1
|
Goriounova AS, Flori Sassano M, Wrennall JA, Tarran R. ELD607 specifically traffics Orai1 to the lysosome leading to inhibition of store operated calcium entry. Cell Calcium 2024; 123:102945. [PMID: 39191091 DOI: 10.1016/j.ceca.2024.102945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024]
Abstract
Orai1 is a plasma membrane Ca2+ channel involved in store operated calcium entry (SOCE). SOCE can regulate cell growth, exocytosis, gene expression and inflammation. We previously found that short palate lung and nasal epithelial clone 1's (SPLUNC1) sixth α-helix (α6) bound Orai1 to inhibit SOCE. SPLUNC1 was not proteolytically stable, so we developed ELD607, an 11 amino acid peptide based on SPLUNC1's α6 region which was more stable and more potent than SPLUNC1/α6. Here, we studied ELD607's mechanism of action. We overexpressed either Orai1-HA or Orai1-YFP in HEK293T cells to probe ELD607-Orai1 interactions by confocal microscopy. We also measured changes in Fluo-4 fluorescence in a multiplate reader as a marker of cytoplasmic Ca2+ levels. ELD607 internalized Orai1 independently of STIM1. Both 15 min and 3 h exposure to ELD607 similarly depleted Orai1 in the plasma membrane. However, 3 h exposure to ELD607 yielded greater inhibition of SOCE. ELD607 continued to colocalize with Orai1 after internalization and this process was dependent on the presence of the ubiquitin ligase NEDD4.2. Similarly, ELD607 increased the colocalization between Orai1 and ubiquitin. ELD607 also increased the colocalization between Orai1 and Rab5 and 7, but not Rab11, suggesting that Orai1 trafficked through early and late but not recycling endosomes. Finally, ELD607 caused Orai1, but not Orai2, Orai3, or STIM1 to traffic to lysosomes. We conclude that ELD607 rapidly binds to Orai1 and works in an identical fashion as full length SPLUNC1 by internalizing Orai1 and sending it to lysosomes, leading to a decrease in SOCE.
Collapse
Affiliation(s)
- Alexandra S Goriounova
- Department of Pharmacology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - M Flori Sassano
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA
| | - Joe A Wrennall
- Department of Cell Biology & Physiology, The University of North Carolina at Chapel Hill, NC 27599, USA
| | - Robert Tarran
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA.
| |
Collapse
|
2
|
Saint-Martin Willer A, Montani D, Capuano V, Antigny F. Orai1/STIMs modulators in pulmonary vascular diseases. Cell Calcium 2024; 121:102892. [PMID: 38735127 DOI: 10.1016/j.ceca.2024.102892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/27/2024] [Accepted: 04/23/2024] [Indexed: 05/14/2024]
Abstract
Calcium (Ca2+) is a secondary messenger that regulates various cellular processes. However, Ca2+ mishandling could lead to pathological conditions. Orai1 is a Ca2+channel contributing to the store-operated calcium entry (SOCE) and plays a critical role in Ca2+ homeostasis in several cell types. Dysregulation of Orai1 contributed to severe combined immune deficiency syndrome, some cancers, pulmonary arterial hypertension (PAH), and other cardiorespiratory diseases. During its activation process, Orai1 is mainly regulated by stromal interacting molecule (STIM) proteins, especially STIM1; however, many other regulatory partners have also been recently described. Increasing knowledge about these regulatory partners provides a better view of the downstream signalling pathways of SOCE and offers an excellent opportunity to decipher Orai1 dysregulation in these diseases. These proteins participate in other cellular functions, making them attractive therapeutic targets. This review mainly focuses on Orai1 regulatory partners in the physiological and pathological conditions of the pulmonary circulation and inflammation.
Collapse
Affiliation(s)
- Anaïs Saint-Martin Willer
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - David Montani
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l'Hypertension Pulmonaire, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Véronique Capuano
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Hôptal Marie Lannelongue, Groupe Hospitalier Paris Saint-Joseph, Le Plessis-Robinson, France
| | - Fabrice Antigny
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999 Hypertension pulmonaire: Physiopathologie et Innovation Thérapeutique, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.
| |
Collapse
|
3
|
Ahmad S, Wrennall JA, Goriounova AS, Sekhri M, Iskarpatyoti JA, Ghosh A, Abdelwahab SH, Voeller A, Rai M, Mahida RY, Krajewski K, Ignar DM, Greenbaum A, Moran TP, Tilley SL, Thickett DR, Sassano MF, Tarran R. Specific Inhibition of Orai1-mediated Calcium Signalling Resolves Inflammation and Clears Bacteria in an Acute Respiratory Distress Syndrome Model. Am J Respir Crit Care Med 2024; 209:703-715. [PMID: 37972349 PMCID: PMC10945054 DOI: 10.1164/rccm.202308-1393oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023] Open
Abstract
Rationale: Acute respiratory distress syndrome (ARDS) has an unacceptably high mortality rate (35%) and is without effective therapy. Orai1 is a Ca2+ channel involved in store-operated Ca2+ entry (SOCE), a process that exquisitely regulates inflammation. Orai1 is considered a druggable target, but no Orai1-specific inhibitors exist to date. Objectives: To evaluate whether ELD607, a first-in-class Orai1 antagonist, can treat ARDS caused by bacterial pneumonia in preclinical models. Methods: ELD607 pharmacology was evaluated in HEK293T cells and freshly isolated immune cells from patients with ARDS. A murine acute lung injury model caused by bacterial pneumonia was then used: mice were infected with Pseudomonas aeruginosa, Staphylococcus aureus, methicillin-resistant S. aureus, or multidrug-resistant P. aeruginosa and then treated with ELD607 intranasally. Measurements and Main Results: ELD607 specifically inhibited SOCE in HEK293T cells with a half-maximal inhibitory concentration of 9 nM. ELD607 was stable in ARDS airway secretions and inhibited SOCE in ARDS immune cells. In vivo, inhaled ELD607 significantly reduced neutrophilia and improved survival. Surprisingly, Orai1 inhibition by ELD607 caused a significant reduction in lung bacteria, including methicillin-resistant S. aureus. ELD607 worked as an immunomodulator that reduced cytokine levels, reduced neutrophilia, and promoted macrophage-mediated resolution of inflammation and clearance of bacteria. Indeed, when alveolar macrophages were depleted with inhaled clodronate, ELD607 was no longer able to resolve inflammation or clear bacteria. Conclusions: These data indicate that specific Orai1 inhibition by ELD607 may be a novel approach to reduce multiorgan inflammation and treat antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Saira Ahmad
- Department of Cell Biology and Physiology
- Eldec Pharmaceuticals, Chapel Hill, North Carolina
| | | | | | | | | | | | | | | | - Mani Rai
- Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina; and
| | - Rahul Y. Mahida
- Birmingham Acute Care Research Group, University of Birmingham, Birmingham, United Kingdom
| | | | | | - Alon Greenbaum
- Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina; and
| | - Timothy P. Moran
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephen L. Tilley
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - David R. Thickett
- Birmingham Acute Care Research Group, University of Birmingham, Birmingham, United Kingdom
| | - M. Flori Sassano
- Department of Cell Biology and Physiology
- Eldec Pharmaceuticals, Chapel Hill, North Carolina
| | - Robert Tarran
- Department of Cell Biology and Physiology
- Eldec Pharmaceuticals, Chapel Hill, North Carolina
| |
Collapse
|
4
|
Ben-Meir E, Perrem L, Shaw M, Ratjen F, Grasemann H. SPLUNC1 as a biomarker of pulmonary exacerbations in children with cystic fibrosis. J Cyst Fibros 2024; 23:288-292. [PMID: 38413298 DOI: 10.1016/j.jcf.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) is an innate defence protein that acts as an anti-microbial agent and regulates airway surface liquid volume through inhibition of the epithelial sodium channel (ENaC). SPLUNC1 levels were found to be reduced in airway secretions of adults with cystic fibrosis (CF). The potential of SPLUNC1 as a biomarker in children with CF is unknown. METHODS We quantified SPLUNC1, interleukin-8 (IL-8) and neutrophil elastase (NE) in sputum of CF children treated with either intravenous antibiotics or oral antibiotics for a pulmonary exacerbation (PEx)s, and in participants of a prospective cohort of CF children with preserved lung function on spirometry, followed over a period of two years. RESULTS Sputum SPLUNC1 levels were significantly lower before compared to after intravenous and oral antibiotic therapy for PEx. In the longitudinal cohort, SPLUNC1 levels were found to be decreased at PEx visits compared to both previous and subsequent stable visits. Higher SPLUNC1 levels at stable visits were associated with longer PEx-free time (hazard ratio 0.85, p = 0.04). SPLUNC1 at PEx visits did not correlate with IL-8 or NE levels in sputum or forced expiratory volume in one second (FEV1) but did correlate with the lung clearance index (LCI) (r=-0.53, p < 0.001). CONCLUSION SPLUNC1 demonstrates promising clinometric properties as a biomarker of PEx in children with CF.
