1
|
Song JH, Kim Y. Beyond the Spirometry: New Diagnostic Modalities in Chronic Obstructive Pulmonary Disease. Tuberc Respir Dis (Seoul) 2025; 88:1-13. [PMID: 39308278 DOI: 10.4046/trd.2024.0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/19/2024] [Indexed: 01/07/2025] Open
Abstract
Spirometry can play a critical role as a gold standard in the diagnosis and treatment of patients with chronic obstructive pulmonary disease (COPD). While the criteria for diagnosis have advanced over time, the Global Initiative for Chronic Obstructive Lung Disease (GOLD) standard of the forced expiratory volume in 1 second/forced vital capacity ratio <0.7 remains the most universally employed metric. However, spirometry cannot be utilized in all situations, and test execution can be difficult for some patients, often showing normal values in the early diagnosis of COPD. Therefore, research on new diagnostic methods is underway. Techniques include whole-body plethysmography for measurement of residual volume and inspiratory capacity and airway resistance, diffusing capacity of carbon monoxide or nitric oxide, impulse oscillometry, infrared time-offlight depth image sensor, diaphragm ultrasonography, which can enable early diagnosis and multifaceted assessment of patients with COPD.
Collapse
Affiliation(s)
- Jin Hwa Song
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Youlim Kim
- Division of Pulmonary and Allergy, Department of Internal Medicine, Konkuk University Hospital, Konkuk University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Zhou T, Zhou X, Ni J, Guan Y, Jiang X, Lin X, Li J, Xia Y, Wang X, Wang Y, Huang W, Tu W, Dong P, Li Z, Liu S, Fan L. A CT-Based Lung Radiomics Nomogram for Classifying the Severity of Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2024; 19:2705-2717. [PMID: 39677830 PMCID: PMC11646399 DOI: 10.2147/copd.s483007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a major global health concern, and while traditional pulmonary function tests are effective, recent radiomics advancements offer enhanced evaluation by providing detailed insights into the heterogeneous lung changes. Purpose To develop and validate a radiomics nomogram based on clinical and whole-lung computed tomography (CT) radiomics features to stratify COPD severity. Patients and Methods One thousand ninety-nine patients with COPD (including 308, 132, and 659 in the training, internal and external validation sets, respectively), confirmed by pulmonary function test, were enrolled from two institutions. The whole-lung radiomics features were obtained after a fully automated segmentation. Thereafter, a clinical model, radiomics signature, and radiomics nomogram incorporating radiomics signature as well as independent clinical factors were constructed and validated. Additionally, receiver-operating characteristic (ROC) curve, area under the ROC curve (AUC), decision curve analysis (DCA), and the DeLong test were used for performance assessment and comparison. Results In comparison with clinical model, both radiomics signature and radiomics nomogram outperformed better on COPD severity (GOLD I-II and GOLD III-IV) in three sets. The AUC of radiomics nomogram integrating age, height and Radscore, was 0.865 (95% CI, 0.818-0.913), 0.851 (95% CI, 0.778-0.923), and 0.781 (95% CI, 0.740-0.823) in three sets, which was the highest among three models (0.857; 0.850; 0.774, respectively) but not significantly different (P > 0.05). Decision curve analysis demonstrated the superiority of the radiomics nomogram in terms of clinical usefulness. Conclusion The present work constructed and verified the novel, diagnostic radiomics nomogram for identifying the severity of COPD, showing the added value of chest CT to evaluate not only the pulmonary structure but also the lung function status.
Collapse
Affiliation(s)
- Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- School of Medical Imaging, Shandong Second Medical University, Weifang, Shandong, People’s Republic of China
| | - Xiuxiu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Jiong Ni
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yu Guan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Xin’ang Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaoqing Lin
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People’s Republic of China
| | - Jie Li
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
- College of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People’s Republic of China
| | - Yi Xia
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Xiang Wang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Yun Wang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Wenjun Huang
- Department of Radiology, The Second People’s Hospital of Deyang, Deyang, Sichuan, People’s Republic of China
| | - Wenting Tu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Peng Dong
- School of Medical Imaging, Shandong Second Medical University, Weifang, Shandong, People’s Republic of China
| | - Zhaobin Li
- Department of Radiation Oncology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, 200233, People’s Republic of China
| | - Shiyuan Liu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| | - Li Fan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Taylor-Blair HC, Siu ACW, Haysom-McDowell A, Kokkinis S, Bani Saeid A, Chellappan DK, Oliver BGG, Paudel KR, De Rubis G, Dua K. The impact of airborne particulate matter-based pollution on the cellular and molecular mechanisms in chronic obstructive pulmonary disease (COPD). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176413. [PMID: 39322084 DOI: 10.1016/j.scitotenv.2024.176413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/26/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Inhalation of particulate matter (PM), one of the many components of air pollution, is associated with the development and exacerbation of chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD). COPD is one of the leading causes of global mortality and morbidity, with a paucity of therapeutic options and a significant contributor to global health expenditure. This review aims to provide a mechanistic understanding of the cellular and molecular pathways that lead to the development of COPD following chronic PM exposure. Our review describes how the inhalation of PM can lead to lung parenchymal destruction and cellular senescence due to chronic pulmonary inflammation and oxidative stress. Following inhalation of PM, significant increases in a range of pro-inflammatory cytokines, mediated by the nuclear factor kappa B pathway are reported. This review also highlights how the inhalation of PM can lead to deleterious chronic oxidative stress persisting in the lung post-exposure. Furthermore, our work summarises how PM inhalation can lead to airway remodelling, with increases in pro-fibrotic cytokines and collagen deposition, typical of COPD. This paper also accentuates the interconnection and possible synergism between the pathophysiological mechanisms leading to COPD. Our work emphasises the serious health consequences of PM exposure on respiratory health. Elucidation of the cellular and molecular mechanisms can provide insight into possible therapeutic options. Finally, this review should serve as a stark reminder of the need for genuine action on air pollution to decrease the associated health burden on our growing global population.
Collapse
Affiliation(s)
- Hudson C Taylor-Blair
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Alexander Chi Wang Siu
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Adam Haysom-McDowell
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Sofia Kokkinis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Ayeh Bani Saeid
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Brian G G Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia; Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, Macquarie Park, NSW 2113, Australia
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, University of Technology Sydney, School of Life Sciences, Faculty of Science, Ultimo, NSW 2007, Australia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
Collapse
|
4
|
Cobb K, Kenyon J, Lu J, Krieger B, Perelas A, Nana-Sinkam P, Kim Y, Rodriguez-Miguelez P. COPD is associated with increased cardiovascular disease risk independent of phenotype. Respirology 2024; 29:1047-1057. [PMID: 39019777 PMCID: PMC11570344 DOI: 10.1111/resp.14799] [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: 03/20/2024] [Accepted: 07/03/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND AND OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide that frequently presents with concomitant cardiovascular diseases. Despite the pathological distinction between individual COPD phenotypes such as emphysema and chronic bronchitis, there is a lack of knowledge about the impact of COPD phenotype on cardiovascular disease risk. Thus, this study aimed to utilize a nationally representative sample to investigate cardiovascular disease prevalence in patients with COPD with emphysema and chronic bronchitis phenotypes. METHODS Data from 31,560 adults including 2504 individuals with COPD, collected as part of the National Health and Nutrition Examination Survey (1999-2018), were examined. RESULTS A significantly increased cardiovascular disease risk, including coronary heart disease, heart failure, myocardial infarction and stroke, was identified in patients with COPD among all disease phenotypes. Particularly, compared to those without COPD, individuals with chronic bronchitis presented with 1.76 (95% CI: 1.41-2.20) times greater odds, individuals with emphysema with 2.31 (95% CI: 1.80-2.96) times greater odds, while those with a concurrent phenotype (combined chronic bronchitis and emphysema) exhibited 2.98 (95% CI: 2.11-4.21) times greater odds of reporting cardiovascular diseases. CONCLUSION Our data confirms that patients with COPD present an elevated risk of developing cardiovascular disease among all phenotypes, with the most marked increase being in those with concurrent chronic bronchitis and emphysema phenotypes. These findings emphasize the need for awareness and appropriate cardiovascular screening in COPD.
