1
|
Lin L, Wu Z, Zhong A, Luo H, Xu W, Luo W. Causal effects of genetically determined blood metabolites on asthma: a bidirectional Mendelian randomization study. J Asthma 2024; 61:1727-1737. [PMID: 39087774 DOI: 10.1080/02770903.2024.2380515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE The observational association between blood metabolites and asthma has been extensively studied. However, it is still unclear whether this association is causal. In this study, we aimed to investigate the causal relationship between blood metabolites and asthma using a bidirectional Mendelian randomization (MR) analysis. Additionally, we aimed to explore the potential mechanisms underlying this relationship. METHODS The study design involved the use of genetic instruments as instrumental variables (IVs) to fulfill the assumptions of MR analysis. The data on 1,091 metabolites and 309 metabolite ratios were obtained from the Canadian Longitudinal Study on Aging (CLSA), while the data on asthma were obtained from the Integrative Epidemiology Unit (IEU) Open GWAS Project. Utilizing the inverse variance-weighted (IVW) method as the primary MR analysis approach, sensitivity tests were conducted to assess the reliability of the findings, which involved employing Cochran's Q and the MR-Egger intercept. Furthermore, Bayesian weighted MR was used to further test the robustness of the results. Additionally, pathway analysis was conducted to explore the metabolic explanations underlying asthma. RESULT In our study, a comprehensive MR Analysis identified 10 metabolites and 6 metabolite ratios significantly associated with the development of asthma (FDR < 0.05). The metabolites included glycerophosphocholines(GPCs), glycerophosphoethanolamines(GPEs), and an unknown metabolite. Of these, 1-arachidonoyl-GPC, 1-myristoyl-2-arachidonoyl-GPC, 1-palmitoyl-2-arachidonoyl-GPC, and 1-(1-enyl-palmitoyl)-2-arachidonoyl-GPC were associated with an increased risk of asthma, whereas 1,2-dilinoleoyl-GPC, 1-palmitoyl-2-linoleoyl-GPC, 1,2-dilinoleoyl-GPE, 1 - oleoyl - 2 - linoleoyl - GPE, 1-palmitoyl-2-linoleoyl-GPE, and X-21470 were found to have a protective effect. No heterogeneity and pleiotropy were observed in the significant metabolites (p > 0.05), and each metabolite exhibited a consistent effect direction across all five methods. BWMR analysis results confirmed the significance and direction of effects across exposures, except for Cholesterol to linoleoyl-arachidonoyl-glycerol ratio(p = 0.673). Pathway analysis suggests that glycerophospholipid metabolism may potentially be a mechanism underlying the development of asthma. CONCLUSION Our MR findings suggest that the identified metabolites and pathways can serve as biomarkers for clinical asthma screening and prevention, while also providing new insights for future mechanistic exploration and drug target selection.
Collapse
Affiliation(s)
- Liyu Lin
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zilun Wu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Anqi Zhong
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haocheng Luo
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjie Xu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wen Luo
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Ramos CDO, Sant'Ana MR, Gonçalves GR, Rios TDS, Nakandakari SCBR, Burger B, Fernandes LGR, Zollner RDL, de Oliveira AN, Ramos RC, da Silva ASR, Pauli JR, de Moura LP, Ropelle ER, Mansour E, Cintra DE. The Effects of High-Fat Diet and Flaxseed Oil-Enriched Diet on the Lung Parenchyma of Obese Mice. Mol Nutr Food Res 2024; 68:e2300050. [PMID: 39205544 DOI: 10.1002/mnfr.202300050] [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: 01/29/2023] [Revised: 05/28/2024] [Indexed: 09/04/2024]
Abstract
Omega-3 (ω3) fatty acids are widely investigated for their anti-inflammatory potential, however, there is little evidence regarding their action in the lung parenchyma in the context of obesity. The objective is to investigate the effects of flaxseed oil (FS), rich in α-linolenic (C18:3 - ω3), on the lungs of obese mice. Mice were fed a high-fat diet (HF) for 8 weeks to induce obesity. Subsequently, a part of these animals received HF containing FS oil for another 8 weeks. The HF consumption induced weight gain and hyperglycemia. The lung parenchyma shows a complete fatty acids profile, compared to the control group (CT). In the lung parenchyma, FS increases the ω3 content and, notwithstanding a reduction in the interleukins (IL) IL1β and IL18 contents compared to HF. However, FS promoted increased alveolar spaces, followed by MCP1 (Monocytes Chemoattractant Protein-1) positive cell infiltration and a dramatic reduction in the anti-inflammatory cytokine, IL10. Despite reducing the pulmonary inflammatory response, the consumption of a food source of ω3 was associated with alterations in the lipid profile and histoarchitecture of the lung parenchyma, which can lead to the development of pulmonary complications. This study brings an alert against the indiscriminate use of ω3 supplements, warranting caution.