Collapse
Affiliation(s)
- E Ben-Meir
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - L Perrem
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - M Shaw
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - F Ratjen
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada
| | - H Grasemann
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; Translational Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada; University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Houston CJ, Alkhatib A, Einarsson GG, Tunney MM, Taggart CC, Downey DG. Diminished airway host innate response in people with cystic fibrosis who experience frequent pulmonary exacerbations. Eur Respir J 2024; 63:2301228. [PMID: 38135443 PMCID: PMC10882324 DOI: 10.1183/13993003.01228-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
RATIONALE Pulmonary exacerbations are clinically impactful events that accelerate cystic fibrosis (CF) lung disease progression. The pathophysiological mechanisms underlying an increased frequency of pulmonary exacerbations have not been explored. OBJECTIVES To compare host immune response during intravenous antibiotic treatment of pulmonary exacerbations in people with CF who have a history of frequent versus infrequent exacerbations. METHODS Adults with CF were recruited at onset of antibiotic treatment of a pulmonary exacerbation and were categorised as infrequent or frequent exacerbators based on their pulmonary exacerbation frequency in the previous 12 months. Clinical parameters, sputum bacterial load and sputum inflammatory markers were measured on day 0, day 5 and at the end of treatment. Shotgun proteomic analysis was performed on sputum using liquid chromatography-mass spectrometry. MEASUREMENTS AND MAIN RESULTS Many sputum proteins were differentially enriched between infrequent and frequent exacerbators (day 0 n=23 and day 5 n=31). The majority of these proteins had a higher abundance in infrequent exacerbators and were secreted innate host defence proteins with antimicrobial, antiprotease and immunomodulatory functions. Several differentially enriched proteins were validated using ELISA and Western blot including secretory leukocyte protease inhibitor (SLPI), lipocalin-1 and cystatin SA. Sputum from frequent exacerbators demonstrated potent ability to cleave exogenous recombinant SLPI in a neutrophil elastase dependent manner. Frequent exacerbators had increased sputum inflammatory markers (interleukin (IL)-1β and IL-8) and total bacterial load compared to infrequent exacerbators. CONCLUSIONS A diminished innate host protein defence may play a role in the pathophysiological mechanisms of frequent CF pulmonary exacerbations. Frequent exacerbators may benefit from therapies targeting this dysregulated host immune response.
Collapse
Affiliation(s)
- Claire J Houston
- Airway Innate Immunity Research Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Aya Alkhatib
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | | | | | - Clifford C Taggart
- Airway Innate Immunity Research Group, Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
- Joint senior authors
| | - Damian G Downey
- Belfast Health and Social Care Trust, Belfast, UK
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
- Joint senior authors
| |
Collapse
|
6
|
Aufy M, Hussein AM, Stojanovic T, Studenik CR, Kotob MH. Proteolytic Activation of the Epithelial Sodium Channel (ENaC): Its Mechanisms and Implications. Int J Mol Sci 2023; 24:17563. [PMID: 38139392 PMCID: PMC10743461 DOI: 10.3390/ijms242417563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Epithelial sodium channel (ENaC) are integral to maintaining salt and water homeostasis in various biological tissues, including the kidney, lung, and colon. They enable the selective reabsorption of sodium ions, which is a process critical for controlling blood pressure, electrolyte balance, and overall fluid volume. ENaC activity is finely controlled through proteolytic activation, a process wherein specific enzymes, or proteases, cleave ENaC subunits, resulting in channel activation and increased sodium reabsorption. This regulatory mechanism plays a pivotal role in adapting sodium transport to different physiological conditions. In this review article, we provide an in-depth exploration of the role of proteolytic activation in regulating ENaC activity. We elucidate the involvement of various proteases, including furin-like convertases, cysteine, and serine proteases, and detail the precise cleavage sites and regulatory mechanisms underlying ENaC activation by these proteases. We also discuss the physiological implications of proteolytic ENaC activation, focusing on its involvement in blood pressure regulation, pulmonary function, and intestinal sodium absorption. Understanding the mechanisms and consequences of ENaC proteolytic activation provides valuable insights into the pathophysiology of various diseases, including hypertension, pulmonary disorders, and various gastrointestinal conditions. Moreover, we discuss the potential therapeutic avenues that emerge from understanding these mechanisms, offering new possibilities for managing diseases associated with ENaC dysfunction. In summary, this review provides a comprehensive discussion of the intricate interplay between proteases and ENaC, emphasizing the significance of proteolytic activation in maintaining sodium and fluid balance in both health and disease.
Collapse
Affiliation(s)
- Mohammed Aufy
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Ahmed M. Hussein
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Zoology, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Tamara Stojanovic
- Programme for Proteomics, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Christian R. Studenik
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
| | - Mohamed H. Kotob
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria; (A.M.H.); (M.H.K.)