Collapse
Affiliation(s)
- Kolton Cobb
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University
| | - Jonathan Kenyon
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University
| | - Juan Lu
- Division of Epidemiology, Virginia Commonwealth University
| | - Benjamin Krieger
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University
| | - Apostolos Perelas
- Division of Pulmonary and Critical Care, Virginia Commonwealth University
| | | | - Youngdeok Kim
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University
| | - Paula Rodriguez-Miguelez
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University
- Division of Pulmonary and Critical Care, Virginia Commonwealth University
| |
Collapse
|
5
|
Shlobin OA, Adir Y, Barbera JA, Cottin V, Harari S, Jutant EM, Pepke-Zaba J, Ghofrani HA, Channick R. Pulmonary hypertension associated with lung diseases. Eur Respir J 2024; 64:2401200. [PMID: 39209469 PMCID: PMC11525344 DOI: 10.1183/13993003.01200-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 09/04/2024]
Abstract
Pulmonary hypertension (PH) associated with chronic lung disease (CLD) is both common and underrecognised. The presence of PH in the setting of lung disease has been consistently shown to be associated with worse outcomes. Recent epidemiological studies have advanced understanding of the heterogeneity of this patient population and shown that defining both the specific type of CLD as well as the severity of PH (i.e. deeper phenotyping) is necessary to inform natural history and prognosis. A systematic diagnostic approach to screening and confirmation of suspected PH in CLD is recommended. Numerous uncontrolled studies and one phase 3 randomised, controlled trial have suggested a benefit in treating PH in some patients with CLD, specifically those with fibrotic interstitial lung disease (ILD). However, other studies in diseases such as COPD-PH showed adverse outcomes with some therapies. Given the expanding list of approved pharmacological treatments for pulmonary arterial hypertension, developing a treatment algorithm for specific phenotypes of CLD-PH is required. This article will summarise existing data in COPD, ILD and other chronic lung diseases, and provide recommendations for classification of CLD-PH and approach to the diagnosis and management of these challenging patients.
Collapse
Affiliation(s)
- Oksana A Shlobin
- Advanced Lung Disease and Transplant Program, Inova Schar Heart and Vascular Institute, Inova Fairfax Hospital, Falls Church, VA, USA
| | - Yochai Adir
- Pulmonary Division, Lady Davis Carmel Medical Center, Faculty of Medicine Technion Institute of Technology, Haifa, Israel
| | - Joan A Barbera
- Department of Pulmonary Medicine, Hospital Clínic-IDIBAPS, University of Barcelona; Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Vincent Cottin
- Department of Respiratory Medicine, National Reference Centre for Rare Pulmonary Diseases, ERN-LUNG, Louis Pradel Hospital, Hospices Civils de Lyon and UMR 754, INRAE, Claude Bernard University Lyon 1, Lyon, France
| | - Sergio Harari
- Unità Operativa di Pneumologia e Terapia Semi-Intensiva Respiratoria, MultiMedica IRCCS, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Etienne-Marie Jutant
- Respiratory Department, Centre Hospitalier Universitaire de Poitiers, INSERM CIC 1402, IS-ALIVE Research Group, University of Poitiers, Poitiers, France
| | - Joanna Pepke-Zaba
- Pulmonary Vascular Diseases Unit, Royal Papworth Hospital, University of Cambridge, Cambridge, UK
| | - Hossein-Ardeschir Ghofrani
- Justus-Liebig University Giessen, ECCPS, Kerckhoff-Klinik Bad Nauheim, Giessen, Germany
- Imperial College London, London, UK
| | - Richard Channick
- Pulmonary Vascular Disease Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
6
|
Dal Negro RW, Turco P, Povero M. Single-Breath Simultaneous Measurement of DL NO and DL CO as Predictor of the Emphysema Component in COPD - A Retrospective Observational Study. Int J Chron Obstruct Pulmon Dis 2024; 19:2123-2133. [PMID: 39351081 PMCID: PMC11439895 DOI: 10.2147/copd.s467138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/15/2024] [Indexed: 10/04/2024] Open
Abstract
Background Chronic Obstructive Pulmonary Disease (COPD) is a respiratory condition characterized by heterogeneous abnormalities of the airways and lung parenchyma that cause different clinical presentations. The assessment of the prevailing pathogenetic components underlying COPD is not usually pursued in daily practice, also due to technological limitations and cost. Aim To assess non-invasively the lung emphysema component of COPD by the simultaneous measurement of DLNO and DLCO via a single-breath (sDLNO and sDLCO). Methods COPD patients aged ≥40 years of both genders were recruited consecutively and labelled by computed tomography as "with significant" emphysema (>10% of CT lung volume) or "with negligible" emphysema otherwise. Current lung function tests such as sDLNO, sDLCO and Vc (the lung capillary blood volume) were measured. All possible subsets of independent spirometric and diffusive parameters were tested as predictors of emphysema, and their predicted power compared to each parameter alone by ROC analysis and area under the curve (AUC). Results Thirty-one patients with "significant emphysema" were compared to thirty-one with "negligible emphysema". FEV1 and FEV1/FVC seemed to be the best spirometric predictors (AUC 0.80 and 0.81, respectively), while sDLCO and Vc had the highest predicted power among diffusive parameters (AUC 0.92 and 0.94, respectively). sDLCO and Vc values were the parameters most correlated to the extent of CT emphysema. Six subsets of independent predictors were identified and included at least one spirometric and one diffusive parameter. According to goodness-to-fit scores (AIC, BIC, log-likelihood and pseudo R2), RV coupled with sDLCO or Vc proved the best predictors of emphysema. Conclusion When investigating the parenchymal destructive component due to emphysema occurring in COPD, sDLNO, sDLCO and Vc do enhance the predictive power of current spirometric measures substantially. sDLNO, sDLCO and Vc contribute to phenotype of the main pathogenetic components of COPD easily and with high sensitivity. Organizational problems, radiation exposure, time and costs could be reduced, while personalized and precision medicine could be noticeably implemented.
Collapse
Affiliation(s)
- Roberto W Dal Negro
- National Centre for Respiratory Pharmacoeconomics and Pharmacoepidemiology - CESFAR, Verona, Italy
| | - Paola Turco
- National Centre for Respiratory Pharmacoeconomics and Pharmacoepidemiology - CESFAR, Verona, Italy
| | | |
Collapse
|
7
|
Konigsberg IR, Vu T, Liu W, Litkowski EM, Pratte KA, Vargas LB, Gilmore N, Abdel-Hafiz M, Manichaikul A, Cho MH, Hersh CP, DeMeo DL, Banaei-Kashani F, Bowler RP, Lange LA, Kechris KJ. Proteomic networks and related genetic variants associated with smoking and chronic obstructive pulmonary disease. BMC Genomics 2024; 25:825. [PMID: 39223457 PMCID: PMC11370252 DOI: 10.1186/s12864-024-10619-1] [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: 02/08/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Studies have identified individual blood biomarkers associated with chronic obstructive pulmonary disease (COPD) and related phenotypes. However, complex diseases such as COPD typically involve changes in multiple molecules with interconnections that may not be captured when considering single molecular features. METHODS Leveraging proteomic data from 3,173 COPDGene Non-Hispanic White (NHW) and African American (AA) participants, we applied sparse multiple canonical correlation network analysis (SmCCNet) to 4,776 proteins assayed on the SomaScan v4.0 platform to derive sparse networks of proteins associated with current vs. former smoking status, airflow obstruction, and emphysema quantitated from high-resolution computed tomography scans. We then used NetSHy, a dimension reduction technique leveraging network topology, to produce summary scores of each proteomic network, referred to as NetSHy scores. We next performed a genome-wide association study (GWAS) to identify variants associated with the NetSHy scores, or network quantitative trait loci (nQTLs). Finally, we evaluated the replicability of the networks in an independent cohort, SPIROMICS. RESULTS We identified networks of 13 to 104 proteins for each phenotype and exposure in NHW and AA, and the derived NetSHy scores significantly associated with the variable of interests. Networks included known (sRAGE, ALPP, MIP1) and novel molecules (CA10, CPB1, HIS3, PXDN) and interactions involved in COPD pathogenesis. We observed 7 nQTL loci associated with NetSHy scores, 4 of which remained after conditional analysis. Networks for smoking status and emphysema, but not airflow obstruction, demonstrated a high degree of replicability across race groups and cohorts. CONCLUSIONS In this work, we apply state-of-the-art molecular network generation and summarization approaches to proteomic data from COPDGene participants to uncover protein networks associated with COPD phenotypes. We further identify genetic associations with networks. This work discovers protein networks containing known and novel proteins and protein interactions associated with clinically relevant COPD phenotypes across race groups and cohorts.
