1
|
Kim H, Kim SJ. 3D Bioprinting of Pig Macrophages and Human Cells Discovered the P2Y14 Receptor as a Mediator of Xenogenic Immune Responses. Immunol Invest 2024:1-16. [PMID: 39356134 DOI: 10.1080/08820139.2024.2411388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
BACKGROUND The survival rate of pig lung xenotransplantation (PLXTx) recipients is severely limited by intense xenogenic immune responses, necessitating further insights into xenogeneic immunity and the development of models to study the PLXTx immune response. METHODS We identified regulators of PLXTx immune response Using Gene ontology analysis. We assessed the metabolic changes and protein levels in 3D4/31 pig alveolar macrophages (PAMs) through flow cytometry and immunoblotting. To induce a xenogenic immune response, we co-cultured 3D4/31-PAMs with A549 human alveolar epithelial cells and evaluated cytokine expression using qRT-PCR. RESULTS Gene ontology analysis identified STAT1 and alveolar macrophages as contributors to lung autoimmunity and transplant rejection. In 3D4/31-PAMs, phorbol myristate acetate-induced glycogen accumulation and cyclooxygenase-2 expression were inhibited by the P2Y14 inhibitor PPTN. Co-culturing 3D4/31-PAMs with A549 human alveolar epithelial cells via 3D bioprinting resulted in a more pronounced inflammatory response than 2D co-culture, with increased expression of genes related to the P2Y14 cascade and inflammation. This inflammatory gene expression was prevented by PPTN treatment. CONCLUSION Based on these results, we propose alginate bioprinting as an in vitro model for PLXTx and suggest that P2Y14 is a key regulator of xenogeneic immune responses in PAMs.
Collapse
Affiliation(s)
- Hyungkuen Kim
- Department of Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, Republic of Korea
| | - Sung-Jo Kim
- Department of Biotechnology, College of Life and Health Sciences, Hoseo University, Asan, Republic of Korea
| |
Collapse
|
2
|
Sun Q, Jiang N, Yao R, Song Y, Li Z, Wang W, Chen J, Guo W. An agonist of the adenosine A 2A receptor, CGS21680, promotes corneal epithelial wound healing via the YAP signalling pathway. Br J Pharmacol 2024; 181:3779-3795. [PMID: 38877785 DOI: 10.1111/bph.16468] [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: 07/04/2023] [Revised: 11/17/2023] [Accepted: 11/29/2023] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The adenosine A2A receptor (A2AR) is involved in various physiological and pathological processes in the eye; however, the role of the A2AR signalling in corneal epithelial wound healing is not known. Here, the expression, therapeutic effects and signalling mechanism of A2AR in corneal epithelial wound healing were investigated using the A2AR agonist CGS21680. EXPERIMENTAL APPROACH A2AR localization and expression during wound healing in the murine cornea were determined by immunofluorescence staining, quantitative reverse transcription polymerase chain reaction (RT-qPCR) and western blotting. The effect of CGS21680 on corneal epithelial wound healing in the lesioned corneal and cultured human corneal epithelial cells (hCECs) by modulating cellular proliferation and migration was critically evaluated. The role of Hippo-YAP signalling in mediating the CGS21680 effect on wound healing by pharmacological inhibition of YAP signalling was explored. KEY RESULTS A2AR expression was up-regulated after corneal epithelial injury. Topical administration of CGS21680 dose-dependently promoted corneal epithelial wound healing in the injured corneal epithelium by promoting cellular proliferation. Furthermore, CGS21680 accelerated the cellular proliferation and migration of hCECs in vitro. A2AR activation promoted early up-regulation and later down-regulation of YAP signalling molecules, and pharmacological inhibition of YAP signalling reverted CGS21680-mediated wound healing effect in vivo and in vitro. CONCLUSION AND IMPLICATIONS A2AR activation promotes wound healing by enhancing cellular proliferation and migration through the YAP signalling pathway. A2ARs play an important role in the maintenance of corneal epithelium integrity and may represent a novel therapeutic target for facilitating corneal epithelial wound healing.
Collapse
Affiliation(s)
- Qiuqin Sun
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Nan Jiang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Rui Yao
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yue Song
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zewen Li
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei Wang
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Jiangfan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei Guo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
3
|
Lairion F, Carbia C, Chiesa IM, Saporito-Magriña C, Borda N, Lazarowski A, Repetto MG. Uridine Diphosphate Glucose (UDP-G) Activates Oxidative Stress and Respiratory Burst in Isolated Neutrophils. Pharmaceuticals (Basel) 2023; 16:1501. [PMID: 37895972 PMCID: PMC10609875 DOI: 10.3390/ph16101501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The extracellular purinergic agonist uridine diphosphate glucose (UDP-G) activates chemotaxis of human neutrophils (PMN) and the recruitment of PMN at the lung level, via P2Y14 purinergic receptor signaling. This effect is similar to the activation of PMN with N-formyl-methionyl-leucyl-phenylalanine (fMLP), a mechanism that also triggers the production of superoxide anion and hydrogen peroxide via the NADPH oxidase system. However, the effects of UDP-G on this system have not been studied. Defects in the intracellular phagocyte respiratory burst (RB) cause recurrent infections, immunodeficiency, and chronic and severe diseases in affected patients, often with sepsis and hypoxia. The extracellular activation of PMN by UDP-G could affect the RB and oxidative stress (OS) in situations of inflammation, infection and/or sepsis. The association of PMNs activation by UDP-G with OS and RB was studied. OS was evaluated by measuring spontaneous chemiluminescence (CL) of PMNs with a scintillation photon counter, and RB by measuring oxygen consumption with an oxygen Clark electrode at 37 °C, in non-stimulated cells and after activation (15 min) with lipopolysaccharides (LPS, 2 µg/mL), phorbol myristate acetate (PMA, 20 ng/mL), or UDP-G (100 μM). The stimulation index (SI) was calculated in order to establish the activation effect of the three agonists. After stimulation with LPS or PMA, the activated PMNs (0.1 × 106 cells/mL) showed an increase in CL (35%, p < 0.05 and 56%, p < 0.01, SI of 1.56 and 2.20, respectively). Contrariwise, the stimulation with UDP-G led to a decreased CL in a dose-dependent manner (60%, 25 μM, p < 0.05; 90%, 50-150 μM, p < 0.001). Nonetheless, despite the lack of oxidative damage, UDP-G triggered RB (SI 1.8) in a dose-dependent manner (38-50%, 100-200 μM, p < 0.0001). UDP-G is able to trigger NADPH oxidase activation in PMNs. Therefore, the prevention of OS and oxidative damage observed upon PMN stimulation with UDP-G indicates an antioxidant property of this molecule which is likely due to the activation of antioxidant defenses. Altogether, LPS and UDP-G have a synergistic effect, suggesting a key role in infection and/or sepsis.
Collapse
Affiliation(s)
- Fabiana Lairion
- Cátedra de Química General e Inorgánica, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (F.L.); (I.M.C.); (C.S.-M.)
- Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Tecnológicas (IBIMOL, UBA-CONICET), Buenos Aires 1113AAD, Argentina
| | - Claudio Carbia
- Cátedra de Bioquímica Clínica II-Área Hematología, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (C.C.); (N.B.)
| | - Iris Maribel Chiesa
- Cátedra de Química General e Inorgánica, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (F.L.); (I.M.C.); (C.S.-M.)
| | - Christian Saporito-Magriña
- Cátedra de Química General e Inorgánica, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (F.L.); (I.M.C.); (C.S.-M.)
- Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Tecnológicas (IBIMOL, UBA-CONICET), Buenos Aires 1113AAD, Argentina
| | - Natalia Borda
- Cátedra de Bioquímica Clínica II-Área Hematología, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (C.C.); (N.B.)
| | - Alberto Lazarowski
- Cátedra de Bioquímica Clínica II-Área Hematología, Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (C.C.); (N.B.)
| | - Marisa Gabriela Repetto
- Cátedra de Química General e Inorgánica, Departamento de Ciencias Químicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires 1113AAD, Argentina; (F.L.); (I.M.C.); (C.S.-M.)
- Instituto de Bioquímica y Medicina Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Tecnológicas (IBIMOL, UBA-CONICET), Buenos Aires 1113AAD, Argentina
| |
Collapse
|
4
|
Ferrari D, Rubini M, Burns JS. The Potential of Purinergic Signaling to Thwart Viruses Including SARS-CoV-2. Front Immunol 2022; 13:904419. [PMID: 35784277 PMCID: PMC9248768 DOI: 10.3389/fimmu.2022.904419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/05/2022] [Indexed: 01/18/2023] Open
Abstract
A long-shared evolutionary history is congruent with the multiple roles played by purinergic signaling in viral infection, replication and host responses that can assist or hinder viral functions. An overview of the involvement of purinergic signaling among a range of viruses is compared and contrasted with what is currently understood for SARS-CoV-2. In particular, we focus on the inflammatory and antiviral responses of infected cells mediated by purinergic receptor activation. Although there is considerable variation in a patient's response to SARS-CoV-2 infection, a principle immediate concern in Coronavirus disease (COVID-19) is the possibility of an aberrant inflammatory activation causing diffuse lung oedema and respiratory failure. We discuss the most promising potential interventions modulating purinergic signaling that may attenuate the more serious repercussions of SARS-CoV-2 infection and aspects of their implementation.
Collapse
Affiliation(s)
- Davide Ferrari
- Section of Microbiology and Applied Pathology, University of Ferrara, Ferrara, Italy
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Michele Rubini
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Jorge S. Burns
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
5
|
Kountz TS, Biyasheva A, Schleimer RP, Prakriya M. Extracellular Nucleotides and Histamine Suppress TLR3- and RIG-I-Mediated Release of Antiviral IFNs from Human Airway Epithelial Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2390-2402. [PMID: 35459743 PMCID: PMC9444327 DOI: 10.4049/jimmunol.2101085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/03/2022] [Indexed: 05/17/2023]
Abstract
Respiratory viruses stimulate the release of antiviral IFNs from the airway epithelium. Previous studies have shown that asthmatic patients show diminished release of type I and type III IFNs from bronchial epithelia. However, the mechanism of this suppression is not understood. In this study, we report that extracellular nucleotides and histamine, which are elevated in asthmatic airways, strongly inhibit release of type I and type III IFNs from human bronchial airway epithelial cells (AECs). Specifically, ATP, UTP, and histamine all inhibited the release of type I and type III IFNs from AECs induced by activation of TLR3, retinoic acid-inducible gene I (RIG-I), or cyclic GMP-AMP synthase-STING. This inhibition was at least partly mediated by Gq signaling through purinergic P2Y2 and H1 receptors, but it did not involve store-operated calcium entry. Pharmacological blockade of protein kinase C partially reversed inhibition of IFN production. Conversely, direct activation of protein kinase C with phorbol esters strongly inhibited TLR3- and RIG-I-mediated IFN production. Inhibition of type I and type III IFNs by ATP, UTP, histamine, and the proteinase-activated receptor 2 (PAR2) receptor agonist SLIGKV also occurred in differentiated AECs grown at an air-liquid interface, indicating that the suppression is conserved following mucociliary differentiation. Importantly, histamine and, more strikingly, ATP inhibited type I IFN release from human airway cells infected with live influenza A virus or rhinovirus 1B. These results reveal an important role for extracellular nucleotides and histamine in attenuating the induction of type I and III IFNs from AECs and help explain the molecular basis of the suppression of IFN responses in asthmatic patients.