- Department of Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
7
|
Iannuzo N, Welfley H, Li NC, Johnson MDL, Rojas-Quintero J, Polverino F, Guerra S, Li X, Cusanovich DA, Langlais PR, Ledford JG. CC16 drives VLA-2-dependent SPLUNC1 expression. Front Immunol 2023; 14:1277582. [PMID: 38053993 PMCID: PMC10694244 DOI: 10.3389/fimmu.2023.1277582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
Rationale CC16 (Club Cell Secretory Protein) is a protein produced by club cells and other non-ciliated epithelial cells within the lungs. CC16 has been shown to protect against the development of obstructive lung diseases and attenuate pulmonary pathogen burden. Despite recent advances in understanding CC16 effects in circulation, the biological mechanisms of CC16 in pulmonary epithelial responses have not been elucidated. Objectives We sought to determine if CC16 deficiency impairs epithelial-driven host responses and identify novel receptors expressed within the pulmonary epithelium through which CC16 imparts activity. Methods We utilized mass spectrometry and quantitative proteomics to investigate how CC16 deficiency impacts apically secreted pulmonary epithelial proteins. Mouse tracheal epithelial cells (MTECS), human nasal epithelial cells (HNECs) and mice were studied in naïve conditions and after Mp challenge. Measurements and main results We identified 8 antimicrobial proteins significantly decreased by CC16-/- MTECS, 6 of which were validated by mRNA expression in Severe Asthma Research Program (SARP) cohorts. Short Palate Lung and Nasal Epithelial Clone 1 (SPLUNC1) was the most differentially expressed protein (66-fold) and was the focus of this study. Using a combination of MTECs and HNECs, we found that CC16 enhances pulmonary epithelial-driven SPLUNC1 expression via signaling through the receptor complex Very Late Antigen-2 (VLA-2) and that rCC16 given to mice enhances pulmonary SPLUNC1 production and decreases Mycoplasma pneumoniae (Mp) burden. Likewise, rSPLUNC1 results in decreased Mp burden in mice lacking CC16 mice. The VLA-2 integrin binding site within rCC16 is necessary for induction of SPLUNC1 and the reduction in Mp burden. Conclusion Our findings demonstrate a novel role for CC16 in epithelial-driven host defense by up-regulating antimicrobials and define a novel epithelial receptor for CC16, VLA-2, through which signaling is necessary for enhanced SPLUNC1 production.
Collapse
Affiliation(s)
- Natalie Iannuzo
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Holly Welfley
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | | | | | | | | | - Stefano Guerra
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona, Tucson, AZ, United States
| | - Xingnan Li
- Department of Medicine, Division of Genetics, Genomics, and Precision Medicine, University of Arizona, Tucson, AZ, United States
| | - Darren A. Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| | - Paul R. Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, United States
| | - Julie G. Ledford
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, Tucson, AZ, United States
| |
Collapse
|
8
|
Wang L, Cao JB, Xia BB, Li YJ, Zhang X, Mo GX, Wang RJ, Guo SQ, Zhang YQ, Xiao K, Zhu GF, Liu PF, Song LC, Ma XH, Xiang PC, Wang J, Liu YH, Xie F, Zhang XD, Li XX, Sun WL, Cao Y, Wang KF, Zhang WH, Zhao WC, Yan P, Chen JC, Yang YW, Yu ZK, Tang JS, Xiao L, Zhou JM, Xie LX, Wang J. Metatranscriptome of human lung microbial communities in a cohort of mechanically ventilated COVID-19 Omicron patients. Signal Transduct Target Ther 2023; 8:432. [PMID: 37949875 PMCID: PMC10638395 DOI: 10.1038/s41392-023-01684-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/21/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
The Omicron variant of the severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) infected a substantial proportion of Chinese population, and understanding the factors underlying the severity of the disease and fatality is valuable for future prevention and clinical treatment. We recruited 64 patients with invasive ventilation for COVID-19 and performed metatranscriptomic sequencing to profile host transcriptomic profiles, plus viral, bacterial, and fungal content, as well as virulence factors and examined their relationships to 28-day mortality were examined. In addition, the bronchoalveolar lavage fluid (BALF) samples from invasive ventilated hospital/community-acquired pneumonia patients (HAP/CAP) sampled in 2019 were included for comparison. Genomic analysis revealed that all Omicron strains belong to BA.5 and BF.7 sub-lineages, with no difference in 28-day mortality between them. Compared to HAP/CAP cohort, invasive ventilated COVID-19 patients have distinct host transcriptomic and microbial signatures in the lower respiratory tract; and in the COVID-19 non-survivors, we found significantly lower gene expressions in pathways related viral processes and positive regulation of protein localization to plasma membrane, higher abundance of opportunistic pathogens including bacterial Alloprevotella, Caulobacter, Escherichia-Shigella, Ralstonia and fungal Aspergillus sydowii and Penicillium rubens. Correlational analysis further revealed significant associations between host immune responses and microbial compositions, besides synergy within viral, bacterial, and fungal pathogens. Our study presents the relationships of lower respiratory tract microbiome and transcriptome in invasive ventilated COVID-19 patients, providing the basis for future clinical treatment and reduction of fatality.
Collapse
Affiliation(s)
- Lin Wang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Jia-Bao Cao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bin-Bin Xia
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yue-Juan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuan Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Faculty of Biological Science and Technology, Baotou Teacher's College, Baotou, 014030, China
| | - Guo-Xin Mo
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Rui-Juan Wang
- Department of Respiratory Medicine, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Si-Qi Guo
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yu-Qing Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kun Xiao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Guang-Fa Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Peng-Fei Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Li-Cheng Song
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Xi-Hui Ma
- Respiratory Research Institute, Department of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Ping-Chao Xiang
- Shougang hospital of Peking University, Beijing, 100144, China
| | - Jiang Wang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Yu-Hong Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Fei Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Xu-Dong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiang-Xin Li
- Department of Respiratory Medicine, Beijing Changping Hospital, Beijing, 102200, China
| | - Wan-Lu Sun
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yan Cao
- Pulmonary research institute, Senior Department of Respiratory and Critical Care Medicine, the 8th medical center of Chinese PLA general hospital, Beijing, 100091, China
| | - Kai-Fei Wang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wen-Hui Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei-Chao Zhao
- Department of Respiratory Medicine, PLA Strategic Support Force Medical Center, Beijing, 100101, China
| | - Peng Yan
- China Aerospace Science & Industry Corporation 731 hospital, Beijing, 100074, China
| | - Ji-Chao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Beijing, 100049, China
| | - Yu-Wei Yang
- Respiratory Research Institute, Department of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Zhong-Kuo Yu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Jing-Si Tang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Xiao
- Respiratory Research Institute, Department of Pulmonary & Critical Care Medicine, Beijing Key Laboratory of OTIR, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Jie-Min Zhou
- Vision Medicals Center for Infectious Diseases, Guangzhou, 510700, China
| | - Li-Xin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China.