Collapse
Affiliation(s)
- Iain R Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Thao Vu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Weixuan Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Elizabeth M Litkowski
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Luciana B Vargas
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Niles Gilmore
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Mohamed Abdel-Hafiz
- Department of Computer Science and Engineering, University of Colorado - Denver, Denver, CO, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Farnoush Banaei-Kashani
- Department of Computer Science and Engineering, University of Colorado - Denver, Denver, CO, USA
| | | | - Leslie A Lange
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Katerina J Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA.
| |
Collapse
|
8
|
Nauck S, Pohl M, Jobst BJ, Melzig C, Meredig H, Weinheimer O, Triphan S, von Stackelberg O, Konietzke P, Kauczor HU, Heußel CP, Wielpütz MO, Biederer J. Phenotyping of COPD with MRI in comparison to same-day CT in a multi-centre trial. Eur Radiol 2024; 34:5597-5609. [PMID: 38345607 PMCID: PMC11364611 DOI: 10.1007/s00330-024-10610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/07/2023] [Accepted: 12/24/2023] [Indexed: 08/31/2024]
Abstract
OBJECTIVES A prospective, multi-centre study to evaluate concordance of morphologic lung MRI and CT in chronic obstructive pulmonary disease (COPD) phenotyping for airway disease and emphysema. METHODS A total of 601 participants with COPD from 15 sites underwent same-day morpho-functional chest MRI and paired inspiratory-expiratory CT. Two readers systematically scored bronchial wall thickening, bronchiectasis, centrilobular nodules, air trapping and lung parenchyma defects in each lung lobe and determined COPD phenotype. A third reader acted as adjudicator to establish consensus. Inter-modality and inter-reader agreement were assessed using Cohen's kappa (im-κ and ir-κ). RESULTS The mean combined MRI score for bronchiectasis/bronchial wall thickening was 4.5/12 (CT scores, 2.2/12 for bronchiectasis and 6/12 for bronchial wall thickening; im-κ, 0.04-0.3). Expiratory right/left bronchial collapse was observed in 51 and 47/583 on MRI (62 and 57/599 on CT; im-κ, 0.49-0.52). Markers of small airways disease on MRI were 0.15/12 for centrilobular nodules (CT, 0.34/12), 0.94/12 for air trapping (CT, 0.9/12) and 7.6/12 for perfusion deficits (CT, 0.37/12 for mosaic attenuation; im-κ, 0.1-0.41). The mean lung defect score on MRI was 1.3/12 (CT emphysema score, 5.8/24; im-κ, 0.18-0.26). Airway-/emphysema/mixed COPD phenotypes were assigned in 370, 218 and 10 of 583 cases on MRI (347, 218 and 34 of 599 cases on CT; im-κ, 0.63). For all examined features, inter-reader agreement on MRI was lower than on CT. CONCLUSION Concordance of MRI and CT for phenotyping of COPD in a multi-centre setting was substantial with variable inter-modality and inter-reader concordance for single diagnostic key features. CLINICAL RELEVANCE STATEMENT MRI of lung morphology may well serve as a radiation-free imaging modality for COPD in scientific and clinical settings, given that its potential and limitations as shown here are carefully considered. KEY POINTS • In a multi-centre setting, MRI and CT showed substantial concordance for phenotyping of COPD (airway-/emphysema-/mixed-type). • Individual features of COPD demonstrated variable inter-modality concordance with features of pulmonary hypertension showing the highest and bronchiectasis showing the lowest concordance. • For all single features of COPD, inter-reader agreement was lower on MRI than on CT.
Collapse
Affiliation(s)
- Sebastian Nauck
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany.
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany.
| | - Moritz Pohl
- Institute of Medical Biometry, University Hospital of Heidelberg, Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Bertram J Jobst
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Claudius Melzig
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Hagen Meredig
- Department of Neuroradiology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany
| | - Oliver Weinheimer
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Simon Triphan
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Oyunbileg von Stackelberg
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Philip Konietzke
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Hans-Ulrich Kauczor
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Claus P Heußel
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at the University of Heidelberg, Röntgenstraße 1, 69126, Heidelberg, Germany
| | - Mark O Wielpütz
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
| | - Jürgen Biederer
- Department of Diagnostic and Interventional Radiology, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Im Neuenheimer Feld 130.3, 69120, Heidelberg, Germany
- Faculty of Medicine, University of Latvia, Raina bulvaris 19, Riga, LV-1586, Latvia
- Faculty of Medicine, Christian-Albrechts-Universität zu Kiel, 24098, Kiel, Germany
| |
Collapse
|
9
|
Bernabeu-Mora R, Valera-Novella E, Bernabeu-Serrano ET, Soler-Cataluña JJ, Calle-Rubio M, Medina-Mirapeix F. Five-Repetition Sit-to-Stand Test as Predictor of Mortality in High Risk COPD Patients. Arch Bronconeumol 2024:S0300-2896(24)00312-0. [PMID: 39245610 DOI: 10.1016/j.arbres.2024.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE To determine if adding performance on the five-repetition sit-to-stand test (5-STS) to chronic obstructive pulmonary disease (COPD) high-risk criteria, proposed by the Spanish COPD guidelines (GesEPOC), affects mortality prognosis. METHODS Observational study of COPD outpatients involved prospective follow-up for 5 years. Patients were classified based on 5-STS performance and risk criteria proposed by GesEPOC version 2021. Outcome measures were 5-year mortality timing and rate. Kaplan-Meier curves and univariate and multivariate Cox proportional-hazard analyses, analysis of variance, and univariate and multivariate linear and logistic regression models were used. RESULTS One hundred and thirty-seven patients were included. Mean age was 66±8.3 years, and 87.6% were men. Of them, 115 (83.9%) were classified as high risk, 43 (34.4%) of whom had poor performance on the 5-STS. Overall mortality at 5 years was 27% and was significantly higher in the high-risk (29.6%) compared with the low-risk (13.6%) group. Among high-risk patients, mortality at 5 years was significantly worse with poor 5-STS performance (60.5%) compared with non-poor performance (11.1%). Poor performance on the 5-STS was independently associated with increased 5-year mortality risk (HR 4.70; 95% CI: 1.96-11.27) in a model adjusted for history of heart disease and dyspnea. CONCLUSION Among high-risk COPD patients, those with poor performance on the 5-STS have a significantly higher mortality at 5 years than those with non-poor 5-STS performance.
Collapse
Affiliation(s)
- Roberto Bernabeu-Mora
- Department of Pneumology, Hospital General Universitario Morales Meseguer, Murcia, Spain; Department of Internal Medicine, University of Murcia, Murcia, Spain; Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Elisa Valera-Novella
- Department of Physical Therapy, University of Murcia, Murcia, Spain; Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain.
| | | | - Juan José Soler-Cataluña
- Department of Pneumology, Hospital Arnau de Vilanova, Valencia, Spain; University of Valencia, Valencia, Spain
| | - Myriam Calle-Rubio
- Department of Pneumology, Hospital Clínico San Carlos, Madrid, Spain; Complutense University of Madrid, Murcia, Spain; Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IDISCC), Madrid, Spain
| | - Francesc Medina-Mirapeix
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain; Department of Physical Therapy, University of Murcia, Murcia, Spain
| |
Collapse
|
10
|
Eryüksel E, Tunca Z, Mercancı Z, Kılıç SS, Kocakaya D, Akdeniz E, Öztop NE, Çetin E, Akkoç T. Stem cell treatment reduces T cell apoptosis in COPD patients with chronic bronchitis but not with emphysema. Tissue Cell 2024; 89:102452. [PMID: 38986345 DOI: 10.1016/j.tice.2024.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a prevalent and preventable condition. Mesenchymal stem cell (MSC) therapy is being explored to aid in the regeneration of lung cells and airway structure, aiming to restore lung function. AIM To examine varied responses of MSCs when cultured with peripheral blood mononuclear cells (PBMCs) from different COPD phenotypes, patients were grouped into ACOS, emphysema, and chronic bronchitis categories. METHODS PBMCs from these groups and controls were co-cultured with MSCs derived from dental follicles, revealing differing rates of apoptosis among COPD phenotypes compared to controls. RESULTS While the chronic bronchitis group exhibited the least lymphocyte viability (p<0.01), introducing MSCs notably enhanced viability across all phenotypes except emphysema, with the chronic bronchitis group showing the most improvement (p<0.05). CONCLUSION Stem cell therapy might reduce peripheral lymphocyte apoptosis in COPD, with varying responses based on phenotype, necessitating further research to understand mechanisms and optimize tailored therapies for each COPD subtype.