Collapse
Affiliation(s)
- Timothy S Kountz
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL; and
| | - Assel Biyasheva
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL; and
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
6
|
Endogenous pannexin1 channels form functional intercellular cell-cell channels with characteristic voltage-dependent properties. Proc Natl Acad Sci U S A 2022; 119:e2202104119. [PMID: 35486697 PMCID: PMC9171361 DOI: 10.1073/pnas.2202104119] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pannexin1 is a glycoprotein that has been shown to form functional plasma membrane channels and mediate many cellular signaling pathways. However, the formation and function of pannexin1-based intercellular cell–cell channels in mammalian cells and vertebrate tissue is a question of substantial debate. This work provides robust electrophysiological evidence to demonstrate that endogenously expressed human pannexin1 forms cell–cell channels and lays the groundwork for studying a potential new type of electrical synapses between many mammalian cell types that endogenously express pannexin1. The occurrence of intercellular channels formed by pannexin1 has been challenged for more than a decade. Here, we provide an electrophysiological characterization of exogenous human pannexin1 (hPanx1) cell–cell channels expressed in HeLa cells knocked out for connexin45. The observed hPanx1 cell–cell channels show two phenotypes: O-state and S-state. The former displayed low transjunctional voltage (Vj) sensitivity and single-channel conductance of ∼175 pS, with a substate of ∼35 pS; the latter showed a peculiar dynamic asymmetry in Vj dependence and single-channel conductance identical to the substate conductance of the O-state. S-state hPanx1 cell–cell channels were also identified between TC620 cells, a human oligodendroglioma cell line that endogenously expresses hPanx1. In these cells, dye and electrical coupling increased with temperature and were strongly reduced after hPanx1 expression was knocked down by small interfering RNA or inhibited with Panx1 mimetic inhibitory peptide. Moreover, cell–cell coupling was augmented when hPanx1 levels were increased with a doxycycline-inducible expression system. Application of octanol, a connexin gap junction (GJ) channel inhibitor, was not sufficient to block electrical coupling between HeLa KO Cx45-hPanx1 or TC620 cell pairs. In silico studies suggest that several arginine residues inside the channel pore may be neutralized by hydrophobic interactions, allowing the passage of DAPI, consistent with dye coupling observed between TC620 cells. These findings demonstrate that endogenously expressed hPanx1 forms intercellular cell–cell channels and their unique properties resemble those described in innexin-based GJ channels. Since Panx1 is ubiquitously expressed, finding conditions to recognize Panx1 cell–cell channels in different cell types might require special attention.
Collapse
|
7
|
Werder RB, Ullah MA, Rahman MM, Simpson J, Lynch JP, Collinson N, Rittchen S, Rashid RB, Sikder MAA, Handoko HY, Curren BF, Sebina I, Hartel G, Bissell A, Ngo S, Yarlagadda T, Hasnain SZ, Lu W, Sohal SS, Martin M, Bowler S, Burr LD, Martinez LO, Robaye B, Spann K, Ferreira MAR, Phipps S. Targeting the P2Y13 Receptor Suppresses IL-33 and HMGB1 Release and Ameliorates Experimental Asthma. Am J Respir Crit Care Med 2021; 205:300-312. [PMID: 34860143 DOI: 10.1164/rccm.202009-3686oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The alarmins IL-33 and HMGB1 (high mobility group box 1) contribute to type-2 inflammation and asthma pathogenesis. OBJECTIVES To determine whether P2Y13 receptor (P2Y13-R), a purinergic G protein-coupled receptor (GPCR) and risk allele for asthma, regulates the release of IL-33 and HMGB1. METHODS Bronchial biopsies were obtained from healthy and asthmatic subjects. Primary human airway epithelial cells (AECs), primary mouse (m)AECs, or C57Bl/6 mice were inoculated with various aeroallergens or respiratory viruses, and the nuclear-to-cytoplasmic translocation and release of alarmins measured by immunohistochemistry and ELISA. The role of P2Y13-R in AEC function and in the onset, progression, and an exacerbation of experimental asthma, was assessed using pharmacological antagonists and P2Y13-R gene-deleted mice. MEASUREMENTS AND MAIN RESULTS Aeroallergen-exposure induced the extracellular release of ADP and ATP, nucleotides that activate P2Y13-R. ATP, ADP, aeroallergen (house dust mite, cockroach or Alternaria) or virus exposure induced the nuclear-to-cytoplasmic translocation and subsequent release of IL-33 and HMGB1, and this response was ablated by genetic deletion or pharmacological antagonism of P2Y13. In mice, prophylactic or therapeutic P2Y13-R blockade attenuated asthma onset, and critically, ablated the severity of a rhinovirus-associated exacerbation in a high-fidelity experimental model of chronic asthma. Moreover, P2Y13-R antagonism derepressed antiviral immunity, increasing IFN-λ production and decreasing viral copies in the lung. CONCLUSIONS We identify P2Y13-R as a novel gatekeeper of the nuclear alarmins IL-33 and HMGB1, and demonstrate that the targeting of this GPCR via genetic deletion or treatment with a small-molecule antagonist protects against the onset and exacerbations of experimental asthma.
Collapse
Affiliation(s)
- Rhiannon B Werder
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia.,Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, United States.,Boston University School of Medicine, 12259, The Pulmonary Center and Department of Medicine, Boston, Massachusetts, United States
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Jennifer Simpson
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia.,National Institute of Allergy and Infectious Diseases, 35037, Barrier Immunity Section, Laboratory of Viral Diseases, Bethesda, Maryland, United States
| | - Jason P Lynch
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,Harvard Medical School, 1811, Department of Microbiology, Boston, Massachusetts, United States
| | - Natasha Collinson
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Sonja Rittchen
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,Medical University of Graz, 31475, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Graz, Steiermark, Austria
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Herlina Y Handoko
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Bodie F Curren
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Gunter Hartel
- QIMR Berghofer, 56362, Brisbane, Queensland, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Sylvia Ngo
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Tejasri Yarlagadda
- Queensland University of Technology Faculty of Health, 110544, Kelvin Grove, Queensland, Australia
| | - Sumaira Z Hasnain
- Mater Medical Research Institute, 200098, Brisbane, Queensland, Australia
| | - Wenying Lu
- University of Tasmania, 3925, Respiratory Translational Research Group, Launceston , Tasmania, Australia
| | - Sukhwinder S Sohal
- University of Tasmania , Respiratory Translational Research Group, Launceston , Tasmania, Australia
| | - Megan Martin
- Mater Health Services, Respiratory Medicine, South Brisbane, Queensland, Australia
| | - Simon Bowler
- Mater Health Services, Respiratory Medicine, South Brisbane, Queensland, Australia
| | - Lucy D Burr
- UQ School of Medicine, Brisbane, Queensland, Australia
| | - Laurent O Martinez
- University of Toulouse, 137668, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Bernard Robaye
- Université Libre de Bruxelles, 26659, IRIBHM, Bruxelles, Belgium
| | - Kirsten Spann
- Queensland University of Technology, 1969, School of Biomedical Sciences, Brisbane, Queensland, Australia
| | - Manuel A R Ferreira
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia;
| |
Collapse
|
8
|
Li Y, Tang XX. Abnormal Airway Mucus Secretion Induced by Virus Infection. Front Immunol 2021; 12:701443. [PMID: 34650550 PMCID: PMC8505958 DOI: 10.3389/fimmu.2021.701443] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
The airway mucus barrier is a primary defensive layer at the airway surface. Mucins are the major structural components of airway mucus that protect the respiratory tract. Respiratory viruses invade human airways and often induce abnormal mucin overproduction and airway mucus secretion, leading to airway obstruction and disease. The mechanism underlying the virus-induced abnormal airway mucus secretion has not been fully studied so far. Understanding the mechanisms by which viruses induce airway mucus hypersecretion may open new avenues to treatment. In this article, we elaborate the clinical and experimental evidence that respiratory viruses cause abnormal airway mucus secretion, review the underlying mechanisms, and also discuss the current research advance as well as potential strategies to treat the abnormal airway mucus secretion caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Yao Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Laboratory, Bio-island, Guangzhou, China
| |
Collapse
|
9
|
Lee DF, Lethem MI, Lansley AB. A comparison of three mucus-secreting airway cell lines (Calu-3, SPOC1 and UNCN3T) for use as biopharmaceutical models of the nose and lung. Eur J Pharm Biopharm 2021; 167:159-174. [PMID: 34332033 PMCID: PMC8422164 DOI: 10.1016/j.ejpb.2021.07.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/09/2021] [Accepted: 07/22/2021] [Indexed: 12/19/2022]
Abstract
The aim of this work was to compare three existing mucus-secreting airway cell lines for use as models of the airways to study drug transport in the presence of mucus. Each cell line secreted mature, glycosylated mucins, evidenced by the enzyme-linked lectin assay. The secretagogue, adenylyl-imidodiphosphate, increased mucin secretion in SPOC1 (3.5-fold) and UNCN3T (1.5-fold) cells but not in Calu-3 cells. In a novel mucus-depleted (MD) model the amount of mucus in the non-depleted wells was 3-, 8- and 4-fold higher than in the mucus-depleted wells of the Calu-3, SPOC1 and UNCN3T cells respectively. The permeability of 'high mucus' cells to testosterone was significantly less in SPOC1 and UNCN3T cells (P < 0.05) but not Calu-3 cells. Mucin secretion and cytokine release were investigated as indicators of drug irritancy in the SPOC1 and UNCN3T cell lines. A number of inhaled drugs significantly increased mucin secretion at high concentrations and the release of IL-6 and IL-8 from SPOC1 or UNCN3T cells (P < 0.05). SPOC1 and UNCN3T cell lines are better able to model the effect of mucus on drug absorption than the Calu-3 cell line and are proposed for use in assessing drug-mucus interactions in inhaled drug and formulation development.
Collapse
Affiliation(s)
- Diane F Lee
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK; School of Veterinary Medicine, University of Surrey, Guildford GU2 7AL, UK(1).
| | - Michael I Lethem
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK
| | - Alison B Lansley
- School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK.
| |
Collapse
|
10
|
Mucus Release and Airway Constriction by TMEM16A May Worsen Pathology in Inflammatory Lung Disease. Int J Mol Sci 2021; 22:ijms22157852. [PMID: 34360618 PMCID: PMC8346050 DOI: 10.3390/ijms22157852] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Activation of the Ca2+ activated Cl− channel TMEM16A is proposed as a treatment in inflammatory airway disease. It is assumed that activation of TMEM16A will induce electrolyte secretion, and thus reduce airway mucus plugging and improve mucociliary clearance. A benefit of activation of TMEM16A was shown in vitro and in studies in sheep, but others reported an increase in mucus production and airway contraction by activation of TMEM16A. We analyzed expression of TMEM16A in healthy and inflamed human and mouse airways and examined the consequences of activation or inhibition of TMEM16A in asthmatic mice. TMEM16A was found to be upregulated in the lungs of patients with asthma or cystic fibrosis, as well as in the airways of asthmatic mice. Activation or potentiation of TMEM16A by the compounds Eact or brevenal, respectively, induced acute mucus release from airway goblet cells and induced bronchoconstriction in mice in vivo. In contrast, niclosamide, an inhibitor of TMEM16A, blocked mucus production and mucus secretion in vivo and in vitro. Treatment of airway epithelial cells with niclosamide strongly inhibited expression of the essential transcription factor of Th2-dependent inflammation and goblet cell differentiation, SAM pointed domain-containing ETS-like factor (SPDEF). Activation of TMEM16A in people with inflammatory airway diseases is likely to induce mucus secretion along with airway constriction. In contrast, inhibitors of TMEM16A may suppress pulmonary Th2 inflammation, goblet cell metaplasia, mucus production, and bronchoconstriction, partially by inhibiting expression of SPDEF.
Collapse
|
11
|
Rasmussen HB, Jürgens G, Thomsen R, Taboureau O, Zeth K, Hansen PE, Hansen PR. Cellular Uptake and Intracellular Phosphorylation of GS-441524: Implications for Its Effectiveness against COVID-19. Viruses 2021; 13:v13071369. [PMID: 34372575 PMCID: PMC8310262 DOI: 10.3390/v13071369] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/22/2022] Open
Abstract
GS-441524 is an adenosine analog and the parent nucleoside of the prodrug remdesivir, which has received emergency approval for treatment of COVID-19. Recently, GS-441524 has been proposed to be effective in the treatment of COVID-19, perhaps even being superior to remdesivir for treatment of this disease. Evaluation of the clinical effectiveness of GS-441524 requires understanding of its uptake and intracellular conversion to GS-441524 triphosphate, the active antiviral substance. We here discuss the potential impact of these pharmacokinetic steps of GS-441524 on the formation of its active antiviral substance and effectiveness for treatment of COVID-19. Available protein expression data suggest that several adenosine transporters are expressed at only low levels in the epithelial cells lining the alveoli in the lungs, i.e., the alveolar cells or pneumocytes from healthy lungs. This may limit uptake of GS-441524. Importantly, cellular uptake of GS-441524 may be reduced during hypoxia and inflammation due to decreased expression of adenosine transporters. Similarly, hypoxia and inflammation may lead to reduced expression of adenosine kinase, which is believed to convert GS-441524 to GS-441524 monophosphate, the perceived rate-limiting step in the intracellular formation of GS-441524 triphosphate. Moreover, increases in extracellular and intracellular levels of adenosine, which may occur during critical illnesses, has the potential to competitively decrease cellular uptake and phosphorylation of GS-441524. Taken together, tissue hypoxia and severe inflammation in COVID-19 may lead to reduced uptake and phosphorylation of GS-441524 with lowered therapeutic effectiveness as a potential outcome. Hypoxia may be particularly critical to the ability of GS-441524 to eliminate SARS-CoV-2 from tissues with low basal expression of adenosine transporters, such as alveolar cells. This knowledge may also be relevant to treatments with other antiviral adenosine analogs and anticancer adenosine analogs as well.
