1
|
Wu X, Zhang G, Du X. Cigarette Smoke Extract Induces MUC5AC Expression Through the ROS/ IP3R/Ca 2+ Pathway in Calu-3 Cells. Int J Chron Obstruct Pulmon Dis 2024; 19:1635-1647. [PMID: 39045541 PMCID: PMC11264152 DOI: 10.2147/copd.s469866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/29/2024] [Indexed: 07/25/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is caused by exposure to noxious external particles, air pollution, and the inhalation of cigarette smoke. Airway mucus hypersecretion particularly mucin5AC (MUC5AC), is a crucial pathological feature of COPD and is associated with its initiation and progression. In this study, we aimed to investigate the effects of cigarette smoke extract (CSE) on MUC5AC expression, particularly the mechanisms by which reactive oxygen species (ROS) induce MUC5AC expression. Methods The effects of CSE on the expression of MUC5AC and mucin5B (MUC5B) were investigated in vitro in Calu-3 cells. MUC5AC and MUC5B expression levels were measured using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), immunofluorescence staining, and enzyme-linked immunosorbent assay (ELISA). Total cellular levels of ROS and Ca2+ were determined using DCFH-DA and Fluo-4 AM. Subsequently, the expression levels of IP3R, IRE1α, p-IRE1α and XBP1s were measured by Western blotting. Gene silencing was achieved by using small-interfering RNAs. Results Our findings revealed that exposure to CSE increased MUC5AC levels and upregulated ROS, IP3R/Ca2+ and unfolded protein response (UPR)-associated factors. In addition, knockdown of IP3R using siRNA decreased CSE-induced Ca2+ production, UPR-associated factors, and MUC5AC expression. Furthermore, 10 mM N-acetyl-l-cysteine (NAC) treatment suppressed the effects of CSE, including ROS generation, IP3R/ Ca2+, UPR activation, and MUC5AC overexpression. Conclusion Our results suggest that ROS regulates CSE-induced UPR and MUC5AC overexpression through IP3R/ Ca2+ signaling. Additionally, we identified NAC as a promising therapeutic agent for mitigating CSE-induced MUC5AC overexpression.
Collapse
Affiliation(s)
- Xiaojuan Wu
- Department of Respiratory Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Guoyue Zhang
- Department of Respiratory Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| | - Xianzhi Du
- Department of Respiratory Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People’s Republic of China
| |
Collapse
|
2
|
Oeler KJ, Blackmon RL, Kreda SM, Robinson T, Ghelardini M, Chapman BS, Tracy J, Hill DB, Oldenburg AL. In situ pulmonary mucus hydration assay using rotational and translational diffusion of gold nanorods with polarization-sensitive optical coherence tomography. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:046004. [PMID: 38690122 PMCID: PMC11060333 DOI: 10.1117/1.jbo.29.4.046004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
Significance Assessing the nanostructure of polymer solutions and biofluids is broadly useful for understanding drug delivery and disease progression and for monitoring therapy. Aim Our objective is to quantify bronchial mucus solids concentration (wt. %) during hypertonic saline (HTS) treatment in vitro via nanostructurally constrained diffusion of gold nanorods (GNRs) monitored by polarization-sensitive optical coherence tomography (PS-OCT). Approach Using PS-OCT, we quantified GNR translational (D T ) and rotational (D R ) diffusion coefficients within polyethylene oxide solutions (0 to 3 wt. %) and human bronchial epithelial cell (hBEC) mucus (0 to 6.4 wt. %). Interpolation of D T and D R data is used to develop an assay to quantify mucus concentration. The assay is demonstrated on the mucus layer of an air-liquid interface hBEC culture during HTS treatment. Results In polymer solutions and mucus, D T and D R monotonically decrease with increasing concentration. D R is more sensitive than D T to changes above 1.5 wt. % of mucus and exhibits less intrasample variability. Mucus on HTS-treated hBEC cultures exhibits dynamic mixing from cilia. A region of hard-packed mucus is revealed by D R measurements. Conclusions The extended dynamic range afforded by simultaneous measurement of D T and D R of GNRs using PS-OCT enables resolving concentration of the bronchial mucus layer over a range from healthy to disease in depth and time during HTS treatment in vitro.
Collapse
Affiliation(s)
- Kelsey J. Oeler
- University of North Carolina at Chapel Hill, Department of Biomedical Engineering, Chapel Hill, North Carolina, United States
| | - Richard L. Blackmon
- Elon University, Department of Engineering, Elon, North Carolina, United States
| | - Silvia M. Kreda
- University of North Carolina at Chapel Hill, Marsico Lung Institute/Cystic Fibrosis/Pulmonary Research and Treatment Center, Chapel Hill, North Carolina, United States
| | - Taylor Robinson
- University of North Carolina at Chapel Hill, Department of Physics and Astronomy, Chapel Hill, North Carolina, United States
| | - Melanie Ghelardini
- North Carolina State University, Department of Materials Science and Engineering, Raleigh, North Carolina, United States
| | - Brian S. Chapman
- North Carolina State University, Department of Materials Science and Engineering, Raleigh, North Carolina, United States
| | - Joseph Tracy
- North Carolina State University, Department of Materials Science and Engineering, Raleigh, North Carolina, United States
| | - David B. Hill
- University of North Carolina at Chapel Hill, Department of Biomedical Engineering, Chapel Hill, North Carolina, United States
- University of North Carolina at Chapel Hill, Marsico Lung Institute/Cystic Fibrosis/Pulmonary Research and Treatment Center, Chapel Hill, North Carolina, United States
| | - Amy L. Oldenburg
- University of North Carolina at Chapel Hill, Department of Biomedical Engineering, Chapel Hill, North Carolina, United States
- University of North Carolina at Chapel Hill, Department of Physics and Astronomy, Chapel Hill, North Carolina, United States
| |
Collapse
|
3
|
Abrami M, Biasin A, Tescione F, Tierno D, Dapas B, Carbone A, Grassi G, Conese M, Di Gioia S, Larobina D, Grassi M. Mucus Structure, Viscoelastic Properties, and Composition in Chronic Respiratory Diseases. Int J Mol Sci 2024; 25:1933. [PMID: 38339210 PMCID: PMC10856136 DOI: 10.3390/ijms25031933] [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: 12/31/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
The respiratory mucus, a viscoelastic gel, effectuates a primary line of the airway defense when operated by the mucociliary clearance. In chronic respiratory diseases (CRDs), such as asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF), the mucus is overproduced and its solid content augments, changing its structure and viscoelastic properties and determining a derangement of essential defense mechanisms against opportunistic microbial (virus and bacteria) pathogens. This ensues in damaging of the airways, leading to a vicious cycle of obstruction and infection responsible for the harsh clinical evolution of these CRDs. Here, we review the essential features of normal and pathological mucus (i.e., sputum in CF, COPD, and asthma), i.e., mucin content, structure (mesh size), micro/macro-rheology, pH, and osmotic pressure, ending with the awareness that sputum biomarkers (mucins, inflammatory proteins and peptides, and metabolites) might serve to indicate acute exacerbation and response to therapies. There are some indications that old and novel treatments may change the structure, viscoelastic properties, and biomarker content of sputum; however, a wealth of work is still needed to embrace these measures as correlates of disease severity in association with (or even as substitutes of) pulmonary functional tests.
Collapse
Affiliation(s)
- Michela Abrami
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I-34127 Trieste, Italy; (M.A.); (A.B.); (M.G.)
| | - Alice Biasin
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I-34127 Trieste, Italy; (M.A.); (A.B.); (M.G.)
| | - Fabiana Tescione
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, P.le E. Fermi 1, I-80055 Portici, Italy; (F.T.); (D.L.)
| | - Domenico Tierno
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, University of Trieste, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (G.G.)
| | - Barbara Dapas
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, I-34127 Trieste, Italy;
| | - Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, I-71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Gabriele Grassi
- Clinical Department of Medical, Surgical and Health Sciences, Cattinara University Hospital, University of Trieste, Strada di Fiume 447, I-34149 Trieste, Italy; (D.T.); (G.G.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, I-71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli 121, I-71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Domenico Larobina
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, P.le E. Fermi 1, I-80055 Portici, Italy; (F.T.); (D.L.)
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Via Valerio 6/A, I-34127 Trieste, Italy; (M.A.); (A.B.); (M.G.)
| |
Collapse
|
4
|
Meziu E, Shehu K, Koch M, Schneider M, Kraegeloh A. Impact of mucus modulation by N-acetylcysteine on nanoparticle toxicity. Int J Pharm X 2023; 6:100212. [PMID: 37771516 PMCID: PMC10522980 DOI: 10.1016/j.ijpx.2023.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023] Open
Abstract
Human respiratory mucus is a biological hydrogel that forms a protective barrier for the underlying epithelium. Modulation of the mucus layer has been employed as a strategy to enhance transmucosal drug carrier transport. However, a drawback of this strategy is a potential reduction of the mucus barrier properties, in particular in situations with an increased exposure to particles. In this study, we investigated the impact of mucus modulation on its protective role. In vitro mucus was produced by Calu-3 cells, cultivated at the air-liquid interface for 21 days and used for further testing as formed on top of the cells. Analysis of confocal 3D imaging data revealed that after 21 days Calu-3 cells secrete a mucus layer with a thickness of 24 ± 6 μm. Mucus appeared to restrict penetration of 500 nm carboxyl-modified polystyrene particles to the upper 5-10 μm of the layer. Furthermore, a mucus modulation protocol using aerosolized N-acetylcysteine (NAC) was developed. This treatment enhanced the penetration of particles through the mucus down to deeper layers by means of the mucolytic action of NAC. These findings were supported by cytotoxicity data, indicating that intact mucus protects the underlying epithelium from particle-induced effects on membrane integrity. The impact of NAC treatment on the protective properties of mucus was probed by using 50 and 100 nm amine-modified and 50 nm carboxyl-modified polystyrene nanoparticles, respectively. Cytotoxicity was only induced by the amine-modified particles in combination with NAC treatment, implying a reduced protective function of modulated mucus. Overall, our data emphasize the importance of integrating an assessment of the protective function of mucus into the development of therapy approaches involving mucus modulation.
