1
|
Park KS, Lässer C, Lötvall J. Extracellular vesicles and the lung: from disease pathogenesis to biomarkers and treatments. Physiol Rev 2025; 105:1733-1821. [PMID: 40125970 DOI: 10.1152/physrev.00032.2024] [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: 08/14/2024] [Revised: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nanosized extracellular vesicles (EVs) are released by all cells to convey cell-to-cell communication. EVs, including exosomes and microvesicles, carry an array of bioactive molecules, such as proteins and RNAs, encapsulated by a membrane lipid bilayer. Epithelial cells, endothelial cells, and various immune cells in the lung contribute to the pool of EVs in the lung microenvironment and carry molecules reflecting their cellular origin. EVs can maintain lung health by regulating immune responses, inducing tissue repair, and maintaining lung homeostasis. They can be detected in lung tissues and biofluids such as bronchoalveolar lavage fluid and blood, offering information about disease processes, and can function as disease biomarkers. Here, we discuss the role of EVs in lung homeostasis and pulmonary diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis, and lung injury. The mechanistic involvement of EVs in pathogenesis and their potential as disease biomarkers are discussed. Finally, the pulmonary field benefits from EVs as clinical therapeutics in severe pulmonary inflammatory disease, as EVs from mesenchymal stem cells attenuate severe respiratory inflammation in multiple clinical trials. Further, EVs can be engineered to carry therapeutic molecules for enhanced and broadened therapeutic opportunities, such as the anti-inflammatory molecule CD24. Finally, we discuss the emerging opportunity of using different types of EVs for treating severe respiratory conditions.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
2
|
Pistenmaa CL, Hoffman EA, Prince MR, Hughes E, Dashnaw S, Lo Cascio CM, Oelsner EC, Shen W, Sun Y, Winther H, Vogel-Claussen J, Wild JM, Tracy RP, Barr RG. Platelet activation and COPD-related clinical and imaging characteristics: The Multi-Ethnic Study of Atherosclerosis (MESA) COPD Study. Respir Med 2025; 241:108058. [PMID: 40147570 DOI: 10.1016/j.rmed.2025.108058] [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: 11/25/2024] [Revised: 02/25/2025] [Accepted: 03/25/2025] [Indexed: 03/29/2025]
Abstract
INTRODUCTION The pulmonary vasculature has been implicated in chronic obstructive pulmonary disease (COPD). Whether platelet activation is associated with COPD-related characteristics is unknown. METHODS The Multi-Ethnic Study of Atherosclerosis (MESA) COPD Study enrolled participants with 10+ pack-years with and without COPD from 2 cohort studies. Platelet activation was measured in platelet-free plasma as von Willebrand factor (vWF), beta-thromboglobulin (BTG) and platelet factor 4 (PF4). Phenotyping included spirometry, percent emphysema-950HU, air trapping on computed tomography (CT), low-ventilated lung volume on 3He-magnetic resonance imaging (MRI) and contrast-enhanced MRI pulmonary perfusion on and off oxygen. Linear and logistic regression adjusted for demographics, anthropometry, platelet count, aspirin use, COPD status and cohort; the final model adjusted for smoking, pack-years, oxygen saturation and hypertension. RESULTS The 116 with vWF were a mean age of 73.5 ± 7.3 years old, 60 % male, 58 % Non-Hispanic White, 28 % Black, 14 % Hispanic/Latino, 23 % smoked currently and 55 % had COPD. vWF was associated with lower FEV1/FVC (-2.0 %/SD vWF, 95 %CI: -3.5, -0.6), and greater percent low-ventilated lung volume on 3He-MRI (6.5 %/SD vWF, 95 %CI: 1.5, 11.5), CT air trapping (3.2 %/SD vWF, 95 %CI: 1.4, 5.1) and heterogeneity (CV) of pulmonary microvascular blood flow (PMBF) and pulmonary microvascular blood volume (PMBV) on oxygen. Higher BTG and PF4 were associated with greater PMBF and PMBV. There was increased odds of COPD with higher PF4 (final model only: OR 1.8/SD PF4, 95 %CI: 1.01, 3.20). CONCLUSIONS Platelet activation was associated with measures of small airways disease and pulmonary microvascular perfusion in this sample of smokers with and without COPD.
Collapse
Affiliation(s)
- Carrie L Pistenmaa
- Columbia University, New York, NY, USA; Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | | | - Wei Shen
- Columbia University, New York, NY, USA
| | | | - Hinrich Winther
- Institute for Diagnostic and Interventional Radiology, Hannover Medical School, Germany; Member of the German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Germany
| | - Jens Vogel-Claussen
- Institute for Diagnostic and Interventional Radiology, Hannover Medical School, Germany; Member of the German Center for Lung Research, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Germany
| | | | | | | |
Collapse
|
3
|
Han N, Qi H, Yin Y, Liu Y, Jin P, Hou Y, Jia Z. The effectiveness of vasodilators on chronic obstructive pulmonary disease: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e39794. [PMID: 39560512 PMCID: PMC11576023 DOI: 10.1097/md.0000000000039794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 08/30/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a complex progressive disease. Some vasodilators have been reported with therapeutic potential to protect vascular function therefore may delay the progression of COPD. METHODS We searched PubMed, Embase, Cochrane Library, Web of Science, OVID and Clinicaltrials.gov database for eligible randomized controlled trials (RCTs) published before January 1, 2024. RCTs which treatment with vasodilators to COPD patients were included. Gas-blood exchange indicators were the primary outcomes, and ventilation function and quality of life indicators were the secondary outcomes. Mean differences with 95% confidence intervals were extracted. Subgroup analysis of vasodilator category and COPD complicated with or without pulmonary hypertension (PH) were performed. The risk of bias was assessed using Cochrane risk of bias tool, and the meta-analysis was conducted. RESULTS Twenty studies with a total sample size of 986 were included. The results showed that the 2 types of drugs in vasodilators included PDE-5 inhibitors could improve DLCO (MD = 6.56 [95% CI (1.74, 11.39)], P = .008) and iNO could reduce PaCO2 (MD = -0.10 [95% CI (-0.17, -0.03)], P = .006). Vasodilators could reduce PaCO2 in COPD complicated with PH (COPD-PH) (MD = -0.10 [95% CI (-0.17, -0.03)], P = .006). There were no statistically significant differences in FEV1 (MD = 0.02 [95% CI (-0.11, 0.16)], P = .74), FEV1% predicted (MD = 0.07 [95% CI (-1.90, 2.05)], P = .94), FEV1/FVC (MD = 0.70 [95% CI (-4.02, 5.42)], P = .77) and VE/VCO2 (MD = -0.17 [95% CI (-2.39, 2.05)], P = .88) levels. The total SGRQ score was significantly lower in vasodilator groups (MD = -5.53 [95% CI (-9.81, -1.24)], P = .01). CONCLUSIONS The therapeutic effects of vasodilators for COPD are controversial. In this meta-analysis, vasodilators have benefits in improving gas-blood exchange function and quality of life in COPD patients. However, vasodilators may have a limited capacity to improve pulmonary function.
Collapse
Affiliation(s)
- Ningxin Han
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Yujie Yin
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Peipei Jin
- Graduate School, Hebei University of Traditional Chinese Medicine, Shijiazhuang, China
| | - Yunlong Hou
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
| | - Zhenhua Jia
- Graduate School, Hebei Medical University, Shijiazhuang, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, China
- Hebei Yiling Hospital, High-level TCM Key Disciplines of National Administration of Traditional Chinese Medicine – Luobing Theory, Shijiazhuang, China
| |
Collapse
|
4
|
Qi H, Hou Y, Han N, Jin P, Guo J, Wang T, Jia Z. Impairment of pulmonary microvascular endothelial cells: The ground for COPD? Chin Med J (Engl) 2024; 137:2369-2371. [PMID: 39227327 PMCID: PMC11441917 DOI: 10.1097/cm9.0000000000003280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Indexed: 09/05/2024] Open
Affiliation(s)
- Hui Qi
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, Hebei 050091, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei 050035, China
- High-Level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, Hebei 050035, China
| | - Yunlong Hou
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, Hebei 050091, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei 050035, China
- High-Level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, Hebei 050035, China
| | - Ningxin Han
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050017, China
| | - Peipei Jin
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050090, China
| | - Jing Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050090, China
| | - Tongxing Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, Hebei 050091, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei 050035, China
- High-Level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, Hebei 050035, China
| | - Zhenhua Jia
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, Hebei 050091, China
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, Hebei 050035, China
- High-Level TCM Key Disciplines of National Administration of Traditional Chinese Medicine-Luobing Theory, Shijiazhuang, Hebei 050035, China
- Hebei Yiling Hospital, Shijiazhuang, Hebei 050090, China
| |
Collapse
|
5
|
Carpi S, Polini B, Nieri D, Doccini S, Conti M, Bazzan E, Pagnini M, Santorelli FM, Cecchini M, Nieri P, Celi A, Neri T. Extracellular Vesicles Induce Nuclear Factor-κB Activation and Interleukin-8 Synthesis through miRNA-191-5p Contributing to Inflammatory Processes: Potential Implications in the Pathogenesis of Chronic Obstructive Pulmonary Disease. Biomolecules 2024; 14:1030. [PMID: 39199417 PMCID: PMC11352467 DOI: 10.3390/biom14081030] [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: 07/08/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/01/2024] Open
Abstract
Extracellular vesicles (EVs) play a pivotal role in a variety of physiologically relevant processes, including lung inflammation. Recent attention has been directed toward EV-derived microRNAs (miRNAs), such as miR-191-5p, particularly in the context of inflammation. Here, we investigated the impact of miR-191-5p-enriched EVs on the activation of NF-κB and the expression of molecules associated with inflammation such as interleukin-8 (IL-8). To this aim, cells of bronchial epithelial origin, 16HBE, were transfected with miR-191-5p mimic and inhibitor and subsequently subjected to stimulations to generate EVs. Then, bronchial epithelial cells were exposed to the obtained EVs to evaluate the activation of NF-κB and IL-8 levels. Additionally, we conducted a preliminary investigation to analyze the expression profiles of miR-191-5p in EVs isolated from the plasma of patients diagnosed with chronic obstructive pulmonary disease (COPD). Our initial findings revealed two significant observations. First, the exposure of bronchial epithelial cells to miR-191-5p-enriched EVs activated the NF-kB signaling and increased the synthesis of IL-8. Second, we discovered the presence of miR-191-5p in peripheral blood-derived EVs from COPD patients and noted a correlation between miR-191-5p levels and inflammatory and functional parameters. Collectively, these data corroborate and further expand the proinflammatory role of EVs, with a specific emphasis on miR-191-5p as a key cargo involved in this process. Consequently, we propose a model in which miR-191-5p, carried by EVs, plays a role in airway inflammation and may contribute to the pathogenesis of COPD.
Collapse
Affiliation(s)
- Sara Carpi
- Department of Health Sciences, University ‘Magna Græcia’ of Catanzaro, 88100 Catanzaro, Italy;
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Beatrice Polini
- Department of Pathology, University of Pisa, 56100 Pisa, Italy;
| | - Dario Nieri
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| | - Stefano Doccini
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (S.D.); (F.M.S.)
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padua, Italy; (M.C.); (E.B.)
| | - Erika Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35122 Padua, Italy; (M.C.); (E.B.)
| | - Marta Pagnini
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| | - Filippo Maria Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy; (S.D.); (F.M.S.)
| | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy;
| | - Alessandro Celi
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| | - Tommaso Neri
- Centre for Cardio-Respiratory Cell Biology, Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, 56100 Pisa, Italy; (D.N.); (M.P.); (T.N.)
| |
Collapse
|
6
|
Gerard SE, Dougherty TM, Nagpal P, Jin D, Han MK, Newell JD, Saha PK, Comellas AP, Cooper CB, Couper D, Fortis S, Guo J, Hansel NN, Kanner RE, Kazeroni EA, Martinez FJ, Motahari A, Paine R, Rennard S, Schroeder JD, Woodruff PG, Barr RG, Smith BM, Hoffman EA. Vessel and Airway Characteristics in One-Year Computed Tomography-defined Rapid Emphysema Progression: SPIROMICS. Ann Am Thorac Soc 2024; 21:1022-1033. [PMID: 38530051 PMCID: PMC11284327 DOI: 10.1513/annalsats.202304-383oc] [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: 04/27/2023] [Accepted: 03/22/2024] [Indexed: 03/27/2024] Open
Abstract
Rationale: Rates of emphysema progression vary in chronic obstructive pulmonary disease (COPD), and the relationships with vascular and airway pathophysiology remain unclear. Objectives: We sought to determine if indices of peripheral (segmental and beyond) pulmonary arterial dilation measured on computed tomography (CT) are associated with a 1-year index of emphysema (EI; percentage of voxels <-950 Hounsfield units) progression. Methods: Five hundred ninety-nine former and never-smokers (Global Initiative for Chronic Obstructive Lung Disease stages 0-3) were evaluated from the SPIROMICS (Subpopulations and Intermediate Outcome Measures in COPD Study) cohort: rapid emphysema progressors (RPs; n = 188, 1-year ΔEI > 1%), nonprogressors (n = 301, 1-year ΔEI ± 0.5%), and never-smokers (n = 110). Segmental pulmonary arterial cross-sectional areas were standardized to associated airway luminal areas (segmental pulmonary artery-to-airway ratio [PAARseg]). Full-inspiratory CT scan-derived total (arteries and veins) pulmonary vascular volume (TPVV) was compared with small vessel volume (radius smaller than 0.75 mm). Ratios of airway to lung volume (an index of dysanapsis and COPD risk) were compared with ratios of TPVV to lung volume. Results: Compared with nonprogressors, RPs exhibited significantly larger PAARseg (0.73 ± 0.29 vs. 0.67 ± 0.23; P = 0.001), lower ratios of TPVV to lung volume (3.21 ± 0.42% vs. 3.48 ± 0.38%; P = 5.0 × 10-12), lower ratios of airway to lung volume (0.031 ± 0.003 vs. 0.034 ± 0.004; P = 6.1 × 10-13), and larger ratios of small vessel volume to TPVV (37.91 ± 4.26% vs. 35.53 ± 4.89%; P = 1.9 × 10-7). In adjusted analyses, an increment of 1 standard deviation in PAARseg was associated with a 98.4% higher rate of severe exacerbations (95% confidence interval, 29-206%; P = 0.002) and 79.3% higher odds of being in the RP group (95% confidence interval, 24-157%; P = 0.001). At 2-year follow-up, the CT-defined RP group demonstrated a significant decline in postbronchodilator percentage predicted forced expiratory volume in 1 second. Conclusions: Rapid one-year progression of emphysema was associated with indices indicative of higher peripheral pulmonary vascular resistance and a possible role played by pulmonary vascular-airway dysanapsis.