Collapse
Affiliation(s)
- Henrik Berg Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, DK-4000 Roskilde, Denmark
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark; (K.Z.); (P.E.H.)
- Correspondence:
| | - Gesche Jürgens
- Clinical Pharmacology Unit, Zealand University Hospital, DK-4000 Roskilde, Denmark;
| | - Ragnar Thomsen
- Section of Forensic Chemistry, Department of Forensic Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark;
| | - Olivier Taboureau
- INSERM U1133, CNRS UMR 8251, Université de Paris, F-75013 Paris, France;
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark; (K.Z.); (P.E.H.)
| | - Poul Erik Hansen
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark; (K.Z.); (P.E.H.)
| | - Peter Riis Hansen
- Department of Cardiology, Herlev and Gentofte Hospital, DK-2900 Hellerup, Denmark;
| |
Collapse
|
12
|
Airway Epithelial Nucleotide Release Contributes to Mucociliary Clearance. Life (Basel) 2021; 11:life11050430. [PMID: 34064654 PMCID: PMC8151306 DOI: 10.3390/life11050430] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/18/2022] Open
Abstract
Mucociliary clearance (MCC) is a dominant component of pulmonary host defense. In health, the periciliary layer (PCL) is optimally hydrated, thus acting as an efficient lubricant layer over which the mucus layer moves by ciliary force. Airway surface dehydration and production of hyperconcentrated mucus is a common feature of chronic obstructive lung diseases such as cystic fibrosis (CF) and chronic bronchitis (CB). Mucus hydration is driven by electrolyte transport activities, which in turn are regulated by airway epithelial purinergic receptors. The activity of these receptors is controlled by the extracellular concentrations of ATP and its metabolite adenosine. Vesicular and conducted pathways contribute to ATP release from airway epithelial cells. In this study, we review the evidence leading to the identification of major components of these pathways: (a) the vesicular nucleotide transporter VNUT (the product of the SLC17A9 gene), the ATP transporter mediating ATP storage in (and release from) mucin granules and secretory vesicles; and (b) the ATP conduit pannexin 1 expressed in non-mucous airway epithelial cells. We further illustrate that ablation of pannexin 1 reduces, at least in part, airway surface liquid (ASL) volume production, ciliary beating, and MCC rates.
Collapse
|
13
|
Lazarowski ER, Boucher RC. Purinergic receptors in airway hydration. Biochem Pharmacol 2021; 187:114387. [PMID: 33358825 DOI: 10.1016/j.bcp.2020.114387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 02/08/2023]
Abstract
Airway epithelial purinergic receptors control key components of the mucociliary clearance (MCC), the dominant component of pulmonary host defense. In healthy airways, the periciliary liquid (PCL) is optimally hydrated, thus acting as an efficient lubricant layer over which the mucus layer moves by ciliary force. When the hydration of the airway surface decreases, the mucus becomes hyperconcentrated, the PCL collapses, and the "thickened" mucus layer adheres to cell surfaces, causing plaque/plug formation. Mucus accumulation is a major contributing factor to the progression of chronic obstructive lung diseases such as cystic fibrosis (CF) and chronic bronchitis (CB). Mucus hydration is regulated by finely tuned mechanisms of luminal Cl- secretion and Na+ absorption with concomitant osmotically driven water flow. These activities are regulated by airway surface liquid (ASL) concentrations of adenosine and ATP, acting on airway epithelial A2B and P2Y2 receptors, respectively. The goal of this article is to provide an overview of our understanding of the role of purinergic receptors in the regulation of airway epithelial ion/fluid transport and the mechanisms of nucleotide release and metabolic activities that contribute to airway surface hydration in healthy and chronically obstructed airways.
Collapse
Affiliation(s)
- Eduardo R Lazarowski
- Marsico Lung Institute/Cystic Fibrosis Center, School of Medicine, University of North Carolina, Chapel Hill, NC, United States.
| | - Richard C Boucher
- Marsico Lung Institute/Cystic Fibrosis Center, School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
14
|
Jing X, Yan W, Zeng H, Cheng W. Qingfei oral liquid alleviates airway hyperresponsiveness and mucus hypersecretion via TRPV1 signaling in RSV-infected asthmatic mice. Biomed Pharmacother 2020; 128:110340. [PMID: 32521453 DOI: 10.1016/j.biopha.2020.110340] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/24/2020] [Accepted: 05/30/2020] [Indexed: 12/14/2022] Open
Abstract
Pediatric asthma is exacerbated by Respiratory Syncytial Virus (RSV) infection, and Transient Receptor Potential Vanilloid 1 (TRPV1) promotes production of inflammatory cytokines and mucus hypersecretion in the pathology of this disease. Our previous research revealed that Qingfei oral liquid (QF) inhibited airway inflammation and mucus hypersecretion in RSV-infected asthmatic mice models and that this may be associated with the TRPV1-regulation of NF-κB and Mucin 5AC (MUC5AC) expression, but the exact mechanism is unknown. In the present study, LC-MS was used for analyzing the chemicals in QF, ovalbumin (OVA)-induced asthmatic mice inhaled RSV three consecutive times to create an RSV-infected asthmatic model. We found treatment from QF alleviated airway hyperresponsiveness (AHR) and reduced congestion, edema, and infiltration of inflammatory cells into pulmonary tissues. Additionally, QF was found to decrease expression of NF-κB and its downstream inflammatory cytokines IL-1β, IL-4, IL-5, and IL-13, as well as a decrease in MUC5AC and pro-inflammatory cytokines in PKC via a reduction in Protein Kinase C-dependent signaling. These findings suggest that QF can alleviate AHR and mucus hypersecretion caused by RSV infection in asthmatic mice, and its mechanism may be associated with the regulation of the TRPV1 signaling pathway.
Collapse
Affiliation(s)
- Xiaoping Jing
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China.
| | - Wuning Yan
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Hairong Zeng
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
| | - Weiwei Cheng
- Department of Traditional Chinese Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai 200040, China
| |
Collapse
|
15
|
Li H, Jiang W, Ye S, Zhou M, Liu C, Yang X, Hao K, Hu Q. P2Y 14 receptor has a critical role in acute gouty arthritis by regulating pyroptosis of macrophages. Cell Death Dis 2020; 11:394. [PMID: 32457291 PMCID: PMC7250907 DOI: 10.1038/s41419-020-2609-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022]
Abstract
Nod-like receptor protein 3 (NLRP3)-mediated pyroptosis has a causal role in the pathogenesis of gout. P2Y14 receptor (P2Y14R) distributed in immune cells including macrophages is a Gi-coupled receptor that inhibits the synthesis of cAMP, which has been regarded as a potential regulator of inflammatory response. Nevertheless, the role of P2Y14R in MSU-induced pyroptosis of macrophages involved in acute gouty arthritis is still unclear. In our present study, P2Y14R knockout (P2Y14R-KO) disrupted MSU-induced histopathologic changes in rat synoviums, accompanied with a significant inhibition of pyroptotic cell death characterized by Caspase-1/PI double-positive and blockade of NLRP3 inflammasome activation in synovial tissues, which was consistent with that observed in in vitro studies. Owing to the interaction of NLRP3 inflammasome and cAMP, we then investigated the effect of adenylate cyclase activator (Forskolin) on macrophage pyroptosis and gout flare caused by MSU stimulation. The reversal effect of Forskolin verified the negative regulatory role of cAMP in MSU-induced pyroptosis. More importantly, adenylate cyclase inhibitor (SQ22536) intervention led to a reversal of protection attributed to P2Y14R deficiency. Findings in air pouch animal models also verified aforementioned experimental results. Our study first identified the role of P2Y14R/cAMP/NLRP3 signaling pathway in acute gouty arthritis, which provides a novel insight into the pathological mechanisms of pyroptosis-related diseases.
Collapse
Affiliation(s)
- Hanwen Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Wenjiao Jiang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shumin Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Mengze Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Chunxiao Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Xiping Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Qinghua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China. .,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
16
|
Role of UDP-Sugar Receptor P2Y 14 in Murine Osteoblasts. Int J Mol Sci 2020; 21:ijms21082747. [PMID: 32326617 PMCID: PMC7216066 DOI: 10.3390/ijms21082747] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The purinergic (P2) receptor P2Y14 is the only P2 receptor that is stimulated by uridine diphosphate (UDP)-sugars and its role in bone formation is unknown. We confirmed P2Y14 expression in primary murine osteoblasts (CB-Ob) and the C2C12-BMP2 osteoblastic cell line (C2-Ob). UDP-glucose (UDPG) had undiscernible effects on cAMP levels, however, induced dose-dependent elevations in the cytosolic free calcium concentration ([Ca2+]i) in CB-Ob, but not C2-Ob cells. To antagonize the P2Y14 function, we used the P2Y14 inhibitor PPTN or generated CRISPR-Cas9-mediated P2Y14 knockout C2-Ob clones (Y14KO). P2Y14 inhibition facilitated calcium signalling and altered basal cAMP levels in both models of osteoblasts. Importantly, P2Y14 inhibition augmented Ca2+ signalling in response to ATP, ADP and mechanical stimulation. P2Y14 knockout or inhibition reduced osteoblast proliferation and decreased ERK1/2 phosphorylation and increased AMPKα phosphorylation. During in vitro osteogenic differentiation, P2Y14 inhibition modulated the timing of osteogenic gene expression, collagen deposition, and mineralization, but did not significantly affect differentiation status by day 28. Of interest, while P2ry14-/- mice from the International Mouse Phenotyping Consortium were similar to wild-type controls in bone mineral density, their tibia length was significantly increased. We conclude that P2Y14 in osteoblasts reduces cell responsiveness to mechanical stimulation and mechanotransductive signalling and modulates osteoblast differentiation.
Collapse
|
17
|
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
|
18
|
Ferrari D, Idzko M, Müller T, Manservigi R, Marconi P. Purinergic Signaling: A New Pharmacological Target Against Viruses? Trends Pharmacol Sci 2018; 39:926-936. [PMID: 30292585 DOI: 10.1016/j.tips.2018.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/11/2022]
Abstract
Viral diseases represent a major global problem in human health, with high morbidity and mortality. Despite recent progress in antiviral treatments, several viral diseases are still not controlled and millions suffer from them every year. It has recently emerged that purinergic signaling participates in viral infection and replication. Furthermore, stimulation of purinergic receptors in infected cells also induces inflammatory and antiviral responses, thus contributing to the host antiviral defense. Here we review the multiple roles played by the purinergic signaling network in cell-virus interactions that can lead either to viral maintenance in the cells or, by contrast, to stronger antiviral responses, and discuss potential future applications of purinergic signaling modulation for the treatment of viral diseases.
Collapse
Affiliation(s)
- Davide Ferrari
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Marco Idzko
- Department of Pneumology, Medical University of Vienna, Vienna, Austria
| | - Tobias Müller
- Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Roberto Manservigi
- Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Peggy Marconi
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
19
|
Chanson M, Watanabe M, O'Shaughnessy EM, Zoso A, Martin PE. Connexin Communication Compartments and Wound Repair in Epithelial Tissue. Int J Mol Sci 2018; 19:ijms19051354. [PMID: 29751558 PMCID: PMC5983803 DOI: 10.3390/ijms19051354] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/26/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022] Open
Abstract
Epithelial tissues line the lumen of tracts and ducts connecting to the external environment. They are critical in forming an interface between the internal and external environment and, following assault from environmental factors and pathogens, they must rapidly repair to maintain cellular homeostasis. These tissue networks, that range from a single cell layer, such as in airway epithelium, to highly stratified and differentiated epithelial surfaces, such as the epidermis, are held together by a junctional nexus of proteins including adherens, tight and gap junctions, often forming unique and localised communication compartments activated for localised tissue repair. This review focuses on the dynamic changes that occur in connexins, the constituent proteins of the intercellular gap junction channel, during wound-healing processes and in localised inflammation, with an emphasis on the lung and skin. Current developments in targeting connexins as corrective therapies to improve wound closure and resolve localised inflammation are also discussed. Finally, we consider the emergence of the zebrafish as a concerted whole-animal model to study, visualise and track the events of wound repair and regeneration in real-time living model systems.
