokines and chemokines that regulate airway inflammation. Specifically, IL-6 upregulates CLB2.0-induced MUC5AC and MUC1 expression. Interestingly, of the tight junction proteins examined, claudin-1 expression was inhibited by CLB2.0. While the overexpression of claudin-1 decreased CLB2.0-induced MUC5AC expression, it increased the expression of the anti-inflammatory mucin, MUC1. CLB2.0-induced IL-6 secretion was mediated by ROS. The ROS scavenger N-acetylcysteine inhibited CLB2.0-induced IL-6 secretion, thereby decreasing the CLB2.0-induced MUC5AC expression, whereas CLB2.0-induced MUC1 expression increased. CLB2.0 activated the ERK1/2 MAPK via a ROS-dependent pathway. ERK1/2 downregulated the claudin-1 and MUC1 expressions, whereas it dramatically increased CLB2.0-induced MUC5AC expression. These findings suggest that CLB2.0-induced ERK1/2 activation acts as a switch for regulating inflammatory conditions though a ROS-dependent pathway. Our data also suggest that secreted IL-6 regulates CLB2.0-induced MUC5AC and MUC1 expression via ROS-mediated downregulation of claudin-1 expression to maintain mucus homeostasis in the airway.
Collapse
Affiliation(s)
- Sang-Su Kim
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Cheol Hong Kim
- Department of Pediatrics, Sungkyunkwan University Samsung Changwon Hospital, Changwon 51353, Korea
| | - Ji Wook Kim
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Hsi Chiang Kung
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Tae Woo Park
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Yu Som Shin
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Ju Deok Kim
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Siejeong Ryu
- Department of Anesthesiology and Pain Medicine, Kosin University College of Medicine, Busan 49267, Korea
| | - Wang-Joon Kim
- Department of Physiology, Kosin University College of Medicine, Busan 49267, Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Don-Eui University, Busan 47227, Korea
| | - Kyoung Seob Song
- Department of Physiology, Kosin University College of Medicine, Busan 49267; Institute of Medicine, Kosin University College of Medicine, Busan 49267, Korea
| |
Collapse
|
243
|
Weiser JN, Ferreira DM, Paton JC. Streptococcus pneumoniae: transmission, colonization and invasion. Nat Rev Microbiol 2018; 16:355-367. [PMID: 29599457 PMCID: PMC5949087 DOI: 10.1038/s41579-018-0001-8] [Citation(s) in RCA: 560] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Streptococcus pneumoniae has a complex relationship with its obligate human host. On the one hand, the pneumococci are highly adapted commensals, and their main reservoir on the mucosal surface of the upper airways of carriers enables transmission. On the other hand, they can cause severe disease when bacterial and host factors allow them to invade essentially sterile sites, such as the middle ear spaces, lungs, bloodstream and meninges. Transmission, colonization and invasion depend on the remarkable ability of S. pneumoniae to evade or take advantage of the host inflammatory and immune responses. The different stages of pneumococcal carriage and disease have been investigated in detail in animal models and, more recently, in experimental human infection. Furthermore, widespread vaccination and the resulting immune pressure have shed light on pneumococcal population dynamics and pathogenesis. Here, we review the mechanistic insights provided by these studies on the multiple and varied interactions of the pneumococcus and its host.