Collapse
Affiliation(s)
- Camila de Oliveira Ramos
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Marcella Ramos Sant'Ana
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Giovana Rios Gonçalves
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Thaiane da Silva Rios
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Susana Castelo Branco Ramos Nakandakari
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Beatriz Burger
- Laboratory of Nutrients and Tissue Repair, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | | | - Ricardo de Lima Zollner
- Laboratory of Translational Immunology, School of Medical Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Arthur Noin de Oliveira
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Rodrigo Catharino Ramos
- Innovare Biomarkers Laboratory, School of Pharmaceutical Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | | | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
- OCRC - Obesity and Comorbidities Research Center, UNICAMP, São Paulo, 13484-350, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
- OCRC - Obesity and Comorbidities Research Center, UNICAMP, São Paulo, 13484-350, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
- OCRC - Obesity and Comorbidities Research Center, UNICAMP, São Paulo, 13484-350, Brazil
| | - Eli Mansour
- Department of Clinical Medicine, School of Medical Sciences, UNICAMP, São Paulo, 13484-350, Brazil
| | - Dennys Esper Cintra
- Nutritional Genomics Laboratory, LabGeN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
- Nutrigenomics and Lipids Research Center, CELN, School of Applied Sciences, UNICAMP, São Paulo, 13484-350, Brazil
- OCRC - Obesity and Comorbidities Research Center, UNICAMP, São Paulo, 13484-350, Brazil
| |
Collapse
|
3
|
Gomez HM, Haw TJ, Ilic D, Robinson P, Donovan C, Croft AJ, Vanka KS, Small E, Carroll OR, Kim RY, Mayall JR, Beyene T, Palanisami T, Ngo DTM, Zosky GR, Holliday EG, Jensen ME, McDonald VM, Murphy VE, Gibson PG, Horvat JC. Landscape fire smoke airway exposure impairs respiratory and cardiac function and worsens experimental asthma. J Allergy Clin Immunol 2024; 154:209-221.e6. [PMID: 38513838 DOI: 10.1016/j.jaci.2024.02.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Millions of people are exposed to landscape fire smoke (LFS) globally, and inhalation of LFS particulate matter (PM) is associated with poor respiratory and cardiovascular outcomes. However, how LFS affects respiratory and cardiovascular function is less well understood. OBJECTIVE We aimed to characterize the pathophysiologic effects of representative LFS airway exposure on respiratory and cardiac function and on asthma outcomes. METHODS LFS was generated using a customized combustion chamber. In 8-week-old female BALB/c mice, low (25 μg/m3, 24-hour equivalent) or moderate (100 μg/m3, 24-hour equivalent) concentrations of LFS PM (10 μm and below [PM10]) were administered daily for 3 (short-term) and 14 (long-term) days in the presence and absence of experimental asthma. Lung inflammation, gene expression, structural changes, and lung function were assessed. In 8-week-old male C57BL/6 mice, low concentrations of LFS PM10 were administered for 3 days. Cardiac function and gene expression were assessed. RESULTS Short- and long-term LFS PM10 airway exposure increased airway hyperresponsiveness and induced steroid insensitivity in experimental asthma, independent of significant changes in airway inflammation. Long-term LFS PM10 airway exposure also decreased gas diffusion. Short-term LFS PM10 airway exposure decreased cardiac function and expression of gene changes relating to oxidative stress and cardiovascular pathologies. CONCLUSIONS We characterized significant detrimental effects of physiologically relevant concentrations and durations of LFS PM10 airway exposure on lung and heart function. Our study provides a platform for assessment of mechanisms that underpin LFS PM10 airway exposure on respiratory and cardiovascular disease outcomes.
Collapse
Affiliation(s)
- Henry M Gomez
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Tatt J Haw
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Dusan Ilic
- Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Peter Robinson
- Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Chantal Donovan
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; School of Life Sciences, University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - Amanda J Croft
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Kanth S Vanka
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; Newcastle Institute for Energy and Resources, University of Newcastle, Callaghan, Australia
| | - Ellen Small
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Olivia R Carroll
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Richard Y Kim
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia; School of Life Sciences, University of Technology Sydney, Faculty of Science, Sydney, Australia
| | - Jemma R Mayall
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Tesfalidet Beyene
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Thava Palanisami
- Global Innovative Centre for Advanced Nanomaterials, University of Newcastle, Callaghan, Australia
| | - Doan T M Ngo
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, Australia; College of Health, Medicine, and Wellbeing, Centre of Excellence Newcastle Cardio-Oncology Research Group, University of Newcastle, Callaghan, Newcastle, Australia
| | - Graeme R Zosky
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart, Australia; College of Health and Medicine, Tasmanian School of Medicine, University of Tasmania, Hobart, Australia
| | - Elizabeth G Holliday
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Megan E Jensen
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Vanessa M McDonald
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Vanessa E Murphy
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Peter G Gibson
- School of Medicine and Public Health, University of Newcastle and Asthma and Breathing Program, Hunter Medical Research Institute, Newcastle, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle and Immune Health Program, Hunter Medical Research Institute, Newcastle, Australia.
| |
Collapse
|
4
|
Budden KF, Shukla SD, Bowerman KL, Vaughan A, Gellatly SL, Wood DLA, Lachner N, Idrees S, Rehman SF, Faiz A, Patel VK, Donovan C, Alemao CA, Shen S, Amorim N, Majumder R, Vanka KS, Mason J, Haw TJ, Tillet B, Fricker M, Keely S, Hansbro N, Belz GT, Horvat J, Ashhurst T, van Vreden C, McGuire H, Fazekas de St Groth B, King NJC, Crossett B, Cordwell SJ, Bonaguro L, Schultze JL, Hamilton-Williams EE, Mann E, Forster SC, Cooper MA, Segal LN, Chotirmall SH, Collins P, Bowman R, Fong KM, Yang IA, Wark PAB, Dennis PG, Hugenholtz P, Hansbro PM. Faecal microbial transfer and complex carbohydrates mediate protection against COPD. Gut 2024; 73:751-769. [PMID: 38331563 DOI: 10.1136/gutjnl-2023-330521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a major cause of global illness and death, most commonly caused by cigarette smoke. The mechanisms of pathogenesis remain poorly understood, limiting the development of effective therapies. The gastrointestinal microbiome has been implicated in chronic lung diseases via the gut-lung axis, but its role is unclear. DESIGN Using an in vivo mouse model of cigarette smoke (CS)-induced COPD and faecal microbial transfer (FMT), we characterised the faecal microbiota using metagenomics, proteomics and metabolomics. Findings were correlated with airway and systemic inflammation, lung and gut histopathology and lung function. Complex carbohydrates were assessed in mice using a high resistant starch diet, and in 16 patients with COPD using a randomised, double-blind, placebo-controlled pilot study of inulin supplementation. RESULTS FMT alleviated hallmark features of COPD (inflammation, alveolar destruction, impaired lung function), gastrointestinal pathology and systemic immune changes. Protective effects were additive to smoking cessation, and transfer of CS-associated microbiota after antibiotic-induced microbiome depletion was sufficient to increase lung inflammation while suppressing colonic immunity in the absence of CS exposure. Disease features correlated with the relative abundance of Muribaculaceae, Desulfovibrionaceae and Lachnospiraceae family members. Proteomics and metabolomics identified downregulation of glucose and starch metabolism in CS-associated microbiota, and supplementation of mice or human patients with complex carbohydrates improved disease outcomes. CONCLUSION The gut microbiome contributes to COPD pathogenesis and can be targeted therapeutically.