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
9
|
Ghosh A, Coakley RD, Alexis NE, Tarran R. Vaping-Induced Proteolysis Causes Airway Surface Dehydration. Int J Mol Sci 2023; 24:15348. [PMID: 37895029 PMCID: PMC10607227 DOI: 10.3390/ijms242015348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Proteases such as neutrophil elastase cleave and activate the epithelial sodium channel (ENaC), causing airway dehydration. Our current study explores the impact of increased protease levels in vapers' airways on ENaC activity and airway dehydration. Human bronchial epithelial cultures (HBECs) were exposed to bronchoalveolar lavage fluid (BALF) from non-smokers, smokers and vapers. Airway surface liquid (ASL) height was measured by confocal microscopy as a marker of hydration. ENaC cleavage was measured by Western blotting. Human peripheral blood neutrophils were treated with a menthol-flavored e-liquid (Juul), and the resulting secretions were added to HBECs. BALF from smokers and vapers significantly and equally increased ENaC activity and decreased ASL height. The ASL height decrease was attenuated by protease inhibitors. Non-smokers' BALF had no effect on ENaC or ASL height. BALF from smokers and vapers, but not non-smokers, induced ENaC cleavage. E-liquid-treated neutrophil secretions cleaved ENaC and decreased ASL height. Our study demonstrated that elevated protease levels in vapers' airways have functional significance since they can activate ENaC, resulting in airway dehydration. Lung dehydration contributes to diseases like cystic fibrosis (CF), chronic obstructive pulmonary disease (COPD) and asthma. Thus, our data predict that vaping, like smoking, will cause airway surface dehydration that likely leads to lung disease.
Collapse
Affiliation(s)
- Arunava Ghosh
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Raymond D. Coakley
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Neil E. Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Robert Tarran
- Division of Genetic, Environmental and Inhalational Disease, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
10
|
Abstract
Cystic fibrosis (CF) pathophysiology is hallmarked by excessive inflammation and the inability to resolve lung infections, contributing to morbidity and eventually mortality. Paradoxically, despite a robust inflammatory response, CF lungs fail to clear bacteria and are susceptible to chronic infections. Impaired mucociliary transport plays a critical role in chronic infection but the immune mechanisms contributing to the adaptation of bacteria to the lung microenvironment is not clear. CFTR modulator therapy has advanced CF life expectancy opening up the need to understand changes in immunity as CF patients age. Here, we have summarized the current understanding of immune dysregulation in CF.
Collapse
Affiliation(s)
- Emanuela M Bruscia
- Department of Pediatrics, Section of Pulmonology, Allergy, Immunology and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Tracey L Bonfield
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
11
|
Wu T, Goriounova AS, Worthington EN, Wrennall JA, Ghosh A, Ahmad S, Flori Sassano M, Tarran R. SPLUNC1 is a negative regulator of the Orai1 Ca 2+ channel. Physiol Rep 2022; 10:e15306. [PMID: 35581745 PMCID: PMC9114653 DOI: 10.14814/phy2.15306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 06/15/2023] Open
Abstract
Orai1 is a ubiquitously-expressed plasma membrane Ca2+ channel that is involved in store-operated Ca2+ entry (SOCE): a fundamental biological process that regulates gene expression, the onset of inflammation, secretion, and the contraction of airway smooth muscle (ASM). During SOCE, Ca2+ leaves the endoplasmic reticulum, which then stimulates a second, amplifying wave of Ca2+ influx through Orai1 into the cytoplasm. Short Palate LUng and Nasal epithelial Clone 1 (SPLUNC1; gene name BPIFA1) is a multi-functional, innate defense protein that is highly abundant in the lung. We have previously reported that SPLUNC1 was secreted from epithelia, where it bound to and inhibited Orai1, leading to reduced SOCE and ASM relaxation. However, the underlying mechanism of action is unknown. Here, we probed the SPLUNC1-Orai1 interactions in ASM and HEK293T cells using biochemical and imaging techniques. We observed that SPLUNC1 caused a conformational change in Orai1, as measured using Forster resonance energy transfer (FRET). SPLUNC1 binding also led to Nedd4-2 dependent ubiquitination of Orai1. Moreover, SPLUNC1 internalized Orai1 to lysosomes, leading to Orai1 degradation. Thus, we conclude that SPLUNC1 is an allosteric regulator of Orai1. Our data indicate that SPLUNC1-mediated Orai1 inhibition could be utilized as a therapeutic strategy to reduce SOCE.
Collapse
Affiliation(s)
- Tongde Wu
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Alexandra S. Goriounova
- Department of PharmacologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Erin N. Worthington
- Divison of PulmonologyDepartment of PediatricsThe University of North Carolina at Chapel HillNorth Carolina27599USA
- Division of Pulmonology, Department of PediatricsCarilion Clinic and Virginia Tech Carilion School of MedicineRoanokeVirginia24016USA
| | - Joe A. Wrennall
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Arunava Ghosh
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Saira Ahmad
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - M. Flori Sassano
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| | - Robert Tarran
- Department of Cell Biology & PhysiologyThe University of North Carolina at Chapel HillNorth Carolina27599USA
| |
Collapse
|
12
|
Woodall M, Reidel B, Kesimer M, Tarran R, Baines DL. Culture with apically applied healthy or disease sputum alters the airway surface liquid proteome and ion transport across human bronchial epithelial cells. Am J Physiol Cell Physiol 2021; 321:C954-C963. [PMID: 34613844 PMCID: PMC8714986 DOI: 10.1152/ajpcell.00234.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Airway secretions contain many signaling molecules and peptides/proteins that are
not found in airway surface liquid (ASL) generated by normal human bronchial
epithelial cells (NHBEs) in vitro. These play a key role in innate defense and
mediate communication between the epithelium, the immune cells, and the external
environment. We investigated how culture of NHBE with apically applied
secretions from healthy or diseased (cystic fibrosis, CF) lungs affected
epithelial function with a view to providing better in vitro models of the in
vivo environment. NHBEs from 6 to 8 different donors were cultured at air-liquid
interface (ALI), with apically applied sputum from normal healthy donors (normal
lung sputum; NLS) or CF donors (CFS) for 2–4 h, 48 h, or with sputum
reapplied over 48 h. Proteomics analysis was carried out on the sputa and on the
NHBE ASL before and after culture with sputa. Transepithelial electrical
resistance (TEER), short circuit current (Isc), and changes to ASL
height were measured. There were 71 proteins common to both sputa but not ASL.
The protease:protease inhibitor balance was increased in CFS compared with NLS
and ASL. Culture of NHBE with sputa for 48 h identified additional factors not
present in NLS, CFS, or ASL alone. Culture with either NLS or CFS for 48 h
increased cystic fibrosis transmembrane regulator (CFTR) activity,
calcium-activated chloride channel (CaCC) activity, and changed ASL height.
These data indicate that culture with healthy or disease sputum changes the
proteomic profile of ASL and ion transport properties of NHBE and this may
increase physiological relevance when using in vitro airway models.