Collapse
Affiliation(s)
| | | | - Prashant Nagpal
- Department of Radiology, University of Wisconsin–Madison, Madison, Wisconsin
| | - Dakai Jin
- Department of Electrical and Computer Engineering
| | | | - John D. Newell
- Roy J. Carver Department of Biomedical Engineering
- Department of Radiology, and
| | - Punam K. Saha
- Department of Electrical and Computer Engineering
- Department of Radiology, and
| | | | - Christopher B. Cooper
- Department of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David Couper
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | | | - Junfeng Guo
- Roy J. Carver Department of Biomedical Engineering
- Department of Radiology, and
| | - Nadia N. Hansel
- Department of Medicine, The Johns Hopkins University, Baltimore, Maryland
| | | | - Ella A. Kazeroni
- Department of Radiology, Medical School, University of Michigan, Ann Arbor, Michigan
| | | | | | | | - Stephen Rennard
- Department of Internal Medicine, University of Nebraska, Omaha, Nebraska
| | | | - Prescott G. Woodruff
- Department of Medicine, University of California, San Francisco, San Francisco, California
| | - R. Graham Barr
- Department of Medicine and
- Department of Epidemiology, College of Medicine, Columbia University, New York, New York; and
| | - Benjamin M. Smith
- Department of Medicine and
- Department of Epidemiology, College of Medicine, Columbia University, New York, New York; and
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Eric A. Hoffman
- Roy J. Carver Department of Biomedical Engineering
- Department of Radiology, and
- Department of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
7
|
Jiang YZ, Huang XR, Chang J, Zhou Y, Huang XT. SIRT1: An Intermediator of Key Pathways Regulating Pulmonary Diseases. J Transl Med 2024; 104:102044. [PMID: 38452903 DOI: 10.1016/j.labinv.2024.102044] [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: 08/31/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Silent information regulator type-1 (SIRT1), a nicotinamide adenine dinucleotide+-dependent deacetylase, is a member of the sirtuins family and has unique protein deacetylase activity. SIRT1 participates in physiological as well as pathophysiological processes by targeting a wide range of protein substrates and signalings. In this review, we described the latest progress of SIRT1 in pulmonary diseases. We have introduced the basic information and summarized the prominent role of SIRT1 in several lung diseases, such as acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, lung cancer, and aging-related diseases.
Collapse
Affiliation(s)
- Yi-Zhu Jiang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xin-Ran Huang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Chang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China.
| |
Collapse
|
8
|
Gou Z, Yang H, Wang R, Wang S, Chen Q, Liu Z, Zhang Y. A new frontier in precision medicine: Exploring the role of extracellular vesicles in chronic obstructive pulmonary disease. Biomed Pharmacother 2024; 174:116443. [PMID: 38513597 DOI: 10.1016/j.biopha.2024.116443] [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: 01/29/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory airway disease characterized by progressive respiratory difficulties. It has a high incidence and disability rate worldwide. However, currently there is still a lack of highly effective treatment methods for COPD, only symptom relief is possible. Therefore, there is an urgent need to explore new treatment options. Almost all cells can secrete extracellular vesicles (EVs), which participate in many physiological activities by transporting cargoes and are associated with the pathogenesis of various diseases. Recently, many scholars have extensively studied the relationship between COPD and EVs, which has strongly demonstrated the significant impact of EVs from different sources on the occurrence and development of COPD. Therefore, EVs are a good starting point and new opportunity for the diagnosis and treatment of COPD. In this review, we mainly describe the current mechanisms of EVs in the pathogenesis of COPD, also the relationship between diagnosis, prognosis, and treatment. At the same time, we also introduce some new methods for COPD therapy based on EVs. It is hoped that this article can provide new ideas for future research and contribute to the development of precision medicine.
Collapse
Affiliation(s)
- Zixuan Gou
- Department of Pediateic Respiration, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Hongrun Yang
- Department of Pediateic Respiration, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Ruijia Wang
- Department of Pediateic Respiration, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Shihan Wang
- Department of Pediateic Respiration, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Qirui Chen
- Department of Pediateic Respiration, Children's Medical Center, The First Hospital of Jilin University, Changchun, China
| | - Ziyu Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China.
| | - Ying Zhang
- Department of Pediateic Respiration, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
9
|
Liu S, Tan X, Liu S. The role of extracellular vesicles in COPD and potential clinical value. Respir Res 2024; 25:84. [PMID: 38331841 PMCID: PMC10854156 DOI: 10.1186/s12931-024-02719-z] [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: 11/06/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous lung disease and a major health burden worldwide. Extracellular vesicles (EVs) are nanosized vesicles which possess a lipid bilayer structure that are secreted by various cells. They contain a variety of bioactive substances, which can regulate various physiological and pathological processes and are closely related to the development of diseases. Recently, EVs have emerged as a novel tool for intercellular crosstalk, which plays an essential role in COPD development. This paper reviews the role of EVs in the development of COPD and their potential clinical value, in order to provide a reference for further research on COPD.
Collapse
Affiliation(s)
- Shasha Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaowu Tan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Sha Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
10
|
Afzal A, Khawar MB, Habiba U, Afzal H, Hamid SE, Rafiq M, Abbasi MH, Sheikh N, Abaidullah R, Asif Z, Saeed T. Diagnostic and therapeutic value of EVs in lungs diseases and inflammation. Mol Biol Rep 2023; 51:26. [PMID: 38127201 DOI: 10.1007/s11033-023-09045-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/02/2023] [Indexed: 12/23/2023]
Abstract
Extracellular vesicles (EVs) are membrane-derived messengers which have been playing an important role in the inflammation and pathogenesis of lung diseases. EVs contain varieties of DNA, RNA, and membrane receptors through which they work as a delivery system for bioactive molecules as well as intracellular communicators. EV signaling mediates tumor progression and metastasis. EVs are linked with many diseases and perform a diagnostic role in lung injury and inflammation so are used to diagnose the severity of diseases. EVs containing a variety of biomolecules communicate with the recipient cells during pathophysiological mechanisms thereby acquiring the attention of clinicians toward the diagnostic and therapeutic potential of EVs in different lung diseases. In this review, we summarize the role of EVs in inflammation with an emphasis on their potential as a novel candidate in the diagnostics and therapeutics of chronic obstructive pulmonary disease, asthma, and sarcoidosis.
Collapse
Affiliation(s)
- Ali Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Muhammad Babar Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan.
| | - Ume Habiba
- Department of Zoology, University of Education, Lahore, Pakistan
| | - Hanan Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Syeda Eisha Hamid
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Mussarat Rafiq
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Rimsha Abaidullah
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Zoya Asif
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Tahaa Saeed
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
11
|
Vitucci EC, Carberry CK, Payton A, Herring LE, Mordant AL, McCullough SD, Rager JE. Characterizing the extracellular vesicle proteomic landscape of the human airway using in vitro organotypic multi-cellular models. iScience 2023; 26:108162. [PMID: 37920665 PMCID: PMC10618692 DOI: 10.1016/j.isci.2023.108162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/01/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023] Open
Abstract
Extracellular vesicle (EV)-mediated intercellular communication significantly influences pulmonary cell health and disease, yet in vitro methods to investigate these mechanisms are limited. We hypothesize that organotypic models of the airway can be leveraged to investigate EV-mediated intercellular signaling, focusing on EV proteomic content as a case study. Two in vitro airway culture models were evaluated by mass spectrometry-based proteomics analysis: a tri-culture model consisting of alveolar epithelial, fibroblast, and lung microvascular endothelial cells and a co-culture model of alveolar epithelial and fibroblasts. EVs isolated from the tri-culture model were enriched with EV proteins regulating RNA-to-protein translation. EVs isolated from the co-culture model were enriched with EV biogenesis and extracellular matrix signaling proteins. These model-specific differences suggest that different pulmonary cell types uniquely affect EV composition and the biological pathways influenced by the EV proteome in recipient cells. These findings can inform future studies surrounding EV-related pulmonary disease pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Eva C.M. Vitucci
- Interdisciplinary Faculty of Toxicology, School of Public Health, Texas A&M University, College Station, TX, USA
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
| | - Celeste K. Carberry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexis Payton
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E. Herring
- UNC Proteomics Core Facility, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angie L. Mordant
- UNC Proteomics Core Facility, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shaun D. McCullough
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Chapel Hill, NC, USA
- Exposure and Protection, RTI International, Durham, NC, USA
| | - Julia E. Rager
- Curriculum in Toxicology & Environmental Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- The Center for Environmental Medicine, Asthma and Lung Biology, School of Medicine, The University of North Carolina, Chapel Hill, NC, USA
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Angelini ED, Yang J, Balte PP, Hoffman EA, Manichaikul AW, Sun Y, Shen W, Austin JHM, Allen NB, Bleecker ER, Bowler R, Cho MH, Cooper CS, Couper D, Dransfield MT, Garcia CK, Han MK, Hansel NN, Hughes E, Jacobs DR, Kasela S, Kaufman JD, Kim JS, Lappalainen T, Lima J, Malinsky D, Martinez FJ, Oelsner EC, Ortega VE, Paine R, Post W, Pottinger TD, Prince MR, Rich SS, Silverman EK, Smith BM, Swift AJ, Watson KE, Woodruff PG, Laine AF, Barr RG. Pulmonary emphysema subtypes defined by unsupervised machine learning on CT scans. Thorax 2023; 78:1067-1079. [PMID: 37268414 PMCID: PMC10592007 DOI: 10.1136/thorax-2022-219158] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/03/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Treatment and preventative advances for chronic obstructive pulmonary disease (COPD) have been slow due, in part, to limited subphenotypes. We tested if unsupervised machine learning on CT images would discover CT emphysema subtypes with distinct characteristics, prognoses and genetic associations. METHODS New CT emphysema subtypes were identified by unsupervised machine learning on only the texture and location of emphysematous regions on CT scans from 2853 participants in the Subpopulations and Intermediate Outcome Measures in COPD Study (SPIROMICS), a COPD case-control study, followed by data reduction. Subtypes were compared with symptoms and physiology among 2949 participants in the population-based Multi-Ethnic Study of Atherosclerosis (MESA) Lung Study and with prognosis among 6658 MESA participants. Associations with genome-wide single-nucleotide-polymorphisms were examined. RESULTS The algorithm discovered six reproducible (interlearner intraclass correlation coefficient, 0.91-1.00) CT emphysema subtypes. The most common subtype in SPIROMICS, the combined bronchitis-apical subtype, was associated with chronic bronchitis, accelerated lung function decline, hospitalisations, deaths, incident airflow limitation and a gene variant near DRD1, which is implicated in mucin hypersecretion (p=1.1 ×10-8). The second, the diffuse subtype was associated with lower weight, respiratory hospitalisations and deaths, and incident airflow limitation. The third was associated with age only. The fourth and fifth visually resembled combined pulmonary fibrosis emphysema and had distinct symptoms, physiology, prognosis and genetic associations. The sixth visually resembled vanishing lung syndrome. CONCLUSION Large-scale unsupervised machine learning on CT scans defined six reproducible, familiar CT emphysema subtypes that suggest paths to specific diagnosis and personalised therapies in COPD and pre-COPD.