Collapse
Affiliation(s)
- Kurtis F Budden
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kate L Bowerman
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, The University of Queensland, Brisbane, QLD, Australia
| | - Annalicia Vaughan
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Shaan L Gellatly
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - David L A Wood
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, The University of Queensland, Brisbane, QLD, Australia
| | - Nancy Lachner
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, The University of Queensland, Brisbane, QLD, Australia
| | - Sobia Idrees
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Saima Firdous Rehman
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Alen Faiz
- Respiratory Bioinformatics and Molecular Biology, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Vyoma K Patel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Charlotte A Alemao
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Sj Shen
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Nadia Amorim
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Rajib Majumder
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Kanth S Vanka
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Jazz Mason
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Tatt Jhong Haw
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Bree Tillet
- Frazer Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Simon Keely
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Nicole Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Gabrielle T Belz
- Frazer Institute, University of Queensland, Woolloongabba, QLD, Australia
| | - Jay Horvat
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Thomas Ashhurst
- Sydney Cytometry, Charles Perkins Centre, Centenary Institute and The University of Sydney, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, NSW, Australia
| | - Caryn van Vreden
- Sydney Cytometry, Charles Perkins Centre, Centenary Institute and The University of Sydney, Sydney, NSW, Australia
- Ramaciotti Facility for Human Systems Biology, Charles Perkins Centre and The University of Sydney, Sydney, NSW, Australia
| | - Helen McGuire
- Ramaciotti Facility for Human Systems Biology, Charles Perkins Centre and The University of Sydney, Sydney, NSW, Australia
| | - Barbara Fazekas de St Groth
- Ramaciotti Facility for Human Systems Biology, Charles Perkins Centre and The University of Sydney, Sydney, NSW, Australia
| | - Nicholas J C King
- Sydney Cytometry, Charles Perkins Centre, Centenary Institute and The University of Sydney, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, NSW, Australia
- Ramaciotti Facility for Human Systems Biology, Charles Perkins Centre and The University of Sydney, Sydney, NSW, Australia
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Ben Crossett
- Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, Australia
| | - Stuart J Cordwell
- Sydney Mass Spectrometry, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, Charles Perkins Centre and The University of Sydney, Sydney, NSW, Australia
| | - Lorenzo Bonaguro
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | | | - Elizabeth Mann
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Samuel C Forster
- Centre for Innate Immunity and Infectious Diseases and Department of Molecular and Translational Science, Hudson Institute of Medical Research and Monash University, Melbourne, VIC, Australia
| | - Matthew A Cooper
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Leopoldo N Segal
- Division of Pulmonary and Critical Care Medicine, Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Translational Respiratory Research Laboratory, Singapore
| | - Peter Collins
- Mater Research Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Dietetics & Food Services, Mater Hospital, Brisbane, QLD, Australia
| | - Rayleen Bowman
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Kwun M Fong
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Ian A Yang
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Department of Thoracic Medicine, The Prince Charles Hospital, Chermside, QLD, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Paul G Dennis
- School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Philip Hugenholtz
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, The University of Queensland, Brisbane, QLD, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
5
|
Moonwiriyakit A, Yimnual C, Noitem R, Dinsuwannakol S, Sontikun J, Kaewin S, Worakajit N, Soontornniyomkij V, Muanprasat C. GPR120/FFAR4 stimulation attenuates airway remodeling and suppresses IL-4- and IL-13-induced airway epithelial injury via inhibition of STAT6 and Akt. Biomed Pharmacother 2023; 168:115774. [PMID: 37924784 DOI: 10.1016/j.biopha.2023.115774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Airway remodeling is associated with severity and treatment insensitivity in asthma. This study aimed to investigate the effects of G protein-coupled receptor 120 (GPR120) stimulation on alleviating allergic inflammation and remodeling of airway epithelium. RESEARCH DESIGN AND METHODS Ovalbumin (OVA)-challenged BALB/c mice and type-2-cytokine (IL-4 and IL-13)-exposed 16HBE human bronchial epithelial cells were treated with GSK137647A, a selective GPR120 agonist. Markers of allergic inflammation and airway remodeling were determined. RESULTS GSK137647A attenuated inflammation and mucus secretion in airway epithelium of OVA-challenged mice. Stimulation of GPR120 in 16HBE suppressed expression of asthma-associated cytokines and cytokine-induced expression of pathogenic mucin-MUC5AC. These effects were abolished by co-treatment with AH7614, a GPR120 antagonist. Moreover, GPR120 stimulation in 16HBE cells reduced expression of fibrotic markers including fibronectin protein and ACTA2 mRNA and inhibited epithelial barrier leakage induced by type-2 inflammation via rescuing expression of zonula occludens-1 protein. Furthermore, GPR120 stimulation prevented the cytokine-induced airway epithelial remodeling via suppression of STAT6 and Akt phosphorylation. CONCLUSIONS Our findings suggest that GPR120 activation alleviates allergic inflammation and remodeling of airway epithelium partly through inhibition of STAT6 and Akt. GPR120 may represent a novel therapeutic target for diseases associated with remodeling of airway epithelium, including asthma.