Collapse
Affiliation(s)
- Enkeleda Meziu
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Kristela Shehu
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Marcus Koch
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics & Pharmaceutical Technology, Saarland University, 66123 Saarbrücken, Germany
| | - Annette Kraegeloh
- INM – Leibniz Institute for New Materials, 66123 Saarbrücken, Germany
| |
Collapse
|
5
|
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
|
6
|
Noronha-Matos JB, Pinto-Cardoso R, Bessa-Andrês C, Magalhães-Cardoso MT, Ferreirinha F, Costa MA, Marinhas J, Freitas R, Lemos R, Vilaça A, Oliveira A, Pelletier J, Sévigny J, Correia-de-Sá P. Silencing NTPDase3 activity rehabilitates the osteogenic commitment of post-menopausal stem cell bone progenitors. Stem Cell Res Ther 2023; 14:97. [PMID: 37076930 PMCID: PMC10116749 DOI: 10.1186/s13287-023-03315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Endogenously released adenine and uracil nucleotides favour the osteogenic commitment of bone marrow-derived mesenchymal stromal cells (BM-MSCs) through the activation of ATP-sensitive P2X7 and UDP-sensitive P2Y6 receptors. Yet, these nucleotides have their osteogenic potential compromised in post-menopausal (Pm) women due to overexpression of nucleotide metabolizing enzymes, namely NTPDase3. This prompted us to investigate whether NTPDase3 gene silencing or inhibition of its enzymatic activity could rehabilitate the osteogenic potential of Pm BM-MSCs. METHODS MSCs were harvested from the bone marrow of Pm women (69 ± 2 years old) and younger female controls (22 ± 4 years old). The cells were allowed to grow for 35 days in an osteogenic-inducing medium in either the absence or the presence of NTPDase3 inhibitors (PSB 06126 and hN3-B3s antibody); pre-treatment with a lentiviral short hairpin RNA (Lenti-shRNA) was used to silence the NTPDase3 gene expression. Immunofluorescence confocal microscopy was used to monitor protein cell densities. The osteogenic commitment of BM-MSCs was assessed by increases in the alkaline phosphatase (ALP) activity. The amount of the osteogenic transcription factor Osterix and the alizarin red-stained bone nodule formation. ATP was measured with the luciferin-luciferase bioluminescence assay. The kinetics of the extracellular ATP (100 µM) and UDP (100 µM) catabolism was assessed by HPLC RESULTS: The extracellular catabolism of ATP and UDP was faster in BM-MSCs from Pm women compared to younger females. The immunoreactivity against NTPDase3 increased 5.6-fold in BM-MSCs from Pm women vs. younger females. Selective inhibition or transient NTPDase3 gene silencing increased the extracellular accumulation of adenine and uracil nucleotides in cultured Pm BM-MSCs. Downregulation of NTPDase3 expression or activity rehabilitated the osteogenic commitment of Pm BM-MSCs measured as increases in ALP activity, Osterix protein cellular content and bone nodule formation; blockage of P2X7 and P2Y6 purinoceptors prevented this effect. CONCLUSIONS Data suggest that NTPDase3 overexpression in BM-MSCs may be a clinical surrogate of the osteogenic differentiation impairment in Pm women. Thus, besides P2X7 and P2Y6 receptors activation, targeting NTPDase3 may represent a novel therapeutic strategy to increase bone mass and reduce the osteoporotic risk of fractures in Pm women.
Collapse
Affiliation(s)
- José Bernardo Noronha-Matos
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal.
| | - Rui Pinto-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Catarina Bessa-Andrês
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Maria Teresa Magalhães-Cardoso
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Fátima Ferreirinha
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
| | - Maria Adelina Costa
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal
- Departamento de Química, Instituto de Ciências Biomédicas Abel Salazar - Universidade Do Porto (ICBAS-UP), 4050-313, Porto, Portugal
| | - José Marinhas
- Serviço de Ortopedia e Traumatologia, Centro Hospitalar de Gaia - Espinho, 4434-502, Vila Nova de Gaia, Portugal
| | - Rolando Freitas
- Serviço de Ortopedia e Traumatologia, Centro Hospitalar de Gaia - Espinho, 4434-502, Vila Nova de Gaia, Portugal
| | - Rui Lemos
- Serviço de Ortopedia e Traumatologia, Centro Hospitalar de Gaia - Espinho, 4434-502, Vila Nova de Gaia, Portugal
| | - Adélio Vilaça
- Serviço de Ortopedia, Centro Hospitalar Universitário de Santo António, 4099-001, Porto, Portugal
| | - António Oliveira
- Serviço de Ortopedia, Centro Hospitalar Universitário de Santo António, 4099-001, Porto, Portugal
| | - Julie Pelletier
- Centre de Recherche en Rhumatologie et Immunologie, University Laval, 2325, rue de l'Université Québec, Québec, G1V 0A6, Canada
| | - Jean Sévigny
- Centre de Recherche en Rhumatologie et Immunologie, University Laval, 2325, rue de l'Université Québec, Québec, G1V 0A6, Canada
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS) - Universidade do Porto (UP), R. Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
- Center for Drug Discovery and Innovative Medicines (MedInUP), Porto, Portugal.
| |
Collapse
|
7
|
Akamatsu Y, Akagi T, Sumitomo T, Takahara Y, Akiyama S, Kawabata S, Akashi M. Construction of Human Three-Dimensional Lung Model Using Layer-by-Layer Method. Tissue Eng Part C Methods 2023; 29:95-102. [PMID: 36721369 DOI: 10.1089/ten.tec.2022.0184] [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: 02/02/2023] Open
Abstract
The respiratory tract is one of the frontline barriers for biological defense. Lung epithelial intercellular adhesions provide protection from bacterial and viral infections and prevent invasion into deep tissues by pathogens. Dysfunction of lung epithelial intercellular adhesion caused by pathogens is associated with development of several diseases, such as acute respiratory distress syndrome, pneumonia, and asthma. To elucidate the pathological mechanism of respiratory infections, two-dimensional cell cultures and animal models are commonly used, although are not useful for evaluating host specificity or human biological response. With the rapid progression and worldwide spread of severe acute respiratory syndrome coronavirus-2, there is increasing interest in the development of a three-dimensional (3D) in vitro lung model for analyzing interactions between pathogens and hosts. However, some models possess unclear epithelial polarity or insufficient barrier functions and need the use of complex technologies, have high cost, and long cultivation terms. We previously reported about the fabrication of 3D cellular multilayers using a layer-by-layer (LbL) cell coating technique with extracellular matrix protein, fibronectin (FN), and gelatin (G). In the present study, such a LbL cell coating technique was utilized to construct a human 3D lung model in which a monolayer of the human lower airway epithelial adenocarcinoma cell line Calu-3 cells was placed on 3D-cellular multilayers composed of FN-G-coated human primary pulmonary fibroblast cells. The 3D lung model thus constructed demonstrated an epithelial-fibroblast layer that maintained uniform thickness until 7 days of incubation. Moreover, expressions of E-cadherin, ZO-1, and mucin in the epithelial layer were observed by immunohistochemical staining. Epithelial barrier integrity was evaluated using transepithelial electrical resistance values. The results indicate that the present constructed human 3D lung model is similar to human lung tissues and also features epithelial polarity and a barrier function, thus is considered useful for evaluating infection and pathological mechanisms related to pneumonia and several pathogens. Impact statement A novel in vitro model of lung tissue was established. Using a layer-by-layer cell coating technique, a three-dimensional cultured lung model was constructed. The present novel model was shown to have epithelial polarity and chemical barrier functions. This model may be useful for investigating interaction pathogens and human biology.
Collapse
Affiliation(s)
- Yukako Akamatsu
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan.,Division of Special Care Dentistry, Osaka University Dental Hospital, Osaka, Japan
| | - Takami Akagi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Tomoko Sumitomo
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yuki Takahara
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan.,Department of Fixed Prosthodontics, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Shigehisa Akiyama
- Division of Special Care Dentistry, Osaka University Dental Hospital, Osaka, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan.,Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Mitsuru Akashi
- Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
8
|
Wang JN, Fan H, Song JT. Targeting purinergic receptors to attenuate inflammation of dry eye. Purinergic Signal 2023; 19:199-206. [PMID: 35218451 PMCID: PMC9984584 DOI: 10.1007/s11302-022-09851-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/04/2022] [Indexed: 11/28/2022] Open
Abstract
Inflammation is one of the potential factors to cause the damage of ocular surface in dry eye disease (DED). Increasing evidence indicated that purinergic A1, A2A, A3, P2X4, P2X7, P2Y1, P2Y2, and P2Y4 receptors play an important role in the regulation of inflammation in DED: A1 adenosine receptor (A1R) is a systemic pro-inflammatory factor; A2AR is involved in the activation of the MAPK/NF-kB pathway; A3R combined with inhibition of adenylate cyclase and regulation of the mitogen-activated protein kinase (MAPK) pathway leads to regulation of transcription; P2X4 promotes receptor-associated activation of pro-inflammatory cytokines and inflammatory vesicles; P2X7 promotes inflammasome activation and release of pro-inflammatory cytokines IL-1β and IL-18; P2Y receptors affect the phospholipase C(PLC)/IP3/Ca2+ signaling pathway and mucin secretion. These suggested that purinergic receptors would be promising targets to control the inflammation of DED in the future.
Collapse
Affiliation(s)
- Jia-Ning Wang
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hua Fan
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jian-Tao Song
- Eye Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
9
|
Sudduth ER, Kolewe EL, Graf J, Yu Y, Somma J, Fromen CA. Nebulization of Model Hydrogel Nanoparticles to Macrophages at the Air-Liquid Interface. FRONTIERS IN CHEMICAL ENGINEERING 2023; 4:1086031. [PMID: 37859802 PMCID: PMC10586456 DOI: 10.3389/fceng.2022.1086031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Nanoparticle evaluation within the pulmonary airspace has increasingly important implications for human health, with growing interest from drug delivery, environmental, and toxicology fields. While there have been widespread investigations of nanoparticle physiochemical properties following many routes of administration, nanoparticle behavior at the air-liquid interface (ALI) is less well-characterized. In this work, we fabricate two formulations of poly(ethylene)-glycol diacrylate (PEGDA)-based model nanoparticles to establish an in vitro workflow allowing evaluation of nanoparticle charge effects at the ALI. Both cationic and anionic PEGDA formulations were synthesized with similar hydrodynamic diameters around ~225 nm and low polydispersity, with expected surface charges corresponding with the respective functional co-monomer. We find that both formulations are readily nebulized from an aqueous suspension in a commercial Aeroneb® Lab Nebulizer, but the aqueous delivery solution served to slightly increase the overall hydrodynamic and geometric size of the cationic particle formulation. However, nanoparticle loading at 50 μg/ml of either formulation did not influence the resultant aerosol diameter from the nebulizer. To assess aerosol delivery in vitro, we designed a 3D printed adapter capable of ensuring aerosol delivery to transwell 24-well culture plates. Nanoparticle uptake by macrophages was compared between traditional cell culture techniques and that of ALI-cultured macrophages following aerosol delivery. Cell viability was unaffected by nanoparticle delivery using either method. However, only traditional cell culture methods demonstrated significant uptake that was dependent on the nanoparticle surface charge. Concurrently, ALI culture resulted in lower metabolic activity of macrophages than those in traditional cell culture, leading to lower overall nanoparticle uptake at ALI. Overall, this work demonstrates that base-material similarities between both particle formulations provide an expected consistency in aerosol delivery regardless of the nanoparticle surface charge and provides an important workflow that enables a holistic evaluation of aerosolizable nanoparticles.
Collapse
Affiliation(s)
- Emma R. Sudduth
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Emily L. Kolewe
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Jodi Graf
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Yinkui Yu
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Joaquina Somma
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
10
|
Abstract
Coronavirus disease 2019 (COVID-19) is a worldwide pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has affected millions of lives. Individuals who survive severe COVID-19 can experience sustained respiratory symptoms that persist for months after initial infection. In other airway diseases, abnormal airway mucus contributes to sustained airway symptoms. However, the impact of SARS-CoV-2 on airway mucus has received limited attention. In the current review, we assess literature describing the impact of SARS-CoV-2 on airway pathophysiology with specific emphasis on mucus production. Accumulating evidence suggests that the 2 major secreted airway mucin glycoproteins, MUC5AC and MUC5B, are abnormal in some patients with COVID-19. Aberrations in MUC5AC or MUC5B in response to SARS-CoV-2 infection are likely due to inflammation, though the responsible mechanisms have yet to be determined. Thus, we also provide a proposed model highlighting mechanisms that can contribute to acute and sustained mucus abnormalities in SARS-CoV-2, with an emphasis on inflammatory cells and mediators, including mast cells and histamine. Last, we bring to light the challenges of studying abnormal mucus production in SARS-CoV-2 infections and discuss the strengths and limitations of model systems commonly used to study COVID-19. The evidence to date suggests that ferrets, nonhuman primates, and cats may have advantages over other models to investigate mucus in COVID-19.