Collapse
Affiliation(s)
- Maximillian Woodall
- Institute for Infection and Immunity, St George's, University of London, Cranmer Terrace, Tooting, London, United Kingdom
| | - Boris Reidel
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Deborah L Baines
- Institute for Infection and Immunity, St George's, University of London, Cranmer Terrace, Tooting, London, United Kingdom
| |
Collapse
|
13
|
Althaus M, Lawong RY. Proteolytic ENaC activation in health and disease-a complicated puzzle. Pflugers Arch 2021; 474:177-179. [PMID: 34799769 PMCID: PMC8766370 DOI: 10.1007/s00424-021-02644-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Mike Althaus
- Institute for Functional Gene Analytics, Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Von-Liebig-Strasse 20, 53359, Rheinbach, Germany.
| | - Rene Yufenyuy Lawong
- Institute for Functional Gene Analytics, Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Von-Liebig-Strasse 20, 53359, Rheinbach, Germany
| |
Collapse
|
14
|
Bingle CD, Bingle L. SPLUNC1 comes of age? Predicting acute exacerbations in cystic fibrosis. Eur Respir J 2021; 58:58/5/2101569. [PMID: 34764214 DOI: 10.1183/13993003.01569-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 06/08/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Colin D Bingle
- Dept of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Lynne Bingle
- Academic Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| |
Collapse
|
15
|
Keir HR, Shoemark A, Huang JTJ, Chalmers JD. SPLUNC1 is a novel marker of disease severity and airway infection in bronchiectasis. Eur Respir J 2021; 58:13993003.01840-2021. [PMID: 34413156 DOI: 10.1183/13993003.01840-2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/05/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Holly R Keir
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK.,Royal Brompton Hospital, Primary Ciliary Dyskinesia Centre, Paediatric Respiratory Medicine, London, UK
| | | | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
16
|
Abstract
The Epithelial Na+ Channel, ENaC, comprised of 3 subunits (αβγ, or sometimes δβγENaC), plays a critical role in regulating salt and fluid homeostasis in the body. It regulates fluid reabsorption into the blood stream from the kidney to control blood volume and pressure, fluid absorption in the lung to control alveolar fluid clearance at birth and maintenance of normal airway surface liquid throughout life, and fluid absorption in the distal colon and other epithelial tissues. Moreover, recent studies have also revealed a role for sodium movement via ENaC in nonepithelial cells/tissues, such as endothelial cells in blood vessels and neurons. Over the past 25 years, major advances have been made in our understanding of ENaC structure, function, regulation, and role in human disease. These include the recently solved three-dimensional structure of ENaC, ENaC function in various tissues, and mutations in ENaC that cause a hereditary form of hypertension (Liddle syndrome), salt-wasting hypotension (PHA1), or polymorphism in ENaC that contributes to other diseases (such as cystic fibrosis). Moreover, great strides have been made in deciphering the regulation of ENaC by hormones (e.g., the mineralocorticoid aldosterone, glucocorticoids, vasopressin), ions (e.g., Na+ ), proteins (e.g., the ubiquitin-protein ligase NEDD4-2, the kinases SGK1, AKT, AMPK, WNKs & mTORC2, and proteases), and posttranslational modifications [e.g., (de)ubiquitylation, glycosylation, phosphorylation, acetylation, palmitoylation]. Characterization of ENaC structure, function, regulation, and role in human disease, including using animal models, are described in this article, with a special emphasis on recent advances in the field. © 2021 American Physiological Society. Compr Physiol 11:1-29, 2021.
Collapse
Affiliation(s)
- Daniela Rotin
- The Hospital for Sick Children, and The University of Toronto, Toronto, Canada
| | - Olivier Staub
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Khanal S, Webster M, Niu N, Zielonka J, Nunez M, Chupp G, Slade MD, Cohn L, Sauler M, Gomez JL, Tarran R, Sharma L, Dela Cruz CS, Egan M, Laguna T, Britto CJ. SPLUNC1: a novel marker of cystic fibrosis exacerbations. Eur Respir J 2021; 58:13993003.00507-2020. [PMID: 33958427 DOI: 10.1183/13993003.00507-2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/29/2021] [Indexed: 11/05/2022]
Abstract
Acute pulmonary Exacerbations (AE) are episodes of clinical worsening in cystic fibrosis (CF), often precipitated by infection. Timely detection is critical to minimise morbidity and lung function declines associated with acute inflammation during AE. Based on our previous observations that airway protein Short Palate Lung Nasal epithelium Clone 1 (SPLUNC1) is regulated by inflammatory signals, we investigated the use of SPLUNC1 fluctuations to diagnose and predict AE in CF.We enrolled CF participants from two independent cohorts to measure AE markers of inflammation in sputum and recorded clinical outcomes for a 1-year follow-up period.SPLUNC1 levels were high in healthy controls (n=9, 10.7 μg mL-1), and significantly decreased in CF participants without AE (n=30, 5.7 μg mL-1, p=0.016). SPLUNC1 levels were 71.9% lower during AE (n=14, 1.6 μg mL-1, p=0.0034) regardless of age, sex, CF-causing mutation, or microbiology findings. Cytokines Il-1β and TNFα were also increased in AE, whereas lung function did not consistently decrease. Stable CF participants with lower SPLUNC1 levels were much more likely to have an AE at 60 days (HR: 11.49, Standard Error: 0.83, p=0.0033). Low-SPLUNC1 stable participants remained at higher AE risk even one year after sputum collection (HR: 3.21, Standard Error: 0.47, p=0.0125). SPLUNC1 was downregulated by inflammatory cytokines and proteases increased in sputum during AE.In acute CF care, low SPLUNC1 levels could support a decision to increase airway clearance or to initiate pharmacological interventions. In asymptomatic, stable patients, low SPLUNC1 levels could inform changes in clinical management to improve long-term disease control and clinical outcomes in CF.
Collapse
Affiliation(s)
- Sara Khanal
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Megan Webster
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Naiqian Niu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jana Zielonka
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Myra Nunez
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Martin D Slade
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lauren Cohn
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marie Egan
- Division of Pediatric Pulmonology, Allergy, Immunology, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Theresa Laguna
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Liu Q, Wang Z, Zhang W. The Multifunctional Roles of Short Palate, Lung, and Nasal Epithelium Clone 1 in Regulating Airway Surface Liquid and Participating in Airway Host Defense. J Interferon Cytokine Res 2021; 41:139-148. [PMID: 33885339 DOI: 10.1089/jir.2020.0141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Short palate, lung, and nasal epithelium clone 1 (SPLUNC1) is a kind of secretory protein, and gets expressed abundantly in normal respiratory epithelium of humans. As a natural immune molecule, SPLUNC1 is proved to be involved in inflammatory response and airway host defense. This review focuses on summarizing and discussing the role of SPLUNC1 in regulating airway surface liquid (ASL) and participating in airway host defense. PubMed and MEDLINE were used for searching and identifying the data in this review. The domain of bactericidal/permeability-increasing protein in SPLUNC1 and the α-helix, α4, are essential for SPLUNC1 to exert biological activities. As a natural innate immune molecule, SPLUNC1 plays a significant role in inflammatory response and airway host defense. Its special expression patterns are not only observed in physiological conditions, but also in some respiratory diseases. The mechanisms of SPLUNC1 in airway host defense include modulating ASL volume, acting as a surfactant protein, inhibiting biofilm formation, as well as regulating ASL compositions, such as LL-37, mucins, Neutrophil elastase, and inflammatory cytokines. Besides, potential correlations are found among these different mechanisms, especially among different ASL compositions, which should be further explored in more systematical frameworks. In this review, we summarize the structural characteristics and expression patterns of SPLUNC1 briefly, and mainly discuss the mechanisms of SPLUNC1 exerted in host defense, aiming to provide a theoretical basis and a novel target for future studies and clinical treatments.