Collapse
Affiliation(s)
- Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chantapol Yimnual
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Rattikarn Noitem
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sasiwimol Dinsuwannakol
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Jenjira Sontikun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Suchada Kaewin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nichakorn Worakajit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand; Translational Medicine Graduate Program, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Virawudh Soontornniyomkij
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
6
|
Schleich F, Bougard N, Moermans C, Sabbe M, Louis R. Cytokine-targeted therapies for asthma and COPD. Eur Respir Rev 2023; 32:32/168/220193. [PMID: 37076177 PMCID: PMC10113955 DOI: 10.1183/16000617.0193-2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/23/2023] [Indexed: 04/21/2023] Open
Abstract
Asthma affects over 300 million people worldwide and its prevalence is increasing. COPD is the third leading cause of death globally. Asthma and COPD are complex inflammatory diseases of the airways in which impaired host defences lead to increased susceptibility to pathogens, pollutants and allergens. There is a constant interplay between host and the environment. Environmental exposures can alter the lung microbiome and influence the development of sensitisation by disrupting normal immunoregulation. The underlying airway inflammation in severe asthma is heterogeneous, with upregulation of type 2 cytokines in most cases but increased neutrophilic inflammation and activated T-helper 17 mediated immunity in others. COPD may also comprise several different phentoypes that are driven by different molecular mechanisms or endotypes. This disease heterogeneity is affected by comorbidities, treatments and environmental exposures. Recent intervention trials have shed light on the pathways beyond type 2 inflammation that can lead to beneficial outcomes versus potentially deleterious effects. We have made a great deal of progress over the last 10 years in terms of immunology and the pathophysiology of asthma and this has led to the development of novel treatments and major improvements in severe asthma outcomes. In COPD, however, no targeted treatments have demonstrated great improvements. This article reviews the mechanism of action and efficacy of the available biologics in asthma and COPD.
Collapse
Affiliation(s)
- Florence Schleich
- Respiratory Medicine, CHU of Liege, Belgium
- GIGA I3, University of Liege, Belgium
| | | | | | - Mare Sabbe
- Respiratory Medicine, CHU of Liege, Belgium
| | - Renaud Louis
- Respiratory Medicine, CHU of Liege, Belgium
- GIGA I3, University of Liege, Belgium
| |
Collapse
|
7
|
Shi L, Dai X, Yan F, Lin Y, Lin L, Zhang Y, Zeng Y, Chen X. Novel lipidomes profile and clinical phenotype identified in pneumoconiosis patients. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2023; 42:55. [PMID: 37322561 DOI: 10.1186/s41043-023-00400-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 06/09/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Pneumoconiosis is a group of occupational lung diseases caused by the inhalation of mineral dust in the lungs, leading to lung dysfunction. Patients with pneumoconiosis are usually accompanied by weight loss, which suggests a lipid metabolism disorder. Recent progress in lipidomics uncovered detailed lipid profiles that play important roles in respiratory diseases, such as asthma, lung cancer and lung injury. The purpose of this study was to shed light on the different expression of lipidome between pneumoconiosis and healthy, hoping to bring new ideas for the diagnosis and treatment of pneumoconiosis. METHODOLOGY This non-matching case-control study was performed among 96 subjects (48 outpatients with male pneumoconiosis and 48 healthy volunteers), data of clinical phenotypes were recorded, and plasma biochemistry (lipidomic profiles) was tested for both pneumoconiosis patients and healthy controls. A total of 426 species in 11 lipid classes were analyzed by high-performance liquid chromatography coupled with triple quadrupole tandem mass spectrometry (HPLC-QqQ-MS) for the cases and controls. We also analyzed the correlation of lipid profiles with clinical phenomes from pneumoconiosis patients by expression quantitative trait locus (eQTL) model to evaluate trans-nodules between lipidomic profiles and clinical phenomes. All visually re-checked data were analyzed using appropriate statistical tools (t-test or one-way ANOVA test) on SPSS. RESULTS Compared with healthy people, 26 significantly increased (> 1.5-fold) and 30 decreased lipid elements (< 2/threefold) in patients with pneumoconiosis were identified (P values all < 0.05). The majority of those elevated lipid elements were phosphatidylethanolamines (PEs), and the minority were free fatty acids (FFAs), while phosphatidylcholines (PCs) and lysophosphatidylcholines (lysoPCs) declined in pneumoconiosis. Clinical trans-omics analyses demonstrated that phenomes in pneumoconiosis connections with multiple lipids, which showed that pH, lung function, mediastinal lymph node calcification, and complication were highly correlated with lipid elements. Furthermore, up-regulated PE was corresponded to pH, smoking history and mediastinal lymph node calcification. PC was corresponded to dust exposure history, BMI and mediastinal lymph node calcification. CONCLUSION We found altered lipid panels between male pneumoconiosis patients and healthy people by qualitatively and quantitatively measured plasma lipidomic profiles. The trans-omic analysis between clinical phenomes and lipidomes might have the potential to uncover the heterogeneity of lipid metabolism of pneumoconiosis patients and to screen out clinically significant phenome-based lipid panels.