Collapse
|
11
|
Esther CR, O'Neal WK, Anderson WH, Kesimer M, Ceppe A, Doerschuk CM, Alexis NE, Hastie AT, Barr RG, Bowler RP, Wells JM, Oelsner EC, Comellas AP, Tesfaigzi Y, Kim V, Paulin LM, Cooper CB, Han MK, Huang YJ, Labaki WW, Curtis JL, Boucher RC. Identification of Sputum Biomarkers Predictive of Pulmonary Exacerbations in COPD. Chest 2022; 161:1239-1249. [PMID: 34801592 PMCID: PMC9131049 DOI: 10.1016/j.chest.2021.10.049] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/15/2021] [Accepted: 10/29/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Improved understanding of the pathways associated with airway pathophysiologic features in COPD will identify new predictive biomarkers and novel therapeutic targets. RESEARCH QUESTION Which physiologic pathways are altered in the airways of patients with COPD and will predict exacerbations? STUDY DESIGN AND METHODS We applied a mass spectrometric panel of metabolomic biomarkers related to mucus hydration and inflammation to sputa from the multicenter Subpopulations and Intermediate Outcome Measures in COPD Study. Biomarkers elevated in sputa from patients with COPD were evaluated for relationships to measures of COPD disease severity and their ability to predict future exacerbations. RESULTS Sputum supernatants from 980 patients were analyzed: 77 healthy nonsmokers, 341 smokers with preserved spirometry, and 562 patients with COPD (178 with Global Initiative on Chronic Obstructive Lung Disease [GOLD] stage 1 disease, 303 with GOLD stage 2 disease, and 81 with GOLD stage 3 disease) were analyzed. Biomarkers from multiple pathways were elevated in COPD and correlated with sputum neutrophil counts. Among the most significant analytes (false discovery rate, 0.1) were sialic acid, hypoxanthine, xanthine, methylthioadenosine, adenine, and glutathione. Sialic acid and hypoxanthine were associated strongly with measures of disease severity, and elevation of these biomarkers was associated with shorter time to exacerbation and improved prediction models of future exacerbations. INTERPRETATION Biomarker evaluation implicated pathways involved in mucus hydration, adenosine metabolism, methionine salvage, and oxidative stress in COPD airway pathophysiologic characteristics. Therapies that target these pathways may be of benefit in COPD, and a simple model adding sputum-soluble phase biomarkers improves prediction of pulmonary exacerbations. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT01969344; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- Charles R Esther
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| | - Wanda K O'Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wayne H Anderson
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mehmet Kesimer
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Agathe Ceppe
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Claire M Doerschuk
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Neil E Alexis
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Annette T Hastie
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC
| | - R Graham Barr
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
| | | | - J Michael Wells
- Lung Health Center, Division of Pulmonary Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, AL
| | - Elizabeth C Oelsner
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Alejandro P Comellas
- Division of Pulmonary, Critical Care and Occupational Medicine, University of Iowa, Iowa City, IA
| | - Yohannes Tesfaigzi
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Victor Kim
- Pulmonary and Critical Care Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Laura M Paulin
- Department of Medicine and Epidemiology, Dartmouth-Hitchcock Medical Center, Geisel School of Medicine, Hanover, NH
| | - Christopher B Cooper
- Department of Medicine and Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan Ann Arbor, Ann Arbor, MI
| | - Yvonne J Huang
- Division of Pulmonary and Critical Care Medicine, University of Michigan Ann Arbor, Ann Arbor, MI
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan Ann Arbor, Ann Arbor, MI
| | - Jeffrey L Curtis
- Division of Pulmonary and Critical Care Medicine, University of Michigan Ann Arbor, Ann Arbor, MI; Medicine Service, VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
12
|
Sonntag T, Rapp M, Didier P, Lebeau L, Pons F, Casset A. Mucus-producing epithelial models for investigating the activity of gene delivery systems in the lung. Int J Pharm 2021; 614:121423. [PMID: 34958896 DOI: 10.1016/j.ijpharm.2021.121423] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/09/2021] [Accepted: 12/21/2021] [Indexed: 11/18/2022]
Abstract
Inhaled transfection particles have to penetrate the mucus layer lining the airways to successfully deliver their therapeutic nucleic acid payload to target cells in the underlying epithelium. However, the in vitro models used for evaluating gene carrier efficiency often disregard this viscous defensive barrier. In this study, the two mucus-secreting cell lines NCI-H292 and Calu-3 were selected to develop a series of epithelial models displaying gradual mucus production. In NCI-H292 models, a gradual increase in the MUC5AC mucin was obtained after cell exposure to inducers. In Calu-3 models, MUC5AC production increased as a function of culture duration (3, 7, 14 days) at the air-liquid interface (ALI). Six DOPC-derived cationic lipids were designed and their pDNA delivery activity was evaluated to validate these cellular models. The strongest impairment of the lipid delivery activity was observed in the Calu-3 14-d ALI model. The MUC5AC production in this model was the greatest and the mucus layer was 20 µm thick. The mucus exhibited a solid viscoelastic behaviour, and represented a major hindrance to lipoplex diffusion. The Calu-3 14-d ALI model will be highly useful for accurate evaluation of gene carriers intended for airway administration and characterization of their interactions with the mucus.
Collapse
Affiliation(s)
- Thomas Sonntag
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Mickael Rapp
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Pascal Didier
- Laboratoire de Bioimagerie et Pathologies, UMR 7021, CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Luc Lebeau
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Françoise Pons
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France
| | - Anne Casset
- Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, CNRS-Université de Strasbourg, Faculté de Pharmacie, Illkirch, France.
| |
Collapse
|
13
|
Terakosolphan W, Altharawi A, Poonprasartporn A, Harvey RD, Forbes B, Chan KLA. In vitro Fourier transform infrared spectroscopic study of the effect of glycerol on the uptake of beclomethasone dipropionate in living respiratory cells. Int J Pharm 2021; 609:121118. [PMID: 34560211 DOI: 10.1016/j.ijpharm.2021.121118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/01/2022]
Abstract
The quantification of drug in living cells is of increasing interest in pharmaceutical research because of its importance in understanding drug efficacy and toxicity. Label-free in situ measurement methods are advantageous for their ability to obtain chemical and time profiles without the need of labelling or extraction steps. We have previously shown that Fourier transform infrared (FTIR) spectroscopy has the potential to quantify drug in situ within living cells at micromolar level when a simple solution of drug was added to the medium. The purpose of this study was to demonstrate that the approach can evaluate more complex systems such as the effect of membrane modification by a formulation on drug uptakes. The inhaled corticosteroid, beclomethasone dipropionate (BDP), in Calu-3 respiratory epithelial cells in the absence and presence of glycerol, an excipient in some inhaled medicines was used as the model system. The FTIR method was first validated for limit of detection (LOD) and quantification (LOQ) according to published guidelines and the LOQ was found to be ∼ 20 μM, good enough to quantify BDP in the living cell. The uptake of BDP by living Calu-3 cells was found to be reduced in the presence of glycerol as expected due to the stiffening of the cell membrane by the presence of glycerol in the formulation. This study demonstrates the valuable analytical capability of live-cell FTIR to study the effect of formulation on drug transport in lungs and to evaluate drug availability to intracellular targets. We conclude that FTIR has potential to contribute widely at the frontier of live-cell studies.
Collapse
Affiliation(s)
- Wachirun Terakosolphan
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, United Kingdom
| | - Ali Altharawi
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, United Kingdom; Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Richard D Harvey
- Department of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, Althanstraße 14 (UZA II), 1090 Wien, Austria
| | - Ben Forbes
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, United Kingdom
| | - K L Andrew Chan
- Institute of Pharmaceutical Science, King's College London, London SE1 9NH, United Kingdom.
| |
Collapse
|
14
|
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
|
15
|
Reyna-Jeldes M, Díaz-Muñoz M, Madariaga JA, Coddou C, Vázquez-Cuevas FG. Autocrine and paracrine purinergic signaling in the most lethal types of cancer. Purinergic Signal 2021; 17:345-370. [PMID: 33982134 PMCID: PMC8410929 DOI: 10.1007/s11302-021-09785-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer comprises a collection of diseases that occur in almost any tissue and it is characterized by an abnormal and uncontrolled cell growth that results in tumor formation and propagation to other tissues, causing tissue and organ malfunction and death. Despite the undeniable improvement in cancer diagnostics and therapy, there is an urgent need for new therapeutic and preventive strategies with improved efficacy and fewer side effects. In this context, purinergic signaling emerges as an interesting candidate as a cancer biomarker or therapeutic target. There is abundant evidence that tumor cells have significant changes in the expression of purinergic receptors, which comprise the G-protein coupled P2Y and AdoR families of receptors and the ligand-gated ion channel P2X receptors. Tumor cells also exhibit changes in the expression of nucleotidases and other enzymes involved in nucleotide metabolism, and the concentrations of extracellular nucleotides are significantly higher than those observed in normal cells. In this review, we will focus on the potential role of purinergic signaling in the ten most lethal cancers (lung, breast, colorectal, liver, stomach, prostate, cervical, esophagus, pancreas, and ovary), which together are responsible for more than 5 million annual deaths.
Collapse
Affiliation(s)
- M Reyna-Jeldes
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
- Núcleo para el Estudio del Cáncer a nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta, Chile
| | - M Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, México
| | - J A Madariaga
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
- Núcleo para el Estudio del Cáncer a nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta, Chile
| | - C Coddou
- Departamento de Ciencias Biomédicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile.
- Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile.
- Núcleo para el Estudio del Cáncer a nivel Básico, Aplicado y Clínico, Universidad Católica del Norte, Antofagasta, Chile.
| | - F G Vázquez-Cuevas
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, México.
| |
Collapse
|
16
|
Dang Y, van Heusden C, Nickerson V, Chung F, Wang Y, Quinney NL, Gentzsch M, Randell SH, Moulton HM, Kole R, Ni A, Juliano RL, Kreda SM. Enhanced delivery of peptide-morpholino oligonucleotides with a small molecule to correct splicing defects in the lung. Nucleic Acids Res 2021; 49:6100-6113. [PMID: 34107015 PMCID: PMC8216463 DOI: 10.1093/nar/gkab488] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary diseases offer many targets for oligonucleotide therapeutics. However, effective delivery of oligonucleotides to the lung is challenging. For example, splicing mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect a significant cohort of Cystic Fibrosis (CF) patients. These individuals could potentially benefit from treatment with splice switching oligonucleotides (SSOs) that can modulate splicing of CFTR and restore its activity. However, previous studies in cell culture used oligonucleotide transfection methods that cannot be safely translated in vivo. In this report, we demonstrate effective correction of a splicing mutation in the lung of a mouse model using SSOs. Moreover, we also demonstrate effective correction of a CFTR splicing mutation in a pre-clinical CF patient-derived cell model. We utilized a highly effective delivery strategy for oligonucleotides by combining peptide-morpholino (PPMO) SSOs with small molecules termed OECs. PPMOs distribute broadly into the lung and other tissues while OECs potentiate the effects of oligonucleotides by releasing them from endosomal entrapment. The combined PPMO plus OEC approach proved to be effective both in CF patient cells and in vivo in the mouse lung and thus may offer a path to the development of novel therapeutics for splicing mutations in CF and other lung diseases.