Collapse
Affiliation(s)
- Qingluan Liu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhicheng Wang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
19
|
Sala V, Cnudde SJ, Murabito A, Massarotti A, Hirsch E, Ghigo A. Therapeutic peptides for the treatment of cystic fibrosis: Challenges and perspectives. Eur J Med Chem 2021; 213:113191. [PMID: 33493828 DOI: 10.1016/j.ejmech.2021.113191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/21/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023]
Abstract
Cystic fibrosis (CF) is the most common amongst rare genetic diseases, affecting more than 70.000 people worldwide. CF is characterized by a dysfunctional chloride channel, termed cystic fibrosis conductance regulator (CFTR), which leads to the production of a thick and viscous mucus layer that clogs the lungs of CF patients and traps pathogens, leading to chronic infections and inflammation and, ultimately, lung damage. In recent years, the use of peptides for the treatment of respiratory diseases, including CF, has gained growing interest. Therapeutic peptides for CF include antimicrobial peptides, inhibitors of proteases, and modulators of ion channels, among others. Peptides display unique features that make them appealing candidates for clinical translation, like specificity of action, high efficacy, and low toxicity. Nevertheless, the intrinsic properties of peptides, together with the need of delivering these compounds locally, e.g. by inhalation, raise a number of concerns in the development of peptide therapeutics for CF lung disease. In this review, we discuss the challenges related to the use of peptides for the treatment of CF lung disease through inhalation, which include retention within mucus, proteolysis, immunogenicity and aggregation. Strategies for overcoming major shortcomings of peptide therapeutics will be presented, together with recent developments in peptide design and optimization, including computational analysis and high-throughput screening.
Collapse
Affiliation(s)
- Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Sophie Julie Cnudde
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Murabito
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alberto Massarotti
- Department of Pharmaceutical Science, University of Piemonte Orientale "A. Avogadro", Largo Donegani 2, 28100, Novara, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy; Kither Biotech S.r.l., Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, 10126, Torino, Italy; Kither Biotech S.r.l., Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
20
|
Salman S, Shah FH, Chaudhry M, Tariq M, Akbar MY, Adnan M. In silico analysis of protein/peptide-based inhalers against SARS-CoV-2. Future Virol 2020. [PMCID: PMC7543042 DOI: 10.2217/fvl-2020-0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: Peptide/protein-based inhalers are excessively used to treat respiratory disorders. The molecular docking was performed for these inhalers including human neutralizing S230 light chain-antibody (monoclonal antibodies [mAbs]), alpha-1-antitrypsin (AAT), short-palate-lung and nasal-epithelial clone-1-derived peptides (SPLUNC1) and dornase-alfa (DA) against spike glycoprotein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to assess their inhibitory activity. Materials & methods: HawkDock was used to dock these biologics against SARS-CoV-2 spike-glycoprotein. Results: Results showed that DA, AAT and mAb were quite active against spike glycoprotein with a binding free energy of -26.35 and -22.94 kcal/mol. Conclusion: mAB and AAT combined with DA can be used in the treatment of coronavirus disease of 2019 as a potential anti-SARS-CoV-2 agent.
Collapse
Affiliation(s)
- Saad Salman
- Department of Pharmacy, The University of Lahore, Islamabad Campus, Islamabad, Federal 44000, Pakistan
| | - Fahad Hassan Shah
- Centre of Biotechnology & Microbiology, University of Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Maham Chaudhry
- Department of Pharmacy, The University of Lahore, Islamabad Campus, Islamabad, Federal 44000, Pakistan
| | - Muniba Tariq
- Diet & Nutritional Sciences, The University of Lahore, Islamabad Campus, Islamabad, Federal 44000, Pakistan
| | - Muhammad Yasir Akbar
- Department of Bioinformatics, Quaid-e-Azam University, Islamabad, Federal 44000, Pakistan
| | - Muhammad Adnan
- Department of Pharmacy, The University of Lahore, Islamabad Campus, Islamabad, Federal 44000, Pakistan
| |
Collapse
|
21
|
Olivença DV, Voit EO, Pinto FR. ENaC regulation by phospholipids and DGK explained through mathematical modeling. Sci Rep 2020; 10:13952. [PMID: 32811866 PMCID: PMC7435262 DOI: 10.1038/s41598-020-70630-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/21/2020] [Indexed: 01/16/2023] Open
Abstract
Cystic fibrosis is a condition caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). It is also thought to increase the activity of epithelial sodium channels (ENaC). The altered function of these ion channels is one of the causes of the thick dehydrated mucus that characterizes the disease and is partially responsible for recurrent pulmonary infections and inflammation events that ultimately destroy the lungs of affected subjects. Phosphoinositides are signaling lipids that regulate numerous cellular processes and membrane proteins, including ENaC. Inhibition of diacylglycerol kinase (DGK), an enzyme of the phosphoinositide pathway, reduces ENaC function. We propose a computational analysis that is based on the combination of two existing mathematical models: one representing the dynamics of phosphoinositides and the other explaining how phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) influences ENaC activity and, consequently, airway surface liquid. This integrated model permits, for the first time, a detailed assessment of the intricate interactions between DGK and ENaC and is consistent with available literature data. In particular, the computational approach allows comparisons of two competing hypotheses regarding the regulation of ENaC. The results strongly suggest that the regulation of ENaC is primarily exerted through the control of PI(4,5)P2 production by type-I phosphatidylinositol-4-phosphate 5-kinase (PIP5KI), which in turn is controlled by phosphatidic acid (PA), the product of the DGK reaction.