Collapse
|
244
|
Honkova K, Rossnerova A, Pavlikova J, Svecova V, Klema J, Topinka J, Milcova A, Libalova H, Choi H, Veleminsky M, Sram RJ, Rossner P. Gene expression profiling in healthy newborns from diverse localities of the Czech Republic. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:401-415. [PMID: 29602183 DOI: 10.1002/em.22184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/19/2018] [Accepted: 02/23/2018] [Indexed: 06/08/2023]
Abstract
Prenatal exposure to air pollution is associated with intrauterine growth restriction and low birth weight. Gene expression changes in newborns in relation to air pollution have not been sufficiently studied. We analyzed whole genome expression in cord blood leukocytes of 202 newborns from diverse localities of the Czech Republic, differing among other factors in levels of air pollution: the district of Karvina (characterized by higher concentration of air pollutants) and Ceske Budejovice (lower air pollution levels). We aimed to identify differentially expressed genes (DEGs) and pathways in relation to locality and concentration of air pollutants. We applied the linear model to identify the specific DEGs and the correlation analysis, to investigate the relationship between the concentrations of air pollutants and gene expression data. An analysis of biochemical pathways and gene set enrichment was also performed. In general, we observed modest changes of gene expression, mostly attributed to the effect of the locality. The highest number of DEGs was found in samples from the district of Karvina. A pathway analysis revealed a deregulation of processes associated with cell growth, apoptosis or cellular homeostasis, immune response-related processes or oxidative stress response. The association between concentrations of air pollutants and gene expression changes was weak, particularly for samples collected in Karvina. In summary, as we did not find a direct effect of exposure to air pollutants, we assume that the general differences in the environment, rather than actual concentrations of individual pollutants, represent a key factor affecting gene expression changes at delivery. Environ. Mol. Mutagen. 59:401-415, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Katerina Honkova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Rossnerova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jitka Pavlikova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vlasta Svecova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Klema
- Czech Technical University, Prague, Czech Republic
| | - Jan Topinka
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Alena Milcova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Helena Libalova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Hyunok Choi
- School of Public Health, University at Albany, Rensselaer, New York
| | - Milos Veleminsky
- Faculty of Health and Social Studies, University of South Bohemia, Ceske Budejovice, Czech Republic
| | - Radim J Sram
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Rossner
- Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
245
|
Role of mucins in lung homeostasis: regulated expression and biosynthesis in health and disease. Biochem Soc Trans 2018; 46:707-719. [PMID: 29802217 DOI: 10.1042/bst20170455] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 01/02/2023]
Abstract
In humans and mice, the first line of innate defense against inhaled pathogens and particles in the respiratory tract is airway mucus. The primary solid components of the mucus layer are the mucins MUC5AC and MUC5B, polymeric glycoproteins whose changes in abundance and structure can dramatically affect airway defense. Accordingly, MUC5AC/Muc5ac and MUC5B/Muc5b are tightly regulated at a transcriptional level by tissue-specific transcription factors in homeostasis and in response to injurious and inflammatory triggers. In addition to modulated levels of mucin gene transcription, translational and post-translational biosynthetic processes also exert significant influence upon mucin function. Mucins are massive macromolecules with numerous functional domains that contribute to their structural composition and biophysical properties. Single MUC5AC and MUC5B apoproteins have molecular masses of >400 kDa, and von Willebrand factor D-like as well as other cysteine-rich domain segments contribute to mucin polymerization and flexibility, thus increasing apoprotein length and complexity. Additional domains serve as sites for O-glycosylation, which increase further mucin mass several-fold. Glycosylation is a defining process for mucins that is specific with respect to additions of glycans to mucin apoprotein backbones, and glycan additions influence the physical properties of the mucins via structural modifications as well as charge interactions. Ultimately, through their tight regulation and complex assembly, airway mucins follow the biological rule of 'form fits function' in that their structural organization influences their role in lung homeostatic mechanisms.