Collapse
Affiliation(s)
- Yan Dang
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Catharina van Heusden
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Veronica Nickerson
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Felicity Chung
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Yang Wang
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Nancy L Quinney
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Martina Gentzsch
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Scott H Randell
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| | - Hong M Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Ryszard Kole
- Department of Pharmacology, The University of North Carolina at Chapel Hill, 4010 Genetic Medicine Bldg, Chapel Hill, NC 27599, USA
| | - Aiguo Ni
- Initos Pharmaceuticals, LLC, Chapel Hill, NC 27514, USA
| | | | - Silvia M Kreda
- Marsico Lung Institute/Cystic Fibrosis Center, The University of North Carolina at Chapel Hill, 6009 Thurston Bowles Bldg, Chapel Hill NC 27599-7248, USA
| |
Collapse
|
17
|
Yimnual C, Satitsri S, Ningsih BNS, Rukachaisirikul V, Muanprasat C. A fungus-derived purpactin A as an inhibitor of TMEM16A chloride channels and mucin secretion in airway epithelial cells. Biomed Pharmacother 2021; 139:111583. [PMID: 33901875 DOI: 10.1016/j.biopha.2021.111583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
TMEM16A is a Ca2+-activated Cl- channel involved in mucus secretion in inflamed airways and proposed as a drug target for diseases associated with mucus hypersecretion including asthma. This study aimed to identify novel inhibitors of TMEM16A-mediated Cl- secretion in airway epithelial cells from a collection of compounds isolated from fungi indigenous in Thailand and examine its potential utility in mitigating airway mucus secretion using Calu-3 cells as a study model. Screening of > 400 fungal metabolites revealed purpactin A isolated from a soil-derived fungus Penicillium aculeatum PSU-RSPG105 as an inhibitor of TMEM16A-mediated Cl- transport with an IC50 value of ~2 µM. A consistent inhibitory effect of purpactin A on TMEM16A were observed regardless of TMEM16A activators or in the presence of an inhibitor of Ca2+/calmodulin-dependent protein kinase II (CaMKII), a negative regulator of TMEM16A. In addition, purpactin A did not affect cell viability, epithelial barrier integrity and activities of membrane transport proteins essential for maintaining airway hydration including CFTR Cl- channels and apical BK K+ channels. Intriguingly, purpactin A prevented a Ca2+-induced mucin release in cytokine-treated airway cells. Taken together, purpactin A represents the first class of TMEM16A inhibitor derived from fungus, which may be beneficial for the treatment of diseases associated with mucus hypersecretion.
Collapse
Affiliation(s)
- Chantapol Yimnual
- Department of Physiology, Faculty of Science, Mahidol University, Rajathevi, Bangkok 10400, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand
| | - Saravut Satitsri
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand
| | - Baiq Nila Sari Ningsih
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Vatcharin Rukachaisirikul
- Division of Physical Science and Center of Excellence for Innovation in Chemistry, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla 90112, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bang Phli, Samut Prakarn 10540, Thailand.
| |
Collapse
|
18
|
Sweeter JM, Kudrna K, Hunt K, Thomes P, Dickey BF, Brody SL, Dickinson JD. Autophagy of mucin granules contributes to resolution of airway mucous metaplasia. Sci Rep 2021; 11:13037. [PMID: 34158522 PMCID: PMC8219712 DOI: 10.1038/s41598-021-91932-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/01/2021] [Indexed: 12/21/2022] Open
Abstract
Exacerbations of muco-obstructive airway diseases such as COPD and asthma are associated with epithelial changes termed mucous metaplasia (MM). Many molecular pathways triggering MM have been identified; however, the factors that regulate resolution are less well understood. We hypothesized that the autophagy pathway is required for resolution of MM by eliminating excess non-secreted intracellular mucin granules. We found increased intracellular levels of mucins Muc5ac and Muc5b in mice deficient in autophagy regulatory protein, Atg16L1, and that this difference was not due to defects in the known baseline or stimulated mucin secretion pathways. Instead, we found that, in mucous secretory cells, Lc3/Lamp1 vesicles colocalized with mucin granules particularly adjacent to the nucleus, suggesting that some granules were being eliminated in the autophagy pathway rather than secreted. Using a mouse model of MM resolution, we found increased lysosomal proteolytic activity that peaked in the days after mucin production began to decline. In purified lysosomal fractions, Atg16L1-deficient mice had reduced proteolytic degradation of Lc3 and Sqstm1 and persistent accumulation of mucin granules associated with impaired resolution of mucous metaplasia. In normal and COPD derived human airway epithelial cells (AECs), activation of autophagy by mTOR inhibition led to a reduction of intracellular mucin granules in AECs. Our findings indicate that during peak and resolution phases of MM, autophagy activity rather than secretion is required for elimination of some remaining mucin granules. Manipulation of autophagy activation offers a therapeutic target to speed resolution of MM in airway disease exacerbations.
Collapse
Affiliation(s)
- J M Sweeter
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Kudrna
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Hunt
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Thomes
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - B F Dickey
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - S L Brody
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - J D Dickinson
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
19
|
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
|
20
|
Sanchez-Guzman D, Boland S, Brookes O, Mc Cord C, Lai Kuen R, Sirri V, Baeza Squiban A, Devineau S. Long-term evolution of the epithelial cell secretome in preclinical 3D models of the human bronchial epithelium. Sci Rep 2021; 11:6621. [PMID: 33758289 PMCID: PMC7988136 DOI: 10.1038/s41598-021-86037-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/10/2021] [Indexed: 01/31/2023] Open
Abstract
The human bronchial epithelium is the first line of defense against atmospheric particles, pollutants, and respiratory pathogens such as the novel SARS-CoV-2. The epithelial cells form a tight barrier and secrete proteins that are major components of the mucosal immune response. Functional in vitro models of the human lung are essential for screening the epithelial response and assessing the toxicity and barrier crossing of drugs, inhaled particles, and pollutants. However, there is a lack of models to investigate the effect of chronic exposure without resorting to animal testing. Here, we developed a 3D model of the human bronchial epithelium using Calu-3 cell line and demonstrated its viability and functionality for 21 days without subculturing. We investigated the effect of reduced Fetal Bovine Serum supplementation in the basal medium and defined the minimal supplementation needed to maintain a functional epithelium, so that the amount of exogenous serum proteins could be reduced during drug testing. The long-term evolution of the epithelial cell secretome was fully characterized by quantitative mass spectrometry in two preclinical models using Calu-3 or primary NHBE cells. 408 common secreted proteins were identified while significant differences in protein abundance were observed with time, suggesting that 7-10 days are necessary to establish a mature secretome in the Calu-3 model. The associated Reactome pathways highlight the role of the secreted proteins in the immune response of the bronchial epithelium. We suggest this preclinical 3D model can be used to evaluate the long-term toxicity of drugs or particles on the human bronchial epithelium, and subsequently to investigate their effect on the epithelial cell secretions.
Collapse
Affiliation(s)
| | - Sonja Boland
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | - Oliver Brookes
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | - Claire Mc Cord
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | - René Lai Kuen
- Cellular and Molecular Imaging Facility, US25 Inserm-3612 CNRS, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Valentina Sirri
- Université de Paris, BFA, UMR 8251, CNRS, 75013, Paris, France
| | | | | |
Collapse
|
21
|
P2Y14 Receptor as a Target for Neutrophilia Attenuation in Severe COVID-19 Cases: From Hematopoietic Stem Cell Recruitment and Chemotaxis to Thrombo-inflammation. Stem Cell Rev Rep 2021; 17:241-252. [PMID: 33575962 PMCID: PMC7877512 DOI: 10.1007/s12015-021-10129-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
The global SARS-CoV-2 pandemic starting in 2019 has already reached more than 2.3 million deaths. Despite the scientific community’s efforts to investigate the COVID-19 disease, a drug for effectively treating or curing patients yet needs to be discovered. Hematopoietic stem cells (HSC) differentiating into immune cells for defense express COVID-19 entry receptors, and COVID-19 infection hinders their differentiation. The importance of purinergic signaling in HSC differentiation and innate immunity has been recognized. The metabotropic P2Y14 receptor subtype, activated by UDP-glucose, controls HSC differentiation and mobilization. Thereon, the exacerbated activation of blood immune cells amplifies the inflammatory state observed in COVID-19 patients, specially through the continuous release of reactive oxygen species and extracellular neutrophil traps (NETs). Further, the P2Y14 subtype, robustly inhibits the infiltration of neutrophils into various epithelial tissues, including lungs and kidneys. Here we discuss findings suggesting that antagonism of the P2Y14 receptor could prevent the progression of COVID-19-induced systemic inflammation, which often leads to severe illness and death cases. Considering the modulation of neutrophil recruitment of extreme relevance for respiratory distress and lung failure prevention, we propose that P2Y14 receptor inhibition by its selective antagonist PPTN could limit neutrophil recruitment and NETosis, hence limiting excessive formation of oxygen reactive species and proteolytic activation of the kallikrein-kinin system and subsequent bradykinin storm in the alveolar septa of COVID-19 patients.
Collapse
|
22
|
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
|
23
|
Grau-Bartual S, Al-Jumaily AM. Prediction of upper airway dryness and optimal continuous positive airway pressure conditions. J Biomech 2020; 112:110037. [PMID: 32947068 PMCID: PMC7481124 DOI: 10.1016/j.jbiomech.2020.110037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 10/25/2022]
Abstract
Continuous positive airway pressure is the most effective long-term treatment for obstructive sleep apnoea, which is a sleeping disorder characterized by pauses in breathing during sleep. It introduces pressurized atmospheric air into the respiratory system in order to maintain open airways without blockage. Some continuous positive airway pressure devices incorporate a convective heat transfer humidifier to overcome dryness. However, many side effects, including the unacceptable excess of water droplets in the air supply line, have been reported and improvements are essential for better patient's comfort and acceptance of the therapy. The excess of water droplets is attributed to the qualitative rather than the quantitative approach of determining the rise in temperature and humidity of the inspired air. Therefore, a human upper airway mathematical model is developed to predict the heat and water transfer variation between normal breathing and continuous positive airway pressure conditions and determine the optimal input temperature and relative humidity in the continuous positive airway pressure humidifier.
Collapse
Affiliation(s)
- Sandra Grau-Bartual
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand
| | - Ahmed M Al-Jumaily
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand.
| |
Collapse
|
24
|
Reid AT, Nichol KS, Chander Veerati P, Moheimani F, Kicic A, Stick SM, Bartlett NW, Grainge CL, Wark PAB, Hansbro PM, Knight DA. Blocking Notch3 Signaling Abolishes MUC5AC Production in Airway Epithelial Cells from Individuals with Asthma. Am J Respir Cell Mol Biol 2020; 62:513-523. [PMID: 31922915 DOI: 10.1165/rcmb.2019-0069oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In asthma, goblet cell numbers are increased within the airway epithelium, perpetuating the production of mucus that is more difficult to clear and results in airway mucus plugging. Notch1, Notch2, or Notch3, or a combination of these has been shown to influence the differentiation of airway epithelial cells. How the expression of specific Notch isoforms differs in fully differentiated adult asthmatic epithelium and whether Notch influences mucin production after differentiation is currently unknown. We aimed to quantify different Notch isoforms in the airway epithelium of individuals with severe asthma and to examine the impact of Notch signaling on mucin MUC5AC. Human lung sections and primary bronchial epithelial cells from individuals with and without asthma were used in this study. Primary bronchial epithelial cells were differentiated at the air-liquid interface for 28 days. Notch isoform expression was analyzed by Taqman quantitative PCR. Immunohistochemistry was used to localize and quantify Notch isoforms in human airway sections. Notch signaling was inhibited in vitro using dibenzazepine or Notch3-specific siRNA, followed by analysis of MUC5AC. NOTCH3 was highly expressed in asthmatic airway epithelium compared with nonasthmatic epithelium. Dibenzazepine significantly reduced MUC5AC production in air-liquid interface cultures of primary bronchial epithelial cells concomitantly with suppression of NOTCH3 intracellular domain protein. Specific knockdown using NOTCH3 siRNA recapitulated the dibenzazepine-induced reduction in MUC5AC. We demonstrate that NOTCH3 is a regulator of MUC5AC production. Increased NOTCH3 signaling in the asthmatic airway epithelium may therefore be an underlying driver of excess MUC5AC production.