Collapse
Affiliation(s)
- Emel Eryüksel
- Pulmonary and Critical Care, Faculty of Medicine, Marmara University, Turkey.
| | - Zeynep Tunca
- Department of Immunology, Faculty of Medicine, Marmara University, Turkey; Department of Pediatric Allergy-Immunology, Faculty of Medicine, Marmara University, Turkey
| | - Zeynep Mercancı
- Pulmonary and Critical Care, Faculty of Medicine, Marmara University, Turkey
| | - Sabriye Senem Kılıç
- Department of Immunology, Faculty of Medicine, Marmara University, Turkey; Department of Pediatric Allergy-Immunology, Faculty of Medicine, Marmara University, Turkey
| | - Derya Kocakaya
- Pulmonary and Critical Care, Faculty of Medicine, Marmara University, Turkey
| | - Esra Akdeniz
- Department of Medical Education, Faculty of Medicine, Marmara University, Turkey
| | - Nur Ecem Öztop
- Department of Immunology, Faculty of Medicine, Marmara University, Turkey
| | - Esin Çetin
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Tunç Akkoç
- Department of Immunology, Faculty of Medicine, Marmara University, Turkey; Department of Pediatric Allergy-Immunology, Faculty of Medicine, Marmara University, Turkey
| |
Collapse
|
11
|
Koopman M, Posthuma R, Vanfleteren LEGW, Simons SO, Franssen FME. Lung Hyperinflation as Treatable Trait in Chronic Obstructive Pulmonary Disease: A Narrative Review. Int J Chron Obstruct Pulmon Dis 2024; 19:1561-1578. [PMID: 38974815 PMCID: PMC11227310 DOI: 10.2147/copd.s458324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024] Open
Abstract
Lung hyperinflation (LH) is a common clinical feature in patients with chronic obstructive pulmonary disease (COPD). It results from a combination of reduced elastic lung recoil as a consequence of irreversible destruction of lung parenchyma and expiratory airflow limitation. LH is an important determinant of morbidity and mortality in COPD, partially independent of the degree of airflow limitation. Therefore, reducing LH has become a major target in the treatment of COPD over the last decades. Advances were made in the diagnostics of LH and several effective interventions became available. Moreover, there is increasing evidence suggesting that LH is not only an isolated feature in COPD but rather part of a distinct clinical phenotype that may require a more integrated management. This narrative review focuses on the pathophysiology and adverse consequences of LH, the assessment of LH with lung function measurements and imaging techniques and highlights LH as a treatable trait in COPD. Finally, several suggestions regarding future studies in this field are made.
Collapse
Affiliation(s)
- Maud Koopman
- Research and Development, Ciro+, Horn, the Netherlands
- NUTRIM, Institute of Nutrition and Translational Research in Metabolism, University Maastricht, Maastricht, the Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Rein Posthuma
- Research and Development, Ciro+, Horn, the Netherlands
- NUTRIM, Institute of Nutrition and Translational Research in Metabolism, University Maastricht, Maastricht, the Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Lowie E G W Vanfleteren
- COPD Center, Institute of Medicine, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Sami O Simons
- NUTRIM, Institute of Nutrition and Translational Research in Metabolism, University Maastricht, Maastricht, the Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Frits M E Franssen
- Research and Development, Ciro+, Horn, the Netherlands
- NUTRIM, Institute of Nutrition and Translational Research in Metabolism, University Maastricht, Maastricht, the Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| |
Collapse
|
12
|
Self AA, Mesarwi OA. Intermittent Versus Sustained Hypoxemia from Sleep-disordered Breathing: Outcomes in Patients with Chronic Lung Disease and High Altitude. Sleep Med Clin 2024; 19:327-337. [PMID: 38692756 DOI: 10.1016/j.jsmc.2024.02.011] [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] [Indexed: 05/03/2024]
Abstract
In a variety of physiologic and pathologic states, people may experience both chronic sustained hypoxemia and intermittent hypoxemia ("combined" or "overlap" hypoxemia). In general, hypoxemia in such instances predicts a variety of maladaptive outcomes, including excess cardiovascular disease or mortality. However, hypoxemia may be one of the myriad phenotypic effects in such states, making it difficult to ascertain whether adverse outcomes are primarily driven by hypoxemia, and if so, whether these effects are due to intermittent versus sustained hypoxemia.
Collapse
Affiliation(s)
- Alyssa A Self
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, University of California, San Diego, 9500 Gilman Drive Mail Code 0623A, La Jolla, CA 92093, USA
| | - Omar A Mesarwi
- Division of Pulmonary, Critical Care, and Sleep Medicine and Physiology, University of California, San Diego, 9500 Gilman Drive Mail Code 0623A, La Jolla, CA 92093, USA.
| |
Collapse
|
13
|
Rodrigues Pereira RP, Mazzali Pessoa Martins AM, Mendes de Carvalho IT, Kel de Souza LD, Francao P, Gomes CM, Bernardes RDP, Meyer KF, Fonseca EMGOD, Machado MG, Tanaka C. Clinical phenotyping of children with nocturnal enuresis: A key classification to improve the approach. J Pediatr Urol 2024; 20:384.e1-384.e9. [PMID: 38508980 DOI: 10.1016/j.jpurol.2024.01.019] [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: 01/05/2023] [Revised: 12/01/2023] [Accepted: 01/19/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION The literature shows that nocturnal enuresis is not an isolated phenomenon of urinary loss during sleep, but encompasses a set of systemic clinical manifestations that significantly influence children's quality of life and development. However, the understanding of the clinical and physiological relationship of these systemic manifestations remains a clinical challenge. The recognition of these manifestations and their subsequent categorisation, may provide better insights into integrated clinical manifestations, facilitating the understanding of pathophysiological mechanisms, and promote increased assertiveness in the assessment and the selection of appropriate therapies. OBJECTIVE The aim of this study is to develop a phenotyping model for children with nocturnal enuresis based on evidence. METHODS This study presents a clinical phenotyping model for children with nocturnal enuresis based on an analytical and methodological review of the literature, about nocturnal enuresis and its associated clinical manifestations. There was a bibliometric analysis carried out to better analyse outcomes. After reading and analysing the literature, the clinical manifestations were categorised into domains and submitted to the validation of an expert committee with extensive experience in their specific area of expertise. A visual representation of the categorised model was developed to make the phenotyping concept easily understandable to all professionals. RESULTS The clinical manifestations related to nocturnal enuresis have been categorised according to frequency and relation found in the literature and validation by an expert committee and the development of the phenotyping model for children with nocturnal enuresis was completed. CONCLUSION The present study developed an evidence-based phenotyping model for children with nocturnal enuresis.