Collapse
Affiliation(s)
- Marc Chanson
- Department of Pediatrics and Cell Physiology & Metabolism, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland.
| | - Masakatsu Watanabe
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan.
| | - Erin M O'Shaughnessy
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| | - Alice Zoso
- Department of Pediatrics and Cell Physiology & Metabolism, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland.
| | - Patricia E Martin
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK.
| |
Collapse
|
20
|
Fois G, Winkelmann VE, Bareis L, Staudenmaier L, Hecht E, Ziller C, Ehinger K, Schymeinsky J, Kranz C, Frick M. ATP is stored in lamellar bodies to activate vesicular P2X 4 in an autocrine fashion upon exocytosis. J Gen Physiol 2017; 150:277-291. [PMID: 29282210 PMCID: PMC5806682 DOI: 10.1085/jgp.201711870] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
Abstract
P2X4 receptor activation facilitates secretion of pulmonary surfactant from secretory vesicles called lamellar bodies in alveolar epithelial cells. Fois et al. reveal that P2X4 receptors on the lamellar body membranes are activated by ATP stored within the vesicles themselves upon vesicle exocytosis. Vesicular P2X4 receptors are known to facilitate secretion and activation of pulmonary surfactant in the alveoli of the lungs. P2X4 receptors are expressed in the membrane of lamellar bodies (LBs), large secretory lysosomes that store lung surfactant in alveolar type II epithelial cells, and become inserted into the plasma membrane after exocytosis. Subsequent activation of P2X4 receptors by adenosine triphosphate (ATP) results in local fusion-activated cation entry (FACE), facilitating fusion pore dilation, surfactant secretion, and surfactant activation. Despite the importance of ATP in the alveoli, and hence lung function, the origin of ATP in the alveoli is still elusive. In this study, we demonstrate that ATP is stored within LBs themselves at a concentration of ∼1.9 mM. ATP is loaded into LBs by the vesicular nucleotide transporter but does not activate P2X4 receptors because of the low intraluminal pH (5.5). However, the rise in intravesicular pH after opening of the exocytic fusion pore results in immediate activation of vesicular P2X4 by vesicular ATP. Our data suggest a new model in which agonist (ATP) and receptor (P2X4) are located in the same intracellular compartment (LB), protected from premature degradation (ATP) and activation (P2X4), and ideally placed to ensure coordinated and timely receptor activation as soon as fusion occurs to facilitate surfactant secretion.
Collapse
Affiliation(s)
- Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Lara Bareis
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Elena Hecht
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Charlotte Ziller
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | | | - Jürgen Schymeinsky
- Immunology and Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Christine Kranz
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
21
|
Wong MKS, Tsukada T, Ogawa N, Pipil S, Ozaki H, Suzuki Y, Iwasaki W, Takei Y. A sodium binding system alleviates acute salt stress during seawater acclimation in eels. ZOOLOGICAL LETTERS 2017; 3:22. [PMID: 29255617 PMCID: PMC5727781 DOI: 10.1186/s40851-017-0081-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 10/11/2017] [Indexed: 05/28/2023]
Abstract
BACKGROUND Teleosts transiting from freshwater (FW) to seawater (SW) environments face an immediate osmotic stress from ion influxes and water loss, but some euryhaline species such as eels can maintain a stable plasma osmolality during early SW exposure. The time course changes in the gene expression, protein abundance, and localization of key ion transporters suggested that the reversal of the ion transport systems was gradual, and we investigate how eels utilize a Na-binding strategy to slow down the ion invasion and complement the transporter-mediated osmoregulation. RESULTS Using an electron probe micro-analyzer, we localized bound Na in various eel tissues in response to SW transfer, suggesting that the Na-binding molecules were produced to sequester excess ionic Na+ to negate its osmotic potential, thus preventing acute cellular dehydration. Mucus cells were acutely activated in digestive tract, gill, and skin after SW transfer, producing Na-binding molecule-containing mucus layers that fence off high osmolality of SW. Using gel filtration HPLC, some molecules at 18 kDa were found to bind Na in the luminal secretion of esophagus and intestine, and higher binding was associated with SW transfer. Transcriptome and protein interaction results indicated that downregulation of Notch and β-catenin pathways, and dynamic changes in TGFβ pathways in intestine were involved during early SW transition, supporting the observed histological changes on epithelial desquamation and increased mucus production. CONCLUSIONS The timing for the activation of the Na-binding mechanism to alleviate the adverse osmotic gradient was temporally complementary to the subsequent remodeling of branchial ionocytes and transporting epithelia of the digestive tract. The strategy to manipulate the osmotic potential of Na+ by specific binding molecules is similar to the osmotically inactive Na described in human skin and muscle. The Na-binding molecules provide a buffer to tolerate the salinity changes, which is advantageous to the estuary and migrating fishes. Our data pave the way to identify this unknown class of molecules and open a new area of vertebrate osmoregulation research.
Collapse
Affiliation(s)
- Marty Kwok Shing Wong
- Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa City, Japan
| | - Takehiro Tsukada
- Department of Biomolecular Science, Toho University, Funabashi City, Japan
| | - Nobuhiro Ogawa
- Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa City, Japan
| | - Supriya Pipil
- Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa City, Japan
| | - Haruka Ozaki
- Department of Computational Biology, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Japan
- Bioinformatics Research Unit, Advanced Center for Computing and Communication, RIKEN, Wako City, Japan
| | - Yutaka Suzuki
- Department of Computational Biology, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Japan
| | - Wataru Iwasaki
- Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa City, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa City, Japan
| | - Yoshio Takei
- Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa City, Japan
| |
Collapse
|
22
|
Droguett K, Rios M, Carreño DV, Navarrete C, Fuentes C, Villalón M, Barrera NP. An autocrine ATP release mechanism regulates basal ciliary activity in airway epithelium. J Physiol 2017; 595:4755-4767. [PMID: 28422293 PMCID: PMC5509870 DOI: 10.1113/jp273996] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 04/10/2017] [Indexed: 01/07/2023] Open
Abstract
KEY POINTS Extracellular ATP, in association with [Ca2+ ]i regulation, is required to maintain basal ciliary beat frequency. Increasing extracellular ATP levels increases ciliary beating in airway epithelial cells, maintaining a sustained response by inducing the release of additional ATP. Extracellular ATP levels in the millimolar range, previously associated with pathophysiological conditions of the airway epithelium, produce a transient arrest of ciliary activity. The regulation of ciliary beat frequency is dependent on ATP release by hemichannels (connexin/pannexin) and P2X receptor activation, the blockage of which may even stop ciliary movement. The force exerted by cilia, measured by atomic force microscopy, is reduced following extracellular ATP hydrolysis. This result complements the current understanding of the ciliary beating regulatory mechanism, with special relevance to inflammatory diseases of the airway epithelium that affect mucociliary clearance. ABSTRACT Extracellular nucleotides, including ATP, are locally released by the airway epithelium and stimulate ciliary activity in a [Ca2+ ]i -dependent manner after mechanical stimulation of ciliated cells. However, it is unclear whether the ATP released is involved in regulating basal ciliary activity and mediating changes in ciliary activity in response to chemical stimulation. In the present study, we evaluated ciliary beat frequency (CBF) and ciliary beating forces in primary cultures from mouse tracheal epithelium, using videomicroscopy and atomic force microscopy (AFM), respectively. Extracellular ATP levels and [Ca2+ ]i were measured by luminometric and fluorimetric assays, respectively. Uptake of ethidium bromide was measured to evaluate hemichannel functionality. We show that hydrolysis of constitutive extracellular ATP levels with apyrase (50 U ml-1 ) reduced basal CBF by 45% and ciliary force by 67%. The apyrase effect on CBF was potentiated by carbenoxolone, a hemichannel inhibitor, and oxidized ATP, an antagonist used to block P2X7 receptors, which reduced basal CBF by 85%. Additionally, increasing extracellular ATP levels (0.1-100 μm) increased CBF, maintaining a sustained response that was suppressed in the presence of carbenoxolone. We also show that high levels of ATP (1 mm), associated with inflammatory conditions, lowered basal CBF by reducing [Ca2+ ]i and hemichannel functionality. In summary, we provide evidence indicating that airway epithelium ATP release is the molecular autocrine mechanism regulating basal ciliary activity and is also the mediator of the ciliary response to chemical stimulation.
Collapse
Affiliation(s)
- Karla Droguett
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Mariana Rios
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Daniela V. Carreño
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Camilo Navarrete
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Christian Fuentes
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Manuel Villalón
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| | - Nelson P. Barrera
- Department of Physiology, Faculty of Biological SciencesPontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
23
|
Evaluation of Connexin 43 Redistribution and Endocytosis in Astrocytes Subjected to Ischemia/Reperfusion or Oxygen-Glucose Deprivation and Reoxygenation. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5064683. [PMID: 28424784 PMCID: PMC5382357 DOI: 10.1155/2017/5064683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 02/23/2017] [Accepted: 03/08/2017] [Indexed: 01/08/2023]
Abstract
Connexin 43 (Cx43) is the major component protein in astrocytic gap junction communication. Recent studies have shown the cellular processes of gap junction internalization and degradation, but many details remain unknown. This study investigated the distribution of Cx43 and its mechanism after ischemic insult. Astrocyte culture system and a model of ischemia/reperfusion (IR) or oxygen-glucose deprivation and reoxygenation (OGDR) were established. Cx43 distribution was observed by laser scanning confocal microscopy under different cultivation conditions. Western blot and RT-PCR assays were applied to quantify Cx43 and MAPRE1 (microtubule-associated protein RP/EB family member 1) expression at different time points. The total number of Cx43 was unchanged in the normal and IR/OGDR groups, but Cx43 particles in the cytoplasm of the IR/OGDR group were significantly greater than that of the normal group. Particles in the cytoplasm were significantly fewer after endocytosis was blocked by dynasore. There was no difference among the groups at each time point regarding protein or gene expression of MAPRE1. We concluded that internalization of Cx43 into the cytoplasm occurred during ischemia, which was partially mediated through endocytosis, not by the change of Cx43 quantity. Moreover, internalization was not related to microtubule transport.
Collapse
|
24
|
Lee HJ, Lee CJ. Effects of Homogentisic Acid and Natural Products Derived from Pinellia ternata on Secretion, Production and Gene Expression of MUC5AC Mucin from Cultured Airway Epithelial Cells. ACTA ACUST UNITED AC 2017. [DOI: 10.20307/nps.2017.23.1.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hyun Jae Lee
- Department of Health Management and Smith Liberal Arts College, Sahmyook University, Seoul, Korea
| | - Choong Jae Lee
- Department of Pharmacology, School of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
25
|
Shishikura Y, Koarai A, Aizawa H, Yamaya M, Sugiura H, Watanabe M, Hashimoto Y, Numakura T, Makiguti T, Abe K, Yamada M, Kikuchi T, Hoshikawa Y, Okada Y, Ichinose M. Extracellular ATP is involved in dsRNA-induced MUC5AC production via P2Y2R in human airway epithelium. Respir Res 2016; 17:121. [PMID: 27677339 PMCID: PMC5039824 DOI: 10.1186/s12931-016-0438-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/20/2016] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND In response to tissue damage or inflammation, adenosine-5'-triphosphate (ATP) is released into the extracellular compartment and has been demonstrated to augment inflammation via purinergic P2 receptors (P2Rs). Recently, ATP has been shown to be increased in the airways of COPD patients. In the present study, we examined the possible involvement of extracellular ATP in airway mucus hypersecretion during viral-induced COPD exacerbations. METHODS The involvement of extracellular ATP in the release of a major airway mucin, MUC5AC, and its signal pathway was examined after stimulation with polyinosine-polycytidylic acid [poly(I:C)], a synthetic analog of dsRNA to mimic viral infection, and rhinovirus (RV) infection in NCI-H292 cells and differentiated airway epithelial cells from COPD patients. RESULTS Treatment with poly(I:C) significantly increased the amount of extracellular ATP and induced MUC5AC release in NCI-H292 cells. Pre-treatment with a pannexin channel inhibitor, carbenoxolone (CBX), reduced the amount of extracellular ATP and suppressed MUC5AC release from poly(I:C)-treated cells. Pre-treatment with the P2R antagonist suramin significantly reduced the expression and release of MUC5AC. The inhibitory effects of CBX and suramin on the release of ATP and/or MUC5AC were replicated with RV infection. Pre-treatment with suramin also significantly reduced the expression and amount of extracellular EGFR ligands and the phosphorylation of EGFR and ERK in poly(I:C)-treated cells. In addition, pre-treatment with a P2Y2 receptor siRNA significantly suppressed the poly(I:C)-potentiated EGFR ligands and MUC5AC release. After poly(I:C) stimulation, the expression of MUC5AC in the differentiated cells from COPD patients was significantly higher than those from healthy subjects and the values of MUC5AC expression were inversely related with forced expiratory volume in 1 s (FEV1) % predicted. The inhibitory effects of CBX and suramin on poly(I:C)-potentiated MUC5AC expression were confirmed in differentiated airway epithelium from COPD patients. CONCLUSIONS These results demonstrate that dsRNA induces the release of ATP via pannexin channel and that the extracellular ATP is involved in the expression and release of MUC5AC, mainly via P2Y2R, in an autocrine manner. Modulation of this pathway could be a therapeutic target for viral-induced mucus hypersecretion in COPD exacerbations.