Collapse
Affiliation(s)
- Daniel V. Olivença
- Faculty of Sciences, BioISI – Biosystems and Integrative Sciences Institute, University of Lisboa, 1749-016 Lisbon, Portugal
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Atlanta, GA 30332-2000 USA
| | - Eberhard O. Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 950 Atlantic Drive, Atlanta, GA 30332-2000 USA
| | - Francisco R. Pinto
- Faculty of Sciences, BioISI – Biosystems and Integrative Sciences Institute, University of Lisboa, 1749-016 Lisbon, Portugal
| |
Collapse
|
22
|
Bartel S, Deshane J, Wilkinson T, Gabrielsson S. Extracellular Vesicles as Mediators of Cellular Cross Talk in the Lung Microenvironment. Front Med (Lausanne) 2020; 7:326. [PMID: 32850874 PMCID: PMC7417309 DOI: 10.3389/fmed.2020.00326] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/03/2020] [Indexed: 12/27/2022] Open
Abstract
The human lung is a complex tissue subdivided into several regions that differ in size, function, and resident cell types. Despite years of intensive research, we still do not fully understand the cross talk between these different regions and diverse cell populations in the lung and how this is altered in the development of chronic respiratory disease. The discovery of extracellular vesicles (EVs), small membrane vesicles released from cells for intercellular communication, has added another layer of complexity to cellular cross talk in the complex lung microenvironment. EVs from patients with chronic obstructive pulmonary disease, asthma, or sarcoidosis have been shown to carry microRNAs, proteins, and lipids that may contribute to inflammation or tissue degeneration. Here, we summarize the contribution of these small vesicles in the interplay of several different cell types in the lung microenvironment, with a focus on the development of chronic respiratory diseases. Although there are already many studies demonstrating the adverse effects of EVs in the diseased lung, we still have substantial knowledge gaps regarding the concrete role of EV involvement in lung disease, which should be addressed in future studies.
Collapse
Affiliation(s)
- Sabine Bartel
- Department of Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jessy Deshane
- Pulmonary Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tom Wilkinson
- Clinical and Experimental Science, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Susanne Gabrielsson
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Ahmad S, Kim CSK, Tarran R. The SPLUNC1-βENaC complex prevents Burkholderia cenocepacia invasion in normal airway epithelia. Respir Res 2020; 21:190. [PMID: 32680508 PMCID: PMC7368771 DOI: 10.1186/s12931-020-01454-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic fibrosis (CF) patients are extremely vulnerable to Burkholderia cepacia complex (Bcc) infections. However, the underlying etiology is poorly understood. We tested the hypothesis that short palate lung and nasal epithelial clone 1 (SPLUNC1)-epithelial sodium channel (ENaC) interactions at the plasma membrane are required to reduce Bcc burden in normal airways. To determine if SPLUNC1 was needed to reduce Bcc burden in the airways, SPLUNC1 knockout mice and their wild-type littermates were infected with B. cenocepacia strain J2315. SPLUNC1 knockout mice had increased bacterial burden in the lungs compared to wild-type littermate mice. SPLUNC1-knockdown primary human bronchial epithelia (HBECs) were incubated with J2315, which resulted in increased bacterial burden compared to non-transduced HBECs. We next determined the interaction of the SPLUNC1-ENaC complex during J2315 infection. SPLUNC1 remained at the apical plasma membrane of normal HBECs but less was present at the apical plasma membrane of CF HBECs. Additionally, SPLUNC1-βENaC complexes reduced intracellular J2315 burden. Our data indicate that (i) secreted SPLUNC1 is required to reduce J2315 burden in the airways and (ii) its interaction with ENaC prevents cellular invasion of J2315.
Collapse
Affiliation(s)
- Saira Ahmad
- Department of Cell Biology and Physiology, The University of North Carolina, Marsico Lung Insitute, 115 Mason Farm Rd CB 7545, UNC, Chapel Hill, NC, 27599, USA
| | - Christine Seul Ki Kim
- Department of Cell Biology and Physiology, The University of North Carolina, Marsico Lung Insitute, 115 Mason Farm Rd CB 7545, UNC, Chapel Hill, NC, 27599, USA
- Present address: Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Robert Tarran
- Department of Cell Biology and Physiology, The University of North Carolina, Marsico Lung Insitute, 115 Mason Farm Rd CB 7545, UNC, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
24
|
Herman M, Tarran R. E-cigarettes, nicotine, the lung and the brain: multi-level cascading pathophysiology. J Physiol 2020; 598:5063-5071. [PMID: 32515030 DOI: 10.1113/jp278388] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Tobacco smoking is highly addictive and causes respiratory disease, cardiovascular disease and multiple types of cancer. Electronic-cigarettes (e-cigarettes) are non-combustible tobacco alternatives that aerosolize nicotine and flavouring agents in a propylene glycol-vegetable glycerine vehicle. They were originally envisaged as a tobacco cessation aid, but whether or not they help people to quit tobacco use is controversial. In this review, we have compared and contrasted what is known regarding the effects of nicotine on the lungs vs. the effects of nicotine in the brain in the context of addiction. Critically, both combustible tobacco products and e-cigarettes contain nicotine, a highly addictive, plant-derived alkaloid that binds to nicotinic acetylcholine receptors (nAChRs). Nicotine's reinforcing properties are primarily mediated by activation of the brain's mesolimbic reward circuitry and release of the neurotransmitter dopamine that contribute to the development of addiction. Moreover, nicotine addiction drives repeated intake that results in chronic pulmonary exposure to either tobacco smoke or e-cigarettes despite negative respiratory symptoms. Beyond the brain, nAChRs are also highly expressed in peripheral neurons, epithelia and immune cells, where their activation may cause harmful effects. Thus, nicotine, a key ingredient of both conventional and electronic cigarettes, produces neurological effects that drive addiction and may damage the lungs in the process, producing a complex, multilevel pathological state. We conclude that vaping needs to be studied by multi-disciplinary teams that include pulmonary and neurophysiologists as well as behaviourists and addiction specialists to fully understand their impact on human physiology.
Collapse
Affiliation(s)
- Melissa Herman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
25
|
Morrison CB, Markovetz MR, Ehre C. Mucus, mucins, and cystic fibrosis. Pediatr Pulmonol 2019; 54 Suppl 3:S84-S96. [PMID: 31715083 PMCID: PMC6853602 DOI: 10.1002/ppul.24530] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023]
Abstract
Cystic fibrosis (CF) is both the most common and most lethal genetic disease in the Caucasian population. CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene and is characterized by the accumulation of thick, adherent mucus plaques in multiple organs, of which the lungs, gastrointestinal tract and pancreatic ducts are the most commonly affected. A similar pathogenesis cascade is observed in all of these organs: loss of CFTR function leads to altered ion transport, consisting of decreased chloride and bicarbonate secretion via the CFTR channel and increased sodium absorption via epithelial sodium channel upregulation. Mucosa exposed to changes in ionic concentrations sustain severe pathophysiological consequences. Altered mucus biophysical properties and weakened innate defense mechanisms ensue, furthering the progression of the disease. Mucins, the high-molecular-weight glycoproteins responsible for the viscoelastic properties of the mucus, play a key role in the disease but the actual mechanism of mucus accumulation is still undetermined. Multiple hypotheses regarding the impact of CFTR malfunction on mucus have been proposed and are reviewed here. (a) Dehydration increases mucin monomer entanglement, (b) defective Ca2+ chelation compromises mucin expansion, (c) ionic changes alter mucin interactions, and (d) reactive oxygen species increase mucin crosslinking. Although one biochemical change may dominate, it is likely that all of these mechanisms play some role in the progression of CF disease. This article discusses recent findings on the initial cause(s) of aberrant mucus properties in CF and examines therapeutic approaches aimed at correcting mucus properties.