Collapse
Affiliation(s)
- Rita Pavione Rodrigues Pereira
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil; LIM 54 - Laboratório de Investigação em Fisioterapia, Hospital das Clínicas da Faculdade de Medicina da Universidade De São Paulo, Sao Paulo, SP, Brazil.
| | - Aline Mari Mazzali Pessoa Martins
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil; LIM 54 - Laboratório de Investigação em Fisioterapia, Hospital das Clínicas da Faculdade de Medicina da Universidade De São Paulo, Sao Paulo, SP, Brazil.
| | - Isabela Teixeira Mendes de Carvalho
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil; LIM 54 - Laboratório de Investigação em Fisioterapia, Hospital das Clínicas da Faculdade de Medicina da Universidade De São Paulo, Sao Paulo, SP, Brazil.
| | - Luana Daniele Kel de Souza
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil; LIM 54 - Laboratório de Investigação em Fisioterapia, Hospital das Clínicas da Faculdade de Medicina da Universidade De São Paulo, Sao Paulo, SP, Brazil.
| | - Patricia Francao
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil; LIM 54 - Laboratório de Investigação em Fisioterapia, Hospital das Clínicas da Faculdade de Medicina da Universidade De São Paulo, Sao Paulo, SP, Brazil.
| | - Cristiano Mendes Gomes
- Divisão de Urologia do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil.
| | | | | | - Eliane Maria Garcez Oliveira da Fonseca
- Departamento de Pediatria, Núcleo de Disfunção Miccional, Faculdade de Ciências Médicas da Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil; Departamento de Pediatria da Escola de Medicina Souza Marques, Rio de Janeiro, Brazil.
| | - Marcos Giannetti Machado
- Divisão de Urologia do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil.
| | - Clarice Tanaka
- Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional da Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil; LIM 54 - Laboratório de Investigação em Fisioterapia, Hospital das Clínicas da Faculdade de Medicina da Universidade De São Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
14
|
Short B, Delaney C, Johnston W, Litherland GJ, Lockhart JC, Williams C, Mackay WG, Ramage G. Informed development of a multi-species biofilm in chronic obstructive pulmonary disease. APMIS 2024; 132:336-347. [PMID: 38379455 DOI: 10.1111/apm.13386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
Recent evidence indicates that microbial biofilm aggregates inhabit the lungs of COPD patients and actively contribute towards chronic colonization and repeat infections. However, there are no contextually relevant complex biofilm models for COPD research. In this study, a meta-analysis of the lung microbiome in COPD was used to inform development of an optimized biofilm model composed of genera highly associated with COPD. Bioinformatic analysis showed that although diversity matrices of COPD microbiomes were similar to healthy controls, and internal compositions made it possible to accurately differentiate between these cohorts (AUC = 0.939). Genera that best defined these patients included Haemophilus, Moraxella and Streptococcus. Many studies fail to account for fungi; therefore, Candida albicans was included in the creation of an interkingdom biofilm model. These organisms formed a biofilm capable of tolerating high concentrations of antimicrobial therapies with no significant reductions in viability. However, combined therapies of antibiotics and an antifungal resulted in significant reductions in viable cells throughout the biofilm (p < 0.05). This biofilm model is representative of the COPD lung microbiome and results from in vitro antimicrobial challenge experiments indicate that targeting both bacteria and fungi in these interkingdom communities will be required for more positive clinical outcomes.
Collapse
Affiliation(s)
- Bryn Short
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, UK
| | - Christopher Delaney
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, UK
| | - William Johnston
- Safeguarding Health through Infection Prevention (SHIP) Research Group, Research Centre for Health, Glasgow Caledonian University, Glasgow, UK
| | - Gary J Litherland
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley, UK
- Hamilton International Technology Park, Glasgow, UK
| | - John C Lockhart
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley, UK
- Hamilton International Technology Park, Glasgow, UK
| | - Craig Williams
- Microbiology Department, Lancaster Royal Infirmary, University of Lancaster, Lancaster, UK
| | - William G Mackay
- Institute of Biomedical and Environmental Health Research, School of Health and Life Sciences, University of the West of Scotland, Paisley, UK
- Hamilton International Technology Park, Glasgow, UK
| | - Gordon Ramage
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences (MVLS), University of Glasgow, Glasgow, UK
- Safeguarding Health through Infection Prevention (SHIP) Research Group, Research Centre for Health, Glasgow Caledonian University, Glasgow, UK
| |
Collapse
|
15
|
Li F, Zhang X, Comellas AP, Hoffman EA, Graham MM, Lin CL. Exploratory Study on COPD Phenotypes and their Progression: Integrating SPECT and qCT Imaging Analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.10.24305577. [PMID: 38645219 PMCID: PMC11030493 DOI: 10.1101/2024.04.10.24305577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background The objective of this study is to understand chronic obstructive pulmonary disease (COPD) phenotypes and their progressions by quantifying heterogeneities of lung ventilation from the single photon emission computed tomography (SPECT) images and establishing associations with the quantitative computed tomography (qCT) imaging-based clusters and variables. Methods Eight COPD patients completed a longitudinal study of three visits with intervals of about a year. CT scans of these subjects at residual volume, functional residual capacity, and total lung capacity were taken for all visits. The functional and structural qCT-based variables were derived, and the subjects were classified into the qCT-based clusters. In addition, the SPECT variables were derived to quantify the heterogeneity of lung ventilation. The correlations between the key qCT-based variables and SPECT-based variables were examined. Results The SPECT-based coefficient of variation (CVTotal), a measure of ventilation heterogeneity, showed strong correlations (|r| ≥ 0.7) with the qCT-based functional small airway disease percentage (fSAD%Total) and emphysematous tissue percentage (Emph%Total) in the total lung on cross-sectional data. As for the two-year changes, the SPECT-based maximum tracer concentration (TCmax), a measure of hot spots, exhibited strong negative correlations with fSAD%Total, Emph%Total, average airway diameter in the left upper lobe, and airflow distribution in the middle and lower lobes. Conclusion Small airway disease is highly associated with the heterogeneity of ventilation in COPD lungs. TCmax is a more sensitive functional biomarker for COPD progression than CVTotal. Besides fSAD%Total and Emph%Total, segmental airways narrowing and imbalanced ventilation between upper and lower lobes may contribute to the development of hot spots over time.
Collapse
Affiliation(s)
- Frank Li
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
- IIHR-Hydroscience & Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Xuan Zhang
- IIHR-Hydroscience & Engineering, University of Iowa, Iowa City, Iowa, USA
- Department of Mechanical Engineering, University of Iowa, Iowa City, Iowa, USA
| | | | - Eric A. Hoffman
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
- Department of Radiology, University of Iowa, Iowa City, Iowa, USA
| | | | - Ching-Long Lin
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
- IIHR-Hydroscience & Engineering, University of Iowa, Iowa City, Iowa, USA
- Department of Mechanical Engineering, University of Iowa, Iowa City, Iowa, USA
- Department of Radiology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
16
|
Correnti S, Preianò M, Gamboni F, Stephenson D, Pelaia C, Pelaia G, Savino R, D'Alessandro A, Terracciano R. An integrated metabo-lipidomics profile of induced sputum for the identification of novel biomarkers in the differential diagnosis of asthma and COPD. J Transl Med 2024; 22:301. [PMID: 38521955 PMCID: PMC10960495 DOI: 10.1186/s12967-024-05100-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/15/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Due to their complexity and to the presence of common clinical features, differentiation between asthma and chronic obstructive pulmonary disease (COPD) can be a challenging task, complicated in such cases also by asthma-COPD overlap syndrome. The distinct immune/inflammatory and structural substrates of COPD and asthma are responsible for significant differences in the responses to standard pharmacologic treatments. Therefore, an accurate diagnosis is of central relevance to assure the appropriate therapeutic intervention in order to achieve safe and effective patient care. Induced sputum (IS) accurately mirrors inflammation in the airways, providing a more direct picture of lung cell metabolism in comparison to those specimen that reflect analytes in the systemic circulation. METHODS An integrated untargeted metabolomics and lipidomics analysis was performed in IS of asthmatic (n = 15) and COPD (n = 22) patients based on Ultra-High-Pressure Liquid Chromatography-Mass Spectrometry (UHPLC-MS) and UHPLC-tandem MS (UHPLC-MS/MS). Partial Least Squares-Discriminant Analysis (PLS-DA) was applied to resulting dataset. The analysis of main enriched metabolic pathways and the association of the preliminary metabolites/lipids pattern identified to clinical parameters of asthma/COPD differentiation were explored. Multivariate ROC analysis was performed in order to determine the discriminatory power and the reliability of the putative biomarkers for diagnosis between COPD and asthma. RESULTS PLS-DA indicated a clear separation between COPD and asthmatic patients. Among the 15 selected candidate biomarkers based on Variable Importance in Projection scores, putrescine showed the highest score. A differential IS bio-signature of 22 metabolites and lipids was found, which showed statistically significant variations between asthma and COPD. Of these 22 compounds, 18 were decreased and 4 increased in COPD compared to asthmatic patients. The IS levels of Phosphatidylethanolamine (PE) (34:1), Phosphatidylglycerol (PG) (18:1;18:2) and spermine were significantly higher in asthmatic subjects compared to COPD. CONCLUSIONS This is the first pilot study to analyse the IS metabolomics/lipidomics signatures relevant in discriminating asthma vs COPD. The role of polyamines, of 6-Hydroxykynurenic acid and of D-rhamnose as well as of other important players related to the alteration of glycerophospholipid, aminoacid/biotin and energy metabolism provided the construction of a diagnostic model that, if validated on a larger prospective cohort, might be used to rapidly and accurately discriminate asthma from COPD.