Collapse
Affiliation(s)
- Yutaka Shishikura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Akira Koarai
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Hiroyuki Aizawa
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8575 Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Mika Watanabe
- Department of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Yuichiro Hashimoto
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Tadahisa Numakura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Tomonori Makiguti
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Kyoko Abe
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Mituhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachidori, Chuo-ku, Niigata 951-8510 Japan
| | - Yasushi Hoshikawa
- Department of Thoracic Surgery, Fujita Health University School of Medicine, Toyoake, 470-1192 Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery Institute of Development, Aging and Cancer Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575 Japan
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574 Japan
| |
Collapse
|
26
|
Janssen WJ, Stefanski AL, Bochner BS, Evans CM. Control of lung defence by mucins and macrophages: ancient defence mechanisms with modern functions. Eur Respir J 2016; 48:1201-1214. [PMID: 27587549 DOI: 10.1183/13993003.00120-2015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 06/12/2016] [Indexed: 12/14/2022]
Abstract
Owing to the need to balance the requirement for efficient respiration in the face of tremendous levels of exposure to endogenous and environmental challenges, it is crucial for the lungs to maintain a sustainable defence that minimises damage caused by this exposure and the detrimental effects of inflammation to delicate gas exchange surfaces. Accordingly, epithelial and macrophage defences constitute essential first and second lines of protection that prevent the accumulation of potentially harmful agents in the lungs, and under homeostatic conditions do so effectively without inducing inflammation. Though epithelial and macrophage-mediated defences are seemingly distinct, recent data show that they are linked through their shared reliance on airway mucins, in particular the polymeric mucin MUC5B. This review highlights our understanding of novel mechanisms that link mucus and macrophage defences. We discuss the roles of phagocytosis and the effects of factors contained within mucus on phagocytosis, as well as newly identified roles for mucin glycoproteins in the direct regulation of leukocyte functions. The emergence of this nascent field of glycoimmunobiology sets forth a new paradigm for considering how homeostasis is maintained under healthy conditions and how it is restored in disease.
Collapse
Affiliation(s)
- William J Janssen
- Dept of Medicine, National Jewish Health, Denver, CO, USA Dept of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Bruce S Bochner
- Dept of Medicine, Division of Allergy-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Christopher M Evans
- Dept of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
27
|
Ferreira MAR, Jansen R, Willemsen G, Penninx B, Bain LM, Vicente CT, Revez JA, Matheson MC, Hui J, Tung JY, Baltic S, Le Souëf P, Montgomery GW, Martin NG, Robertson CF, James A, Thompson PJ, Boomsma DI, Hopper JL, Hinds DA, Werder RB, Phipps S. Gene-based analysis of regulatory variants identifies 4 putative novel asthma risk genes related to nucleotide synthesis and signaling. J Allergy Clin Immunol 2016; 139:1148-1157. [PMID: 27554816 DOI: 10.1016/j.jaci.2016.07.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND Hundreds of genetic variants are thought to contribute to variation in asthma risk by modulating gene expression. Methods that increase the power of genome-wide association studies (GWASs) to identify risk-associated variants are needed. OBJECTIVE We sought to develop a method that aggregates the evidence for association with disease risk across expression quantitative trait loci (eQTLs) of a gene and use this approach to identify asthma risk genes. METHODS We developed a gene-based test and software package called EUGENE that (1) is applicable to GWAS summary statistics; (2) considers both cis- and trans-eQTLs; (3) incorporates eQTLs identified in different tissues; and (4) uses simulations to account for multiple testing. We applied this approach to 2 published asthma GWASs (combined n = 46,044) and used mouse studies to provide initial functional insights into 2 genes with novel genetic associations. RESULTS We tested the association between asthma and 17,190 genes that were found to have cis- and/or trans-eQTLs across 16 published eQTL studies. At an empirical FDR of 5%, 48 genes were associated with asthma risk. Of these, for 37, the association was driven by eQTLs located in established risk loci for allergic disease, including 6 genes not previously implicated in disease cause (eg, LIMS1, TINF2, and SAFB). The remaining 11 significant genes represent potential novel genetic associations with asthma. The association with 4 of these replicated in an independent GWAS: B4GALT3, USMG5, P2RY13, and P2RY14, which are genes involved in nucleotide synthesis or nucleotide-dependent cell activation. In mouse studies, P2ry13 and P2ry14-purinergic receptors activated by adenosine 5-diphosphate and UDP-sugars, respectively-were upregulated after allergen challenge, notably in airway epithelial cells, eosinophils, and neutrophils. Intranasal exposure with receptor agonists induced the release of IL-33 and subsequent eosinophil infiltration into the lungs. CONCLUSION We identified novel associations between asthma and eQTLs for 4 genes related to nucleotide synthesis/signaling and demonstrated the power of gene-based analyses of GWASs.
Collapse
Affiliation(s)
| | - Rick Jansen
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije University Amsterdam, Amsterdam, The Netherlands
| | - Brenda Penninx
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Lisa M Bain
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Joana A Revez
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Melanie C Matheson
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Jennie Hui
- PathWest Laboratory Medicine of Western Australia, Nedlands, Australia; School of Population Health, University of Western Australia, Nedlands, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Australia; Busselton Population Medical Research Foundation, Sir Charles Gairdner Hospital, Nedlands, Australia
| | | | - Svetlana Baltic
- Institute for Respiratory Health, Harry Perkins Institute of Medical Research, Nedlands, Australia
| | - Peter Le Souëf
- School of Paediatrics and Child Health, Princess Margaret Hospital for Children, Subiaco, Australia
| | | | | | - Colin F Robertson
- Respiratory Medicine, Murdoch Children's Research Institute, Melbourne, Australia
| | - Alan James
- Busselton Population Medical Research Foundation, Sir Charles Gairdner Hospital, Nedlands, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia; Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Nedlands, Australia
| | - Philip J Thompson
- Institute for Respiratory Health, Harry Perkins Institute of Medical Research, Nedlands, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije University Amsterdam, Amsterdam, The Netherlands
| | - John L Hopper
- Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | | | - Rhiannon B Werder
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Simon Phipps
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
28
|
Sesma JI, Weitzer CD, Livraghi-Butrico A, Dang H, Donaldson S, Alexis NE, Jacobson KA, Harden TK, Lazarowski ER. UDP-glucose promotes neutrophil recruitment in the lung. Purinergic Signal 2016; 12:627-635. [PMID: 27421735 DOI: 10.1007/s11302-016-9524-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/05/2016] [Indexed: 10/21/2022] Open
Abstract
In addition to their role in glycosylation reactions, UDP-sugars are released from cells and activate widely distributed cell surface P2Y14 receptors (P2Y14R). However, the physiological/pathophysiological consequences of UDP-sugar release are incompletely defined. Here, we report that UDP-glucose levels are abnormally elevated in lung secretions from patients with cystic fibrosis (CF) as well as in a mouse model of CF-like disease, the βENaC transgenic (Tg) mouse. Instillation of UDP-glucose into wild-type mouse tracheas resulted in enhanced neutrophil lung recruitment, and this effect was nearly abolished when UDP-glucose was co-instilled with the P2Y14R antagonist PPTN [4-(piperidin-4-yl)-phenyl)-7-(4-(trifluoromethyl)-phenyl-2-naphthoic acid]. Importantly, administration of PPTN to βENaC-Tg mice reduced neutrophil lung inflammation. These results suggest that UDP-glucose released into the airways acts as a local mediator of neutrophil inflammation.
Collapse
Affiliation(s)
- Juliana I Sesma
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina School of Medicine, 6007 Thurston-Bowles Building, CB 7248, Chapel Hill, NC, 27599-7248, USA
| | - Clarissa D Weitzer
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Alessandra Livraghi-Butrico
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina School of Medicine, 6007 Thurston-Bowles Building, CB 7248, Chapel Hill, NC, 27599-7248, USA
| | - Hong Dang
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina School of Medicine, 6007 Thurston-Bowles Building, CB 7248, Chapel Hill, NC, 27599-7248, USA
| | - Scott Donaldson
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina School of Medicine, 6007 Thurston-Bowles Building, CB 7248, Chapel Hill, NC, 27599-7248, USA
| | - Neil E Alexis
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kenneth A Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - T Kendall Harden
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Eduardo R Lazarowski
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina School of Medicine, 6007 Thurston-Bowles Building, CB 7248, Chapel Hill, NC, 27599-7248, USA.
| |
Collapse
|
29
|
Upregulation of TMEM16A Protein in Bronchial Epithelial Cells by Bacterial Pyocyanin. PLoS One 2015; 10:e0131775. [PMID: 26121472 PMCID: PMC4486680 DOI: 10.1371/journal.pone.0131775] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 06/06/2015] [Indexed: 01/22/2023] Open
Abstract
Induction of mucus hypersecretion in the airway epithelium by Th2 cytokines is associated with the expression of TMEM16A, a Ca2+-activated Cl- channel. We asked whether exposure of airway epithelial cells to bacterial components, a condition that mimics the highly infected environment occurring in cystic fibrosis (CF), also results in a similar response. In cultured human bronchial epithelial cells, treatment with pyocyanin or with a P. aeruginosa culture supernatant caused a significant increase in TMEM16A function. The Ca2+-dependent Cl- secretion, triggered by stimulation with UTP, was particularly enhanced by pyocyanin in cells from CF patients. Increased expression of TMEM16A protein and of MUC5AC mucin by bacterial components was demonstrated by immunofluorescence in CF and non-CF cells. We also investigated TMEM16A expression in human bronchi by immunocytochemistry. We found increased TMEM16A staining in the airways of CF patients. The strongest signal was observed in CF submucosal glands. Our results suggest that TMEM16A expression/function is upregulated in CF lung disease, possibly as a response towards the presence of bacteria in the airways.
Collapse
|
30
|
Lazarowski ER, Harden TK. UDP-Sugars as Extracellular Signaling Molecules: Cellular and Physiologic Consequences of P2Y14 Receptor Activation. Mol Pharmacol 2015; 88:151-60. [PMID: 25829059 DOI: 10.1124/mol.115.098756] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 03/31/2015] [Indexed: 12/15/2022] Open
Abstract
UDP-sugars, which are indispensable for protein glycosylation reactions in cellular secretory pathways, also act as important extracellular signaling molecules. We discuss here the broadly expressed P2Y14 receptor, a G-protein-coupled receptor targeted by UDP sugars, and the increasingly diverse set of physiologic responses discovered recently functioning downstream of this receptor in many epithelia as well as in immune, inflammatory, and other cells.