Collapse
Affiliation(s)
- Liyong Shi
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Xiaofang Dai
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Furong Yan
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yujun Lin
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Lianshun Lin
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yongquan Zhang
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Xiaoyang Chen
- Department of Pulmonary and Critical Care Medicine, Respiratory Medicine Center of Fujian Province, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| |
Collapse
|
8
|
Huang T, Long Y, Ou Y, Li J, Huang Y, Gao J. Association between circulating fatty acid metabolites and asthma risk: a two-sample bidirectional Mendelian randomization study. BMC Med Genomics 2023; 16:112. [PMID: 37221513 DOI: 10.1186/s12920-023-01545-4] [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: 10/11/2022] [Accepted: 05/13/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Fatty acids are involved in a wide range of immunological responses in humans. Supplementation of polyunsaturated fatty acids has been reported to help alleviate symptoms and airway inflammation in asthma patients, whereas the effects of fatty acids on the actual risk of asthma remain controversial. This study comprehensively investigated the causal effects of serum fatty acids on asthma risk using two-sample bidirectional Mendelian Randomization (MR) analysis. METHODS Genetic variants strongly associated with 123 circulating fatty acid metabolites were extracted as instrumental variables, and a large GWAS data of asthma was used to test effects of the metabolites on this outcome. The inverse-variance weighted method was used for primary MR analysis. The weighted median, MR-Egger regression, MR-PRESSO, and leave-one-out analyses were utilized to evaluate heterogeneity and pleiotropy. Potential confounders were adjusted by performing multivariable MR analyses. Reverse MR analysis was also conducted to estimate the causal effect of asthma on candidate fatty acid metabolites. Further, we performed colocalization analysis to examine the pleiotropy of variants within the fatty acid desaturase 1 (FADS1) locus between the significant metabolite traits and the risk of asthma. Cis-eQTL-MR and colocalization analysis were also performed to determine the association between RNA expression of FADS1 and asthma. RESULTS Genetically instrumented higher average number of methylene groups was causally associated with a lower risk of asthma in primary MR analysis, while inversely, the higher ratio of bis-allylic groups to double bonds and the higher ratio of bis-allylic groups to total fatty acids, were associated with higher probabilities of asthma. Consistent results were obtained in multivariable MR when adjusted for potential confounders. However, these effects were completely eliminated after SNPs correlated with the FADS1 gene were excluded. The reverse MR also found no causal association. The colocalization analysis suggested that the three candidate metabolite traits and asthma likely share causal variants within the FADS1 locus. In addition, the cis-eQTL-MR and colocalization analyses demonstrated a causal association and shared causal variants between FADS1 expression and asthma. CONCLUSIONS Our study supports a negative association between several PUFA traits and the risk of asthma. However, this association is largely attributed to the influence of FADS1 polymorphisms. The results of this MR study should be carefully interpreted given the pleiotropy of SNPs associated with FADS1.
Collapse
Affiliation(s)
- Tingting Huang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yichen Long
- Department of Epidemiology, School of Public Health, Southeast University, Jiangsu, Nanjing, China
| | - Yang Ou
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jia Li
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yilin Huang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinming Gao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China.
| |
Collapse
|
9
|
Lam M, Lamanna E, Organ L, Donovan C, Bourke JE. Perspectives on precision cut lung slices-powerful tools for investigation of mechanisms and therapeutic targets in lung diseases. Front Pharmacol 2023; 14:1162889. [PMID: 37261291 PMCID: PMC10228656 DOI: 10.3389/fphar.2023.1162889] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/19/2023] [Indexed: 06/02/2023] Open
Abstract
Precision cut lung slices (PCLS) have emerged as powerful experimental tools for respiratory research. Pioneering studies using mouse PCLS to visualize intrapulmonary airway contractility have been extended to pulmonary arteries and for assessment of novel bronchodilators and vasodilators as therapeutics. Additional disease-relevant outcomes, including inflammatory, fibrotic, and regenerative responses, are now routinely measured in PCLS from multiple species, including humans. This review provides an overview of established and innovative uses of PCLS as an intermediary between cellular and organ-based studies and focuses on opportunities to increase their application to investigate mechanisms and therapeutic targets to oppose excessive airway contraction and fibrosis in lung diseases.
Collapse
Affiliation(s)
- Maggie Lam
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Emma Lamanna
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Institut Pasteur, Unit of Antibodies in Therapy and Pathology, INSERM UMR1222, Paris, France
| | - Louise Organ
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Chantal Donovan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
- Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jane E. Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
10
|
Cheng TY, Chang CC, Luo CS, Chen KY, Yeh YK, Zheng JQ, Wu SM. Targeting Lung-Gut Axis for Regulating Pollution Particle-Mediated Inflammation and Metabolic Disorders. Cells 2023; 12:901. [PMID: 36980242 PMCID: PMC10047528 DOI: 10.3390/cells12060901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Cigarette smoking (CS) or ambient particulate matter (PM) exposure is a risk factor for metabolic disorders, such as insulin resistance (IR), increased plasma triglycerides, hyperglycemia, and diabetes mellitus (DM); it can also cause gut microbiota dysbiosis. In smokers with metabolic disorders, CS cessation decreases the risks of serious pulmonary events, inflammation, and metabolic disorder. This review included recent studies examining the mechanisms underlying the effects of CS and PM on gut microbiota dysbiosis and metabolic disorder development; one of the potential mechanisms is the disruption of the lung-gut axis, leading to gut microbiota dysbiosis, intestinal dysfunction, systemic inflammation, and metabolic disease. Short-chain fatty acids (SCFAs) are the primary metabolites of gut bacteria, which are derived from the fermentation of dietary fibers. They activate G-protein-coupled receptor (GPCR) signaling, suppress histone deacetylase (HDAC) activity, and inhibit inflammation, facilitating the maintenance of gut health and biofunction. The aforementioned gut microbiota dysbiosis reduces SCFA levels. Treatment targeting SCFA/GPCR signaling may alleviate air pollution-associated inflammation and metabolic disorders, which involve lung-gut axis disruption.
Collapse
Affiliation(s)
- Tzu-Yu Cheng
- Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan;
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Cheng Chang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Ching-Shan Luo
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yun-Kai Yeh
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jing-Quan Zheng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (C.-C.C.); (C.-S.L.); (K.-Y.C.); (Y.-K.Y.); (J.-Q.Z.)