Collapse
|
246
|
Erdoğan S, Sağsöz H. Papillary Architecture and Functional Characterization of Mucosubstances in the Sheep Tongue. Anat Rec (Hoboken) 2018; 301:1320-1335. [DOI: 10.1002/ar.23840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Serkan Erdoğan
- Department of Anatomy, Faculty of Veterinary Medicine; Namık Kemal University; Tekirdağ 59030 Turkey
| | - Hakan Sağsöz
- Department of Histology and Embryology, Faculty of Veterinary Medicine; Dicle University; Diyarbakır 21280 Turkey
| |
Collapse
|
247
|
Katamreddy JD, Yalavarthi PR, D SR, Battu S, Peesa JP. Biopharmaceutical insights of particulate emulsified systems - a prospective overview. Lipids Health Dis 2018; 17:112. [PMID: 29747645 PMCID: PMC5946457 DOI: 10.1186/s12944-018-0757-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 04/24/2018] [Indexed: 11/10/2022] Open
Abstract
During the twenty-first century, drug discovery is expanding rapidly and a large number of chemical moieties are recognized. Many of them are poorly soluble and hence related biopharmaceutical constraints are to be addressed systematically. Among novel approaches to resolving biopharmaceutical issues, micro- and nano-emulsified systems serve as the best strategy for delivering both hydrophobic and hydrophilic drugs owing to their greater solubilization and transportation capabilities. Of late, the unique physical and biopharmaceutical properties of these liquid isotropic homogenous systems have gained substantive research importance. In addition nano/micro lipid systems share structural and functional similarity with that of the physiological lipids which offer better tolerance ability in the body. In this context, this article provides information on the historical emergence of particulate emulsified systems, importance and rationale of selection of carriers. It also encompasses the physicochemical principles that are responsible for the elevation of therapeutic outcomes of delivery systems. Detailed and schematic absorption of these drug delivery systems is explained here. Gastro-intestinal biochemistry necessary in the understanding of digestion process, lipolytic products formed, micellar structures, enzymes, transporters, mechanism of cell uptake involved after subsequent oral absorption are also emphasized. In addition, this article also explains disposition and pharmacokinetic properties of emulsified systems with real-time therapeutic research outcomes. The influence of biochemical compositions and biopharmaceutical principles on absorption and disposition patterns of ME/NEs was described in the article for the interest of readers and young researchers.
Collapse
Affiliation(s)
- Jyothshna Devi Katamreddy
- Faculty of Pharmaceutical Sciences, JNTUA, Ananthapuramu, 515002, India. .,Department of Pharmaceutics, Krishna Teja Pharmacy College, Tirupati, 517506, India.
| | | | - Subba Rao D
- Department of Chemical Engineering, JNTUA College of Engineering, Ananthapuramu, 515002, India
| | - Sowjanya Battu
- Department of Pharmaceutics, CMR College of Pharmacy, Hyderabad, 501401, India
| | - Jaya Preethi Peesa
- Department of Pharmaceutical Chemistry, Sree Vidyanikethan College of Pharmacy, Tirupati, 517102, India
| |
Collapse
|
248
|
Samaniego Lopez C, Hebe Martínez J, Uhrig ML, Coluccio Leskow F, Spagnuolo CC. A Highly Sensitive Fluorogenic Probe for Imaging Glycoproteins and Mucine Activity in Live Cells in the Near-Infrared Region. Chemistry 2018; 24:6344-6348. [DOI: 10.1002/chem.201800790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Cecilia Samaniego Lopez
- Departamento de Química Orgánica-CIHIDECAR-CONICET; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - Jimena Hebe Martínez
- IQUIBICEN-CONICET, Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - María Laura Uhrig
- Departamento de Química Orgánica-CIHIDECAR-CONICET; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - Federico Coluccio Leskow
- IQUIBICEN-CONICET, Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - Carla Cecilia Spagnuolo
- Departamento de Química Orgánica-CIHIDECAR-CONICET; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| |
Collapse
|
249
|
De Rose V, Molloy K, Gohy S, Pilette C, Greene CM. Airway Epithelium Dysfunction in Cystic Fibrosis and COPD. Mediators Inflamm 2018; 2018:1309746. [PMID: 29849481 PMCID: PMC5911336 DOI: 10.1155/2018/1309746] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/15/2018] [Accepted: 02/01/2018] [Indexed: 12/22/2022] Open
Abstract
Cystic fibrosis is a genetic disease caused by mutations in the CFTR gene, whereas chronic obstructive pulmonary disease (COPD) is mainly caused by environmental factors (mostly cigarette smoking) on a genetically susceptible background. Although the etiology and pathogenesis of these diseases are different, both are associated with progressive airflow obstruction, airway neutrophilic inflammation, and recurrent exacerbations, suggesting common mechanisms. The airway epithelium plays a crucial role in maintaining normal airway functions. Major molecular and morphologic changes occur in the airway epithelium in both CF and COPD, and growing evidence suggests that airway epithelial dysfunction is involved in disease initiation and progression in both diseases. Structural and functional abnormalities in both airway and alveolar epithelium have a relevant impact on alteration of host defences, immune/inflammatory response, and the repair process leading to progressive lung damage and impaired lung function. In this review, we address the evidence for a critical role of dysfunctional airway epithelial cells in chronic airway inflammation and remodelling in CF and COPD, highlighting the common mechanisms involved in the epithelial dysfunction as well as the similarities and differences of the two diseases.