Collapse
Affiliation(s)
- Serena Correnti
- Department of Health Sciences, Magna Græcia University, 88100, Catanzaro, Italy.
| | | | - Fabia Gamboni
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Girolamo Pelaia
- Department of Health Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Rocco Savino
- Department of Medical and Surgical Sciences, Magna Græcia University, 88100, Catanzaro, Italy
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Rosa Terracciano
- Department of Experimental and Clinical Medicine, Magna Græcia University, 88100, Catanzaro, Italy.
| |
Collapse
|
17
|
Alupo P, Mugenyi L, Katagira W, Kayongo A, Nalunjogi J, Siddharthan T, Hurst JR, Kirenga B, Jones R. Characteristics and phenotypes of a COPD cohort from referral hospital clinics in Uganda. BMJ Open Respir Res 2024; 11:e001816. [PMID: 38490695 PMCID: PMC10946361 DOI: 10.1136/bmjresp-2023-001816] [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: 05/08/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024] Open
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a heterogeneous condition with varied clinical and pathophysiological characteristics. Although there is increasing evidence that COPD in low-income and middle-income countries may have different clinical characteristics from that in high-income countries, little is known about COPD phenotypes in these settings. We describe the clinical characteristics and risk factor profile of a COPD population in Uganda. METHODS We cross sectionally analysed the baseline clinical characteristics of 323 patients with COPD aged 30 years and above who were attending 2 national referral outpatient facilities in Kampala, Uganda between July 2019 and March 2021. Logistic regression was used to determine factors associated with spirometric disease severity. RESULTS The median age was 62 years; 51.1% females; 93.5% scored COPD Assessment Test >10; 63.8% modified medical research council (mMRC) >2; 71.8% had wheezing; 16.7% HIV positive; 20.4% had a history of pulmonary tuberculosis (TB); 50% with blood eosinophilic count >3%, 51.7% had 3 or more exacerbations in the past year. Greater severity by Global initiative for Chronic Obstructive Lung Disease (GOLD) stage was inversely related to age (aOR=0.95, 95% CI 0.92 to 0.97), and obesity compared with underweight (aOR=0.25, 95% CI 0.07 to 0.82). Regarding clinical factors, more severe airflow obstruction was associated with SPO2 <93% (aOR=3.79, 95% CI 2.05 to 7.00), mMRC ≥2 (aOR=2.21, 95% CI 1.08 to 4.53), and a history of severe exacerbations (aOR=2.64, 95% CI 1.32 to 5.26). CONCLUSION Patients with COPD in this population had specific characteristics and risk factor profiles including HIV and TB meriting tailored preventative approaches. Further studies are needed to better understand the pathophysiological mechanisms at play and the therapeutic implications of these findings.
Collapse
Affiliation(s)
- Patricia Alupo
- Lung Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Levicatus Mugenyi
- Lung Institute, Makerere University College of Health Sciences, Kampala, Uganda
- Statistics Department, MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Winceslaus Katagira
- Lung Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Alex Kayongo
- Lung Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Joanitah Nalunjogi
- Lung Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Trishul Siddharthan
- Division of Pulmonary, Critical care and Sleep medicine, University of Miami School of Medicine, Miami, Florida, USA
| | - John R Hurst
- UCL Respiratory, University College London, London, UK
| | - Bruce Kirenga
- Lung Institute, Makerere University College of Health Sciences, Kampala, Uganda
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | | |
Collapse
|
18
|
Teixeira EM, Ribeiro CO, Lopes AJ, de Melo PL. Respiratory Oscillometry and Functional Performance in Different COPD Phenotypes. Int J Chron Obstruct Pulmon Dis 2024; 19:667-682. [PMID: 38464561 PMCID: PMC10924760 DOI: 10.2147/copd.s446085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/27/2024] [Indexed: 03/12/2024] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) phenotypes may introduce different characteristics that need to be known to improve treatment. Respiratory oscillometry provides a detailed analysis and may offer insight into the pathophysiology of COPD. In this paper, we used this method to evaluate the differences in respiratory mechanics of COPD phenotypes. Patients and Methods This study investigated a sample of 83 volunteers, being divided into control group (CG = 20), emphysema (n = 23), CB (n = 20) and asthma-COPD overlap syndrome (ACOS, n = 20). These analyses were performed before and after bronchodilator (BD) use. Functional capacity was evaluated using the Glittre‑ADL test, handgrip strength and respiratory pressures. Results Initially it was observed that oscillometry provided a detailed description of the COPD phenotypes, which was consistent with the involved pathophysiology. A correlation between oscillometry and functional capacity was observed (r=-0.541; p = 0.0001), particularly in the emphysema phenotype (r = -0.496, p = 0.031). BD response was different among the studied phenotypes. This resulted in an accurate discrimination of ACOS from CB [area under the receiver operating curve (AUC) = 0.84] and emphysema (AUC = 0.82). Conclusion These results offer evidence that oscillatory indices may enhance the comprehension and identification of COPD phenotypes, thereby potentially improving the support provided to these patients.
Collapse
Affiliation(s)
- Elayne Moura Teixeira
- Biomedical Instrumentation Laboratory, Institute of Biology and Faculty of Engineering, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caroline Oliveira Ribeiro
- Biomedical Instrumentation Laboratory, Institute of Biology and Faculty of Engineering, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Agnaldo José Lopes
- Pulmonary Function Laboratory, Pedro Ernesto University Hospital, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Pulmonary Rehabilitation Laboratory, Augusto Motta University Center, Rio de Janeiro, Brazil
| | - Pedro Lopes de Melo
- Biomedical Instrumentation Laboratory, Institute of Biology and Faculty of Engineering, State University of Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Clinical and Experimental Research in Vascular Biology - Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
He ZJ, Chu C, Dickson R, Okuda K, Cai LH. A gel-coated air-liquid-interface culture system with tunable substrate stiffness matching healthy and diseased lung tissues. Am J Physiol Lung Cell Mol Physiol 2024; 326:L292-L302. [PMID: 38252871 PMCID: PMC11280679 DOI: 10.1152/ajplung.00153.2023] [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: 05/15/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Since its invention in the late 1980s, the air-liquid-interface (ALI) culture system has been the standard in vitro model for studying human airway biology and pulmonary diseases. However, in a conventional ALI system, cells are cultured on a porous plastic membrane that is much stiffer than human airway tissues. Here, we develop a gel-ALI culture system by simply coating the plastic membrane with a thin layer of hydrogel with tunable stiffness matching that of healthy and fibrotic airway tissues. We determine the optimum gel thickness that does not impair the transport of nutrients and biomolecules essential to cell growth. We show that the gel-ALI system allows human bronchial epithelial cells (HBECs) to proliferate and differentiate into pseudostratified epithelium. Furthermore, we discover that HBECs migrate significantly faster on hydrogel substrates with stiffness matching that of fibrotic lung tissues, highlighting the importance of mechanical cues in human airway remodeling. The developed gel-ALI system provides a facile approach to studying the effects of mechanical cues in human airway biology and in modeling pulmonary diseases.NEW & NOTEWORTHY In a conventional ALI system, cells are cultured on a plastic membrane that is much stiffer than human airway tissues. We develop a gel-ALI system by coating the plastic membrane with a thin layer of hydrogel with tunable stiffness matching that of healthy and fibrotic airway tissues. We discover that human bronchial epithelial cells migrate significantly faster on hydrogel substrates with pathological stiffness, highlighting the importance of mechanical cues in human airway remodeling.