Collapse
Affiliation(s)
- Eduardo R Lazarowski
- Departments of Medicine (E.R.L.) and Pharmacology (T.K.H.), University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - T Kendall Harden
- Departments of Medicine (E.R.L.) and Pharmacology (T.K.H.), University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
31
|
Azroyan A, Cortez-Retamozo V, Bouley R, Liberman R, Ruan YC, Kiselev E, Jacobson KA, Pittet MJ, Brown D, Breton S. Renal intercalated cells sense and mediate inflammation via the P2Y14 receptor. PLoS One 2015; 10:e0121419. [PMID: 25799465 PMCID: PMC4370445 DOI: 10.1371/journal.pone.0121419] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/01/2015] [Indexed: 12/31/2022] Open
Abstract
Uncontrolled inflammation is one of the leading causes of kidney failure. Pro-inflammatory responses can occur in the absence of infection, a process called sterile inflammation. Here we show that the purinergic receptor P2Y14 (GPR105) is specifically and highly expressed in collecting duct intercalated cells (ICs) and mediates sterile inflammation in the kidney. P2Y14 is activated by UDP-glucose, a damage-associated molecular pattern molecule (DAMP) released by injured cells. We found that UDP-glucose increases pro-inflammatory chemokine expression in ICs as well as MDCK-C11 cells, and UDP-glucose activates the MEK1/2-ERK1/2 pathway in MDCK-C11 cells. These effects were prevented following inhibition of P2Y14 with the small molecule PPTN. Tail vein injection of mice with UDP-glucose induced the recruitment of neutrophils to the renal medulla. This study identifies ICs as novel sensors, mediators and effectors of inflammation in the kidney via P2Y14.
Collapse
Affiliation(s)
- Anie Azroyan
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Virna Cortez-Retamozo
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Richard Bouley
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rachel Liberman
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ye Chun Ruan
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Evgeny Kiselev
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kenneth A. Jacobson
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Mikael J. Pittet
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Dennis Brown
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sylvie Breton
- Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- Program in Membrane Biology/Nephrology Division, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
32
|
Takahara N, Ito S, Furuya K, Naruse K, Aso H, Kondo M, Sokabe M, Hasegawa Y. Real-time imaging of ATP release induced by mechanical stretch in human airway smooth muscle cells. Am J Respir Cell Mol Biol 2015; 51:772-82. [PMID: 24885163 DOI: 10.1165/rcmb.2014-0008oc] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway smooth muscle (ASM) cells within the airway walls are continually exposed to mechanical stimuli, and exhibit various functions in response to these mechanical stresses. ATP acts as an extracellular mediator in the airway. Moreover, extracellular ATP is considered to play an important role in the pathophysiology of asthma and chronic obstructive pulmonary disease. However, it is not known whether ASM cells are cellular sources of ATP secretion in the airway. We therefore investigated whether mechanical stretch induces ATP release from ASM cells. Mechanical stretch was applied to primary human ASM cells cultured on a silicone chamber coated with type I collagen using a stretching apparatus. Concentrations of ATP in cell culture supernatants measured by luciferin-luciferase bioluminescence were significantly elevated by cyclic stretch (12 and 20% strain). We further visualized the stretch-induced ATP release from the cells in real time using a luminescence imaging system, while acquiring differential interference contrast cell images with infrared optics. Immediately after a single uniaxial stretch for 1 second, strong ATP signals were produced by a certain population of cells and spread to surrounding spaces. The cyclic stretch-induced ATP release was significantly reduced by inhibitors of Ca(2+)-dependent vesicular exocytosis, 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetraacetoxymethyl ester, monensin, N-ethylmaleimide, and bafilomycin. In contrast, the stretch-induced ATP release was not inhibited by a hemichannel blocker, carbenoxolone, or blockade of transient receptor potential vanilloid 4 by short interfering RNA transfection or ruthenium red. These findings reveal a novel property of ASM cells: mechanically induced ATP release may be a cellular source of ATP in the airway.
Collapse
|
33
|
Dynamic changes in the Streptococcus pneumoniae transcriptome during transition from biofilm formation to invasive disease upon influenza A virus infection. Infect Immun 2014; 82:4607-19. [PMID: 25135685 DOI: 10.1128/iai.02225-14] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae is a leading cause of infectious disease globally. Nasopharyngeal colonization occurs in biofilms and precedes infection. Prior studies have indicated that biofilm-derived pneumococci are avirulent. However, influenza A virus (IAV) infection releases virulent pneumococci from biofilms in vitro and in vivo. Triggers of dispersal include IAV-induced changes in the nasopharynx, such as increased temperature (fever) and extracellular ATP (tissue damage). We used whole-transcriptome shotgun sequencing (RNA-seq) to compare the S. pneumoniae transcriptome in biofilms, bacteria dispersed from biofilms after exposure to IAV, febrile-range temperature, or ATP, and planktonic cells grown at 37°C. Compared with biofilm bacteria, actively dispersed S. pneumoniae, which were more virulent in invasive disease, upregulated genes involved in carbohydrate metabolism. Enzymatic assays for ATP and lactate production confirmed that dispersed pneumococci exhibited increased metabolism compared to those in biofilms. Dispersed pneumococci also upregulated genes associated with production of bacteriocins and downregulated colonization-associated genes related to competence, fratricide, and the transparent colony phenotype. IAV had the largest impact on the pneumococcal transcriptome. Similar transcriptional differences were also observed when actively dispersed bacteria were compared with avirulent planktonic bacteria. Our data demonstrate complex changes in the pneumococcal transcriptome in response to IAV-induced changes in the environment. Our data suggest that disease is caused by pneumococci that are primed to move to tissue sites with altered nutrient availability and to protect themselves from the nasopharyngeal microflora and host immune response. These data help explain pneumococcal virulence after IAV infection and have important implications for studies of S. pneumoniae pathogenesis.
Collapse
|
34
|
Conner GE, Ivonnet P, Gelin M, Whitney P, Salathe M. H2O2 stimulates cystic fibrosis transmembrane conductance regulator through an autocrine prostaglandin pathway, using multidrug-resistant protein-4. Am J Respir Cell Mol Biol 2014; 49:672-9. [PMID: 23742099 DOI: 10.1165/rcmb.2013-0156oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) activity is essential for the maintenance of airway surface liquid depth, and therefore mucociliary clearance. Reactive oxygen species, increased during inflammatory airway diseases, alter CFTR activity. Here, H2O2 levels in the surface liquid of normal human bronchial epithelial cultures differentiated at the air-liquid interface were estimated, and H2O2-mediated changes in CFTR activity were examined. In Ussing chambers, H2O2-induced anion currents were sensitive to the CFTR inhibitors CFTRinh172 and GlyH-101. These currents were absent in cells from patients with cystic fibrosis. Responses to greater than 500 μM H2O2 were transient. Cyclooxygenase inhibitors blocked the H2O2 response, as did EP1 and EP4 receptor antagonists. A multidrug-resistant protein (MRP) inhibitor and short hairpin RNA directed against MRP4 blocked H2O2 responses. EP1 and EP4 agonists mimicked H2O2 in both control and MRP4 knockdown cells. Thus, H2O2 activates the synthesis, export, and binding of prostanoids via EP4 and, interestingly, EP1 receptors in normal, differentiated human airway epithelial cells to activate cyclic adenosine monophosphate pathways that in turn activate CFTR channels in the apical membrane.
Collapse
Affiliation(s)
- Gregory E Conner
- 1 Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, and
| | | | | | | | | |
Collapse
|
35
|
Okada SF, Ribeiro CMP, Sesma JI, Seminario-Vidal L, Abdullah LH, van Heusden C, Lazarowski ER, Boucher RC. Inflammation promotes airway epithelial ATP release via calcium-dependent vesicular pathways. Am J Respir Cell Mol Biol 2013; 49:814-20. [PMID: 23763446 DOI: 10.1165/rcmb.2012-0493oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
ATP in airway surface liquid (ASL) controls mucociliary clearance functions via the activation of airway epithelial purinergic receptors. However, abnormally elevated ATP levels have been reported in inflamed airways, suggesting that excessive ATP in ASL contributes to airway inflammation. Despite these observations, little is known about the mechanisms of ATP accumulation in the ASL covering inflamed airways. In this study, links between cystic fibrosis (CF)-associated airway inflammation and airway epithelial ATP release were investigated. Primary human bronchial epithelial (HBE) cells isolated from CF lungs exhibited enhanced IL-8 secretion after 6 to 11 days, but not 28 to 35 days, in culture, compared with normal HBE cells. Hypotonic cell swelling-promoted ATP release was increased in 6- to 11-day-old CF HBE cells compared with non-CF HBE cells, but returned to normal values after 28 to 35 days in culture. The exposure of non-CF HBE cells to airway secretions isolated from CF lungs, namely, sterile supernatants of mucopurulent material (SMM), also caused enhanced IL-8 secretion and increased ATP release. The SMM-induced increase in ATP release was sensitive to Ca(2+) chelation and vesicle trafficking/exocytosis inhibitors, but not to pannexin inhibition. Transcript levels of the vesicular nucleotide transporter, but not pannexin 1, were up-regulated after SMM exposure. SMM-treated cultures displayed increased basal mucin secretion, but mucin secretion was not enhanced in response to hypotonic challenge after the exposure of cells to either vehicle or SMM. We propose that CF airway inflammation up-regulates the capacity of airway epithelia to release ATP via Ca(2+)-dependent vesicular mechanisms not associated with mucin granule secretion.
Collapse
Affiliation(s)
- Seiko F Okada
- 1 Cystic Fibrosis/Pulmonary Research and Treatment Center, Department of Medicine, and
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Miklavc P, Thompson KE, Frick M. A new role for P2X4 receptors as modulators of lung surfactant secretion. Front Cell Neurosci 2013; 7:171. [PMID: 24115920 PMCID: PMC3792447 DOI: 10.3389/fncel.2013.00171] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/12/2013] [Indexed: 12/17/2022] Open
Abstract
In recent years, P2X receptors have attracted increasing attention as regulators of exocytosis and cellular secretion. In various cell types, P2X receptors have been found to stimulate vesicle exocytosis directly via Ca(2+) influx and elevation of the intracellular Ca(2+) concentration. Recently, a new role for P2X4 receptors as regulators of secretion emerged. Exocytosis of lamellar bodies (LBs), large storage organelles for lung surfactant, results in a local, fusion-activated Ca(2+) entry (FACE) in alveolar type II epithelial cells. FACE is mediated via P2X4 receptors that are located on the limiting membrane of LBs and inserted into the plasma membrane upon exocytosis of LBs. The localized Ca(2+) influx at the site of vesicle fusion promotes fusion pore expansion and facilitates surfactant release. In addition, this inward-rectifying cation current across P2X4 receptors mediates fluid resorption from lung alveoli. It is hypothesized that the concomitant reduction in the alveolar lining fluid facilitates insertion of surfactant into the air-liquid interphase thereby "activating" it. These findings constitute a novel role for P2X4 receptors in regulating vesicle content secretion as modulators of the secretory output during the exocytic post-fusion phase.
Collapse
Affiliation(s)
- Pika Miklavc
- Institute of General Physiology, University of Ulm Ulm, Germany
| | | | | |
Collapse
|
37
|
Roflumilast inhibits respiratory syncytial virus infection in human differentiated bronchial epithelial cells. PLoS One 2013; 8:e69670. [PMID: 23936072 PMCID: PMC3720563 DOI: 10.1371/journal.pone.0069670] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/11/2013] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes acute exacerbations in COPD and asthma. RSV infects bronchial epithelial cells (HBE) that trigger RSV associated lung pathology. This study explores whether the phosphodiesterase 4 (PDE4) inhibitor Roflumilast N-oxide (RNO), alters RSV infection of well-differentiated HBE (WD-HBE) in vitro. WD-HBE were RSV infected in the presence or absence of RNO (0.1-100 nM). Viral infection (staining of F and G proteins, nucleoprotein RNA level), mRNA of ICAM-1, ciliated cell markers (digital high speed videomicroscopy, β-tubulin immunofluorescence, Foxj1 and Dnai2 mRNA), Goblet cells (PAS), mRNA of MUC5AC and CLCA1, mRNA and protein level of IL-13, IL-6, IL-8, TNFα, formation of H2O2 and the anti-oxidative armamentarium (mRNA of Nrf2, HO-1, GPx; total antioxidant capacity (TAC) were measured at day 10 or 15 post infection. RNO inhibited RSV infection of WD-HBE, prevented the loss of ciliated cells and markers, reduced the increase of MUC5AC and CLCA1 and inhibited the increase of IL-13, IL-6, IL-8, TNFα and ICAM-1. Additionally RNO reversed the reduction of Nrf2, HO-1 and GPx mRNA levels and consequently restored the TAC and reduced the H2O2 formation. RNO inhibits RSV infection of WD-HBE cultures and mitigates the cytopathological changes associated to this virus.