Collapse
Affiliation(s)
- Elsa D Angelini
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- LTCI, Institut Polytechnique de Paris, Telecom Paris, Palaiseau, France
- NIHR Imperial Biomedical Research Centre, ITMAT Data Science Group, Imperial College, London, UK
| | - Jie Yang
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Pallavi P Balte
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Eric A Hoffman
- Departments of Radiology, Medicine and Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Yifei Sun
- Department of Biostatistics, Columbia University Irving Medical Center, New York, New York, USA
| | - Wei Shen
- Department of Pediatrics, Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York, USA
- Columbia Magnetic Resonance Research Center (CMRRC), Columbia University Irving Medical Center, New York, New York, USA
| | - John H M Austin
- Department of Radiology, Columbia University Irving Medical Center, New York, New York, USA
| | - Norrina B Allen
- Institute for Public Health and Medicine (IPHAM) - Center for Epidemiology and Population Health, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Eugene R Bleecker
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Russell Bowler
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - David Couper
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Christine Kim Garcia
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - MeiLan K Han
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nadia N Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Emlyn Hughes
- Department of Physics, Columbia University, New York, New York, USA
| | - David R Jacobs
- Division of Epidemiology and Community Public Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Silva Kasela
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
- New York Genome Center, New York, New York, USA
| | - Joel Daniel Kaufman
- Departments of Environmental & Occupational Health Sciences, Medicine, and Epidemiology, University of Washington, Seattle, Washington, USA
| | - John Shinn Kim
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Tuuli Lappalainen
- Department of Systems Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Joao Lima
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel Malinsky
- Department of Biostatistics, Columbia University Irving Medical Center, New York, New York, USA
| | - Fernando J Martinez
- Department of Medicine, Cornell University Joan and Sanford I Weill Medical College, New York, New York, USA
| | - Elizabeth C Oelsner
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Victor E Ortega
- Department of Pulmonary Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Robert Paine
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Wendy Post
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tess D Pottinger
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Martin R Prince
- Department of Radiology, Cornell University Joan and Sanford I Weill Medical College, New York, New York, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Benjamin M Smith
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Andrew J Swift
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, UK
| | - Karol E Watson
- Department of Medicine, University of California, Los Angeles, California, USA
| | - Prescott G Woodruff
- Department of Medicine, University of California, San Francisco, California, USA
| | - Andrew F Laine
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
- Columbia Magnetic Resonance Research Center (CMRRC), Columbia University Irving Medical Center, New York, New York, USA
- Department of Radiology, Columbia University Irving Medical Center, New York, New York, USA
| | - R Graham Barr
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
- Department of Epidemiology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
13
|
Kalia V, Baccarelli AA, Happel C, Hollander JA, Jukic AM, McAllister KA, Menon R, Merrick BA, Milosavljevic A, Ravichandran LV, Roth ME, Subramanian A, Tyson FL, Worth L, Shaughnessy DT. Seminar: Extracellular Vesicles as Mediators of Environmental Stress in Human Disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:104201. [PMID: 37861803 PMCID: PMC10588739 DOI: 10.1289/ehp12980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs), membrane-bound particles containing a variety of RNA types, DNA, proteins, and other macromolecules, are now appreciated as an important means of communication between cells and tissues, both in normal cellular physiology and as a potential indicator of cellular stress, environmental exposures, and early disease pathogenesis. Extracellular signaling through EVs is a growing field of research for understanding fundamental mechanisms of health and disease and for the potential for biomarker discovery and therapy development. EVs are also known to play important roles in mediating the effects of exposure to environmental stress. OBJECTIVES This seminar addresses the application of new tools and approaches for EV research, developed in part through the National Institutes of Health (NIH) Extracellular RNA Communication Program, and reflects presentations and discussions from a workshop held 27-28 September 2021 by the National Institute of Environmental Health Sciences (NIEHS) and the National Center for Advancing Translational Sciences (NCATS) on "Extracellular Vesicles, Exosomes, and Cell-Cell Signaling in Response to Environmental Stress." The panel of experts discussed current research on EVs and environmental exposures, highlighted recent advances in EV isolation and characterization, and considered research gaps and opportunities toward identifying and characterizing the roles for EVs in environmentally related diseases, as well as the current challenges and opportunities in this field. DISCUSSION The authors discuss the application of new experimental models, particularly organ-on-chip (OOC) systems and in vitro approaches and how these have the potential to extend findings in population-based studies of EVs in exposure-related diseases. Given the complex challenges of identifying cell-specific EVs related to environmental exposures, as well as the general heterogeneity and variability in EVs in blood and other accessible biological samples, there is a critical need for rigorous reporting of experimental methods and validation studies. The authors note that these efforts, combined with cross-disciplinary approaches, would ensure that future research efforts in environmental health studies on EV biomarkers are rigorous and reproducible. https://doi.org/10.1289/EHP12980.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Christine Happel
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), U.S. Department of Health and Human Services (DHHS), Bethesda, Maryland, USA
| | - Jonathan A. Hollander
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Anne Marie Jukic
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Kimberly A. McAllister
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Bruce A. Merrick
- Division of Translational Toxicology, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | | | - Lingamanaidu V. Ravichandran
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Matthew E. Roth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Anita Subramanian
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Frederick L. Tyson
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Leroy Worth
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Daniel T. Shaughnessy
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
14
|
Raoof S, Shah M, Braman S, Agrawal A, Allaqaband H, Bowler R, Castaldi P, DeMeo D, Fernando S, Hall CS, Han MK, Hogg J, Humphries S, Lee HY, Lee KS, Lynch D, Machnicki S, Mehta A, Mehta S, Mina B, Naidich D, Naidich J, Ohno Y, Regan E, van Beek EJR, Washko G, Make B. Lung Imaging in COPD Part 2: Emerging Concepts. Chest 2023; 164:339-354. [PMID: 36907375 PMCID: PMC10475822 DOI: 10.1016/j.chest.2023.02.049] [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: 10/06/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/13/2023] Open
Abstract
The diagnosis, prognostication, and differentiation of phenotypes of COPD can be facilitated by CT scan imaging of the chest. CT scan imaging of the chest is a prerequisite for lung volume reduction surgery and lung transplantation. Quantitative analysis can be used to evaluate extent of disease progression. Evolving imaging techniques include micro-CT scan, ultra-high-resolution and photon-counting CT scan imaging, and MRI. Potential advantages of these newer techniques include improved resolution, prediction of reversibility, and obviation of radiation exposure. This article discusses important emerging techniques in imaging patients with COPD. The clinical usefulness of these emerging techniques as they stand today are tabulated for the benefit of the practicing pulmonologist.
Collapse
Affiliation(s)
- Suhail Raoof
- Northwell Health, Lenox Hill Hospital, New York, NY.
| | - Manav Shah
- Northwell Health, Lenox Hill Hospital, New York, NY
| | - Sidney Braman
- Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | | | | | - Dawn DeMeo
- Brigham and Women's Hospital, Boston, MA
| | | | | | | | - James Hogg
- University of British Columbia, Vancouver, BC, Canada
| | | | - Ho Yun Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Health Sciences and Technology, Sungkyunkwan University, ChangWon, South Korea
| | - Kyung Soo Lee
- Sungkyunkwan University School of Medicine, Samsung ChangWon Hospital, ChangWon, South Korea
| | | | | | | | | | - Bushra Mina
- Northwell Health, Lenox Hill Hospital, New York, NY
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Stern RA, Lawrence J, Wolfson JM, Li L, Koutrakis P. Radon sampling methodologies: A case for accurate, accessible measurements using household instruments. JOURNAL OF THE AIR & WASTE MANAGEMENT ASSOCIATION (1995) 2023; 73:519-524. [PMID: 37311115 PMCID: PMC11380572 DOI: 10.1080/10962247.2023.2223149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/26/2023] [Accepted: 05/10/2023] [Indexed: 06/15/2023]
Abstract
Radon is a prevalent carcinogenic gas and the leading cause of lung cancer in the United States besides smoking. As the residential environment is the primary source of radon exposure, accessible and accurate measurements of radon in this environment are essential. However, no radon monitors have been evaluated that are inexpensive enough for regular household use. In this study, we examine two household-grade, continuous monitoring devices, the Ecosense RadonEye and EcoQube. We compare them to two research-grade instruments, the Durridge Company Rad7 and the Rad Elec Inc. E-PERM. In our study, the Ecosense household radon monitors performed accurately and can be used by homeowners and researchers alike as an affordable and reliable radon sensor.Implications: The ability of homeowners and renters to regularly monitor the radon levels inside their home is an important preventative health measure. However, low-cost instrumentation is needed that can provide accurate radon measurements. In this study, we show that the affordable Ecosense continuous monitors produce results that are in line with expensive research-grade instruments in a residential environment, over a range of concentrations. The Ecosense monitors may be suitable for home use, and they may provide a solution that can be used by policymakers and home-dwellers alike to improve regular radon monitoring in residences.
Collapse
Affiliation(s)
- Rebecca A. Stern
- Department of Environmental Health, Harvard T.H. Chan School of Public Heath, Boston, MA 02115 USA
| | - Joy Lawrence
- Department of Environmental Health, Harvard T.H. Chan School of Public Heath, Boston, MA 02115 USA
| | - Jack M. Wolfson
- Department of Environmental Health, Harvard T.H. Chan School of Public Heath, Boston, MA 02115 USA
| | - Longxiang Li
- Department of Environmental Health, Harvard T.H. Chan School of Public Heath, Boston, MA 02115 USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Heath, Boston, MA 02115 USA
| |
Collapse
|
16
|
Tinè M, Neri T, Biondini D, Bernardinello N, Casara A, Conti M, Minniti M, Cosio MG, Saetta M, Celi A, Nieri D, Bazzan E. Do Circulating Extracellular Vesicles Strictly Reflect Bronchoalveolar Lavage Extracellular Vesicles in COPD? Int J Mol Sci 2023; 24:ijms24032966. [PMID: 36769286 PMCID: PMC9918055 DOI: 10.3390/ijms24032966] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Cell-derived extracellular vesicles (EVs) found in the circulation and body fluids contain biomolecules that could be used as biomarkers for lung and other diseases. EVs from bronchoalveolar lavage (BAL) might be more informative of lung abnormalities than EVs from blood, where information might be diluted. To compare EVs' characteristics in BAL and blood in smokers with and without COPD. Same-day BAL and blood samples were obtained in 9 nonsmokers (NS), 11 smokers w/o COPD (S), and 9 with COPD (SCOPD) (FEV1: 59 ± 3% pred). After differential centrifugation, EVs (200-500 nm diameter) were identified by flow cytometry and labeled with cell-type specific antigens: CD14 for macrophage-derived EVs, CD326 for epithelial-derived EVs, CD146 for endothelial-derived EVs, and CD62E for activated-endothelial-derived EVs. In BAL, CD14-EVs were increased in S compared to NS [384 (56-567) vs. 172 (115-282) events/μL; p = 0.007] and further increased in SCOPD [619 (224-888)] compared to both S (p = 0.04) and NS (p < 0.001). CD326-EVs were increased in S [760 (48-2856) events/μL, p < 0.001] and in SCOPD [1055 (194-11,491), p < 0.001] when compared to NS [15 (0-68)]. CD146-EVs and CD62E-EVs were similar in the three groups. In BAL, significant differences in macrophage and epithelial-derived EVs can be clearly detected between NS, S and SCOPD, while these differences were not found in plasma. This suggests that BAL is a better medium than blood to study EVs in lung diseases.
Collapse
Affiliation(s)
- Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Tommaso Neri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy
- Correspondence:
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Nicol Bernardinello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Alvise Casara
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Maria Conti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Marianna Minniti
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC H3A 0G4, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Alessandro Celi
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy
| | - Dario Nieri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| |
Collapse
|
17
|
Zhang L, Jiang F, Xie Y, Mo Y, Zhang X, Liu C. Diabetic endothelial microangiopathy and pulmonary dysfunction. Front Endocrinol (Lausanne) 2023; 14:1073878. [PMID: 37025413 PMCID: PMC10071002 DOI: 10.3389/fendo.2023.1073878] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 02/17/2023] [Indexed: 04/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread metabolic condition with a high global morbidity and mortality rate that affects the whole body. Their primary consequences are mostly caused by the macrovascular and microvascular bed degradation brought on by metabolic, hemodynamic, and inflammatory variables. However, research in recent years has expanded the target organ in T2DM to include the lung. Inflammatory lung diseases also impose a severe financial burden on global healthcare. T2DM has long been recognized as a significant comorbidity that influences the course of various respiratory disorders and their disease progress. The pathogenesis of the glycemic metabolic problem and endothelial microangiopathy of the respiratory disorders have garnered more attention lately, indicating that the two ailments have a shared history. This review aims to outline the connection between T2DM related endothelial cell dysfunction and concomitant respiratory diseases, including Coronavirus disease 2019 (COVID-19), asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
- Lanlan Zhang
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lanlan Zhang, ; Xin Zhang, ; Chuntao Liu,
| | - Faming Jiang
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Yingying Xie
- Department of Nephrology, Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yan Mo
- Department of Neurology Medicine, The Aviation Industry Corporation of China (AVIC) 363 Hospital, Chengdu, China
| | - Xin Zhang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lanlan Zhang, ; Xin Zhang, ; Chuntao Liu,
| | - Chuntao Liu
- Department of Respiratory and Critical Care Medicine, Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- *Correspondence: Lanlan Zhang, ; Xin Zhang, ; Chuntao Liu,
| |
Collapse
|
18
|
Extracellular Vesicles' Role in the Pathophysiology and as Biomarkers in Cystic Fibrosis and COPD. Int J Mol Sci 2022; 24:ijms24010228. [PMID: 36613669 PMCID: PMC9820204 DOI: 10.3390/ijms24010228] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/03/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
In keeping with the extraordinary interest and advancement of extracellular vesicles (EVs) in pathogenesis and diagnosis fields, we herein present an update to the knowledge about their role in cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD). Although CF and COPD stem from a different origin, one genetic and the other acquired, they share a similar pathophysiology, being the CF transmembrane conductance regulator (CFTR) protein implied in both disorders. Various subsets of EVs, comprised mainly of microvesicles (MVs) and exosomes (EXOs), are secreted by various cell types that are either resident or attracted in the airways during the onset and progression of CF and COPD lung disease, representing a vehicle for metabolites, proteins and RNAs (especially microRNAs), that in turn lead to events as such neutrophil influx, the overwhelming of proteases (elastase, metalloproteases), oxidative stress, myofibroblast activation and collagen deposition. Eventually, all of these pathomechanisms lead to chronic inflammation, mucus overproduction, remodeling of the airways, and fibrosis, thus operating a complex interplay among cells and tissues. The detection of MVs and EXOs in blood and biological fluids coming from the airways (bronchoalveolar lavage fluid and sputum) allows the consideration of EVs and their cargoes as promising biomarkers for CF and COPD, although clinical expectations have yet to be fulfilled.