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
11
|
Budden KF, Gellatly SL, Vaughan A, Amorim N, Horvat JC, Hansbro NG, Wood DLA, Hugenholtz P, Dennis PG, Wark PAB, Hansbro PM. Probiotic Bifidobacterium longum subsp. longum Protects against Cigarette Smoke-Induced Inflammation in Mice. Int J Mol Sci 2022; 24:252. [PMID: 36613693 PMCID: PMC9820259 DOI: 10.3390/ijms24010252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
Bifidobacterium are prominent gut commensals that produce the short-chain fatty acid (SCFA) acetate, and they are often used as probiotics. Connections between the gut and the lung, termed the gut-lung axis, are regulated by the microbiome. The gut-lung axis is increasingly implicated in cigarette smoke-induced diseases, and cigarette smoke exposure has been associated with depletion of Bifidobacterium species. In this study, we assessed the impact of acetate-producing Bifidobacterium longum subsp. longum (WT) and a mutant strain with an impaired acetate production capacity (MUT) on cigarette smoke-induced inflammation. The mice were treated with WT or MUT B. longum subsp. longum and exposed to cigarette smoke for 8 weeks before assessments of lung inflammation, lung tissue gene expression and cecal SCFAs were performed. Both strains of B. longum subsp. longum reduced lung inflammation, inflammatory cytokine expression and adhesion factor expression and alleviated cigarette smoke-induced depletion in caecum butyrate. Thus, the probiotic administration of B. longum subsp. longum, irrespective of its acetate-producing capacity, alleviated cigarette smoke-induced inflammation and the depletion of cecal butyrate levels.
Collapse
Affiliation(s)
- Kurtis F. Budden
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Shaan L. Gellatly
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Annalicia Vaughan
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Nadia Amorim
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Jay C. Horvat
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Nicole G. Hansbro
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| | - David L. A. Wood
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Paul G. Dennis
- School of Earth and Environmental Sciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Peter A. B. Wark
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs and Immune Health Research Program, Hunter Medical Research Institute and The University of Newcastle, Callaghan, NSW 2308, Australia
- Centre for Inflammation, School of Life Sciences, Faculty of Science, Centenary Institute and University of Technology Sydney, Sydney, NSW 2007, Australia
| |
Collapse
|
12
|
How Arrestins and GRKs Regulate the Function of Long Chain Fatty Acid Receptors. Int J Mol Sci 2022; 23:ijms232012237. [PMID: 36293091 PMCID: PMC9602559 DOI: 10.3390/ijms232012237] [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: 09/08/2022] [Revised: 10/03/2022] [Accepted: 10/08/2022] [Indexed: 11/16/2022] Open
Abstract
FFA1 and FFA4, two G protein-coupled receptors that are activated by long chain fatty acids, play crucial roles in mediating many biological functions in the body. As a result, these fatty acid receptors have gained considerable attention due to their potential to be targeted for the treatment of type-2 diabetes. However, the relative contribution of canonical G protein-mediated signalling versus the effects of agonist-induced phosphorylation and interactions with β-arrestins have yet to be fully defined. Recently, several reports have highlighted the ability of β-arrestins and GRKs to interact with and modulate different functions of both FFA1 and FFA4, suggesting that it is indeed important to consider these interactions when studying the roles of FFA1 and FFA4 in both normal physiology and in different disease settings. Here, we discuss what is currently known and show the importance of understanding fully how β-arrestins and GRKs regulate the function of long chain fatty acid receptors.
Collapse
|
13
|
Tang X, Hou Y, Schwartz TW, Haeggström JZ. Metabolite G-protein coupled receptor signaling: Potential regulation of eicosanoids. Biochem Pharmacol 2022; 204:115208. [PMID: 35963340 DOI: 10.1016/j.bcp.2022.115208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/19/2022]
Abstract
Eicosanoids are a family of bioactive compounds derived from arachidonic acid (AA) that play pivotal roles in physiology and disease, including inflammatory conditions of multiple organ systems. The biosynthesis of eicosanoids requires a series of catalytic steps that are controlled by designated enzymes, which can be regulated by inflammatory and stress signals via transcriptional and translational mechanisms. In the past decades, evidence have emerged indicating that G-protein coupled receptors (GPCRs) can sense extracellular metabolites, and regulate inflammatory responses including eicosanoid production. This review focuses on the recent advances of metabolite GPCRs research, their role in regulation of eicosanoid biosynthesis, and the link to pathophysiological conditions.
Collapse
Affiliation(s)
- Xiao Tang
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| | - Yaolin Hou
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Thue W Schwartz
- Section for Metabolic Receptology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; Laboratory for Molecular Pharmacology, Department for Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jesper Z Haeggström
- Division of Physiological Chemistry II, Biomedicum 9A, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden.
| |
Collapse
|
14
|
Martínez-Morales JC, Solís KH, Romero-Ávila MT, Reyes-Cruz G, García-Sáinz JA. Cell Trafficking and Function of G Protein-coupled Receptors. Arch Med Res 2022; 53:451-460. [PMID: 35835604 DOI: 10.1016/j.arcmed.2022.06.008] [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: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022]
Abstract
The G protein-coupled receptors (GPCRs) are plasma membrane proteins that function as sensors of changes in the internal and external milieux and play essential roles in health and disease. They are targets of hormones, neurotransmitters, local hormones (autacoids), and a large proportion of the drugs currently used as therapeutics and for "recreational" purposes. Understanding how these receptors signal and are regulated is fundamental for progress in areas such as physiology and pharmacology. This review will focus on what is currently known about their structure, the molecular events that trigger their signaling, and their trafficking to endosomal compartments. GPCR phosphorylation and its role in desensitization (signaling switching) are also discussed. It should be mentioned that the volume of information available is enormous given the large number and variety of GPCRs. However, knowledge is fragmentary even for the most studied receptors, such as the adrenergic receptors. Therefore, we attempt to present a panoramic view of the field, conscious of the risks and limitations (such as oversimplifications and incorrect generalizations). We hope this will provoke further research in the area. It is currently accepted that GPCR internalization plays a role signaling events. Therefore, the processes that allow them to internalize and recycle back to the plasma membrane are briefly reviewed. The functions of cytoskeletal elements (mainly actin filaments and microtubules), the molecular motors implicated in receptor trafficking (myosin, kinesin, and dynein), and the GTPases involved in GPCR internalization (dynamin) and endosomal sorting (Rab proteins), are discussed. The critical role phosphoinositide metabolism plays in regulating these events is also depicted.