Collapse
Affiliation(s)
- Andrew T Reid
- School of Biomedical Sciences and Pharmacy.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and
| | - Kristy S Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and.,School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Punnam Chander Veerati
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and.,School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
| | - Fatemeh Moheimani
- School of Biomedical Sciences and Pharmacy.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and
| | - Anthony Kicic
- School of Paediatrics and Child Health.,Telethon Kids Institute, and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia.,Occupation and Environment, School of Public Health, Curtin University, Bentley, Western Australia, Australia
| | - Stephen M Stick
- School of Paediatrics and Child Health.,Telethon Kids Institute, and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| | - Nathan W Bartlett
- School of Biomedical Sciences and Pharmacy.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and
| | - Chris L Grainge
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and.,School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia; and
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and.,School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia; and
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, and.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
25
|
Ramsey KA, Chen ACH, Radicioni G, Lourie R, Martin M, Broomfield A, Sheng YH, Hasnain SZ, Radford-Smith G, Simms LA, Burr L, Thornton DJ, Bowler SD, Livengood S, Ceppe A, Knowles MR, Noone PG, Donaldson SH, Hill DB, Ehre C, Button B, Alexis NE, Kesimer M, Boucher RC, McGuckin MA. Airway Mucus Hyperconcentration in Non-Cystic Fibrosis Bronchiectasis. Am J Respir Crit Care Med 2020; 201:661-670. [PMID: 31765597 DOI: 10.1164/rccm.201906-1219oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Rationale: Non-cystic fibrosis bronchiectasis is characterized by airway mucus accumulation and sputum production, but the role of mucus concentration in the pathogenesis of these abnormalities has not been characterized.Objectives: This study was designed to: 1) measure mucus concentration and biophysical properties of bronchiectasis mucus; 2) identify the secreted mucins contained in bronchiectasis mucus; 3) relate mucus properties to airway epithelial mucin RNA/protein expression; and 4) explore relationships between mucus hyperconcentration and disease severity.Methods: Sputum samples were collected from subjects with bronchiectasis, with and without chronic erythromycin administration, and healthy control subjects. Sputum percent solid concentrations, total and individual mucin concentrations, osmotic pressures, rheological properties, and inflammatory mediators were measured. Intracellular mucins were measured in endobronchial biopsies by immunohistochemistry and gene expression. MUC5B (mucin 5B) polymorphisms were identified by quantitative PCR. In a replication bronchiectasis cohort, spontaneously expectorated and hypertonic saline-induced sputa were collected, and mucus/mucin concentrations were measured.Measurements and Main Results: Bronchiectasis sputum exhibited increased percent solids, total and individual (MUC5B and MUC5AC) mucin concentrations, osmotic pressure, and elastic and viscous moduli compared with healthy sputum. Within subjects with bronchiectasis, sputum percent solids correlated inversely with FEV1 and positively with bronchiectasis extent, as measured by high-resolution computed tomography, and inflammatory mediators. No difference was detected in MUC5B rs35705950 SNP allele frequency between bronchiectasis and healthy individuals. Hypertonic saline inhalation acutely reduced non-cystic fibrosis bronchiectasis mucus concentration by 5%.Conclusions: Hyperconcentrated airway mucus is characteristic of subjects with bronchiectasis, likely contributes to disease pathophysiology, and may be a target for pharmacotherapy.
Collapse
Affiliation(s)
- Kathryn A Ramsey
- Marsico Lung Institute.,Department of Pediatrics, Pediatric Respiratory Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alice C H Chen
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute, Translational Research Institute, and.,School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | | | - Rohan Lourie
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute, Translational Research Institute, and.,Department of Anatomical Pathology, Mater Misericordiae Limited, South Brisbane, Queensland, Australia
| | - Megan Martin
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Queensland, Australia
| | - Amy Broomfield
- Department of Anatomical Pathology, Mater Misericordiae Limited, South Brisbane, Queensland, Australia
| | - Yong H Sheng
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute, Translational Research Institute, and
| | - Sumaira Z Hasnain
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute, Translational Research Institute, and
| | - Graham Radford-Smith
- Inflammatory Bowel Diseases Research Laboratory, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Lisa A Simms
- Inflammatory Bowel Diseases Research Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | - Lucy Burr
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute, Translational Research Institute, and.,Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Queensland, Australia
| | - David J Thornton
- Wellcome Trust Centre for Cell-Matrix Research, Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; and
| | - Simon D Bowler
- Department of Respiratory Medicine, Mater Adult Hospital, South Brisbane, Queensland, Australia
| | | | | | | | | | | | - David B Hill
- Marsico Lung Institute.,Department of Physics and Astronomy, and
| | | | | | - Neil E Alexis
- Center for Environmental Medicine, Asthma and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | - Michael A McGuckin
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute, Translational Research Institute, and.,Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Grau-Bartual S, Al-Jumaily AM, Young PM, Traini D, Ghadiri M. Effect of continuous positive airway pressure treatment on permeability, inflammation and mucus production of human epithelial cells. ERJ Open Res 2020; 6:00327-2019. [PMID: 32537464 PMCID: PMC7276533 DOI: 10.1183/23120541.00327-2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/17/2020] [Indexed: 11/05/2022] Open
Abstract
Continuous positive airway pressure (CPAP) therapy is the gold standard treatment for obstructive sleep apnoea, which affects millions of people worldwide. However, this therapy normally results in symptoms such as dryness, sneezing, rhinorrhoea, post-nasal drip, nasal congestion and epistaxis in the upper airways. Using bronchial epithelial (Calu-3) and nasal epithelial (RPMI 2650) cells in an in vitro respiratory model, this study, for the first time, investigates the effect of CPAP positive pressure on the human respiratory epithelial mechanisms that regulate upper airways lubrication characteristics. To understand how the epithelium and mucus are affected by this therapy, several parameters were determined before and after positive pressure application. This work demonstrates that the positive pressure not only compresses the cells, but also reduces their permeability and mucus secretion rate, thus drying the airway surface liquid layer and altering the mucus/water ratio. It is also observed that the respiratory epithelia is equally inflamed without or with external humidification during CPAP application. These findings clearly identify the causes of the side-effects reported by patients under CPAP therapy.
Collapse
Affiliation(s)
- Sandra Grau-Bartual
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand
| | - Ahmed M Al-Jumaily
- Institute of Biomedical Technologies, Auckland University of Technology, Auckland, New Zealand
| | - Paul M Young
- Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Daniela Traini
- Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | - Maliheh Ghadiri
- Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| |
Collapse
|
27
|
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
|
28
|
Alpha-tocopherol exerts protective function against the mucotoxicity of particulate matter in amphibian and human goblet cells. Sci Rep 2020; 10:6224. [PMID: 32277121 PMCID: PMC7148342 DOI: 10.1038/s41598-020-63085-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 03/25/2020] [Indexed: 01/04/2023] Open
Abstract
Exposure to particulate matter (PM) in ambient air is known to increase the risk of cardiovascular disorders and mortality. The cytotoxicity of PM is mainly due to the abnormal increase of reactive oxygen species (ROS), which damage cellular components such as DNA, RNA, and proteins. The correlation between PM exposure and human disorders, including mortality, is based on long-term exposure. In this study we have investigated acute responses of mucus-secreting goblet cells upon exposure to PM derived from a heavy diesel engine. To this end, we employed the mucociliary epithelium of amphibian embryos and human Calu-3 cells to examine PM mucotoxicity. Our data suggest that acute exposure to PM significantly impairs mucus secretion and results in the accumulation of mucus vesicles in the cytoplasm of goblet cells. RNA-seq analysis revealed that acute responses to PM exposure significantly altered gene expression patterns; however, known regulators of mucus production and the secretory pathway were not significantly altered. Interestingly, pretreatment with α-tocopherol nearly recovered the hyposecretion of mucus from both amphibian and human goblet cells. We believe this study demonstrates the mucotoxicity of PM and the protective function of α-tocopherol on mucotoxicity caused by acute PM exposure from heavy diesel engines.
Collapse
|
29
|
Lodes N, Seidensticker K, Perniss A, Nietzer S, Oberwinkler H, May T, Walles T, Hebestreit H, Hackenberg S, Steinke M. Investigation on Ciliary Functionality of Different Airway Epithelial Cell Lines in Three-Dimensional Cell Culture. Tissue Eng Part A 2019; 26:432-440. [PMID: 31696788 PMCID: PMC7187987 DOI: 10.1089/ten.tea.2019.0188] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Three-dimensional respiratory tissue models have been generated using, for example, human primary airway epithelial cells (hAEC) or respective cell lines. To investigate ciliopathies, such as primary ciliary dyskinesia, the presence of functional kinocilia in vitro is an essential prerequisite. Since access to hAEC of healthy donors is limited, we aimed to identify a respiratory epithelial cell line that is capable to display functional kinocilia on at least 60% of the apical surface. Thus, we cultured four different human respiratory cell lines with human primary airway fibroblasts under airlift conditions, characterized the morphology, and analyzed ciliary function. Only one of the tested cell lines showed beating kinocilia; however, <10% of the whole surface was covered and ciliary beating was undirected. Positive control tissue models using hAEC and fibroblasts displayed expected directed ciliary beating pattern around 11 Hz. Our data show that the available cell lines are not suitable for basic and applied research questions whenever functional kinocilia are required and that, rather, hAEC- or human induced pluripotent stem cell-derived tissue models need to be generated.
Collapse
Affiliation(s)
- Nina Lodes
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany.,Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Katharina Seidensticker
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Alexander Perniss
- Institute for Anatomy and Cell Biology, German Center for Lung Research DZL, Justus-Liebig-University Giessen, Giessen, Germany
| | - Sarah Nietzer
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany.,Translational Center Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
| | - Heike Oberwinkler
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | - Thorsten Walles
- Department of Thoracic Surgery, University Medicine Magdeburg, Magdeburg, Germany
| | - Helge Hebestreit
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Stephan Hackenberg
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Maria Steinke
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany.,Translational Center Regenerative Therapies, Fraunhofer Institute for Silicate Research ISC, Würzburg, Germany
| |
Collapse
|
30
|
Gupta R, Radicioni G, Abdelwahab S, Dang H, Carpenter J, Chua M, Mieczkowski PA, Sheridan JT, Randell SH, Kesimer M. Intercellular Communication between Airway Epithelial Cells Is Mediated by Exosome-Like Vesicles. Am J Respir Cell Mol Biol 2019; 60:209-220. [PMID: 30230353 DOI: 10.1165/rcmb.2018-0156oc] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Airway epithelium structure/function can be altered by local inflammatory/immune signals, and this process is called epithelial remodeling. The mechanism by which this innate response is regulated, which causes mucin/mucus overproduction, is largely unknown. Exosomes are nanovesicles that can be secreted and internalized by cells to transport cellular cargo, such as proteins, lipids, and miRNA. The objective of this study was to understand the role exosomes play in airway remodeling through cell-cell communication. We used two different human airway cell cultures: primary human tracheobronchial (HTBE) cells, and a cultured airway epithelial cell line (Calu-3). After intercellular exosomal transfer, comprehensive proteomic and genomic characterization of cell secretions and exosomes was performed. Quantitative proteomics and exosomal miRNA analysis profiles indicated that the two cell types are fundamentally distinct. HTBE cell secretions were typically dominated by fundamental innate/protective proteins, including mucin MUC5B, and Calu-3 cell secretions were dominated by pathology-associated proteins, including mucin MUC5AC. After exosomal transfer/intake, approximately 20% of proteins, including MUC5AC and MUC5B, were significantly altered in HTBE secretions. After exosome transfer, approximately 90 miRNAs (∼4%) were upregulated in HTBE exosomes, whereas Calu-3 exosomes exhibited a preserved miRNA profile. Together, our data suggest that the transfer of exosomal cargo between airway epithelial cells significantly alters the qualitative and quantitative profiles of airway secretions, including mucin hypersecretion, and the miRNA cargo of exosomes in target cells. This finding indicates that cellular information can be carried between airway epithelial cells via exosomes, which may play an important role in airway biology and epithelial remodeling.