Collapse
|
19
|
Burke H, Cellura D, Freeman A, Hicks A, Ostridge K, Watson A, Williams NP, Spalluto CM, Staples KJ, Wilkinson TMA. Pulmonary EV miRNA profiles identify disease and distinct inflammatory endotypes in COPD. Front Med (Lausanne) 2022; 9:1039702. [PMID: 36590967 PMCID: PMC9797812 DOI: 10.3389/fmed.2022.1039702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/28/2022] [Indexed: 12/16/2022] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a heterogeneous condition without effective disease modifying therapies. Identification of novel inflammatory endotype markers such as extracellular vesicles (EVs), which are important intercellular messengers carrying microRNA (miRNA), may enable earlier diagnosis and disease stratification for a targeted treatment approach. Our aim was to identify differentially expressed EV miRNA in the lungs of COPD patients compared with healthy ex-smokers and determine whether they can help define inflammatory COPD endotypes. Methods EV miRNA were isolated and sequenced from ex-smoking COPD patients and healthy ex-smoker bronchoalveolar lavage fluid. Results were validated with RT-qPCR and compared to differential inflammatory cell counts. Results Expression analysis identified five upregulated miRNA in COPD (miR-223-3p, miR-2110, miR-182-5p, miR-200b-5p and miR-625-3p) and three downregulated miRNA (miR-138-5p, miR-338-3p and miR-204-5p), all with a log2 fold change of >1/-1, FDR < 0.05. These miRNAs correlated with disease defining characteristics such as FEF 25-75% (a small airways disease measure) and DLCO% (a surrogate measure of emphysema). Receiver operator curve analysis demonstrated miR-2110, miR-223-3p, and miR-182-5p showed excellent combinatory predictive ability (AUC 0.91, p < 0.0001) in differentiating between health and mild COPD. Furthermore, miR-223-3p and miR-338-3p correlated with airway eosinophilia and were able to distinguish "pure eosinophilic" COPD from other airway inflammatory subtypes (AUC 0.94 and 0.85, respectively). Discussion This is the first study to identify differentially expressed miRNA in COPD bronchoalveolar lavage fluid EVs. These findings suggest specific lung derived EV miRNA are a strong predictor of disease presence even in mild COPD. Furthermore, specific miRNA correlated with inflammatory cell numbers in COPD, and may have a role in defining inflammatory endotypes for future treatment stratification.
Collapse
Affiliation(s)
- Hannah Burke
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Doriana Cellura
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Anna Freeman
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Alex Hicks
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Kris Ostridge
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alastair Watson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Nicholas P. Williams
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Karl J. Staples
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Tom M. A. Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| |
Collapse
|
20
|
Dong S, Koutrakis P, Li L, Coull BA, Schwartz J, Kosheleva A, Zanobetti A. Synergistic Effects of Particle Radioactivity (Gross β Activity) and Particulate Matter ≤2.5 μm Aerodynamic Diameter on Cardiovascular Disease Mortality. J Am Heart Assoc 2022; 11:e025470. [PMID: 36197036 PMCID: PMC9673676 DOI: 10.1161/jaha.121.025470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022]
Abstract
Background Although the effects of fine particulate matter (particulate matter ≤2.5 μm aerodynamic diameter [PM2.5]) on cardiovascular disease (CVD) morbidity and mortality are well established, little is known about the CVD health effects of particle radioactivity. In addition, there are still questions about which of the PM2.5 physical, chemical, or biological properties are mostly responsible for its toxicity. Methods and Results We investigated the association between particle radioactivity, measured as gross β activity from highly resolved spatiotemporal predictions, and mortality for CVD, myocardial infarction, stroke, and all-cause nonaccidental mortality in Massachusetts (2001-2015). Within both difference-in-differences model and generalized linear mixed model frameworks, we fit both single-exposure and 2-exposure models adjusting for PM2.5 and examined the interaction between PM2.5 and gross β activity. We found significant associations between gross β activity and PM2.5 and each mortality cause. Using difference-in-differences and adjusting for PM2.5, we found the highest associations with myocardial infarction (rate ratio, 1.16 [95% CI, 1.08-1.24]) and stroke (rate ratio, 1.11 [95% CI, 1.04-1.18]) for an interquartile range increase (0.055 millibecquerels per cubic meter) in gross β activity. We found a significant positive interaction between PM2.5 and gross β activity, with higher associations between PM2.5 and mortality at a higher level of gross β activity. We also observed that the associations varied across age groups. The results were comparable between the 2 statistical methods also with and without adjusting for PM2.5. Conclusions This is the first study that, using highly spatiotemporal predictions of gross β-activity, provides evidence that particle radioactivity increases CVD mortality and enhances PM2.5 CVD mortality. Therefore, particle radioactivity can be an important property of PM2.5 that must be further investigated. Addressing this important question can lead to cost-effective air-quality regulations.
Collapse
Affiliation(s)
- Shuxin Dong
- Department of Environmental Health, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| | - Petros Koutrakis
- Department of Environmental Health, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| | - Longxiang Li
- Department of Environmental Health, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| | - Brent A. Coull
- Department of Biostatistics, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| | - Joel Schwartz
- Department of Environmental Health, T.H. Chan School of Public HealthHarvard UniversityBostonMA
- Department of Epidemiology, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| | - Anna Kosheleva
- Department of Environmental Health, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| | - Antonella Zanobetti
- Department of Environmental Health, T.H. Chan School of Public HealthHarvard UniversityBostonMA
| |
Collapse
|
21
|
Bronchial Asthma as a Cardiovascular Risk Factor: A Prospective Observational Study. Biomedicines 2022; 10:biomedicines10102614. [PMID: 36289876 PMCID: PMC9599703 DOI: 10.3390/biomedicines10102614] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: Asthma as a chronic inflammatory disorder has been suggested as a risk factor for endothelial dysfunction (ED), but studies on the association between asthma and cardiovascular disease (CVD) risk are limited. Background: We assessed associations of ED with the severity of asthma, eosinophilic inflammation, lung function, and asthma control. Methods: 52 young asthmatics (median age of 25.22 years) and 45 healthy individuals were included. Demographic, clinical, and laboratory findings were recorded. We evaluated microvascular responsiveness by recording the reactive hyperemia index (RHI) indicating post-occlusive peripheral endothelium-dependent changes in vascular tone using the Itamar Medical EndoPAT2000. VCAM-1, ADMA, high-sensitive CRP (hsCRP), and E-selectin were measured. Results: Asthmatics had considerably lower RHI values (p < 0.001) with a dynamic decreasing trend by asthma severity and higher hsCRP levels (p < 0.001). A substantial increase in hsCRP and E-selectin with asthma severity (p < 0.05) was also observed. We confirmed a higher body mass index (BMI) in asthmatics (p < 0.001), especially in women and in severe asthma. Conclusions: We demonstrated the progression of CVD in asthmatics and the association of the ongoing deterioration of ED with the inflammatory severity, suggesting that the increased risk of CVD in young asthmatics is dependent on disease severity. The underlying mechanisms of risk factors for CVD and disease control require further study.
Collapse
|
22
|
Machine-learning algorithms for asthma, COPD, and lung cancer risk assessment using circulating microbial extracellular vesicle data and their application to assess dietary effects. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1586-1595. [PMID: 36180580 PMCID: PMC9534896 DOI: 10.1038/s12276-022-00846-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/17/2022] [Accepted: 07/12/2022] [Indexed: 11/08/2022]
Abstract
Although mounting evidence suggests that the microbiome has a tremendous influence on intractable disease, the relationship between circulating microbial extracellular vesicles (EVs) and respiratory disease remains unexplored. Here, we developed predictive diagnostic models for COPD, asthma, and lung cancer by applying machine learning to microbial EV metagenomes isolated from patient serum and coded by their accumulated taxonomic hierarchy. All models demonstrated high predictive strength with mean AUC values ranging from 0.93 to 0.99 with various important features at the genus and phylum levels. Application of the clinical models in mice showed that various foods reduced high-fat diet-associated asthma and lung cancer risk, while COPD was minimally affected. In conclusion, this study offers a novel methodology for respiratory disease prediction and highlights the utility of serum microbial EVs as data-rich features for noninvasive diagnosis. Artificial intelligence (AI) has enabled researchers to intercept microbial messages bearing clinically useful information about of a variety of respiratory disorders. The organisms that comprise our microbiome communicate via the release of tiny, biomolecule-laden membrane bubbles called ‘extracellular vesicles’ (EVs) into the bloodstream. EVs are also influenced by human disease. South Korean researchers led by Yoon-Keun Kim of MD Healthcare, Seoul, and Young-Koo Jee of Dankook University College of Medicine, Cheonan, have used an AI algorithm to assemble EV-based profiles that can discriminate between healthy people and those with conditions like asthma or lung cancer. Their analysis of 1727 patient serum samples revealed microbial signatures that enabled accurate diagnosis of several respiratory disorders. Preliminary experiments in mice suggest that certain dietary changes could help shift the microbiome of high-risk individuals towards a healthier profile.
Collapse
|
23
|
Abstract
ABSTRACT Extracellular vesicles (EVs) are anuclear particles composed of lipid bilayers that contain nucleic acids, proteins, lipids, and organelles. EVs act as an important mediator of cell-to-cell communication by transmitting biological signals or components, including lipids, proteins, messenger RNAs, DNA, microRNAs, organelles, etc, to nearby or distant target cells to activate and regulate the function and phenotype of target cells. Under physiological conditions, EVs play an essential role in maintaining the homeostasis of the pulmonary milieu but they can also be involved in promoting the pathogenesis and progression of various respiratory diseases including chronic obstructive pulmonary disease, asthma, acute lung injury/acute respiratory distress syndrome, idiopathic pulmonary fibrosis (IPF), and pulmonary artery hypertension. In addition, in multiple preclinical studies, EVs derived from mesenchymal stem cells (EVs) have shown promising therapeutic effects on reducing and repairing lung injuries. Furthermore, in recent years, researchers have explored different methods for modifying EVs or enhancing EVs-mediated drug delivery to produce more targeted and beneficial effects. This article will review the characteristics and biogenesis of EVs and their role in lung homeostasis and various acute and chronic lung diseases and the potential therapeutic application of EVs in the field of clinical medicine.
Collapse
|
24
|
Xu Y, Liang T, Ma Y, Xie S, Sun H, Wang L, Xu Y. Strain Analysis in Patients at High-Risk for COPD Using Four-Dimensional Dynamic-Ventilation CT. Int J Chron Obstruct Pulmon Dis 2022; 17:1121-1130. [PMID: 35573658 PMCID: PMC9094643 DOI: 10.2147/copd.s360770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/01/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To quantitatively identify abnormal lung motion in chronic obstructive pulmonary disease (COPD) using strain analysis, and further clarify the potential differences of deformation in COPD with different severity of airflow limitation. Materials and Methods Totally, 53 patients at high-risk for COPD were enrolled in this study. All CT examinations were performed on a 320-row MDCT scanner, and strain measurement based on dynamic-ventilation CT data was performed with a computational fluid dynamics analysis software (Micro Vec V3.6.2). The strain-related parameters derived from the whole expiration phase (PSmax-all, PSmean-all, Speedmax-all ), the first 2s of expiration phase (PSmax2s, PSmean2s, Speedmax2s ) were divided respectively by the changes in lung volume to adjust for the degree of expiration. Spearman rank correlation analysis was used to evaluate associations between the strain-related parameters and various spirometric parameters. Comparisons of the strain-related parameters between COPD and non-COPD patients, between GOLD I (mild airflow restriction) and GOLD II-IV (moderate to severe airflow restriction) were made using the Mann-Whitney U-test. Receiver-operating characteristic (ROC) analysis was performed to evaluate the diagnostic performance of the strain-related parameters for COPD. P <0.05 was considered statistically significant. Results Strain-related parameters demonstrated positive correlations with spirometric parameters (ρ=0.275~0.687, P<0.05), suggesting that heterogeneity in lung motion was related to abnormal spirometric results. Strain-related parameters can quantitatively distinguish COPD from non-COPD patients with moderate diagnostic significance with the AUC values ranged from 0.821 to 0.894. Furthermore, parameters of the whole expiration phase (PSmax-all, Speedmax-all) demonstrated significant differences (P=0.005; P=0.04) between COPD patients with mild and moderate to severe airflow limitation. Conclusion Strain-related parameters derived from dynamic-ventilation CT data covering the whole lung associated with lung function changes in COPD, reflecting the severity of airflow limitation in some degree, even though its utility in severe COPD patients remains to be investigated.
Collapse
Affiliation(s)
- Yanyan Xu
- Department of Radiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People’s Republic of China
| | - Tian Liang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yanhui Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Sheng Xie
- Department of Radiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, People’s Republic of China
| | - Hongliang Sun
- Department of Radiology, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Lei Wang
- Beijing MicroVec. Inc., Beijing, People’s Republic of China
| | - Yinghao Xu
- Canon Medical Systems, Beijing, People’s Republic of China
| |
Collapse
|
25
|
Neri T, Celi A, Tinè M, Bernardinello N, Cosio MG, Saetta M, Nieri D, Bazzan E. The Emerging Role of Extracellular Vesicles Detected in Different Biological Fluids in COPD. Int J Mol Sci 2022; 23:ijms23095136. [PMID: 35563528 PMCID: PMC9101666 DOI: 10.3390/ijms23095136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 01/08/2023] Open
Abstract
The pathogenesis of chronic obstructive pulmonary disease (COPD) is characterized by complex cellular and molecular mechanisms, not fully elucidated so far. It involves inflammatory cells (monocytes/macrophages, neutrophils, lymphocytes), cytokines, chemokines and, probably, new players yet to be clearly identified and described. Chronic local and systemic inflammation, lung aging and cellular senescence are key pathological events in COPD development and progression over time. Extracellular vesicles (EVs), released by virtually all cells both as microvesicles and exosomes into different biological fluids, are involved in intercellular communication and, therefore, represent intriguing players in pathobiological mechanisms (including those characterizing aging and chronic diseases); moreover, the role of EVs as biomarkers in different diseases, including COPD, is rapidly gaining recognition. In this review, after recalling the essential steps of COPD pathogenesis, we summarize the current evidence on the roles of EVs collected in different biological mediums as biomarkers in COPD and as potential players in the specific mechanisms leading to disease development. We will also briefly review the data on EV as potential therapeutic targets and potential therapeutic agents.