Collapse
Affiliation(s)
- Virginia De Rose
- Department of Clinical and Biological Sciences, University of Torino, A.O.U. S. Luigi Gonzaga, Regione Gonzole 10, 10043 Orbassano, Torino, Italy
| | - Kevin Molloy
- Department of Medicine, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Sophie Gohy
- Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Université Catholique de Louvain (UCL), Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Charles Pilette
- Institute of Experimental and Clinical Research, Pole of Pneumology, ENT and Dermatology, Université Catholique de Louvain (UCL), Brussels, Belgium
- Department of Pneumology, Cliniques Universitaires St-Luc, Brussels, Belgium
| | - Catherine M. Greene
- Lung Biology Group, Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland
| |
Collapse
|
250
|
Li H, Guo D, Zhang L, Feng X. Glycyrrhizin attenuates histamine-mediated MUC5AC upregulation, inflammatory cytokine production, and aquaporin 5 downregulation through suppressing the NF-κB pathway in human nasal epithelial cells. Chem Biol Interact 2018; 285:21-26. [PMID: 29452068 DOI: 10.1016/j.cbi.2018.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/29/2018] [Accepted: 02/12/2018] [Indexed: 01/19/2023]
Abstract
Allergic rhinitis (AR) is a chronic respiratory inflammatory disease. Glycyrrhizin is a main bioactive component of the licorice root extract and exhibits anti-inflammatory activity. However, the role of glycyrrhizin in AR has not been studied. The aim of the present study was to investigate the effect of glycyrrhizin on histamine-induced human nasal epithelial cells (HNEpCs). Here, we found that glycyrrhizin (20 or 40 μM) inhibited histamine-induced the mRNA expression and secretion of mucin 5 subtype AC (MUC5AC), interleukin (IL)-6 and IL-8 in HNEpCs. The expression levels of aquaporin 5 (AQP5) and phosphorylated cyclic adenosine monophosphate-responsive element binding protein (p-CREB) were decreased by histamine in HNEpCs and increased in cells treated with glycyrrhizin. The glycyrrhizin treatment inhibited histamine-induced expressions of p-NF-κB p65 and p-IκBα in HNEpCs, indicating that glycyrrhizin inhibited the activation of NF-κB pathway in histamine-induced HNEpCs. In addition, inhibition of the NF-κB pathway exhibited the similar effect with glycyrrhizin on histamine-induced HNEpCs. In summary, the results showed that glycyrrhizin reversed the effect of histamine on MUC5AC expression, inflammatory cytokine production, and AQP5 expression in HNEpCs, and the NF-κB pathway was involved in the effect. Glycyrrhizin might be used for complementary and alternative therapeutics of AR.
Collapse
Affiliation(s)
- Haixia Li
- Department of Otolaryngology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan, China.
| | - Dandan Guo
- Department of Otolaryngology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan, China
| | - Liangran Zhang
- Department of Otolaryngology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan, China
| | - Xiao Feng
- Department of Otolaryngology, Huaihe Hospital of Henan University, Kaifeng 475000, Henan, China
| |
Collapse
|