Collapse
Affiliation(s)
- Richa Gupta
- 1 Department of Pathology and Laboratory Medicine.,2 Marsico Lung Institute
| | - Giorgia Radicioni
- 1 Department of Pathology and Laboratory Medicine.,2 Marsico Lung Institute
| | - Sabri Abdelwahab
- 1 Department of Pathology and Laboratory Medicine.,2 Marsico Lung Institute
| | | | - Jerome Carpenter
- 1 Department of Pathology and Laboratory Medicine.,2 Marsico Lung Institute
| | | | | | - John T Sheridan
- 1 Department of Pathology and Laboratory Medicine.,2 Marsico Lung Institute
| | - Scott H Randell
- 2 Marsico Lung Institute.,4 Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Mehmet Kesimer
- 1 Department of Pathology and Laboratory Medicine.,2 Marsico Lung Institute
| |
Collapse
|
31
|
Atanasova KR, Reznikov LR. Strategies for measuring airway mucus and mucins. Respir Res 2019; 20:261. [PMID: 31752894 PMCID: PMC6873701 DOI: 10.1186/s12931-019-1239-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
Mucus secretion and mucociliary transport are essential defense mechanisms of the airways. Deviations in mucus composition and secretion can impede mucociliary transport and elicit airway obstruction. As such, mucus abnormalities are hallmark features of many respiratory diseases, including asthma, cystic fibrosis and chronic obstructive pulmonary disease (COPD). Studying mucus composition and its physical properties has therefore been of significant interest both clinically and scientifically. Yet, measuring mucus production, output, composition and transport presents several challenges. Here we summarize and discuss the advantages and limitations of several techniques from five broadly characterized strategies used to measure mucus secretion, composition and mucociliary transport, with an emphasis on the gel-forming mucins. Further, we summarize advances in the field, as well as suggest potential areas of improvement moving forward.
Collapse
Affiliation(s)
- Kalina R Atanasova
- Department of Physiological Sciences, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA
| | - Leah R Reznikov
- Department of Physiological Sciences, University of Florida, 1333 Center Drive, PO Box 100144, Gainesville, FL, 32610, USA.
| |
Collapse
|
32
|
Chen Y, Kumar RK, Thomas PS, Herbert C. Th1/17-Biased Inflammatory Environment Associated with COPD Alters the Response of Airway Epithelial Cells to Viral and Bacterial Stimuli. Mediators Inflamm 2019; 2019:7281462. [PMID: 31534438 PMCID: PMC6732592 DOI: 10.1155/2019/7281462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/25/2019] [Accepted: 07/22/2019] [Indexed: 01/20/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by airway inflammation associated with a Th1/17-biased cytokine environment. Acute exacerbations of COPD (AECOPD) are most often triggered by respiratory infections, which elicit an exaggerated inflammatory response in these patients, via poorly defined mechanisms. We investigated the responses of airway epithelial cells (AECs) to infective stimuli in COPD and the effects of the Th1/17-biased environment on these responses. Cytokine expression was assessed following exposure to virus-like stimuli (poly I:C or imiquimod) or bacterial LPS. The effects of pretreatment with Th1/17 cytokines were evaluated in both primary AECs and the Calu-3 AEC cell line. We found that poly I:C induced increased expression of the proinflammatory cytokines IL1β, IL6, CXCL8, and TNF and IFN-β1 in AECs from both control subjects and COPD patients. Expression of IL1β in response to all 3 stimuli was significantly enhanced in COPD AECs. Primary AECs pretreated with Th1/17 cytokines exhibited enhanced expression of mRNA for proinflammatory cytokines in response to poly I:C. Similarly, Calu-3 cells responded to virus-like/bacterial stimuli with increased expression of proinflammatory cytokines, and a Th1/17 environment significantly enhanced their expression. Furthermore, increased expression of pattern recognition receptors for viruses (TLR3, TLR7, IFIH1, and DDX58) was induced by Th1/17 cytokines, in both primary AECs and Calu-3 cells. These findings suggest that the Th1/17-biased environment associated with COPD may enhance the proinflammatory cytokine response of AECs to viral and bacterial infections and that increased signaling via upregulated receptors may contribute to exaggerated inflammation in virus-induced AECOPD.
Collapse
Affiliation(s)
- Yifan Chen
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney 2052, Australia
| | - Rakesh K. Kumar
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney 2052, Australia
| | - Paul S. Thomas
- Prince of Wales Clinical School, Faculty of Medicine, UNSW Sydney, Sydney 2052, Australia
- Department of Respiratory Medicine, Prince of Wales Hospital, Sydney 2031, Australia
| | - Cristan Herbert
- Mechanisms of Disease and Translational Research, School of Medical Sciences, UNSW Sydney, Sydney 2052, Australia
| |
Collapse
|
33
|
Grygorczyk R, Boudreault F, Tan JJ, Ponomarchuk O, Sokabe M, Furuya K. Mechanosensitive ATP release in the lungs: New insights from real-time luminescence imaging studies. CURRENT TOPICS IN MEMBRANES 2019; 83:45-76. [PMID: 31196610 DOI: 10.1016/bs.ctm.2019.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Extracellular ATP and other nucleotides are important autocrine/paracrine mediators that stimulate purinergic receptors and regulate diverse processes in the normal lungs. They are also associated with pathogenesis of a number of respiratory diseases and clinical complications including acute respiratory distress syndrome and ventilator induced lung injury. Mechanical forces are major stimuli for cellular ATP release but precise mechanisms responsible for this release are still debated. The present review intends to provide the current state of knowledge of the mechanisms of ATP release in the lung. Putative pathways of the release, including the contribution of cell membrane injury and cell lysis are discussed addressing their strength, weaknesses and missing evidence that requires future study. We also provide an overview of the recent technical advances in studying cellular ATP release in vitro and ex vivo. Special attention is given to new insights into lung ATP release obtained with the real-time luminescence ATP imaging. This includes recent data on stretch-induced mechanosensitive ATP release in a model and primary cells of lung alveoli in vitro as well as inflation-induced ATP release in airspaces and pulmonary blood vessels of lungs, ex vivo.
Collapse
Affiliation(s)
- Ryszard Grygorczyk
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.
| | - Francis Boudreault
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Ju Jing Tan
- Department of Medicine, Université de Montréal, Montréal, QC, Canada; Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Olga Ponomarchuk
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada; Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| | - Kishio Furuya
- Mechanobiology Laboratory, Nagoya University, Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
34
|
Evert C, Loesekann T, Bhat G, Shajahan A, Sonon R, Azadi P, Hunter RC. Generation of 13C-Labeled MUC5AC Mucin Oligosaccharides for Stable Isotope Probing of Host-Associated Microbial Communities. ACS Infect Dis 2019; 5:385-393. [PMID: 30623643 DOI: 10.1021/acsinfecdis.8b00296] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stable isotope probing (SIP) has emerged as a powerful tool to address key questions about microbiota structure and function. To date, diverse isotopically labeled substrates have been used to characterize in situ growth activity of specific bacterial taxa and have revealed the flux of bioavailable substrates through microbial communities associated with health and disease. A major limitation to the growth of the field is the dearth of biologically relevant "heavy" labeled substrates. Mucin glycoproteins, for example, comprise an abundant source of carbon in the gut, oral cavity, respiratory tract, and other mucosal surfaces but are not commercially available. Here, we describe a method to incorporate a 13C-labeled monosaccharide into MUC5AC, a predominant mucin in both gastrointestinal and airway environments. Using the lung adenocarcinoma cell line, Calu-3, polarized cell cultures grown in 13C-labeled d-glucose resulted in liberal mucin production on the apical surface. Mucins were isolated by size-exclusion chromatography, and O-linked glycans were released by β-elimination, permethylated, and analyzed by electrospray ionization tandem mass spectrometry (ESI-MS/MS) and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) techniques. We demonstrate a 98.7% incorporation of 13C in the heterogeneous O-linked oligosaccharides that make up >80% of mucin dry weight. These "heavy" labeled glycoproteins represent a valuable tool for probing in vivo activity of host-associated bacterial communities and their interactions with the mucosal barrier. The continued expansion of labeled substrates for use in SIP will eventually allow bacterial taxa that degrade host compounds to be identified, with long-term potential for improved health and disease management.
Collapse
Affiliation(s)
- Clayton Evert
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Tina Loesekann
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Ganapati Bhat
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Roberto Sonon
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Ryan C. Hunter
- Department of Microbiology & Immunology, University of Minnesota, 689 23rd Avenue SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
35
|
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
|
36
|
Webster MJ, Tarran R. Slippery When Wet: Airway Surface Liquid Homeostasis and Mucus Hydration. CURRENT TOPICS IN MEMBRANES 2018; 81:293-335. [PMID: 30243435 DOI: 10.1016/bs.ctm.2018.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ability to regulate cell volume is crucial for normal physiology; equally the regulation of extracellular fluid homeostasis is of great importance. Alteration of normal extracellular fluid homeostasis contributes to the development of several diseases including cystic fibrosis. With regard to the airway surface liquid (ASL), which lies apically on top of airway epithelia, ion content, pH, mucin and protein abundance must be tightly regulated. Furthermore, airway epithelia must be able to switch from an absorptive to a secretory state as required. A heterogeneous population of airway epithelial cells regulate ASL solute and solvent composition, and directly secrete large mucin molecules, antimicrobials, proteases and soluble mediators into the airway lumen. This review focuses on how epithelial ion transport influences ASL hydration and ASL pH, with a specific focus on the roles of anion and cation channels and exchangers. The role of ions and pH in mucin expansion is also addressed. With regard to fluid volume regulation, we discuss the roles of nucleotides, adenosine and the short palate lung and nasal epithelial clone 1 (SPLUNC1) as soluble ASL mediators. Together, these mechanisms directly influence ciliary beating and in turn mucociliary clearance to maintain sterility and to detoxify the airways. Whilst all of these components are regulated in normal airways, defective ion transport and/or mucin secretion proves detrimental to lung homeostasis as such we address how defective ion and fluid transport, and a loss of homeostatic mechanisms, contributes to the development of pathophysiologies associated with cystic fibrosis.
Collapse
Affiliation(s)
- Megan J Webster
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Robert Tarran
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
37
|
Xu R, Zhou J, Zhou XD, Li Q, Perelman JM, Kolosov VP. Munc13‑4 mediates human neutrophil elastase‑induced airway mucin5AC hypersecretion by interacting with syntaxin2. Mol Med Rep 2018; 18:1015-1024. [PMID: 29767240 DOI: 10.3892/mmr.2018.9015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 03/27/2018] [Indexed: 11/06/2022] Open
Abstract
The overexpression and hypersecretion of mucus is a hallmark of chronic pulmonary inflammatory disease. Mucin5AC (MUC5AC) is a major component of airway gel‑forming mucin. Members of the Unc13 (Munc13) protein family act as important activators of granule exocytosis from various types of mammalian cells. The present study aimed to determine the role of Munc13 family proteins in MUC5AC secretion via an in vitro study with BEAS‑2B and Calu‑3 cell lines. Reverse transcription‑quantitative polymerase chain reaction and western blotting indicated that stimulation of the cells with 100 nM human neutrophil elastase (hNE) for 1 h did not affect the expression of either unc13 homolog B (Munc13‑2) or unc13 homolog D (Munc13‑4), but immunofluorescence analysis demonstrated that hNE treatment was associated with the recruitment of Munc13‑4 to the plasma membrane. Co‑immunoprecipitation analysis indicated increased binding between Munc13‑4 and syntaxin2 followingh NE stimulation; however, Munc13‑2 formed a stable interaction with syntaxin2 with or without hNE stimulation. Subsequently, Munc13‑2 and Munc13‑4 expression levels were downregulated in BEAS‑2B and Calu‑3 cells using small interfering RNA (siRNA). ELISAs and immunofluorescence analysis were performed to assess MUC5AC secretion and intracellular retention, respectively. Munc13‑2 siRNA transfection did not alter the expression levels of intracellular or secreted MUC5AC following hNE stimulation in either cell line; however, it increased the baseline intracellular levels of MUC5AC and decreased the amount of secreted MUC5AC. Conversely, Munc13‑4 siRNA transfection increased the intracellular levels of MUC5AC and decreased the amount of secreted MUC5AC following hNE stimulation, but did not affect their baseline quantities. The results of the present study indicate that Munc13‑2 may be an essential regulator of basal MUC5AC exocytosis, while Munc13‑4 appears to be a Munc13 protein subtype that may to be sensitive to hNE stimulation during airway MUC5AC hypersecretion.