Collapse
Affiliation(s)
- Tommaso Neri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (T.N.); (A.C.); (D.N.)
| | - Alessandro Celi
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (T.N.); (A.C.); (D.N.)
| | - Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (N.B.); (M.G.C.); (M.S.)
| | - Nicol Bernardinello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (N.B.); (M.G.C.); (M.S.)
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (N.B.); (M.G.C.); (M.S.)
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QC H3A 0G4, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (N.B.); (M.G.C.); (M.S.)
| | - Dario Nieri
- Centro Dipartimentale di Biologia Cellulare Cardiorespiratoria, Dipartimento di Patologia Chirurgica, Medica, Molecolare e dell’Area Critica, Università degli Studi di Pisa, 56124 Pisa, Italy; (T.N.); (A.C.); (D.N.)
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy; (M.T.); (N.B.); (M.G.C.); (M.S.)
- Correspondence: ; Tel.: +39-049-821-3449
| |
Collapse
|
26
|
Zeng Y, Zhao Y, Chen Y, Cai S, Chen P. PECAM EMPs regulate apoptosis in pulmonary microvascular
endothelial cells in COPD by activating the Akt signaling
pathway. Tob Induc Dis 2022; 20:40. [PMID: 35592594 PMCID: PMC9059265 DOI: 10.18332/tid/146959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/01/2021] [Accepted: 02/25/2022] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Endothelial microparticles (EMPs) are partly associated with the progress of chronic obstructive pulmonary disease (COPD). We sought to measure the levels of EMPs in COPD patients and in human pulmonary microvascular endothelial cells (HPMECs) exposed to cigarette smoking extract (CSE) to elucidate the potential mechanisms of their action. METHODS We obtained prospectively blood EMPs from 30 stable COPD patients and 20 non-COPD volunteers. EMP subpopulations were determined by flow cytometry in platelet-free plasma according to the expression of membrane specific antigens. Cell growth, proliferation, apoptosis and the expression of protein kinase B (Akt) in HPMECs after exposure to PECAM EMPs were assessed. After intervention with an antioxidant (Eukarion-134, EUK-134), apoptosis and the expression of Akt in HPMECs were also measured. RESULTS Unlike those of MCAM EMPs, VE-cadherin, PECAM and E-selectin EMP values were significantly higher in the stable COPD patients than in the non-COPD volunteers (p<0.05). Only PECAM EMPs were higher in HPMECs exposed to CSE (p<0.05). Further, in vitro studies showed that the apoptosis rate and expression of cleaved caspase 3/9 in HPMECs increased in a dose- and time-independent manner with PECAM EMPs. The expression of phospho-Akt (p-Akt) decreased in a time-independent manner with PECAM EMPs (p<0.05). Compared with the control group, the early apoptosis rate of HPMECs was higher, and the expression of p-Akt was lower in both the PECAM EMP group and EUK-134 + PECAM EMP group (p<0.05). The apoptosis rate declined markedly, and the expression of p-Akt was higher in the EUK-134 + PECAM EMP group, compared with the PECAM EMPs group (p<0.05). CONCLUSIONS The present results suggest that PECAM EMPs positively regulate apoptosis in HPMECs in COPD, likely by decreasing Akt phosphorylation and can be protected by antioxidants.
Collapse
Affiliation(s)
- Yuqin Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Research Unit of Respiratory Disease, Central South University, Changsha, People’s Republic of China
- Hunan Centre for Evidence-Based Medicine, Changsha, People’s Republic of China
| | - Yiyang Zhao
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Research Unit of Respiratory Disease, Central South University, Changsha, People’s Republic of China
- Hunan Centre for Evidence-Based Medicine, Changsha, People’s Republic of China
| | - Yan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Research Unit of Respiratory Disease, Central South University, Changsha, People’s Republic of China
- Hunan Centre for Evidence-Based Medicine, Changsha, People’s Republic of China
| | - Shan Cai
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Research Unit of Respiratory Disease, Central South University, Changsha, People’s Republic of China
- Hunan Centre for Evidence-Based Medicine, Changsha, People’s Republic of China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Research Unit of Respiratory Disease, Central South University, Changsha, People’s Republic of China
- Hunan Centre for Evidence-Based Medicine, Changsha, People’s Republic of China
| |
Collapse
|
27
|
Gomez N, James V, Onion D, Fairclough LC. Extracellular vesicles and chronic obstructive pulmonary disease (COPD): a systematic review. Respir Res 2022; 23:82. [PMID: 35382831 PMCID: PMC8985325 DOI: 10.1186/s12931-022-01984-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/12/2022] [Indexed: 12/15/2022] Open
Abstract
Background Chronic Obstructive Pulmonary Disease (COPD) is a common inflammatory disease of the airways characterized by irreversible airflow limitation, ranking the third highest cause of death worldwide. Extracellular vesicles (EVs) are important intercellular communication mediators released by cells into their extracellular environment with the capacity to transfer biological signals. EVs involved in COPD hold great potential to understand disease pathogenesis and identify important biomarkers. This systematic review aims to examine all available research on EVs in the pathogenesis and diagnosis of COPD to identify existing knowledge and support further research within the field. Methods Publications were searched using PubMed and EMBASE with the search terms (Exosomes or extracellular vesicles or microvesicles or microparticles or ectosomes) AND (chronic obstructive pulmonary disease or COPD or emphysema or bronchitis). Results Initial search yielded 512 papers of which 142 were manually selected for review and 43 were eligible for analyses. The studies were divided into groups according to the role of EVs in pathogenesis, EV origin and cargo, their role in COPD exacerbations and their diagnostic utility. EVs were found to be involved in the mechanism of pathogenesis of COPD, derived from various cell types, as well as containing modified levels of miRNAs. EVs also varied according to the pathophysiological status of disease, therefore presenting a possible method for COPD diagnosis and progress monitoring. Conclusion The current findings show the limited but good quality research looking at the role of EVs in COPD, demonstrating the need for more studies to better define and provide further insight into the functional characteristics of EV in COPD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01984-0.
Collapse
Affiliation(s)
- Nancy Gomez
- School of Life Sciences, The University of Nottingham, Life Sciences Building, Nottingham, NG7 2RD, UK
| | - Victoria James
- School of Veterinary Medicine and Science, The University of Nottingham, Nottingham, NG7 2UH, UK
| | - David Onion
- School of Life Sciences, The University of Nottingham, Life Sciences Building, Nottingham, NG7 2RD, UK
| | - Lucy C Fairclough
- School of Life Sciences, The University of Nottingham, Life Sciences Building, Nottingham, NG7 2RD, UK.
| |
Collapse
|
28
|
Mahmoudi A, Moadab F, Safdarian E, Navashenaq JG, Rezaee M, Gheibihayat SM. MicroRNAs and Efferocytosis: Implications for Diagnosis and Therapy. Mini Rev Med Chem 2022; 22:2641-2660. [PMID: 35362375 DOI: 10.2174/1389557522666220330150937] [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: 09/24/2021] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
About 10-100 billion cells are generated in the human body in a day, and accordingly, 10-100 billion cells predominantly die for maintaining homeostasis. Dead cells generated by apoptosis are also rapidly engulfed by macrophages (Mθs) to be degraded. In case of the inefficient engulfment of apoptotic cells (ACs) via Mθs, they experience secondary necrosis and thus release intracellular materials, which display damage-associated molecular patterns (DAMPs) and result in diseases. Over the last decades, researchers have also reflected on the significant contribution of microRNAs (miRNAs) to autoimmune diseases through the regulation of Mθs functions. Moreover, miRNAs have shown intricate involvement with completely adjusting basic Mθs functions, such as phagocytosis, inflammation, efferocytosis, tumor promotion, and tissue repair. In this review, the mechanism of efferocytosis containing "Find-Me", "Eat-Me", and "Digest-Me" signals is summarized and the biogenesis of miRNAs is briefly described. Finally, the role of miRNAs in efferocytosis is discussed. It is concluded that miRNAs represent promising treatments and diagnostic targets in impaired phagocytic clearance, which leads to different diseases.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of medical biotechnology and nanotechnology, faculty of medicine, Mashhad University of Medical science, Iran
| | - Fatemeh Moadab
- Medical student, Student Research Committee, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Esmat Safdarian
- Legal Medicine Research Center, Legal Medicine Organization, Tehran Iran
| | | | - Mehdi Rezaee
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran;
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
29
|
Pantazopoulos I, Magounaki K, Kotsiou O, Rouka E, Perlikos F, Kakavas S, Gourgoulianis K. Incorporating Biomarkers in COPD Management: The Research Keeps Going. J Pers Med 2022; 12:jpm12030379. [PMID: 35330379 PMCID: PMC8955907 DOI: 10.3390/jpm12030379] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/17/2022] [Accepted: 02/28/2022] [Indexed: 12/17/2022] Open
Abstract
Globally, chronic obstructive pulmonary disease (COPD) remains a major cause of morbidity and mortality, having a significant socioeconomic effect. Several molecular mechanisms have been related to COPD including chronic inflammation, telomere shortening, and epigenetic modifications. Nowadays, there is an increasing need for novel therapeutic approaches for the management of COPD. These treatment strategies should be based on finding the source of acute exacerbation of COPD episodes and estimating the patient’s own risk. The use of biomarkers and the measurement of their levels in conjunction with COPD exacerbation risk and disease prognosis is considered an encouraging approach. Many types of COPD biomarkers have been identified which include blood protein biomarkers, cellular biomarkers, and protease enzymes. They have been isolated from different sources including peripheral blood, sputum, bronchoalveolar fluid, exhaled air, and genetic material. However, there is still not an exclusive biomarker that is used for the evaluation of COPD but rather a combination of them, and this is attributed to disease complexity. In this review, we summarize the clinical significance of COPD-related biomarkers, their association with disease outcomes, and COPD patients’ management. Finally, we depict the various samples that are used for identifying and measuring these biomarkers.
Collapse
Affiliation(s)
- Ioannis Pantazopoulos
- Department of Emergency Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece
- Correspondence: ; Tel.: +30-6945661525
| | | | - Ourania Kotsiou
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece; (O.K.); (E.R.); (K.G.)
| | - Erasmia Rouka
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece; (O.K.); (E.R.); (K.G.)
| | - Fotis Perlikos
- ICU Department, Henry Dynant Hospital Center, 11526 Athens, Greece;
| | - Sotirios Kakavas
- Critical Care Department, “Sotiria” General Hospital of Chest Diseases, 11527 Athens, Greece;
| | - Konstantinos Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, University of Thessaly, Biopolis, 41500 Larissa, Greece; (O.K.); (E.R.); (K.G.)
| |
Collapse
|
30
|
Serafini FL, Lanuti P, Delli Pizzi A, Procaccini L, Villani M, Taraschi AL, Pascucci L, Mincuzzi E, Izzi J, Chiacchiaretta P, Buca D, Catitti G, Bologna G, Simeone P, Pieragostino D, Caulo M. Diagnostic Impact of Radiological Findings and Extracellular Vesicles: Are We Close to Radiovesicolomics? BIOLOGY 2021; 10:biology10121265. [PMID: 34943180 PMCID: PMC8698452 DOI: 10.3390/biology10121265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 11/23/2022]
Abstract
Simple Summary Over the years, diagnostic tests such as in radiology and flow cytometry have become more and more powerful in the constant struggle against different pathologies, some of which are life-threatening. The possibility of using these “weapons” in a conjugated manner could result in higher healing and prevention rates, and a decrease in late diagnosis diseases. Different correlations among pathologies, extracellular vesicles (EVs), and radiological findings were recently demonstrated by many authors. Together with the increasing importance of “omics” sciences, and artificial intelligence in this new century, the perspective of a new research field called “radiovesicolomics” could be the missing link, enabling a different approach to disease diagnosis and treatment. Abstract Currently, several pathologies have corresponding and specific diagnostic and therapeutic branches of interest focused on early and correct detection, as well as the best therapeutic approach. Radiology never ceases to develop newer technologies in order to give patients a clear, safe, early, and precise diagnosis; furthermore, in the last few years diagnostic imaging panoramas have been extended to the field of artificial intelligence (AI) and machine learning. On the other hand, clinical and laboratory tests, like flow cytometry and the techniques found in the “omics” sciences, aim to detect microscopic elements, like extracellular vesicles, with the highest specificity and sensibility for disease detection. If these scientific branches started to cooperate, playing a conjugated role in pathology diagnosis, what could be the results? Our review seeks to give a quick overview of recent state of the art research which investigates correlations between extracellular vesicles and the known radiological features useful for diagnosis.
Collapse
Affiliation(s)
- Francesco Lorenzo Serafini
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Andrea Delli Pizzi
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
- Correspondence:
| | - Luca Procaccini
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Michela Villani
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Alessio Lino Taraschi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Luca Pascucci
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Erica Mincuzzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Jacopo Izzi
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
| | - Piero Chiacchiaretta
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Davide Buca
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Giulia Catitti
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (P.L.); (D.B.); (G.C.); (G.B.); (P.S.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio”, 66100 Chieti, Italy;
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio”, 66100 Chieti, Italy
| | - Massimo Caulo
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d’Annunzio”, 66100 Chieti, Italy; (F.L.S.); (L.P.); (M.V.); (A.L.T.); (L.P.); (E.M.); (J.I.); (P.C.); (M.C.)