Collapse
Affiliation(s)
- Zhi-Jian He
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Catherine Chu
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Riley Dickson
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Kenichi Okuda
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Li-Heng Cai
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, Virginia, United States
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
20
|
Bell AJ, Pal R, Labaki WW, Hoff BA, Wang JM, Murray S, Kazerooni EA, Galban S, Lynch DA, Humphries SM, Martinez FJ, Hatt CR, Han MK, Ram S, Galban CJ. Local heterogeneity of normal lung parenchyma and small airways disease are associated with COPD severity and progression. Respir Res 2024; 25:106. [PMID: 38419014 PMCID: PMC10903150 DOI: 10.1186/s12931-024-02729-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Small airways disease (SAD) is a major cause of airflow obstruction in COPD patients and has been identified as a precursor to emphysema. Although the amount of SAD in the lungs can be quantified using our Parametric Response Mapping (PRM) approach, the full breadth of this readout as a measure of emphysema and COPD progression has yet to be explored. We evaluated topological features of PRM-derived normal parenchyma and SAD as surrogates of emphysema and predictors of spirometric decline. METHODS PRM metrics of normal lung (PRMNorm) and functional SAD (PRMfSAD) were generated from CT scans collected as part of the COPDGene study (n = 8956). Volume density (V) and Euler-Poincaré Characteristic (χ) image maps, measures of the extent and coalescence of pocket formations (i.e., topologies), respectively, were determined for both PRMNorm and PRMfSAD. Association with COPD severity, emphysema, and spirometric measures were assessed via multivariable regression models. Readouts were evaluated as inputs for predicting FEV1 decline using a machine learning model. RESULTS Multivariable cross-sectional analysis of COPD subjects showed that V and χ measures for PRMfSAD and PRMNorm were independently associated with the amount of emphysema. Readouts χfSAD (β of 0.106, p < 0.001) and VfSAD (β of 0.065, p = 0.004) were also independently associated with FEV1% predicted. The machine learning model using PRM topologies as inputs predicted FEV1 decline over five years with an AUC of 0.69. CONCLUSIONS We demonstrated that V and χ of fSAD and Norm have independent value when associated with lung function and emphysema. In addition, we demonstrated that these readouts are predictive of spirometric decline when used as inputs in a ML model. Our topological PRM approach using PRMfSAD and PRMNorm may show promise as an early indicator of emphysema onset and COPD progression.
Collapse
Affiliation(s)
- Alexander J Bell
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA
| | - Ravi Pal
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA
| | - Wassim W Labaki
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Benjamin A Hoff
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA
| | - Jennifer M Wang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Susan Murray
- School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ella A Kazerooni
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Stefanie Galban
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA
| | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, CO, USA
| | | | | | | | - MeiLan K Han
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sundaresh Ram
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - Craig J Galban
- Department of Radiology, University of Michigan, 109 Zina Pitcher Place BSRB A506, Ann Arbor, MI, 48109-2200, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
Konigsberg IR, Vu T, Liu W, Litkowski EM, Pratte KA, Vargas LB, Gilmore N, Abdel-Hafiz M, Manichaikul AW, Cho MH, Hersh CP, DeMeo DL, Banaei-Kashani F, Bowler RP, Lange LA, Kechris KJ. Proteomic Networks and Related Genetic Variants Associated with Smoking and Chronic Obstructive Pulmonary Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.02.26.24303069. [PMID: 38464285 PMCID: PMC10925350 DOI: 10.1101/2024.02.26.24303069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Studies have identified individual blood biomarkers associated with chronic obstructive pulmonary disease (COPD) and related phenotypes. However, complex diseases such as COPD typically involve changes in multiple molecules with interconnections that may not be captured when considering single molecular features. Methods Leveraging proteomic data from 3,173 COPDGene Non-Hispanic White (NHW) and African American (AA) participants, we applied sparse multiple canonical correlation network analysis (SmCCNet) to 4,776 proteins assayed on the SomaScan v4.0 platform to derive sparse networks of proteins associated with current vs. former smoking status, airflow obstruction, and emphysema quantitated from high-resolution computed tomography scans. We then used NetSHy, a dimension reduction technique leveraging network topology, to produce summary scores of each proteomic network, referred to as NetSHy scores. We next performed genome-wide association study (GWAS) to identify variants associated with the NetSHy scores, or network quantitative trait loci (nQTLs). Finally, we evaluated the replicability of the networks in an independent cohort, SPIROMICS. Results We identified networks of 13 to 104 proteins for each phenotype and exposure in NHW and AA, and the derived NetSHy scores significantly associated with the variable of interests. Networks included known (sRAGE, ALPP, MIP1) and novel molecules (CA10, CPB1, HIS3, PXDN) and interactions involved in COPD pathogenesis. We observed 7 nQTL loci associated with NetSHy scores, 4 of which remained after conditional analysis. Networks for smoking status and emphysema, but not airflow obstruction, demonstrated a high degree of replicability across race groups and cohorts. Conclusions In this work, we apply state-of-the-art molecular network generation and summarization approaches to proteomic data from COPDGene participants to uncover protein networks associated with COPD phenotypes. We further identify genetic associations with networks. This work discovers protein networks containing known and novel proteins and protein interactions associated with clinically relevant COPD phenotypes across race groups and cohorts.
Collapse
Affiliation(s)
- Iain R Konigsberg
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Thao Vu
- Department of Biostatistics and Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Weixuan Liu
- Department of Biostatistics and Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Elizabeth M Litkowski
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
- Department of Medicine, University of Michigan, Ann Arbor, MI
| | | | - Luciana B Vargas
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Niles Gilmore
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Mohamed Abdel-Hafiz
- Department of Computer Science and Engineering, University of Colorado - Denver, Denver, CO
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Craig P Hersh
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dawn L DeMeo
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | - Leslie A Lange
- Department of Biomedical Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| | - Katerina J Kechris
- Department of Biostatistics and Informatics, University of Colorado - Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
22
|
Choe J, Choi HY, Lee SM, Oh SY, Hwang HJ, Kim N, Yun J, Lee JS, Oh YM, Yu D, Kim B, Seo JB. Evaluation of retrieval accuracy and visual similarity in content-based image retrieval of chest CT for obstructive lung disease. Sci Rep 2024; 14:4587. [PMID: 38403628 PMCID: PMC10894863 DOI: 10.1038/s41598-024-54954-5] [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: 06/17/2023] [Accepted: 02/19/2024] [Indexed: 02/27/2024] Open
Abstract
The aim of our study was to assess the performance of content-based image retrieval (CBIR) for similar chest computed tomography (CT) in obstructive lung disease. This retrospective study included patients with obstructive lung disease who underwent volumetric chest CT scans. The CBIR database included 600 chest CT scans from 541 patients. To assess the system performance, follow-up chest CT scans of 50 patients were evaluated as query cases, which showed the stability of the CT findings between baseline and follow-up chest CT, as confirmed by thoracic radiologists. The CBIR system retrieved the top five similar CT scans for each query case from the database by quantifying and comparing emphysema extent and size, airway wall thickness, and peripheral pulmonary vasculatures in descending order from the database. The rates of retrieval of the same pairs of query CT scans in the top 1-5 retrievals were assessed. Two expert chest radiologists evaluated the visual similarities between the query and retrieved CT scans using a five-point scale grading system. The rates of retrieving the same pairs of query CTs were 60.0% (30/50) and 68.0% (34/50) for top-three and top-five retrievals. Radiologists rated 64.8% (95% confidence interval 58.8-70.4) of the retrieved CT scans with a visual similarity score of four or five and at least one case scored five points in 74% (74/100) of all query cases. The proposed CBIR system for obstructive lung disease integrating quantitative CT measures demonstrated potential for retrieving chest CT scans with similar imaging phenotypes. Further refinement and validation in this field would be valuable.
Collapse
Affiliation(s)
- Jooae Choe
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea
| | - Hye Young Choi
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea
- Department of Radiology, Kyung Hee University Hospital at Gangdong, College of Medicine Kyung, Hee University, Seoul, Korea
| | - Sang Min Lee
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea.
| | - Sang Young Oh
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea
| | - Hye Jeon Hwang
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea
| | - Namkug Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea
- Department of Convergence Medicine, Biomedical Engineering Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jihye Yun
- Department of Convergence Medicine, Biomedical Engineering Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Seung Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | - Joon Beom Seo
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, 86 Asanbyeongwon-Gil, Songpa-Gu, 05505, Seoul, Korea
| |
Collapse
|
23
|
Papi A, Faner R, Pavord I, Baraldi F, McDonald VM, Thomas M, Miravitlles M, Roche N, Agustí A. From treatable traits to GETomics in airway disease: moving towards clinical practice. Eur Respir Rev 2024; 33:230143. [PMID: 38232989 DOI: 10.1183/16000617.0143-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/24/2023] [Indexed: 01/19/2024] Open
Abstract
The treatable traits approach represents a strategy for patient management. It is based on the identification of characteristics susceptible to treatments or predictive of treatment response in each individual patient. With the objective of accelerating progress in research and clinical practice relating to such a treatable traits approach, the Portraits event was convened in Barcelona, Spain, in November 2022. Here, while reporting the key concepts that emerged from the discussions during the meeting, we review the current state of the art related to treatable traits and chronic respiratory diseases management, and we describe the possible actions that clinicians can take in clinical practice to implement the treatable traits framework. Furthermore, we explore the new concept of GETomics and the new models of research in the field of COPD.