Collapse
Affiliation(s)
- Rui Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jia Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiang-Dong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570102, P.R. China
| | - Juliy M Perelman
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk 675000, Russia
| | - Victor P Kolosov
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk 675000, Russia
| |
Collapse
|
38
|
Cardenas EI, Breaux K, Da Q, Flores JR, Ramos MA, Tuvim MJ, Burns AR, Rumbaut RE, Adachi R. Platelet Munc13-4 regulates hemostasis, thrombosis and airway inflammation. Haematologica 2018; 103:1235-1244. [PMID: 29674495 PMCID: PMC6029531 DOI: 10.3324/haematol.2017.185637] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/12/2018] [Indexed: 01/15/2023] Open
Abstract
Platelet degranulation is crucial for hemostasis and may participate in inflammation. Exocytosis in platelets is mediated by SNARE proteins and should be controlled by Munc13 proteins. We found that platelets express Munc13-2 and -4. We assessed platelet granule exocytosis in Munc13-2 and -4 global and conditional knockout (KO) mice, and observed that deletion of Munc13-4 ablates dense granule release and indirectly impairs alpha granule exocytosis. We found no exocytic role for Munc13-2 in platelets, not even in the absence of Munc13-4. In vitro, Munc13-4-deficient platelets exhibited defective aggregation at low doses of collagen. In a flow chamber assay, we observed that Munc13-4 acted as a rate-limiting factor in the formation of thrombi. In vivo, we observed a dose-dependency between Munc13-4 expression in platelets and both venous bleeding time and time to arterial thrombosis. Finally, in a model of allergic airway inflammation, we found that platelet-specific Munc13-4 KO mice had a reduction in airway hyper-responsiveness and eosinophilic inflammation. Taken together, our results indicate that Munc13-4-dependent platelet dense granule release plays essential roles in hemostasis, thrombosis and allergic inflammation.
Collapse
Affiliation(s)
- Eduardo I Cardenas
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Monterrey, Mexico
| | - Keegan Breaux
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Da
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jose R Flores
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marco A Ramos
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael J Tuvim
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alan R Burns
- College of Optometry, University of Houston, TX, USA
| | - Rolando E Rumbaut
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Roberto Adachi
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
39
|
Samaniego Lopez C, Hebe Martínez J, Uhrig ML, Coluccio Leskow F, Spagnuolo CC. A Highly Sensitive Fluorogenic Probe for Imaging Glycoproteins and Mucine Activity in Live Cells in the Near-Infrared Region. Chemistry 2018; 24:6344-6348. [DOI: 10.1002/chem.201800790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Cecilia Samaniego Lopez
- Departamento de Química Orgánica-CIHIDECAR-CONICET; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - Jimena Hebe Martínez
- IQUIBICEN-CONICET, Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - María Laura Uhrig
- Departamento de Química Orgánica-CIHIDECAR-CONICET; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - Federico Coluccio Leskow
- IQUIBICEN-CONICET, Departamento de Química Biológica; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| | - Carla Cecilia Spagnuolo
- Departamento de Química Orgánica-CIHIDECAR-CONICET; Facultad de Ciencias Exactas y Naturales; Universidad de Buenos Aires; Int. Guiraldes 2160 1428 Ciudad de Buenos Aires Argentina
| |
Collapse
|
40
|
Sim HJ, Kim SH, Myung KJ, Kwon T, Lee HS, Park TJ. Xenopus: An alternative model system for identifying muco-active agents. PLoS One 2018; 13:e0193310. [PMID: 29470529 PMCID: PMC5823443 DOI: 10.1371/journal.pone.0193310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/08/2018] [Indexed: 12/03/2022] Open
Abstract
The airway epithelium in human plays a central role as the first line of defense against environmental contaminants. Most respiratory diseases such as chronic obstructive pulmonary disease (COPD), asthma, and respiratory infections, disturb normal muco-ciliary functions by stimulating the hypersecretion of mucus. Several muco-active agents have been used to treat hypersecretion symptoms in patients. Current muco-active reagents control mucus secretion by modulating either airway inflammation, cholinergic parasympathetic nerve activities or by reducing the viscosity by cleaving crosslinking in mucin and digesting DNAs in mucus. However, none of the current medication regulates mucus secretion by directly targeting airway goblet cells. The major hurdle for screening potential muco-active agents that directly affect the goblet cells, is the unavailability of in vivo model systems suitable for high-throughput screening. In this study, we developed a high-throughput in vivo model system for identifying muco-active reagents using Xenopus laevis embryos. We tested mucus secretion under various conditions and developed a screening strategy to identify potential muco-regulators. Using this novel screening technique, we identified narasin as a potential muco-regulator. Narasin treatment of developing Xenopus embryos significantly reduced mucus secretion. Furthermore, the human lung epithelial cell line, Calu-3, responded similarly to narasin treatment, validating our technique for discovering muco-active reagents.
Collapse
Affiliation(s)
- Hyo Jung Sim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung-Jae Myung
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Taejoon Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
| | - Hyun-Shik Lee
- College of Natural Sciences, Kyungpook National University, Daegu, South Korea
- * E-mail: (TJP); (HSL)
| | - Tae Joo Park
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, South Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
- * E-mail: (TJP); (HSL)
| |
Collapse
|
41
|
Fate of PEGylated antibody fragments following delivery to the lungs: Influence of delivery site, PEG size and lung inflammation. J Control Release 2017; 272:62-71. [PMID: 29247664 DOI: 10.1016/j.jconrel.2017.12.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/11/2017] [Accepted: 12/13/2017] [Indexed: 12/21/2022]
Abstract
Pulmonary administration of anti-cytokine antibodies offers a targeted therapy in asthma. However, the rapid elimination of proteins from the lungs limits the efficacy of inhaled medications. PEGylation has been shown to increase the residence time of anti-interleukin (IL)-17A and anti-IL-13 antibody fragments in the lungs and to improve their therapeutic efficacy. Yet, little is known about the factors that affect the residence time of PEGylated antibody fragments in the lungs following pulmonary delivery. In this study, we showed that the molecular weight of polyethylene glycol (PEG), 20kDa or 40kDa, had a moderate effect on the residence time of an anti-IL-17A Fab' fragment in the lungs of mice. By contrast, the site of delivery of the anti-IL-17A and anti-IL-13 Fab' fragments within the lungs had a major impact on their residence time, with the deeper the delivery, the more prolonged the residence time. The nature of the Fab' fragment had an influence on its residence time as well and the anti-IL-17A Fab' benefited more from PEGylation than the anti-IL-13 Fab' did. Acute lung inflammation slightly shortened the residence time of the anti-IL-17A and anti-IL-13 Fab' fragments in the lungs but PEGylation was able to prolong their presence in both the healthy and inflamed lungs. Antibody fragments were predominately located within the airway lumen rather than the lung parenchyma. Transport experiments on monolayers of Calu-3 cells and studies of fluorescence recovery after photobleaching in respiratory mucus showed that mechanisms involved in the prolonged presence of PEGylated Fab' in the airway lumen might include binding to the mucus, reduced uptake by respiratory cells and reduced transport across lung epithelia. Finally, using I125-labeled anti-IL-17A Fab', we showed that the protein fragment hardly penetrated into the lungs following subcutaneous injection, as opposed to pulmonary delivery.
Collapse
|
42
|
Airway surface liquid pH is not acidic in children with cystic fibrosis. Nat Commun 2017; 8:1409. [PMID: 29123085 PMCID: PMC5680186 DOI: 10.1038/s41467-017-00532-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 07/05/2017] [Indexed: 01/08/2023] Open
Abstract
Modulation of airway surface liquid (ASL) pH has been proposed as a therapy for cystic fibrosis (CF). However, evidence that ASL pH is reduced in CF is limited and conflicting. The technical challenges associated with measuring ASL pH in vivo have precluded accurate measurements in humans. In order to address this deficiency, ASL pH was measured in vivo in children using a novel luminescent technology integrated with fibre-optic probes. Here we show that ASL pH in children with CF is similar to that of children without CF. Findings were supported by highly controlled direct pH measurements in primary human airway epithelial cell culture models, which also suggest that the potential ASL pH gradient produced by defective apical ion transport is balanced out by paracellular shunting of acid/base. Thus, reduced baseline ASL pH is unlikely to be an important pathobiological factor in early CF lung disease. Modulation of airway surface liquid pH has been proposed as a therapy for cystic fibrosis, but whether pH is indeed altered in cystic fibrosis is controversial. Here, the authors develop a novel fibre-optic based pH measurement technology, and show that pH is not altered in children with cystic fibrosis.
Collapse
|
43
|
Khalafalla FG, Kayani W, Kassab A, Ilves K, Monsanto MM, Alvarez R, Chavarria M, Norman B, Dembitsky WP, Sussman MA. Empowering human cardiac progenitor cells by P2Y 14 nucleotide receptor overexpression. J Physiol 2017; 595:7135-7148. [PMID: 28980705 DOI: 10.1113/jp274980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/27/2017] [Indexed: 01/10/2023] Open
Abstract
KEY POINTS Autologous cardiac progenitor cell (CPC) therapy is a promising approach for treatment of heart failure (HF). There is an unmet need to identify inherent deficits in aged/diseased human CPCs (hCPCs) derived from HF patients in the attempts to augment their regenerative capacity prior to use in the clinical setting. Here we report significant functional correlations between phenotypic properties of hCPCs isolated from cardiac biopsies of HF patients, clinical parameters of patients and expression of the P2Y14 purinergic receptor (P2Y14 R), a crucial detector for extracellular UDP-sugars released during injury/stress. P2Y14 R is downregulated in hCPCs derived from HF patients with lower ejection fraction or diagnosed with diabetes. Augmenting P2Y14 R expression levels in aged/diseased hCPCs antagonizes senescence and improves functional responses. This study introduces purinergic signalling modulation as a potential strategy to rejuvenate and improve phenotypic characteristics of aged/functionally compromised hCPCs prior to transplantation in HF patients. ABSTRACT Autologous cardiac progenitor cell therapy is a promising alternative approach to current inefficient therapies for heart failure (HF). However, ex vivo expansion and pharmacological/genetic modification of human cardiac progenitor cells (hCPCs) are necessary interventions to rejuvenate aged/diseased cells and improve their regenerative capacities. This study was designed to assess the potential of improving hCPC functional capacity by targeting the P2Y14 purinergic receptor (P2Y14 R), which has been previously reported to induce regenerative and anti-senescence responses in a variety of experimental models. c-Kit+ hCPCs were isolated from cardiac biopsies of multiple HF patients undergoing left ventricular assist device implantation surgery. Significant correlations existed between the expression of P2Y14 R in hCPCs and clinical parameters of HF patients. P2Y14 R was downregulated in hCPCs derived from patients with a relatively lower ejection fraction and patients diagnosed with diabetes. hCPC lines with lower P2Y14 R expression did not respond to P2Y14 R agonist UDP-glucose (UDP-Glu) while hCPCs with higher P2Y14 R expression showed enhanced proliferation in response to UDP-Glu stimulation. Mechanistically, UDP-Glu stimulation enhanced the activation of canonical growth signalling pathways ERK1/2 and AKT. Restoring P2Y14 R expression levels in functionally compromised hCPCs via lentiviral-mediated overexpression improved proliferation, migration and survival under stress stimuli. Additionally, P2Y14 R overexpression reversed senescence-associated morphology and reduced levels of molecular markers of senescence p16INK4a , p53, p21 and mitochondrial reactive oxygen species. Findings from this study unveil novel biological roles of the UDP-sugar receptor P2Y14 in hCPCs and suggest purinergic signalling modulation as a promising strategy to improve phenotypic properties of functionally impaired hCPCs.