- Institute of Advanced Biomedical Technologies (ITAB), University “G. d’Annunzio”, 66100 Chieti, Italy
| |
Collapse
|
31
|
Kumar S, Kumar P, Kodidela S, Duhart B, Cernasev A, Nookala A, Kumar A, Singh UP, Bissler J. Racial Health Disparity and COVID-19. J Neuroimmune Pharmacol 2021; 16:729-742. [PMID: 34499313 PMCID: PMC8426163 DOI: 10.1007/s11481-021-10014-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/19/2021] [Indexed: 02/07/2023]
Abstract
The infection by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and resultant coronavirus diseases-19 (COVID-19) disproportionally affects minorities, especially African Americans (AA) compared to the Caucasian population. The AA population is disproportionally affected by COVID-19, in part, because they have high prevalence of underlying conditions such as obesity, diabetes, and hypertension, which are known to exacerbate not only kidney diseases, but also COVID-19. Further, a decreased adherence to COVID-19 guidelines among tobacco smokers could result in increased infection, inflammation, reduced immune response, and lungs damage, leading to more severe form of COVID-19. As a result of high prevalence of underlying conditions that cause kidney diseases in the AA population coupled with tobacco smoking make the AA population vulnerable to severe form of both COVID-19 and kidney diseases. In this review, we describe how tobacco smoking interact with SARS-CoV-2 and exacerbates SARS-CoV-2-induced kidney diseases including renal failure, especially in the AA population. We also explore the role of extracellular vesicles (EVs) in COVID-19 patients who smoke tobacco. EVs, which play important role in tobacco-mediated pathogenesis in infectious diseases, have also shown to be important in COVID-19 pathogenesis and organ injuries including kidney. Further, we explore the potential role of EVs in biomarker discovery and therapeutics, which may help to develop early diagnosis and treatment of tobacco-induced renal injury in COVID-19 patients, respectively.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA.
| | - Prashant Kumar
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, USA
| | - Sunitha Kodidela
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Benjamin Duhart
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Alina Cernasev
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Sciences Center, Nashville, TN, USA
| | | | - Asit Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - John Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN, USA.
| |
Collapse
|
32
|
Takahashi T, Schleimer RP. Epithelial-Cell-Derived Extracellular Vesicles in Pathophysiology of Epithelial Injury and Repair in Chronic Rhinosinusitis: Connecting Immunology in Research Lab to Biomarkers in Clinics. Int J Mol Sci 2021; 22:11709. [PMID: 34769139 PMCID: PMC8583779 DOI: 10.3390/ijms222111709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Epithelial barrier disruption and failure of epithelial repair by aberrant epithelial-mesenchymal transition (EMT)-induced basal cells observed in nasal mucosa of chronic rhinosinusitis (CRS) are speculated to play important roles in disease pathophysiology. Microparticles (MPs) are a type of extracellular vesicle (EV) released by budding or shedding from the plasma membrane of activated or apoptotic cells. MPs are detected in nasal lavage fluids (NLFs) and are now receiving attention as potential biomarkers to evaluate the degree of activation of immune cells and injury of structural cells in nasal mucosa of subjects with sinus disease. There are three types of epithelial-cell-derived MPs, which are defined by the expression of different epithelial specific markers on their surface: EpCAM, E-cadherin, and integrin β6 (ITGB6). When these markers are on MPs that are also carrying canonical EMT/mesenchymal markers (Snail (SNAI1); Slug (SNAI2); alpha-smooth muscle actin (αSMA, ACTA2)) or pro- and anti-coagulant molecules (tissue factor (TF); tissue plasminogen activator (tPA); plasminogen activator inhibitor-1 (PAI-1)), they provide insight as to the roles of epithelial activation for EMT or regulation of coagulation in the underlying disease. In this review, we discuss the potential of epithelial MPs as research tools to evaluate status of nasal mucosae of CRS patients in the lab, as well as biomarkers for management and treatment of CRS in the clinic.
Collapse
Affiliation(s)
- Toru Takahashi
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
| | - Robert P Schleimer
- Division of Allergy-Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA;
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
33
|
Kotlyarov S. Diversity of Lipid Function in Atherogenesis: A Focus on Endothelial Mechanobiology. Int J Mol Sci 2021; 22:11545. [PMID: 34768974 PMCID: PMC8584259 DOI: 10.3390/ijms222111545] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/12/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is one of the most important problems in modern medicine. Its high prevalence and social significance determine the need for a better understanding of the mechanisms of the disease's development and progression. Lipid metabolism and its disorders are one of the key links in the pathogenesis of atherosclerosis. Lipids are involved in many processes, including those related to the mechanoreception of endothelial cells. The multifaceted role of lipids in endothelial mechanobiology and mechanisms of atherogenesis are discussed in this review. Endothelium is involved in ensuring adequate vascular hemodynamics, and changes in blood flow characteristics are detected by endothelial cells and affect their structure and function.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
34
|
Tung NT, Ho SC, Lu YH, Chen TT, Lee KY, Chen KY, Wu CD, Chung KF, Kuo HP, Thao HNX, Dung HB, Thuy TPC, Wu SM, Kou HY, Lee YL, Chuang HC. Association Between Air Pollution and Lung Lobar Emphysema in COPD. Front Med (Lausanne) 2021; 8:705792. [PMID: 34621758 PMCID: PMC8490678 DOI: 10.3389/fmed.2021.705792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
The development of emphysema has been linked to air pollution; however, the association of air pollution with the extent of lobar emphysema remains unclear. This study examined the association of particulate matter <2.5 μm in aerodynamic diameters (PM2.5) (≤2.5 μm), nitrogen dioxide (NO2), and ozone (O3) level of exposure with the presence of emphysema in 86 patients with chronic obstructive pulmonary disease (COPD). Exposure to the air pollution estimated using the land-use regression model was associated with lung function, BODE (a body mass index, degree of obstruction, dyspnea severity, and exercise capacity index) quartiles, and emphysema measured as low-attenuation areas on high-resolution CT (HR-CT) lung scans. Using paraseptal emphysema as the reference group, we observed that a 1 ppb increase in O3 was associated with a 1.798-fold increased crude odds ratio of panlobular emphysema (p < 0.05). We observed that PM2.5 was associated with BODE quartiles, modified Medical Research Council (mMRC) dyspnea score, and exercise capacity (all p < 0.05). We found that PM2.5, NO2, and O3 were associated with an increased degree of upper lobe emphysema and lower lobe emphysema (all p < 0.05). Furthermore, we observed that an increase in PM2.5, NO2, and O3 was associated with greater increases in upper lobe emphysema than in lower lobe emphysema. In conclusion, exposure to O3 can be associated with a higher risk of panlobular emphysema than paraseptal emphysema in patients with COPD. Emphysema severity in lung lobes, especially the upper lobes, may be linked to air pollution exposure in COPD.
Collapse
Affiliation(s)
- Nguyen Thanh Tung
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Otorhinolaryngology Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Hsun Lu
- Department of Radiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chih-Da Wu
- Department of Geomatics, National Cheng Kung University, Tainan City, Taiwan.,National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Han-Pin Kuo
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Huynh Nguyen Xuan Thao
- Otorhinolaryngology Department, Ho Chi Minh City University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Hoang Ba Dung
- Otorhinolaryngology Department, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Tran Phan Chung Thuy
- Otorhinolaryngology Department, Faculty of Medicine, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Yun Kou
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
35
|
Abstract
This commentary reviews the contribution of imaging by CT and MRI to functional assessment in chronic obstructive pulmonary disease (COPD). CT can help individualize the assessment of COPD by quantifying emphysema, air trapping and airway wall thickening, potentially leading to more specific treatments for these distinct components of COPD. Longitudinal changes in these metrics can help assess progression or improvement. On hyperpolarized gas MRI, the apparent diffusion coefficient of provides an index of airspace enlargement reflecting emphysema. Perfusion imaging and measurement of pulmonary vascular volume on non-contrast CT provide insight into the contribution of pulmonary vascular disease to pulmonary impairment. Functional imaging is particularly valuable in detecting early lung dysfunction in subjects with inhalational exposures.
Collapse
Affiliation(s)
- David A Lynch
- Department of Radiology, National Jewish Health, Denver, CO, United States
| |
Collapse
|
36
|
Tung NT, Ho SC, Lu YH, Chen TT, Lee KY, Chen KY, Wu CD, Chung KF, Kuo HP, Thao HNX, Dung HB, Thuy TPC, Wu SM, Kou HY, Lee YL, Chuang HC. Higher alveolar deposition of particulate matter in emphysematous lobes of COPD. ERJ Open Res 2021; 7:00324-2021. [PMID: 34476255 PMCID: PMC8405879 DOI: 10.1183/23120541.00324-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 06/27/2021] [Indexed: 11/24/2022] Open
Abstract
Emphysema can be examined quantitatively on high-resolution computed tomography (HRCT) by measuring the low-attenuation areas of the lung and has been associated with decrease in lung function in patients with COPD [1]. Previous studies have associated levels of air pollution with emphysema severity of the total lung [2, 3]. However, the relationship between inhaled particulate matter (PM) deposition in the lungs and the degree of emphysema at the lung lobar level remains poorly understood. We examined the association of lung lobe-deposited doses of PM2.5 (particles with a 50% cut-off aerodynamic diameter of 2.5 μm) with the extent of emphysema in different lung lobes of COPD subjects. The novelty of this study is that it identified the associations between PM2.5 deposition in the lung and the degree of emphysema in different lung lobes of COPD patients, especially in the right middle lobe and both upper lobeshttps://bit.ly/3k21ri0
Collapse
Affiliation(s)
- Nguyen Thanh Tung
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Otorhinolaryngology Dept, Cho Ray Hospital, Ho Chi Minh City, Vietnam.,These authors contributed equally
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.,These authors contributed equally
| | - Yueh-Hsun Lu
- Dept of Radiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Dept of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Dept of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Dept of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Dept of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Dept of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chih-Da Wu
- Dept of Geomatics, National Cheng Kung University, Tainan, Taiwan.,National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Han-Pin Kuo
- Division of Pulmonary Medicine, Dept of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Hoang Ba Dung
- Otorhinolaryngology Dept, Cho Ray Hospital, Ho Chi Minh City, Vietnam
| | - Tran Phan Chung Thuy
- Otorhinolaryngology Dept, Faculty of Medicine, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Dept of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Division of Pulmonary Medicine, Dept of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Yun Kou
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Dept of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Dept of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
37
|
Lascano J, Katz J, Cearras M, Campos M. Association of Systemic Endothelial-Derived and Platelet-Derived Microparticles With Clinical Outcomes in Chronic Obstructive Pulmonary Disease. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2021; 8:382-395. [PMID: 34062638 PMCID: PMC8428587 DOI: 10.15326/jcopdf.2021.0211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/26/2021] [Indexed: 11/21/2022]
Abstract
PURPOSE Endothelial and platelet microparticles (eMPs and pMPs), markers of cellular activation, dysfunction, or apoptosis, have been associated with multiple cardiovascular conditions. Chronic obstructive pulmonary disease (COPD) is associated with cardiovascular comorbidities and platelet/endothelial dysfunction. We analyzed whether eMPs and pMPs are associated with COPD status and/or severity. PATIENTS AND METHODS A total of 58 COPD patients and 19 controls were enrolled and followed for an average of 1.17 years. Characterization of COPD included lung function, Body mass index-airflow Obstruction-Dyspnea-Exercise (BODE) scores, health-related quality of life, exacerbations, comorbidities, and mortality. Plasma collection to measure eMPs and pMPs via flow cytometry was performed at enrollment as well as during acute exacerbation in 17 participants. We measured pMPs (CD31+, CD41+31+, CD 62P+), eMPs (ULEX lectin+, CD51+, CD54+, CD62E+), the apoptotic CD62E+/CD31+ ratio, and Annexin V MP. RESULTS As a group, COPD participants had no difference in all MP levels studied compared with controls. No significant correlations with diffusion capacity for carbon monoxide, quality of life, and exacerbation status were found in all MPs studied. However, the eMP ULEX and the pMP CD 62P+ were higher among COPD Global initiative for chronic Obstructive Lung Disease (GOLD) stage 3 patients compared to controls. The CD62E+/CD31+ ratio was lower in controls and GOLD stage 1 COPD participants compared with GOLD stage 2/3 COPD participants, suggesting increased apoptosis. eMP ULEX lectin+ decreased during acute exacerbations and pMP41+31+ significantly increased as BODE score increased. CONCLUSIONS After adjusting for comorbidities, most eMPs and pMPs studied do not correlate significantly with COPD status or severity.
Collapse
Affiliation(s)
- Jorge Lascano
- Pulmonary and Critical Care Division, Department of Internal Medicine, UF Health-Shands Hospital, University of Florida, Gainesville, Florida, United States
| | - Jason Katz
- Pulmonary and Critical Care Division, Department of Internal Medicine, UF Health-Shands Hospital, University of Florida, Gainesville, Florida, United States
| | - Martin Cearras
- Advent Health Medical Group, Central Florida Division, Orlando, Florida, United States
| | - Michael Campos
- Pulmonary and Critical Care Division, Department of Internal Medicine, University of Miami Health Systems, Miami, Florida, United States
| |
Collapse
|
38
|
Yamada M. Extracellular vesicles: Their emerging roles in the pathogenesis of respiratory diseases. Respir Investig 2021; 59:302-311. [PMID: 33753011 DOI: 10.1016/j.resinv.2021.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 01/29/2021] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
Alveoli are the basic structure of the lungs, consisting of various types of parenchymal and bone marrow-derived cells including alveolar macrophages. These various types of cells have several important functions; thus, communication between these cells plays an important role in homeostasis as well as in the pathophysiology of diseases in the lungs. For a better understanding of the pathophysiology of lung diseases, researchers have isolated each type of lung cell to investigate the changes in their gene expressions, including their humoral factor or adhesion molecules, to reveal the intercellular communication among these cells. In particular, investigations during the past decade have focused on extracellular vesicles, which are lipid bilayer delimited vesicles released from a cell that can move among various cells and transfer substances, including microRNAs, mRNAs and proteins, thus, functioning as intercellular messengers. Extracellular vesicles can be classified into three general groups: apoptotic bodies, exosomes, and microparticles. Extracellular vesicles, especially exosomes and microparticles, are attracting increasing attention from pulmonologists as tools for understanding pathogenesis and disease diagnosis. Here, we review studies, including our own, on exosomes and microparticles and their roles in both lung homeostasis and the pathogenesis of lung diseases such as idiopathic pulmonary fibrosis, chronic obstructive lung diseases, and acute respiratory distress syndrome. This review also addresses the roles of extracellular vesicles in COVID-19, the current global public health crisis.