Collapse
|
38
|
O'Grady SM, Patil N, Melkamu T, Maniak PJ, Lancto C, Kita H. ATP release and Ca2+ signalling by human bronchial epithelial cells following Alternaria aeroallergen exposure. J Physiol 2013; 591:4595-609. [PMID: 23858006 DOI: 10.1113/jphysiol.2013.254649] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Exposure of human bronchial epithelial (HBE) cells from normal and asthmatic subjects to extracts from Alternaria alternata evoked a rapid and sustained release of ATP with greater efficacy observed in epithelial cells from asthmatic patients. Previously, Alternaria allergens were shown to produce a sustained increase in intracellular Ca2+ concentration ([Ca2+]i) that was dependent on the coordinated activation of specific purinergic receptor (P2Y2 and P2X7) subtypes. In the present study, pretreatment with a cell-permeable Ca2+-chelating compound (BAPTA-AM) significantly inhibited ATP release, indicating dependency on [Ca2+]i. Alternaria-evoked ATP release exhibited a greater peak response and a slightly lower EC50 value in cells obtained from asthmatic donors compared to normal control cells. Furthermore, the maximum increase in [Ca2+]i resulting from Alternaria treatment was greater in cells from asthmatic patients compared to normal subjects. The vesicle transport inhibitor brefeldin A and BAPTA-AM significantly blocked Alternaria-stimulated incorporation of fluorescent lipid (FM1-43)-labelled vesicles into the plasma membrane and ATP release. In addition, inhibiting uptake of ATP into exocytotic vesicles with bafilomycin also reduced ATP release comparable to the effects of brefeldin A and BAPTA-AM. These results indicate that an important mechanism for Alternaria-induced ATP release is Ca2+ dependent and involves exocytosis of ATP. Serine and cysteine protease inhibitors also reduced Alternaria-induced ATP release; however, the sustained increase in [Ca2+]i typically observed following Alternaria exposure appeared to be independent of protease-activated receptor (PAR2) stimulation.
Collapse
Affiliation(s)
- Scott M O'Grady
- S. M. O'Grady: Department of Animal Science, University of Minnesota, 480 Haecker Hall, 1364 Eckles Avenue, St Paul, MN 55108, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Barrett MO, Sesma JI, Ball CB, Jayasekara PS, Jacobson KA, Lazarowski ER, Harden TK. A selective high-affinity antagonist of the P2Y14 receptor inhibits UDP-glucose-stimulated chemotaxis of human neutrophils. Mol Pharmacol 2013; 84:41-9. [PMID: 23592514 PMCID: PMC3684828 DOI: 10.1124/mol.113.085654] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 04/12/2013] [Indexed: 12/31/2022] Open
Abstract
The nucleotide-sugar-activated P2Y14 receptor (P2Y14-R) is highly expressed in hematopoietic cells. Although the physiologic functions of this receptor remain undefined, it has been strongly implicated recently in immune and inflammatory responses. Lack of availability of receptor-selective high-affinity antagonists has impeded progress in studies of this and most of the eight nucleotide-activated P2Y receptors. A series of molecules recently were identified by Gauthier et al. (Gauthier et al., 2011) that exhibited antagonist activity at the P2Y14-R. We synthesized one of these molecules, a 4,7-disubstituted 2-naphthoic acid derivative (PPTN), and studied its pharmacological properties in detail. The concentration-effect curve of UDP-glucose for promoting inhibition of adenylyl cyclase in C6 glioma cells stably expressing the P2Y14-R was shifted to the right in a concentration-dependent manner by PPTN. Schild analyses revealed that PPTN-mediated inhibition followed competitive kinetics, with a KB of 434 pM observed. In contrast, 1 μM PPTN exhibited no agonist or antagonist effect at the P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, or P2Y13 receptors. UDP-glucose-promoted chemotaxis of differentiated HL-60 human promyelocytic leukemia cells was blocked by PPTN with a concentration dependence consistent with the KB determined with recombinant P2Y14-R. In contrast, the chemotactic response evoked by the chemoattractant peptide fMetLeuPhe was unaffected by PPTN. UDP-glucose-promoted chemotaxis of freshly isolated human neutrophils also was blocked by PPTN. In summary, this work establishes PPTN as a highly selective high-affinity antagonist of the P2Y14-R that is useful for interrogating the action of this receptor in physiologic systems.
Collapse
Affiliation(s)
- Matthew O Barrett
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Mitrovic S, Nogueira C, Cantero-Recasens G, Kiefer K, Fernández-Fernández JM, Popoff JF, Casano L, Bard FA, Gomez R, Valverde MA, Malhotra V. TRPM5-mediated calcium uptake regulates mucin secretion from human colon goblet cells. eLife 2013; 2:e00658. [PMID: 23741618 PMCID: PMC3667631 DOI: 10.7554/elife.00658] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/23/2013] [Indexed: 12/23/2022] Open
Abstract
Mucin 5AC (MUC5AC) is secreted by goblet cells of the respiratory tract and, surprisingly, also expressed de novo in mucus secreting cancer lines. siRNA-mediated knockdown of 7343 human gene products in a human colonic cancer goblet cell line (HT29-18N2) revealed new proteins, including a Ca(2+)-activated channel TRPM5, for MUC5AC secretion. TRPM5 was required for PMA and ATP-induced secretion of MUC5AC from the post-Golgi secretory granules. Stable knockdown of TRPM5 reduced a TRPM5-like current and ATP-mediated Ca(2+) signal. ATP-induced MUC5AC secretion depended strongly on Ca(2+) influx, which was markedly reduced in TRPM5 knockdown cells. The difference in ATP-induced Ca(2+) entry between control and TRPM5 knockdown cells was abrogated in the absence of extracellular Ca(2+) and by inhibition of the Na(+)/Ca(2+) exchanger (NCX). Accordingly, MUC5AC secretion was reduced by inhibition of NCX. Thus TRPM5 activation by ATP couples TRPM5-mediated Na(+) entry to promote Ca(2+) uptake via an NCX to trigger MUC5AC secretion. DOI:http://dx.doi.org/10.7554/eLife.00658.001.
Collapse
Affiliation(s)
- Sandra Mitrovic
- Department of Cell and Developmental Biology , Centre for Genomic Regulation , Barcelona , Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sesma JI, Kreda SM, Okada SF, van Heusden C, Moussa L, Jones LC, O'Neal WK, Togawa N, Hiasa M, Moriyama Y, Lazarowski ER. Vesicular nucleotide transporter regulates the nucleotide content in airway epithelial mucin granules. Am J Physiol Cell Physiol 2013; 304:C976-84. [PMID: 23467297 DOI: 10.1152/ajpcell.00371.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Nucleotides within the airway surface liquid promote fluid secretion via activation of airway epithelial purinergic receptors. ATP is stored within and released from mucin granules as co-cargo with mucins, but the mechanism by which ATP, and potentially other nucleotides, enter the lumen of mucin granules is not known. We assessed the contribution of the recently identified SLC17A9 vesicle nucleotide transporter (VNUT) to the nucleotide availability within isolated mucin granules and further examined the involvement of VNUT in mucin granule secretion-associated nucleotide release. RT-PCR and Western blot analyses indicated that VNUT is abundantly expressed in airway epithelial goblet-like Calu-3 cells, migrating as a duplex with apparent mobility of 55 and 60 kDa. Subcellular fractionation studies indicated that VNUT55 was associated with high-density mucin granules, whereas VNUT60 was associated with low-density organelles. Immunofluorescence studies showed that recombinant VNUT localized to mucin granules and other organelles. Mucin granules isolated from VNUT short hairpin RNA-expressing cells exhibited a marked reduction of ATP, ADP, AMP, and UTP levels within granules. Ca(2+)-regulated vesicular ATP release was markedly reduced in these cells, but mucin secretion was not affected. These results suggest that VNUT is the relevant nucleotide transporter responsible for the uptake of cytosolic nucleotides into mucin granules. By controlling the entry of nucleotides into mucin granules, VNUT contributes to the release of purinergic signaling molecules necessary for the proper hydration of co-released mucins.
Collapse
Affiliation(s)
- Juliana I Sesma
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Epithelial cell culture from human adenoids: a functional study model for ciliated and secretory cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:478713. [PMID: 23484122 PMCID: PMC3581098 DOI: 10.1155/2013/478713] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/16/2012] [Accepted: 11/19/2012] [Indexed: 11/23/2022]
Abstract
Background. Mucociliary transport (MCT) is a defense mechanism of the airway. To study the underlying mechanisms of MCT, we have both developed an experimental model of cultures, from human adenoid tissue of ciliated and secretory cells, and characterized the response to local chemical signals that control ciliary activity and the secretion of respiratory mucins in vitro. Materials and Methods. In ciliated cell cultures, ciliary beat frequency (CBF) and intracellular Ca2+ levels were measured in response to ATP, UTP, and adenosine. In secretory cultures, mucin synthesis and secretion were identified by using immunodetection. Mucin content was taken from conditioned medium and analyzed in the presence or absence of UTP. Results. Enriched ciliated cell monolayers and secretory cells were obtained. Ciliated cells showed a basal CBF of 10.7 Hz that increased significantly after exposure to ATP, UTP, or adenosine. Mature secretory cells showed active secretion of granules containing different glycoproteins, including MUC5AC. Conclusion. Culture of ciliated and secretory cells grown from adenoid epithelium is a reproducible and feasible experimental model, in which it is possible to observe ciliary and secretory activities, with a potential use as a model to understand mucociliary transport control mechanisms.
Collapse
|
43
|
Kusu T, Kayama H, Kinoshita M, Jeon SG, Ueda Y, Goto Y, Okumura R, Saiga H, Kurakawa T, Ikeda K, Maeda Y, Nishimura JI, Arima Y, Atarashi K, Honda K, Murakami M, Kunisawa J, Kiyono H, Okumura M, Yamamoto M, Takeda K. Ecto-nucleoside triphosphate diphosphohydrolase 7 controls Th17 cell responses through regulation of luminal ATP in the small intestine. THE JOURNAL OF IMMUNOLOGY 2012; 190:774-83. [PMID: 23241884 DOI: 10.4049/jimmunol.1103067] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Extracellular ATP is released from live cells in controlled conditions, as well as dying cells in inflammatory conditions, and, thereby, regulates T cell responses, including Th17 cell induction. The level of extracellular ATP is closely regulated by ATP hydrolyzing enzymes, such as ecto-nucleoside triphosphate diphosphohydrolases (ENTPDases). ENTPDase1/CD39, which is expressed in immune cells, was shown to regulate immune responses by downregulating the ATP level. In this study, we analyzed the immunomodulatory function of ENTPDase7, which is preferentially expressed in epithelial cells in the small intestine. The targeted deletion of Entpd7 encoding ENTPDase7 in mice resulted in increased ATP levels in the small intestinal lumen. The number of Th17 cells was selectively increased in the small intestinal lamina propria in Entpd7(-/-) mice. Th17 cells were decreased by oral administration of antibiotics or the ATP antagonist in Entpd7(-/-) mice, indicating that commensal microbiota-dependent ATP release mediates the enhanced Th17 cell development in the small intestinal lamina propria of Entpd7(-/-) mice. In accordance with the increased number of small intestinal Th17 cells, Entpd7(-/-) mice were resistant to oral infection with Citrobacter rodentium. Entpd7(-/-) mice suffered from severe experimental autoimmune encephalomyelitis, which was associated with increased numbers of CD4(+) T cells producing both IL-17 and IFN-γ. Taken together, these findings demonstrate that ENTPDase7 controls the luminal ATP level and, thereby, regulates Th17 cell development in the small intestine.