Collapse
Affiliation(s)
- Juan Carlos Martínez-Morales
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - K Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Guadalupe Reyes-Cruz
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, México
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
15
|
Pascoe CD, Basu S, Schwartz J, Fonseca M, Kahnamoui S, Jha A, Dolinsky VW, Halayko AJ. Maternal diabetes promotes offspring lung dysfunction and inflammation in a sex-dependent manner. Am J Physiol Lung Cell Mol Physiol 2022; 322:L373-L384. [PMID: 35043678 DOI: 10.1152/ajplung.00425.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Exposure to maternal diabetes is increasingly recognized as a risk factor for chronic respiratory disease in children. It is currently unclear, however, whether maternal diabetes affects the lung health of male and female offspring equally. This study characterizes the sex-specific impact of a murine model of diet-induced gestational diabetes (GDM) on offspring lung function and airway inflammation. Female adult mice are fed a high-fat (45% kcal) diet for 6-weeks prior to mating. Control offspring are from mothers fed a low fat (10% kcal) diet. Offspring were weaned and fed a chow diet until 10-weeks of age, at which point lung function was measured and lung lavage was collected. Male, but not female offspring exposed to GDM had increased lung compliance and reduced lung resistance at baseline. Female offspring exposed to GDM displayed increased methacholine reactivity and elevated levels of pro-inflammatory cytokines (e.g. interleukin (IL)-1β, IL-5, and CXCL1) in lung lavage. Female GDM offspring also displayed elevated abundance of matrix metalloproteinases (MMP) within their airways, namely MMP-3 and MMP-8. These results indicate disparate effects of maternal diabetes on lung health and airway inflammation of male and female offspring exposed to GDM. Female mice may be at greater risk of inflammatory lung conditions, such as asthma, while male offspring display changes that more closely align with models of chronic obstructive pulmonary disease. In conclusion, there are important sex-based differences in the impact of maternal diabetes on offspring lung health that could signal differences in future disease risk.
Collapse
Affiliation(s)
- Christopher D Pascoe
- Deptartment of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Sujata Basu
- Deptartment of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Jacquie Schwartz
- Deptartment of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Mario Fonseca
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada.,Diabetes Research Envisioned and Accomplished in Manitoba, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Shana Kahnamoui
- Deptartment of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Aruni Jha
- Deptartment of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Vernon W Dolinsky
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada.,Diabetes Research Envisioned and Accomplished in Manitoba, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew John Halayko
- Deptartment of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing Group, The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
16
|
Zheng H, Jin S, Li T, Ying W, Ying B, Chen D, Ning J, Zheng C, Li Y, Li C, Chen C, Li X, Gao H. Metabolomics reveals sex-specific metabolic shifts and predicts the duration from positive to negative in non-severe COVID-19 patients during recovery process. Comput Struct Biotechnol J 2021; 19:1863-1873. [PMID: 33841749 PMCID: PMC8021501 DOI: 10.1016/j.csbj.2021.03.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 02/08/2023] Open
Abstract
Metabolic profiling in COVID-19 patients has been associated with disease severity, but there is no report on sex-specific metabolic changes in discharged survivors. Herein we used an integrated approach of LC-MS-and GC-MS-based untargeted metabolomics to analyze plasma metabolic characteristics in men and women with non-severe COVID-19 at both acute period and 30 days after discharge. The results demonstrate that metabolic alterations in plasma of COVID-19 patients during the recovery and rehabilitation process were presented in a sex specific manner. Overall, the levels of most metabolites were increased in COVID-19 patients after the cure relative to acute period. The major plasma metabolic changes were identified including fatty acids in men and glycerophosphocholines and carbohydrates in women. In addition, we found that women had shorter length of hospitalization than men and metabolic characteristics may contribute to predict the duration from positive to negative in non-severe COVID-19 patients. Collectively, this study shed light on sex-specific metabolic shifts in non-severe COVID-19 patients during the recovery process, suggesting a sex bias in prognostic and therapeutic evaluations based on metabolic profiling.