Collapse
Affiliation(s)
- Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 9808574, Japan.
| |
Collapse
|
39
|
CT Pulmonary Vessels and MRI Ventilation in Chronic Obstructive Pulmonary Disease: Relationship with worsening FEV 1 in the TINCan cohort study. Acad Radiol 2021; 28:495-506. [PMID: 32303446 DOI: 10.1016/j.acra.2020.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE AND OBJECTIVES The relationships between computed tomography (CT) pulmonary vascularity and MRI ventilation is not well-understood in chronic obstructive pulmonary disease (COPD) patients. Our objective was to evaluate CT pulmonary vascular and MRI ventilation measurements in ex-smokers and to investigate their associations and how such measurements change over time. MATERIALS AND METHODS Ninety ex-smokers (n = 41 without COPD 71 ± 10 years and n = 49 COPD 71 ± 8 years) provided written informed-consent to an ethics-board approved protocol and underwent imaging and pulmonary-function-tests twice, 31 ± 7 months apart. 3He MRI was acquired to generate ventilation-defect-percent (VDP). CT measurements of the relative area-of-the-lung with attenuation <-950 Hounsfield units (RA950), pulmonary vascular total-blood-volume (TBV) and percent of vessels with radius < one voxel (PV1) were evaluated. RESULTS At baseline, there were significant differences in RA950 (p = 0.0001), VDP (p = 0.0001), total-blood-volume (p = 0.0001) and PV1 (p = 0.01) between ex-smokers and COPD participants as well as for VDP (p = 0.0001) in COPD participants with and without emphysema. The annual FEV1 change (-40 ± 93 mL/year) was not different among participant subgroups (p = 0.87), but the annual RA950 (p = 0.01) and PV1 (p = 0.007) changes were significantly different in participants with an accelerated annual FEV1 decline as compared to participants with a diminished annual FEV1 decline. There were significant but weak relationships for PV1 with FEV1%pred (p = 0.02), FEV1/FVC (p = 0.001), and log RA950 (p = 0.0001), but not VDP (p=0.20). The mean change in PV1 was also weakly but significantly related to the change in RA950 (p = 0.02). CONCLUSION CT pulmonary vascular measurements were significantly different in ex-smokers and participants with COPD and related to RA950 but not VDP worsening over 2.5 years.
Collapse
|
40
|
Kim JS, Thomashow MA, Yip NH, Burkart KM, Lo Cascio CM, Shimbo D, Barr RG. Randomization to Omega-3 Fatty Acid Supplementation and Endothelial Function in COPD: The COD-Fish Randomized Controlled Trial. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2021; 8. [PMID: 33150779 DOI: 10.15326/jcopdf.8.1.2020.0132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rationale Studies suggest a pathogenic role of endothelial dysfunction in chronic obstructive lung disease (COPD). Omega-3 (n-3) polyunsaturated fatty acid (PUFA) supplementation improves endothelial function in other diseases but has not been examined in COPD. Objective We hypothesized that n-3 PUFA supplementation would improve systemic endothelial function in COPD. We performed a pilot randomized, placebo-controlled, double-blind, phase 2 superiority trial (NCT00835289). Methods Adults with moderate and severe stable COPD (79% with emphysema on computed tomography [CT]) were randomized to high-dose fish oil capsules or placebo daily for 6 months. The primary endpoint was percentage change in brachial artery flow-mediated dilation (FMD) from baseline to 6 months. Secondary endpoints included peripheral arterial tonometry, endothelial microparticles (EMPs), 6-minute walk distance, respiratory symptoms, and pulmonary function. Results Thirty-three of 40 randomized participants completed all measurements. Change in FMD after 6 months did not differ between the fish oil and placebo arms (-1.1%, 95% CI -5.0-2.9, p=0.59). CD31+ EMPs increased in the fish oil arm (0.9%, 95% CI 0.1-1.7, p=0.04). More participants in the fish oil arm reported at least a 4-point improvement in the St George's Respiratory Questionnaire (SGRQ) compared to placebo (8 versus 1; p=0.01). There were no significant changes in other secondary endpoints. There were 4 serious adverse events determined to be unrelated to the study (3 in the fish oil arm and 1 in the placebo arm). Conclusion Randomization to n-3 PUFAs for 6 months did not change systemic endothelial function in COPD. Changes in EMPs and SGRQ suggest n-3 PUFAs might have biologic and clinical effects that warrant further investigation.
Collapse
Affiliation(s)
- John S Kim
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States.,Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Michael A Thomashow
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States.,Kaiser Permanante San Francisco Medical Center, San Francisco, California, United States
| | - Natalie H Yip
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Kristin M Burkart
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Christian M Lo Cascio
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - Daichi Shimbo
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States
| | - R Graham Barr
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States.,Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, New York, New York, United States
| |
Collapse
|
41
|
Gao Y, Raj JU. Extracellular Vesicles as Unique Signaling Messengers: Role in Lung Diseases. Compr Physiol 2020; 11:1351-1369. [PMID: 33294981 DOI: 10.1002/cphy.c200006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed extracellular particles carrying rich cargo such as proteins, lipids, and microRNAs with distinct characteristics of their parental cells. EVs are emerging as an important form of cellular communication with the ability to selectively deliver a kit of directional instructions to nearby or distant cells to modulate their functions and phenotypes. According to their biogenesis, EVs can be divided into two groups: those of endocytic origin are called exosomes and those derived from outward budding of the plasma membrane are called microvesicles (also known as ectosomes or microparticles). Under physiological conditions, EVs are actively involved in maintenance of pulmonary hemostasis. However, EVs can contribute to the pathogenesis of diseases such as chronic obstructive pulmonary disease, asthma, acute lung injury/acute respiratory distress syndrome, interstitial lung disease, and pulmonary arterial hypertension. EVs, especially those derived from mesenchymal/stromal stem cells, can also be beneficial and can curb the development of lung diseases. Novel technologies are continuously being developed to minimize the undesirable effects of EVs and also to engineer EVs so that they may have beneficial effects and can be used as therapeutic agents in lung diseases. © 2021 American Physiological Society. Compr Physiol 11:1351-1369, 2021.
Collapse
Affiliation(s)
- Yuansheng Gao
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - J Usha Raj
- Department of Pediatrics, College of Medicine at Chicago, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
42
|
Bi H, He J, He X, Du J, Chen M, Huang Z, Yang C, Yang L, Li H, Zhou K, Wang Q, He L, Jin Z. Bone marrow stem cells therapy alleviates vascular injury in a chronic obstructive pulmonary disease‑obstructive sleep apnea overlap syndrome rat model. Mol Med Rep 2020; 23:69. [PMID: 33236768 PMCID: PMC7716420 DOI: 10.3892/mmr.2020.11707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/27/2020] [Indexed: 11/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and obstructive sleep apnea (OSA) are highly prevalent potential risk factors for systemic disease. Previous studies have reported that COPD and OSA are major independent risk factors for cardio- or cerebrovascular diseases. The present study aimed to investigate the role of bone marrow mesenchymal stem cells (BMSCs) on vascular injury in a COPD-OSA overlap syndrome (OS) rat model. Rats were randomly divided into three groups: Sham, OS model and BMSC. BMSC localization in major organs was detected via confocal laser fluorescence microscopy, and the aortic tissue pathological changes and related genes were measured using hematoxylin & eosin and Masson staining. Genes associated with vascular endothelial cell injury, including endothelin 1, vascular cell adhesion molecule 1 and endothelial nitric oxide synthase, were detected via reverse transcription-quantitative PCR and western blotting. Apoptosis of vascular endothelial cells was detected using TUNEL and immunofluorescence assays. The endothelial cell marker CD31 in injured vessels was analyzed via immunohistochemistry. BMSCs migrated into the heart, liver, spleen, lung, kidney, brain and aorta in the OS model. The green fluorescence expression of BMSCs demonstrated the highest level in the lung, followed by the aorta. Aortic tissue had a more severe vascular injury and increased apoptosis in the model group compared with the BMSC group. Vascular endothelial cell apoptosis was decreased in the BMSC group compared with the model group. The findings suggested that BMSCs could repair vascular injury by inhibiting endothelial cell damage and apoptosis. These data provide a theoretical basis for the treatment of cardiovascular diseases caused by OS with BMSCs.
Collapse
Affiliation(s)
- Hong Bi
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Jian He
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Xu He
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Junyi Du
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Min Chen
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Zhaoming Huang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Chao Yang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Lijuan Yang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Hang Li
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Kaihua Zhou
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Qing Wang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Lewei He
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Zhixian Jin
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| |
Collapse
|
43
|
Soni S, Garner JL, O'Dea KP, Koh M, Finney L, Tirlapur N, Srikanthan K, Tenda ED, Aboelhassan AM, Singh S, Wilson MR, Wedzicha JA, Kemp SV, Usmani OS, Shah PL, Takata M. Intra-alveolar neutrophil-derived microvesicles are associated with disease severity in COPD. Am J Physiol Lung Cell Mol Physiol 2020; 320:L73-L83. [PMID: 33146567 DOI: 10.1152/ajplung.00099.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite advances in the pathophysiology of chronic obstructive pulmonary disease (COPD), there is a distinct lack of biochemical markers to aid clinical management. Microvesicles (MVs) have been implicated in the pathophysiology of inflammatory diseases including COPD, but their association to COPD disease severity remains unknown. We analyzed different MV populations in plasma and bronchoalveolar lavage fluid (BALF) taken from 62 patients with mild to very severe COPD (51% male; mean age: 65.9 yr). These patients underwent comprehensive clinical evaluation (symptom scores, lung function, and exercise testing), and the capacity of MVs to be clinical markers of disease severity was assessed. We successfully identified various MV subtype populations within BALF [leukocyte, polymorphonuclear leukocyte (PMN; i.e., neutrophil), monocyte, epithelial, and platelet MVs] and plasma (leukocyte, PMN, monocyte, and endothelial MVs) and compared each MV population to disease severity. BALF neutrophil MVs were the only population to significantly correlate with the clinical evaluation scores including forced expiratory volume in 1 s, modified Medical Research Council dyspnea score, 6-min walk test, hyperinflation, and gas transfer. BALF neutrophil MVs, but not neutrophil cell numbers, also strongly correlated with BODE index. We have undertaken, for the first time, a comprehensive evaluation of MV profiles within BALF/plasma of COPD patients. We demonstrate that BALF levels of neutrophil-derived MVs are unique in correlating with a number of key functional and clinically relevant disease severity indexes. Our results show the potential of BALF neutrophil MVs for a COPD biomarker that tightly links a key pathophysiological mechanism of COPD (intra-alveolar neutrophil activation) with clinical severity/outcome.
Collapse
Affiliation(s)
- Sanooj Soni
- Division of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Justin L Garner
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,Chelsea and Westminster Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Kieran P O'Dea
- Division of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Marissa Koh
- Division of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Lydia Finney
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Nikhil Tirlapur
- Division of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Karthi Srikanthan
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,Chelsea and Westminster Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Eric D Tenda
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,Chelsea and Westminster Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Arafa M Aboelhassan
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,Chelsea and Westminster Hospital, Respiratory Medicine, London, United Kingdom
| | - Suveer Singh
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,Chelsea and Westminster Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Michael R Wilson
- Division of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Jadwiga A Wedzicha
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Samuel V Kemp
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom
| | - Omar S Usmani
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Pallav L Shah
- Royal Brompton Hospital, Respiratory Medicine, London, United Kingdom.,Chelsea and Westminster Hospital, Respiratory Medicine, London, United Kingdom.,National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Masao Takata
- Division of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| |
Collapse
|
44
|
Hatabu H, Ohno Y, Gefter WB, Parraga G, Madore B, Lee KS, Altes TA, Lynch DA, Mayo JR, Seo JB, Wild JM, van Beek EJR, Schiebler ML, Kauczor HU. Expanding Applications of Pulmonary MRI in the Clinical Evaluation of Lung Disorders: Fleischner Society Position Paper. Radiology 2020; 297:286-301. [PMID: 32870136 DOI: 10.1148/radiol.2020201138] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Pulmonary MRI provides structural and quantitative functional images of the lungs without ionizing radiation, but it has had limited clinical use due to low signal intensity from the lung parenchyma. The lack of radiation makes pulmonary MRI an ideal modality for pediatric examinations, pregnant women, and patients requiring serial and longitudinal follow-up. Fortunately, recent MRI techniques, including ultrashort echo time and zero echo time, are expanding clinical opportunities for pulmonary MRI. With the use of multicoil parallel acquisitions and acceleration methods, these techniques make pulmonary MRI practical for evaluating lung parenchymal and pulmonary vascular diseases. The purpose of this Fleischner Society position paper is to familiarize radiologists and other interested clinicians with these advances in pulmonary MRI and to stratify the Society recommendations for the clinical use of pulmonary MRI into three categories: (a) suggested for current clinical use, (b) promising but requiring further validation or regulatory approval, and (c) appropriate for research investigations. This position paper also provides recommendations for vendors and infrastructure, identifies methods for hypothesis-driven research, and suggests opportunities for prospective, randomized multicenter trials to investigate and validate lung MRI methods.