Collapse
Affiliation(s)
- Farid G Khalafalla
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Waqas Kayani
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Arwa Kassab
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Kelli Ilves
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Megan M Monsanto
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Roberto Alvarez
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Monica Chavarria
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Benjamin Norman
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | | | - Mark A Sussman
- San Diego Heart Research Institute, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| |
Collapse
|
44
|
The properties of the mucus barrier, a unique gel--how can nanoparticles cross it? Ther Deliv 2016; 7:229-44. [PMID: 27010985 DOI: 10.4155/tde-2015-0002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The key criterion for a nanoparticle drug-delivery system is the ability to produce substantial bioavailability without damaging the physiological protective mechanisms. The main area for drug delivery is the aerodigestive tract. All epithelial surfaces have a membrane-bound layer and in the lung this layer is surmounted by a gel layer. In the gastrointestinal tract the membrane-bound mucin layer is covered by a mucus bilayer. The pore sizes of mucus gels are around 100 to 200 nm. Consequently, only nanoparticles in this size range could potentially penetrate without modification of these layers. To study nanoparticle permeation with results that pertain to in vivo conditions, native mucus mucin preparations must be used. Strategies to increase pores in mucus gels are discussed herein.
Collapse
|
45
|
Furuya K, Tan JJ, Boudreault F, Sokabe M, Berthiaume Y, Grygorczyk R. Real-time imaging of inflation-induced ATP release in the ex vivo rat lung. Am J Physiol Lung Cell Mol Physiol 2016; 311:L956-L969. [PMID: 27638905 DOI: 10.1152/ajplung.00425.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 09/13/2016] [Indexed: 12/25/2022] Open
Abstract
Extracellular ATP and other nucleotides are important autocrine/paracrine mediators that regulate diverse processes critical for lung function, including mucociliary clearance, surfactant secretion, and local blood flow. Cellular ATP release is mechanosensitive; however, the impact of physical stimuli on ATP release during breathing has never been tested in intact lungs in real time and remains elusive. In this pilot study, we investigated inflation-induced ATP release in rat lungs ex vivo by real-time luciferin-luciferase (LL) bioluminescence imaging coupled with simultaneous infrared tissue imaging to identify ATP-releasing sites. With LL solution introduced into air spaces, brief inflation of such edematous lung (1 s, ∼20 cmH2O) induced transient (<30 s) ATP release in a limited number of air-inflated alveolar sacs during their recruitment/opening. Released ATP reached concentrations of ∼10-6 M, relevant for autocrine/paracrine signaling, but it remained spatially restricted to single alveolar sacs or their clusters. ATP release was stimulus dependent: prolonged (100 s) inflation evoked long-lasting ATP release that terminated upon alveoli deflation/derecruitment while cyclic inflation/suction produced cyclic ATP release. With LL introduced into blood vessels, inflation induced transient ATP release in many small patchlike areas the size of alveolar sacs. Findings suggest that inflation induces ATP release in both alveoli and the surrounding blood capillary network; the functional units of ATP release presumably consist of alveolar sacs or their clusters. Our study demonstrates the feasibility of real-time ATP release imaging in ex vivo lungs and provides the first direct evidence of inflation-induced ATP release in lung air spaces and in pulmonary blood capillaries, highlighting the importance of purinergic signaling in lung function.
Collapse
Affiliation(s)
- Kishio Furuya
- Mechanobiology Laboratory, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Ju Jing Tan
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Francis Boudreault
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Masahiro Sokabe
- Mechanobiology Laboratory, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yves Berthiaume
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; and.,Institut de recherches cliniques de Montréal (IRCM), Quebec, Canada
| | - Ryszard Grygorczyk
- Centre de recherche, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; .,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada; and
| |
Collapse
|
46
|
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
|
47
|
Zaidman NA, Panoskaltsis-Mortari A, O'Grady SM. Differentiation of human bronchial epithelial cells: role of hydrocortisone in development of ion transport pathways involved in mucociliary clearance. Am J Physiol Cell Physiol 2016; 311:C225-36. [PMID: 27306366 PMCID: PMC5129773 DOI: 10.1152/ajpcell.00073.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/10/2016] [Indexed: 01/27/2023]
Abstract
Glucocorticoids strongly influence the mucosal-defense functions performed by the bronchial epithelium, and inhaled corticosteroids are critical in the treatment of patients with inflammatory airway diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis. A common pathology associated with these diseases is reduced mucociliary clearance, a defense mechanism involving the coordinated transport of salt, water, and mucus by the bronchial epithelium, ultimately leading to retention of pathogens and particles in the airways and to further disease progression. In the present study we investigated the role of hydrocortisone (HC) in differentiation and development of the ion transport phenotype of normal human bronchial epithelial cells under air-liquid interface conditions. Normal human bronchial epithelial cells differentiated in the absence of HC (HC0) showed significantly less benzamil-sensitive short-circuit current than controls, as well as a reduced response after stimulation with the selective β2-adrenergic receptor agonist salbutamol. Apical membrane localization of epithelial Na(+) channel α-subunits was similarly reduced in HC0 cells compared with controls, supporting a role of HC in the trafficking and density of Na(+) channels in the plasma membrane. Additionally, glucocorticoid exposure during differentiation regulated the transcription of cystic fibrosis transmembrane conductance regulator and β2-adrenergic receptor mRNAs and appeared to be necessary for the expression of cystic fibrosis transmembrane conductance regulator-dependent anion secretion in response to β2-agonists. HC had no significant effect on surface cell differentiation but did modulate the expression of mucin mRNAs. These findings indicate that glucocorticoids support mucosal defense by regulating critical transport pathways essential for effective mucociliary clearance.
Collapse
Affiliation(s)
- Nathan A Zaidman
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota
| | - Angela Panoskaltsis-Mortari
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota; and
| | - Scott M O'Grady
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota; Department of Animal Science, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
48
|
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
|
49
|
Chang AY, Mann TS, McFawn PK, Han L, Dong X, Henry PJ. Investigating the role of MRGPRC11 and capsaicin-sensitive afferent nerves in the anti-influenza effects exerted by SLIGRL-amide in murine airways. Respir Res 2016; 17:62. [PMID: 27215903 PMCID: PMC4877944 DOI: 10.1186/s12931-016-0378-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/15/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The hexapeptide SLIGRL-amide activates protease-activated receptor-2 (PAR-2) and mas-related G protein-coupled receptor C11 (MRGPRC11), both of which are known to be expressed on populations of sensory nerves. SLIGRL-amide has recently been reported to inhibit influenza A (IAV) infection in mice independently of PAR-2 activation, however the explicit roles of MRGPRC11 and sensory nerves in this process are unknown. Thus, the principal aim of this study was to determine whether SLIGRL-amide-induced inhibition of influenza infection is mediated by MRGPRC11 and/or by capsaicin-sensitive sensory nerves. METHODS The inhibitory effect of SLIGRL-amide on IAV infection observed in control mice in vivo was compared to effects produced in mice that did not express MRGPRC11 (mrgpr-cluster∆ (-/-) mice) or had impaired sensory nerve function (induced by chronic pre-treatment with capsaicin). Complementary mechanistic studies using both in vivo and ex vivo approaches investigated whether the anti-IAV activity of SLIGRL-amide was (1) mimicked by either activators of MRGPRC11 (BAM8-22) or by activators (acute capsaicin) or selected mediators (substance P, CGRP) of sensory nerve function, or (2) suppressed by inhibitors of sensory nerve function (e.g. NK1 receptor antagonists). RESULTS SLIGRL-amide and BAM8-22 dose-dependently inhibited IAV infection in mrgpr-cluster∆ (-/-) mice that do not express MRGPRC11. In addition, SLIGRL-amide and BAM8-22 each inhibited IAV infection in capsaicin-pre-treated mice that lack functional sensory nerves. Furthermore, the anti-IAV activity of SLIGRL-amide was not mimicked by the sensory neuropeptides substance P or CGRP, nor blocked by either NK1 (L-703,606, RP67580) and CGRP receptor (CGRP8-37) antagonists. Direct stimulation of airway sensory nerves through acute exposure to the TRPV1 activator capsaicin also failed to mimic SLIGRL-amide-induced inhibition of IAV infectivity. The anti-IAV activity of SLIGRL-amide was mimicked by the purinoceptor agonist ATP, a direct activator of mucus secretion from airway epithelial cells. Additionally, both SLIGRL-amide and ATP stimulated mucus secretion and inhibited IAV infectivity in mouse isolated tracheal segments. CONCLUSIONS SLIGRL-amide inhibits IAV infection independently of MRGPRC11 and independently of capsaicin-sensitive, neuropeptide-releasing sensory nerves, and its secretory action on epithelial cells warrants further investigation.
Collapse
Affiliation(s)
- Amy Y Chang
- School of Medicine and Pharmacology, University of Western Australia, Crawley, WA, 6009, Australia.,School of Anatomy, Physiology & Human Biology, University of Western Australia, Crawley, 6009, WA, Australia
| | - Tracy S Mann
- School of Medicine and Pharmacology, University of Western Australia, Crawley, WA, 6009, Australia
| | - Peter K McFawn
- School of Anatomy, Physiology & Human Biology, University of Western Australia, Crawley, 6009, WA, Australia
| | - Liang Han
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Xinzhong Dong
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Peter J Henry
- School of Medicine and Pharmacology, University of Western Australia, Crawley, WA, 6009, Australia.
| |
Collapse
|
50
|
Bhowmick R, Gappa-Fahlenkamp H. Cells and Culture Systems Used to Model the Small Airway Epithelium. Lung 2016; 194:419-28. [PMID: 27071933 DOI: 10.1007/s00408-016-9875-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/01/2016] [Indexed: 01/28/2023]
Abstract
The pulmonary epithelium is divided into upper, lower, and alveolar (or small) airway epithelia and acts as the mechanical and immunological barrier between the external environment and the underlying submucosa. Of these, the small airway epithelium is the principal area of gas exchange and has high immunological activity, making it a major area of cell biology, immunology, and pharmaceutical research. As animal models do not faithfully represent the human pulmonary system and ex vivo human lung samples have reliability and availability issues, cell lines, and primary cells are widely used as small airway epithelial models. In vitro, these cells are mostly cultured as monolayers (2-dimensional cultures), either media submerged or at air-liquid interface. However, these 2-dimensional cultures lack a three dimension-a scaffolding extracellular matrix, which establishes the intercellular network in the in vivo airway epithelium. Therefore, 3-dimensional cell culture is currently a major area of development, where cells are cultured in a matrix or are cultured in a manner that they develop ECM-like scaffolds between them, thus mimicking the in vivo phenotype more faithfully. This review focuses on the commonly used small airway epithelial cells, their 2-dimensional and 3-dimensional culture techniques, and their comparative phenotype when cultured under these systems.
Collapse
Affiliation(s)
- Rudra Bhowmick
- Department of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA
| | - Heather Gappa-Fahlenkamp
- Department of Chemical Engineering, Oklahoma State University, 420 Engineering North, Stillwater, OK, 74078, USA.
| |
Collapse
|