Collapse
Affiliation(s)
- Takashi Kusu
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mata M, Sarrion I, Armengot M, Carda C, Martinez I, Melero JA, Cortijo J. Respiratory syncytial virus inhibits ciliagenesis in differentiated normal human bronchial epithelial cells: effectiveness of N-acetylcysteine. PLoS One 2012; 7:e48037. [PMID: 23118923 PMCID: PMC3485262 DOI: 10.1371/journal.pone.0048037] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 09/20/2012] [Indexed: 01/23/2023] Open
Abstract
Persistent respiratory syncytial virus (RSV) infections have been associated with the exacerbation of chronic inflammatory diseases, including chronic obstructive pulmonary disease (COPD). This virus infects the respiratory epithelium, leading to chronic inflammation, and induces the release of mucins and the loss of cilia activity, two factors that determine mucus clearance and the increase in sputum volume. These alterations involve reactive oxygen species-dependent mechanisms. The antioxidant N-acetylcysteine (NAC) has proven useful in the management of COPD, reducing symptoms, exacerbations, and accelerated lung function decline. NAC inhibits RSV infection and mucin release in human A549 cells. The main objective of this study was to analyze the effects of NAC in modulating ciliary activity, ciliagenesis, and metaplasia in primary normal human bronchial epithelial cell (NHBEC) cultures infected with RSV. Our results indicated that RSV induced ultrastructural abnormalities in axonemal basal bodies and decreased the expression of β-tubulin as well as two genes involved in ciliagenesis, FOXJ1 and DNAI2. These alterations led to a decrease in ciliary activity. Furthermore, RSV induced metaplastic changes to the epithelium and increased the number of goblet cells and the expression of MUC5AC and GOB5. NAC restored the normal functions of the epithelium, inhibiting ICAM1 expression, subsequent RSV infection through mechanisms involving nuclear receptor factor 2, and the expression of heme oxygenase 1, which correlated with the restoration of the antioxidant capacity, the intracellular H(2)O(2) levels and glutathione content of NHBECs. The results presented in this study support the therapeutic use of NAC for the management of chronic respiratory diseases, including COPD.
Collapse
Affiliation(s)
- Manuel Mata
- Research Foundation of the University General Hospital of Valencia, Valencia, Spain.
| | | | | | | | | | | | | |
Collapse
|
45
|
Burnstock G, Brouns I, Adriaensen D, Timmermans JP. Purinergic signaling in the airways. Pharmacol Rev 2012; 64:834-68. [PMID: 22885703 DOI: 10.1124/pr.111.005389] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Evidence for a significant role and impact of purinergic signaling in normal and diseased airways is now beyond dispute. The present review intends to provide the current state of knowledge of the involvement of purinergic pathways in the upper and lower airways and lungs, thereby differentiating the involvement of different tissues, such as the epithelial lining, immune cells, airway smooth muscle, vasculature, peripheral and central innervation, and neuroendocrine system. In addition to the vast number of well illustrated functions for purinergic signaling in the healthy respiratory tract, increasing data pointing to enhanced levels of ATP and/or adenosine in airway secretions of patients with airway damage and respiratory diseases corroborates the emerging view that purines act as clinically important mediators resulting in either proinflammatory or protective responses. Purinergic signaling has been implicated in lung injury and in the pathogenesis of a wide range of respiratory disorders and diseases, including asthma, chronic obstructive pulmonary disease, inflammation, cystic fibrosis, lung cancer, and pulmonary hypertension. These ostensibly enigmatic actions are based on widely different mechanisms, which are influenced by the cellular microenvironment, but especially the subtypes of purine receptors involved and the activity of distinct members of the ectonucleotidase family, the latter being potential protein targets for therapeutic implementation.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Royal Free Campus, London, UK.
| | | | | | | |
Collapse
|
46
|
Sesma JI, Kreda SM, Steinckwich-Besancon N, Dang H, García-Mata R, Harden TK, Lazarowski ER. The UDP-sugar-sensing P2Y(14) receptor promotes Rho-mediated signaling and chemotaxis in human neutrophils. Am J Physiol Cell Physiol 2012; 303:C490-8. [PMID: 22673622 DOI: 10.1152/ajpcell.00138.2012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The G(i)-coupled P2Y(14) receptor (P2Y(14)-R) is potently activated by UDP-sugars and UDP. Although P2Y(14)-R mRNA is prominently expressed in circulating neutrophils, the signaling pathways and functional responses associated with this receptor are undefined. In this study, we illustrate that incubation of isolated human neutrophils with UDP-glucose resulted in cytoskeleton rearrangement, change of cell shape, and enhanced cell migration. We also demonstrate that UDP-glucose promotes rapid, robust, and concentration-dependent activation of RhoA in these cells. Ecto-nucleotidases expressed on neutrophils rapidly hydrolyzed extracellular ATP, but incubation with UDP-glucose for up to 1 h resulted in negligible metabolism of the nucleotide-sugar. HL60 human promyelocytic leukemia cells do not express the P2Y(14)-R, but neutrophil differentiation of HL60 cells with DMSO resulted in markedly enhanced P2Y(14)-R expression. Accordingly, UDP-glucose, UDP-galactose, and UDP-N-acetylglucosamine promoted Rho activation in differentiated but not in undifferentiated HL60 cells. Stable expression of recombinant human P2Y(14)-R conferred UDP-sugar-promoted responses to undifferentiated HL60 cells. UDP-glucose-promoted RhoA activation also was accompanied by enhanced cell migration in differentiated HL60 cells, and these responses were blocked by Rho kinase inhibitors. These results support the notion that UDP-glucose is a stable and potent proinflammatory mediator that promotes P2Y(14)-R-mediated neutrophil motility via Rho/Rho kinase activation.
Collapse
Affiliation(s)
- Juliana I Sesma
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599-7248, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Lazarowski ER. Vesicular and conductive mechanisms of nucleotide release. Purinergic Signal 2012; 8:359-73. [PMID: 22528679 DOI: 10.1007/s11302-012-9304-9] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 01/21/2012] [Indexed: 12/22/2022] Open
Abstract
Extracellular nucleotides and nucleosides promote a vast range of physiological responses, via activation of cell surface purinergic receptors. Virtually all tissues and cell types exhibit regulated release of ATP, which, in many cases, is accompanied by the release of uridine nucleotides. Given the relevance of extracellular nucleotide/nucleoside-evoked responses, understanding how ATP and other nucleotides are released from cells is an important physiological question. By facilitating the entry of cytosolic nucleotides into the secretory pathway, recently identified vesicular nucleotide and nucleotide-sugar transporters contribute to the exocytotic release of ATP and UDP-sugars not only from endocrine/exocrine tissues, but also from cell types in which secretory granules have not been biochemically characterized. In addition, plasma membrane connexin hemichannels, pannexin channels, and less-well molecularly defined ATP conducting anion channels have been shown to contribute to the release of ATP (and UTP) under a variety of conditions.
Collapse
Affiliation(s)
- Eduardo R Lazarowski
- School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7248, USA.
| |
Collapse
|
48
|
Segovia J, Sabbah A, Mgbemena V, Tsai SY, Chang TH, Berton MT, Morris IR, Allen IC, Ting JPY, Bose S. TLR2/MyD88/NF-κB pathway, reactive oxygen species, potassium efflux activates NLRP3/ASC inflammasome during respiratory syncytial virus infection. PLoS One 2012; 7:e29695. [PMID: 22295065 PMCID: PMC3266238 DOI: 10.1371/journal.pone.0029695] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 12/02/2011] [Indexed: 12/02/2022] Open
Abstract
Human respiratory syncytial virus (RSV) constitute highly pathogenic virus that cause severe respiratory diseases in newborn, children, elderly and immuno-compromised individuals. Airway inflammation is a critical regulator of disease outcome in RSV infected hosts. Although “controlled” inflammation is required for virus clearance, aberrant and exaggerated inflammation during RSV infection results in development of inflammatory diseases like pneumonia and bronchiolitis. Interleukin-1β (IL-1β) plays an important role in inflammation by orchestrating the pro-inflammatory response. IL-1β is synthesized as an immature pro-IL-1β form. It is cleaved by activated caspase-1 to yield mature IL-1β that is secreted extracellularly. Activation of caspase-1 is mediated by a multi-protein complex known as the inflammasome. Although RSV infection results in IL-1β release, the mechanism is unknown. Here in, we have characterized the mechanism of IL-1β secretion following RSV infection. Our study revealed that NLRP3/ASC inflammasome activation is crucial for IL-1β production during RSV infection. Further studies illustrated that prior to inflammasome formation; the “first signal” constitutes activation of toll-like receptor-2 (TLR2)/MyD88/NF-κB pathway. TLR2/MyD88/NF-κB signaling is required for pro-IL-1β and NLRP3 gene expression during RSV infection. Following expression of these genes, two “second signals” are essential for triggering inflammasome activation. Intracellular reactive oxygen species (ROS) and potassium (K+) efflux due to stimulation of ATP-sensitive ion channel promote inflammasome activation following RSV infection. Thus, our studies have underscored the requirement of TLR2/MyD88/NF-κB pathway (first signal) and ROS/potassium efflux (second signal) for NLRP3/ASC inflammasome formation, leading to caspase-1 activation and subsequent IL-1β release during RSV infection.
Collapse
Affiliation(s)
- Jesus Segovia
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Ahmed Sabbah
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Victoria Mgbemena
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Su-Yu Tsai
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Te-Hung Chang
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Michael T. Berton
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Ian R. Morris
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Irving C. Allen
- Department of Microbiology-Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Jenny P.-Y. Ting
- Department of Microbiology-Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Santanu Bose
- Department of Microbiology and Immunology, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
49
|
Jones LC, Moussa L, Fulcher ML, Zhu Y, Hudson EJ, O'Neal WK, Randell SH, Lazarowski ER, Boucher RC, Kreda SM. VAMP8 is a vesicle SNARE that regulates mucin secretion in airway goblet cells. J Physiol 2011; 590:545-62. [PMID: 22144578 DOI: 10.1113/jphysiol.2011.222091] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mucin secretion is an innate defence mechanism, which is noxiously upregulated in obstructive lung diseases (e.g. chronic obstructive pulmonary disease (COPD), cystic fibrosis and asthma). Mucin granule exocytosis is regulated by specific protein complexes, but the SNARE exocytotic core has not been defined in airway goblet cells. In this study, we identify VAMP8 as one of the SNAREs regulating mucin granule exocytosis. VAMP8 mRNA was present in human airway and lung epithelial cells, and deep-sequencing and expression analyses of airway epithelial cells revealed that VAMP8 transcripts were expressed at 10 times higher levels than other VAMP mRNAs. In human airway epithelial cell cultures and freshly excised tissues, VAMP8 immunolocalised mainly to goblet cell mucin granules. The function of VAMP8 in airway mucin secretion was tested by RNA interference techniques. Both VAMP8 short interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs) reduced mucin secretion induced by PAR agonists, neutrophil elastase and ATP in two airway epithelial cell culture models. Notably, basal (non-agonist elicited) mucin secretion was also reduced in these experiments. VAMP8 knockdown was also effective in decreasing mucin secretion in airway epithelial cell cultures with induced mucous metaplasia/mucin hypersecretion. Unlike VAMP8 silencing, knockdown of VAMP2 or VAMP3 did not affect mucin secretion. Importantly, in VAMP8 knock-out (KO) mice with IL-13-induced mucous metaplasia, mucin content in the bronchoalveolar lavage (BAL) and ATP-stimulated mucin secretion in the trachea were reduced compared to WT-matched littermates. Our data indicate that VAMP8 is an essential SNARE in airway mucin granule exocytosis. Reduction of VAMP8 activity/expression may provide a novel therapeutic target to ameliorate airway mucus obstruction in lung diseases.
Collapse
Affiliation(s)
- Lisa C Jones
- Cystic Fibrosis/Pulmonary Research and Treatment Centre, The University of North Carolina at Chapel Hill, 4029A Thurston Bowles Building, Chapel Hill, NC 27599-7248, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Airway epithelia are continuously damaged by airborne pollutants, pathogens and allergens, and they rely on intrinsic mechanisms to restore barrier integrity. Epithelial repair is a multi-step process including cell migration into the wounded area, proliferation, differentiation and matrix deposition. Each step requires the secretion of various molecules, including growth factors, integrins and matrix metalloproteinases. Evidence is emerging that purinergic signaling promotes repair in human airway epithelia. An injury induces ATP release, which binds P2Y(2) receptors (P2Y(2)Rs) to initiate protein kinase C (PKC)-dependent oxidative activation of TNFα-converting enzyme (TACE), which then releases the membrane-bound ligands of the epidermal growth factor receptor (EGFR). The P2Y(2)R- and EGFR-dependent signaling cascades converge to induce mediator release, whereas the latter also induces cytoskeletal rearrangement for cell migration and proliferation. Similar roles for purinergic signaling are reported in pulmonary endothelial cells, smooth muscle cells and fibroblasts. In chronic airway diseases, the aberrant regulation of extracellular purines is implicated in the development of airway remodeling by mucus cell metaplasia and hypersecretion, excess collagen deposition, fibrosis and neovascularization. This chapter describes the crosstalk between these signaling cascades and discusses the impact of deregulated purinergic signaling in chronic lung diseases.
Collapse
|