Collapse
Affiliation(s)
- Hiroto Hatabu
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Yoshiharu Ohno
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Warren B Gefter
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Grace Parraga
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Bruno Madore
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Kyung Soo Lee
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Talissa A Altes
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - David A Lynch
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - John R Mayo
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Joon Beom Seo
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Jim M Wild
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Edwin J R van Beek
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Mark L Schiebler
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | - Hans-Ulrich Kauczor
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| | -
- From the Center for Pulmonary Functional Imaging, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, 75 Francis St, Boston, MA 02115 (H.H.); Department of Radiology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan (Y.O.); Department of Radiology, Penn Medicine, University of Pennsylvania, Philadelphia, Pa (W.B.G.); Department of Medical Biophysics, Western University, London, Canada (G.P.); Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass (B.M.); Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine (SKKU-SOM), Seoul, Korea (K.S.L.); Department of Radiology, University of Missouri, Columbia, Mo (T.A.A.); Department of Radiology, National Jewish Health, Denver, Colo (D.A.L.); Department of Radiology, Vancouver General Hospital and University of British Colombia, Vancouver, Canada (J.R.M.); Department of Radiology, Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea (J.B.S.); Section of Academic Radiology, University of Sheffield, Sheffield, England, United Kingdom (J.M.W.); Edinburgh Imaging, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom (E.J.R.v.B.); Department of Radiology, UW Madison School of Medicine and Public Health, Madison, Wis (M.L.S.); and Diagnostic and Interventional Radiology, University Hospital Heidelberg, Translational Lung Research Center Heidelberg, member of the German Center of Lung Research, Heidelberg, Germany (H.U.K.)
| |
Collapse
|
45
|
Fasting Therapy Contributes to the Improvement of Endothelial Function and Decline in Vascular Injury-Related Markers in Overweight and Obese Individuals via Activating Autophagy of Endothelial Progenitor Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3576030. [PMID: 32802124 PMCID: PMC7403908 DOI: 10.1155/2020/3576030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/19/2020] [Indexed: 11/17/2022]
Abstract
Background High body mass index- (BMI-) related vascular injury contributes to the pathogenesis of the atherosclerotic cardiovascular disease (ASCVD). Rigorous calorie restriction is one of the major lifestyle interventions to reduce vascular risk in overweight or obese individuals. However, the effects of fasting therapy (FT) on vascular function and the mechanism are still unclear. This study was aimed to investigate the impacts of FT on endothelial function, arterial stiffness, and circulating arterial damage parameters in overweight and obese individuals and possible mechanism. Methods Overweight and obese individuals participated in FT intervention (7-day very low calorie diet). Arterial function including brachial arterial flow-mediated dilation (FMD), brachial-ankle pulse wave velocity (baPWV), vascular injury-related markers including trimethylamine N-oxide (TMAO), and leptin and endothelial microparticles (EMPs) were assessed. Endothelial progenitor cells (EPCs) of these participants were isolated and cultured to investigate EPCs function. mRFP-GFP-LC3 confocal microscopy scanning and western blot were carried out to determine autophagy. Results After FT, body weight and BMI significantly decreased (81.76 ± 12.04 vs. 77.51 ± 12.06 kg, P < 0.01; 29.93 ± 2.82 vs. 28.47 ± 2.83 kg/m2, P < 0.01). FT remarkably improved FMD (5.26 ± 1.34 vs. 6.25 ± 1.60%, P=0.01) while baPWV kept unchanged. TMAO and leptin levels decreased (3.96 ± 1.85 vs. 2.73 ± 1.33 μmol/L, P=0.044; 6814 ± 2639 vs. 3563 ± 2668 μmol/L, P < 0.01). EMPs showed a decreased tendency. EPCs function was significantly improved, autophagy fluorescence intensity was enhanced, and the level of Beclin1, Atg5, LC3 II/I also increased after starvation in vitro, and the effects were blocked by autophagy inhibitor. Conclusions Our present study demonstrated for the first time that FT markedly improves endothelial function and reduces the levels of arterial injury markers through improving EPCs function via activating autophagy. These findings provide a novel insight into FT as a lifestyle intervention strategy to promote the maintenance of vascular homeostasis in overweight or obese individuals. The trial was registered with ChiCTR1900024290.
Collapse
|
46
|
Mohan A, Agarwal S, Clauss M, Britt NS, Dhillon NK. Extracellular vesicles: novel communicators in lung diseases. Respir Res 2020; 21:175. [PMID: 32641036 PMCID: PMC7341477 DOI: 10.1186/s12931-020-01423-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
The lung is the organ with the highest vascular density in the human body. It is therefore perceivable that the endothelium of the lung contributes significantly to the circulation of extracellular vesicles (EVs), which include exosomes, microvesicles, and apoptotic bodies. In addition to the endothelium, EVs may arise from alveolar macrophages, fibroblasts and epithelial cells. Because EVs harbor cargo molecules, such as miRNA, mRNA, and proteins, these intercellular communicators provide important insight into the health and disease condition of donor cells and may serve as useful biomarkers of lung disease processes. This comprehensive review focuses on what is currently known about the role of EVs as markers and mediators of lung pathologies including COPD, pulmonary hypertension, asthma, lung cancer and ALI/ARDS. We also explore the role EVs can potentially serve as therapeutics for these lung diseases when released from healthy progenitor cells, such as mesenchymal stem cells.
Collapse
Affiliation(s)
- Aradhana Mohan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Stuti Agarwal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Matthias Clauss
- Division of Pulmonary, Critical Care, Sleep & Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nicholas S Britt
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA.,Division of Infectious Diseases, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Navneet K Dhillon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Kansas Medical Center, Mail Stop 3007, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA. .,Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
47
|
MacNeil JL, Capaldi DPI, Westcott AR, Eddy RL, Barker AL, McCormack DG, Kirby M, Parraga G. Pulmonary Imaging Phenotypes of Chronic Obstructive Pulmonary Disease Using Multiparametric Response Maps. Radiology 2020; 295:227-236. [PMID: 32096708 DOI: 10.1148/radiol.2020191735] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Pulmonary imaging of chronic obstructive pulmonary disease (COPD) has focused on CT or MRI measurements, but these have not been evaluated in combination. Purpose To generate multiparametric response map (mPRM) measurements in ex-smokers with or without COPD by using volume-matched CT and hyperpolarized helium 3 (3He) MRI. Materials and Methods In this prospective study (https://clinicaltrials.gov, NCT02279329), participants underwent MRI and CT and completed pulmonary function tests, questionnaires, and the 6-minute walk test between December 2010 and January 2019. Disease status was determined by using Global initiative for chronic Obstructive Lung Disease (GOLD) criteria. The mPRM voxel values were generated by using co-registered MRI and CT labels. Kruskal-Wallis and Bonferroni tests were used to determine differences across disease severity, and correlations were determined by using Spearman coefficients. Results A total of 175 ex-smokers (mean age, 69 years ± 9 [standard deviation], 108 men) with or without COPD were evaluated. Ex-smokers without COPD had a larger fraction of normal mPRM voxels (60% vs 37%, 20%, and 7% for GOLD I, II, and III/IV disease, respectively; all P ≤ .001) and a smaller fraction of abnormal voxels, including small airways disease (normal CT, not ventilated: 5% vs 6% [not significant], 11%, and 19% [P ≤ .001 for both] for GOLD I, II, and III/IV disease, respectively) and mild emphysema (normal CT, abnormal apparent diffusion coefficient [ADC]: 33% vs 54%, 56%, and 54% for GOLD I, II, and III/IV disease respectively; all P ≤ .001). Normal mPRM measurements were positively correlated with forced expiratory volume in 1 second (FEV1) (r = 0.65, P < .001), the FEV1-to-forced vital capacity ratio (r = 0.81, P < .001), and diffusing capacity (r = 0.75, P < .001) and were negatively correlated with worse quality of life (r = -0.48, P < .001). Abnormal mPRM measurements of small airways disease (normal CT, not ventilated) and mild emphysema (normal CT, abnormal ADC) were negatively correlated with FEV1 (r = -0.65 and -0.42, respectively; P < .001) and diffusing capacity (r = -0.53 and -0.60, respectively; P < .001) and were positively correlated with worse quality of life (r = 0.45 and r = 0.33, respectively; P < .001), both of which were present in ex-smokers without COPD. Conclusion Multiparametric response maps revealed two abnormal structure-function results related to emphysema and small airways disease, both of which were unexpectedly present in ex-smokers with normal spirometry and CT findings. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Jonathan L MacNeil
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - Dante P I Capaldi
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - Andrew R Westcott
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - Rachel L Eddy
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - Andrea L Barker
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - David G McCormack
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - Miranda Kirby
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| | - Grace Parraga
- From the Robarts Research Institute (J.L.M., A.R.W., R.L.E., A.L.B., G.P.), School of Biomedical Engineering (J.L.M., G.P.), Department of Medical Biophysics (A.R.W., R.L.E., A.L.B., G.P.), and Division of Respirology, Department of Medicine (D.G.M., G.P.), Western University, 1151 Richmond St N, London, ON, Canada N6A 5B7; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, Calif (D.P.I.C.); and Department of Physics, Ryerson University, Toronto, Ontario, Canada (M.K.)
| |
Collapse
|
48
|
Green CE. Lung function and endothelial dysfunction: Is there a relationship without the presence of lung disease? Respirology 2020; 25:49-50. [DOI: 10.1111/resp.13573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 11/29/2022]
Affiliation(s)
- Clara E. Green
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of Birmingham Birmingham UK
| |
Collapse
|
49
|
PULLIERO A, PERGOLI L, LA MAESTRA S, MICALE R, CAMOIRANO A, BOLLATI V, IZZOTTI A, DE FLORA S. Extracellular vesicles in biological fluids. A biomarker of exposure to cigarette smoke and treatment with chemopreventive drugs. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2019; 60:E327-E336. [PMID: 31967089 PMCID: PMC6953455 DOI: 10.15167/2421-4248/jpmh2019.60.4.1284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) are released from cells and enter into body fluids thereby providing a toxicological mechanism of cell-cell communication. The present study aimed at assessing (a) the presence of EVs in mouse body fluids under physiological conditions, (b) the effect of exposure of mice to cigarette smoke for 8 weeks, and (c) modulation of smoke-related alterations by the nonsteroidal anti-inflammatory drug celecoxib, a selective cyclooxygenase-2 inhibitor. To this purpose, ICR (CD-1) mice were either unexposed or exposed to cigarette smoke, either treated or untreated with oral celecoxib. EVs, isolated from bronchoalveolar lavage fluid (BALF), blood serum, and urines, were analyzed by nanoparticle tracking analysis and flow cytometry. EVs baseline concentrations in BALF were remarkably high. Larger EVs were detected in urines. Smoking increased EVs concentrations but only in BALF. Celecoxib remarkably increased EVs concentrations in the blood serum of both male and female smoking mice. The concentration of EVs positive for EpCAM, a mediator of cell-cell adhesion in epithelia playing a role in tumorigenesis, was much higher in urines than in BALF, and celecoxib significantly decreased their concentration. Thus, the effects of smoke on EVs concentrations were well detectable in the extracellular environment of the respiratory tract, where they could behave as delivery carriers to target cells. Celecoxib exerted both protective mechanisms in the urinary tract and adverse systemic effects of likely hepatotoxic origin in smoke-exposed mice. Detection of EVs in body fluids may provide an early diagnostic tool and an end-point exploitable for preventive medicine strategies.
Collapse
Affiliation(s)
- A. PULLIERO
- Department of Health Sciences, University of Genoa, Italy
| | - L. PERGOLI
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Italy
| | - S. LA MAESTRA
- Department of Health Sciences, University of Genoa, Italy
| | - R.T. MICALE
- Department of Health Sciences, University of Genoa, Italy
| | - A. CAMOIRANO
- Department of Health Sciences, University of Genoa, Italy
| | - V. BOLLATI
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Italy
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Unit of Occupational Medicine, Milan, Italy
| | - A. IZZOTTI
- Department of Health Sciences, University of Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - S. DE FLORA
- Department of Health Sciences, University of Genoa, Italy
| |
Collapse
|
50
|
O'Farrell HE, Yang IA. Extracellular vesicles in chronic obstructive pulmonary disease (COPD). J Thorac Dis 2019; 11:S2141-S2154. [PMID: 31737342 DOI: 10.21037/jtd.2019.10.16] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterised by chronic inflammation and significant airflow obstruction that is not fully reversible, and is one of the leading causes of morbidity and mortality worldwide. Extracellular vesicles (EVs) (including apoptotic bodies, microvesicles and exosomes) are small membrane-bound vesicles released by nearly all cell types and can be found in various bodily fluids including blood, sputum and urine. EVs are key mediators in cell-cell communication due to their ability to exchange information to recipient cells, influencing physiological and pathological conditions using their bioactive cargo (DNA, RNA, miRNA, proteins and other metabolites). Therefore the main aim of this review is to highlight recent evidence of the potential use of EVs as diagnostic and therapeutic biomarkers for COPD managements, as well as EVs potential role in COPD pathogenesis. As EVs have been under intense investigation as diagnostic and therapeutic biomarkers for lung disease, in relation to COPD, key studies have identified EVs as potential biomarkers to distinguish exacerbations from stable state, and to characterise COPD phenotypes. EVs are also linked to key inflammatory mediators in COPD progression. In addition, bacteria and their EV cargo influence the lung microenvironment. Further recent therapeutic approaches and advances have seen EVs bioengineered as novel drug delivery vehicles, which could potentially have clinical utility for lung diseases such as COPD.
Collapse
Affiliation(s)
- Hannah E O'Farrell
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Ian A Yang
- UQ Thoracic Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Thoracic Program, The Prince Charles Hospital, Metro North Hospital and Health Service, Brisbane, Australia
| |
Collapse
|