Collapse
Affiliation(s)
- Cameron Bradley Morrison
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Matthew Raymond Markovetz
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Camille Ehre
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Division of Pediatric Pulmonology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
26
|
Ahmad S, Gilmore RC, Alexis NE, Tarran R. SPLUNC1 Loses Its Antimicrobial Activity in Acidic Cystic Fibrosis Airway Secretions. Am J Respir Crit Care Med 2019; 200:633-636. [PMID: 31013116 PMCID: PMC6727152 DOI: 10.1164/rccm.201812-2303le] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Saira Ahmad
- The University of North Carolina at Chapel HillChapel Hill, North Carolina
| | - Rodney C. Gilmore
- The University of North Carolina at Chapel HillChapel Hill, North Carolina
| | - Neil E. Alexis
- The University of North Carolina at Chapel HillChapel Hill, North Carolina
| | - Robert Tarran
- The University of North Carolina at Chapel HillChapel Hill, North Carolina
| |
Collapse
|
27
|
Olivença DV, Fonseca LL, Voit EO, Pinto FR. Thickness of the airway surface liquid layer in the lung is affected in cystic fibrosis by compromised synergistic regulation of the ENaC ion channel. J R Soc Interface 2019; 16:20190187. [PMID: 31455163 DOI: 10.1098/rsif.2019.0187] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The lung epithelium is lined with a layer of airway surface liquid (ASL) that is crucial for healthy lung function. ASL thickness is controlled by two ion channels: epithelium sodium channel (ENaC) and cystic fibrosis (CF) transmembrane conductance regulator (CFTR). Here, we present a minimal mathematical model of ENaC, CFTR and ASL regulation that sheds light on the control of ENaC by the short palate lung and nasal epithelial clone 1 (SPLUNC1) protein and by phosphatidylinositol 4,5-biphosphate (PI(4,5)P2). The model, despite its simplicity, yields a good fit to experimental observations and is an effective tool for exploring the interplay between ENaC, CFTR and ASL. Steady-state data and dynamic information constrain the model's parameters without ambiguities. Testing the hypothesis that PI(4,5)P2 protects ENaC from ubiquitination suggests that this protection does not improve the model results and that the control of the ENaC opening probability by PI(4,5)P2 is sufficient to explain all available data. The model analysis further demonstrates that ASL at the steady state is sensitive to small changes in PI(4,5)P2 abundance, particularly in CF conditions, which suggests that manipulation of phosphoinositide metabolism may promote therapeutic benefits for CF patients.
Collapse
Affiliation(s)
- Daniel V Olivença
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisboa, Portugal
| | - Luis L Fonseca
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Eberhard O Voit
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Francisco R Pinto
- Faculty of Sciences, BioISI-Biosystems and Integrative Sciences Institute, University of Lisboa, Lisboa, Portugal
| |
Collapse
|
28
|
Moore PJ, Sesma J, Alexis NE, Tarran R. Tobacco exposure inhibits SPLUNC1-dependent antimicrobial activity. Respir Res 2019; 20:94. [PMID: 31113421 PMCID: PMC6530064 DOI: 10.1186/s12931-019-1066-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 05/06/2019] [Indexed: 11/24/2022] Open
Abstract
Background Tobacco smoke exposure impairs the lung’s innate immune response, leading to an increased risk of chronic infections. SPLUNC1 is a secreted, multifunctional innate defense protein that has antimicrobial activity against Gram negative organisms. We hypothesize that tobacco smoke-induced SPLUNC1 dysfunction contributes to the observed defect in innate immunity in tobacco smokers and that this dysfunction can be used as a potential biomarker of harm. Methods We collected sputum from never-smokers and otherwise healthy smokers. We performed Western blotting to determine SPLUNC1 levels and determined antimicrobial activity against nontypeable Haemophilus influenzae. An in vitro exposure model was utilized to measure the effect of tobacco exposure on human bronchial epithelial culture (HBEC) antimicrobial activity against H. influenzae. The direct effects of cigarette and little cigar smoke exposure on SPLUNC1 function was determined using 24 h growth measurements and LPS binding assays. Results H. influenzae growth in cigarette smoker’s sputum was significantly greater compared to never-smokers sputum over 24 h. HBEC supernatants and lysates contained significantly higher numbers of H. influenzae following chronic cigarette and little cigar smoke exposure compared to air-exposed controls. Furthermore, SPLUNC1’s antimicrobial activity and LPS-binding capability against both H. influenzae and P. aeruginosa was attenuated following cigarette and little cigar exposure. Conclusions These data suggest that cigarette and little cigar exposure impairs SPLUNC1’s antimicrobial ability and that this inhibition may serve as a novel biomarker of harm that can be used to assess the toxicity of commercial tobacco products.
Collapse
Affiliation(s)
- Patrick J Moore
- Marsico Lung Institute, University of North Carolina at Chapel Hill, 7118A Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA.
| | - Juliana Sesma
- CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Robert Tarran
- Marsico Lung Institute, University of North Carolina at Chapel Hill, 7118A Marsico Hall, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA.,Department of Cell Biology & Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
29
|
Musante I, Scudieri P, Venturini A, Guidone D, Caci E, Castellani S, Conese M, Galietta LJV. Peripheral localization of the epithelial sodium channel in the apical membrane of bronchial epithelial cells. Exp Physiol 2019; 104:866-875. [PMID: 30924990 DOI: 10.1113/ep087590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/15/2019] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the precise subcellular localization of the epithelial sodium channel (ENaC) in human airway epithelium? What is the main finding and its importance? ENaC protein has an unexpected localization in the peripheral region of the apical membrane of bronchial epithelial cells, very close to tight junctions. This may be important for the mechanism of Na+ absorption ABSTRACT: The epithelial sodium channel (ENaC) has a key role in absorbing fluid across the human airway epithelium. Altered activity of ENaC may perturb the process of mucociliary clearance, thus impairing the innate defence mechanisms against microbial agents. The proteins forming ENaC are present on the apical membrane of the epithelium. However, their precise localization is unknown. In the present study, we used two antibodies recognizing the α and β ENaC subunits. Both antibodies revealed a restricted localization of ENaC in the peripheral region of the apical membrane of cultured bronchial epithelial cells, close to but not overlapping with tight junctions. In contrast, the cystic fibrosis transmembrane conductance regulator chloride channel was more diffusely expressed on the whole apical membrane. Modulation of ENaC activity by aprotinin or elastase resulted in a decrease or increase in the peripheral localization, respectively. Our results suggest that sodium absorption is mainly occurring close to tight junctions where this cation may be rapidly expelled by the Na+ /K+ pump present in lateral membranes. This arrangement of channels and pumps may limit Na+ build-up in other regions of the cells.
Collapse
Affiliation(s)
- Ilaria Musante
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Paolo Scudieri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Arianna Venturini
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Emanuela Caci
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Stefano Castellani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Luis J V Galietta
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.,Department of Translational Medical Sciences (DISMET), Federico II University of Naples, Naples, Italy
| |
Collapse
|