Collapse
Key Words
- ALT, Alanine aminotransferase
- AP, Acute period (AP)
- APTT, Activated partial thromboplastin time
- BCAAs, Branched‐chain amino acids
- BP, Blood platelet
- CA, Carbamide
- COVID-19
- COVID-19, Novel coronavirus disease 2019
- CRP, C-reactive protein
- DAA, Dehydroascorbic acid
- DD, D-dimer
- DP, Diastolic pressure
- FIB, Fibrinogen
- FP, Follow-up period
- Fatty acid
- GPCs, Glycerophosphocholines
- HGB, Hemoglobin
- LY, Lymphocyte
- Metabolism
- NG, Neutrophilic granulocyte
- NK, Natural killer
- PCT, Procalcitonin
- PLS-DA, Partial least squares-discriminant analysis
- PLSR, Partial least squares regression
- PT, Prothrombin time
- PTC, Phosphatidylcholine
- RDW, Red cell distribution width
- RR, Respiratory rate
- S1P, Sphingosine-1-phosphate
- SARS-CoV
- Sex difference
- TBL, Total B lymphocyte
- TTL, Total T lymphocyte
- WBC, White blood cell
Collapse
Affiliation(s)
- Hong Zheng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ting Li
- Clinical Research Unit, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Weiyang Ying
- Department of Pain Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Binyu Ying
- Department of Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Dong Chen
- Wenzhou Central Hospital, Wenzhou 325015, China
| | - Jie Ning
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Chanfan Zheng
- Clinical Research Unit, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yuping Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, China
| | - Chen Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Chengshui Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Hongchang Gao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325015, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
17
|
Xu S, Schwab A, Karmacharya N, Cao G, Woo J, Kim N, An SS, Panettieri RA, Jude JA. FFAR1 activation attenuates histamine-induced myosin light chain phosphorylation and cortical tension development in human airway smooth muscle cells. Respir Res 2020; 21:317. [PMID: 33256729 PMCID: PMC7708129 DOI: 10.1186/s12931-020-01584-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/22/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Activation of free fatty acid receptors (FFAR1 and FFAR4) which are G protein-coupled receptors (GPCRs) with established (patho)physiological roles in a variety of obesity-related disorders, induce human airway smooth muscle (HASM) cell proliferation and shortening. We reported amplified agonist-induced cell shortening in HASM cells obtained from obese lung donors. We hypothesized that FFAR1 modulate excitation-contraction (EC) coupling in HASM cells and play a role in obesity-associated airway hyperresponsiveness. METHODS In HASM cells pre-treated (30 min) with FFAR1 agonists TAK875 and GW9508, we measured histamine-induced Ca2+ mobilization, myosin light chain (MLC) phosphorylation, and cortical tension development with magnetic twisting cytometry (MTC). Phosphorylation of MLC phosphatase and Akt also were determined in the presence of the FFAR1 agonists or vehicle. In addition, the effects of TAK875 on MLC phosphorylation were measured in HASM cells desensitized to β2AR agonists by overnight salmeterol treatment. The inhibitory effect of TAK875 on MLC phosphorylation was compared between HASM cells from age and sex-matched non-obese and obese human lung donors. The mean measurements were compared using One-Way ANOVA with Dunnett's test for multiple group comparisons or Student's t-test two-group comparison. For cortical tension measurements by magnetic twisted cytometry, mixed effect model using SAS V.9.2 was applied. Means were considered significant when p ≤ 0.05. RESULTS Unexpectedly, we found that TAK875, a synthetic FFAR1 agonist, attenuated histamine-induced MLC phosphorylation and cortical tension development in HASM cells. These physiological outcomes were unassociated with changes in histamine-evoked Ca2+ flux, protein kinase B (AKT) activation, or MLC phosphatase inhibition. Of note, TAK875-mediated inhibition of MLC phosphorylation was maintained in β2AR-desensitized HASM cells and across obese and non-obese donor-derived HASM cells. CONCLUSIONS Taken together, our findings identified the FFAR1 agonist TAK875 as a novel bronchoprotective agent that warrants further investigation to treat difficult-to-control asthma and/or airway hyperreactivity in obesity.
Collapse
Affiliation(s)
- Shengjie Xu
- The Joint Graduate Program in Toxicology, Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Piscataway, USA
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
| | - Anthony Schwab
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
| | - Nikhil Karmacharya
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
| | - Gaoyuan Cao
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
| | - Joanna Woo
- The Joint Graduate Program in Toxicology, Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Piscataway, USA
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
| | - Nicholas Kim
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Steven S An
- The Joint Graduate Program in Toxicology, Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Piscataway, USA
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Reynold A Panettieri
- The Joint Graduate Program in Toxicology, Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Piscataway, USA
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Joseph A Jude
- The Joint Graduate Program in Toxicology, Department of Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Piscataway, USA.
- Rutgers Institute for Translational Medicine & Science, New Brunswick, NJ, 08901, USA.
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
- Pharmacology & Toxicology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Rm: 4276, 89, French Street, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
18
|
Imbalanced serum levels of resolvin E1 (RvE1) and leukotriene B4 (LTB4) in patients with allergic rhinitis. Mol Biol Rep 2020; 47:7745-7754. [PMID: 32960415 DOI: 10.1007/s11033-020-05849-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/10/2020] [Accepted: 09/17/2020] [Indexed: 10/23/2022]
Abstract
Timely and successful resolution of acute inflammation plays a crucial role in preventing the development of chronic airway inflammation in allergic rhinitis (AR). This study intends to assess the serum levels of pro-inflammatory leukotriene B4 (LTB4), anti-inflammatory mediators, including resolvin E1 (RvE1), RvD1, IL-10, and TGF-β, besides mRNA expression level of G-protein coupled receptor 120 (GPR120) and peroxisome proliferator-activated receptor-γ (PPAR-γ) receptors in peripheral blood leukocytes of AR patients. Thirty-seven AR patients and thirty age- and gender-matched healthy subjects were enrolled in this study. The serum levels of LTB4, RvE1, RvD1, IL-10, and TGF-β were measured using enzyme-linked immunosorbent assay (ELISA) technique, and the mRNA expression level of GPR120 and PPAR-γ was assessed by the real-time PCR method. The serum levels of RvE1 and LTB4 were significantly higher in patients with AR than in healthy subjects (P < 0.01 and P < 0.0001, respectively). However, a significantly lower ratio of RvE1 and RvD1 to LTB4 was found in patients with AR relative to healthy subjects (P < 0.05 and P < 0.0001, respectively). Likewise, the serum levels of both IL-10 and TGF-β cytokines were significantly reduced in patients with AR compared to healthy subjects (P < 0.01 and P < 0.0001, respectively). Furthermore, the mRNA expression of PPAR-γ was significantly lower in patients with AR than in healthy subjects (P < 0.05). Our findings indicate that imbalanced pro-resolving lipid mediator RvE1 and pro-inflammatory LTB4 might contribute to the defective airway inflammation-resolution and subsequent progression toward chronic inflammation in AR patients.
Collapse
|
19
|
Fatty acid receptor gives lungs a breather. Nat Rev Drug Discov 2020; 19:672. [PMID: 32879435 DOI: 10.1038/d41573-020-00156-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|