Collapse
Affiliation(s)
- Alberto Papi
- Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Rosa Faner
- University of Barcelona, Biomedicine Department, FCRB-IDIBAPS, Centro de Investigación Biomedica en Red M.P. (CIBER), Barcelona, Spain
| | - Ian Pavord
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Vanessa M McDonald
- School of Nursing and Midwifery, The University of Newcastle, NHMRC Centre of Excellence in Asthma Treatable Traits, Hunter Medical Research Institute Asthma and Breathing Research Programme and Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Mike Thomas
- Primary Care and Population Sciences, University of Southampton, Southampton, UK
| | - Marc Miravitlles
- Pneumology Department Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Nicholas Roche
- Respiratory Medicine Department, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, AP-HP and Université Paris Cité, Paris, France
| | - Alvar Agustí
- University of Barcelona, Hospital Clinic, IDIBAPS and CIBERES, Barcelona, Spain
- Pulmonary Service, Respiratory Institute, Clinic Barcelona, Barcelona, Spain
| |
Collapse
|
24
|
Ware SA, Kliment CR, Giordano L, Redding KM, Rumsey WL, Bates S, Zhang Y, Sciurba FC, Nouraie SM, Kaufman BA. Cell-free DNA levels associate with COPD exacerbations and mortality. Respir Res 2024; 25:42. [PMID: 38238743 PMCID: PMC10797855 DOI: 10.1186/s12931-023-02658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/26/2023] [Indexed: 01/22/2024] Open
Abstract
THE QUESTION ADDRESSED BY THE STUDY Good biological indicators capable of predicting chronic obstructive pulmonary disease (COPD) phenotypes and clinical trajectories are lacking. Because nuclear and mitochondrial genomes are damaged and released by cigarette smoke exposure, plasma cell-free mitochondrial and nuclear DNA (cf-mtDNA and cf-nDNA) levels could potentially integrate disease physiology and clinical phenotypes in COPD. This study aimed to determine whether plasma cf-mtDNA and cf-nDNA levels are associated with COPD disease severity, exacerbations, and mortality risk. MATERIALS AND METHODS We quantified mtDNA and nDNA copy numbers in plasma from participants enrolled in the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE, n = 2,702) study and determined associations with relevant clinical parameters. RESULTS Of the 2,128 participants with COPD, 65% were male and the median age was 64 (interquartile range, 59-69) years. During the baseline visit, cf-mtDNA levels positively correlated with future exacerbation rates in subjects with mild/moderate and severe disease (Global Initiative for Obstructive Lung Disease [GOLD] I/II and III, respectively) or with high eosinophil count (≥ 300). cf-nDNA positively associated with an increased mortality risk (hazard ratio, 1.33 [95% confidence interval, 1.01-1.74] per each natural log of cf-nDNA copy number). Additional analysis revealed that individuals with low cf-mtDNA and high cf-nDNA abundance further increased the mortality risk (hazard ratio, 1.62 [95% confidence interval, 1.16-2.25] per each natural log of cf-nDNA copy number). ANSWER TO THE QUESTION Plasma cf-mtDNA and cf-nDNA, when integrated into quantitative clinical measurements, may aid in improving COPD severity and progression assessment.
Collapse
Affiliation(s)
- Sarah A Ware
- Department of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street BST W1044, Pittsburgh, PA, 15261, USA
| | - Corrine R Kliment
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Luca Giordano
- Department of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street BST W1044, Pittsburgh, PA, 15261, USA
| | - Kevin M Redding
- Department of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street BST W1044, Pittsburgh, PA, 15261, USA
| | - William L Rumsey
- GlaxoSmithKline Respiratory Therapeutic Area Unit, Collegeville, PA, USA
| | - Stewart Bates
- GlaxoSmithKline Respiratory Therapeutic Area Unit, Stevenage, UK
| | - Yingze Zhang
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Frank C Sciurba
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - S Mehdi Nouraie
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- UPMC Montefiore Hospital, NW628 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA.
| | - Brett A Kaufman
- Department of Medicine, Division of Cardiology, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine, 200 Lothrop Street BST W1044, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
25
|
Palazzo A, Makulyte G, Goerhig D, Médard JJ, Gros V, Trottein F, Adnot S, Vindrieux D, Flaman JM, Bernard D. Benidipine calcium channel blocker promotes the death of cigarette smoke-induced senescent cells and improves lung emphysema. Aging (Albany NY) 2023; 15:13581-13592. [PMID: 38095616 PMCID: PMC10756105 DOI: 10.18632/aging.205259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/03/2023] [Indexed: 12/21/2023]
Abstract
Smoking is the main risk factor for many lung diseases including chronic obstructive pulmonary disease. Cigarette smoke (CS) contains carcinogenic and reactive oxygen species that favor DNA mutations and perturb the homeostasis and environment of cells. CS induces lung cell senescence resulting in a stable proliferation arrest and a senescence-associated secretory phenotype. It was recently reported that senescent cell accumulation promotes several lung diseases. In this study, we performed a chemical screen, using an FDA-approved drug library, to identify compounds selectively promoting the death of CS-induced senescent lung cells. Aside from the well-known senolytic, ABT-263, we identified other potentially new senescence-eliminating compounds, including a new class of molecules, the dihydropyridine family of calcium voltage-gated channel (CaV) blockers. Among these blockers, Benidipine, decreased senescent lung cells and ameliorates lung emphysema in a mouse model. The dihydropyridine family of CaV blockers thus constitutes a new class of senolytics that could improve lung diseases. Hence, our work paves the way for further studies on the senolytic activity of CaV blockers in different senescence contexts and age-related diseases.
Collapse
Affiliation(s)
- Alberta Palazzo
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Gabriela Makulyte
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Delphine Goerhig
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Jean-Jacques Médard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Vincent Gros
- Université Paris Est Créteil, INSERM U955, IMRB, Créteil F-94010, France
- AP-HP, Hôpital Henri Mondor, Département de Physiologie-Explorations Fonctionnelles and FHU Senec, Créteil F-94010, France
| | - François Trottein
- Université Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille F-59000, France
| | - Serge Adnot
- Université Paris Est Créteil, INSERM U955, IMRB, Créteil F-94010, France
- AP-HP, Hôpital Henri Mondor, Département de Physiologie-Explorations Fonctionnelles and FHU Senec, Créteil F-94010, France
| | - David Vindrieux
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - Jean-Michel Flaman
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Centre Léon Bérard, Université de Lyon, Lyon, France
- Equipe Labellisée la Ligue Contre le Cancer, Lyon, France
| |
Collapse
|
26
|
Anandan J, Dwivedi DP, Govindaraj V. Clinical phenotypes of COPD and their impact on quality of life: A cross-sectional study. Respir Med 2023; 220:107452. [PMID: 37944828 DOI: 10.1016/j.rmed.2023.107452] [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: 04/28/2023] [Revised: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND A Chronic Obstructive Pulmonary Disease (COPD) phenotype is a single or group of disease characteristics that describe differences between individuals based on clinically important factors such as symptoms, exacerbations, morbidity, and treatment responses. Many studies estimated the prevalence of various phenotypes, but very few studies looked into their quality of life. We aimed to estimate the prevalence of different COPD phenotypes and their disease-specific Health-Related Quality of Life (HRQoL). MATERIALS AND METHODS The prospective study, with a sample size of 136, was conducted between May 2021 and December 2022 in a tertiary teaching institute. Based on their clinical features, COPD patients were classified into 4 different clinical phenotypes, and their disease-specific quality of life was assessed using St. George Respiratory Questionnaire-COPD(SGRQ-c) and COPD Assessment Test (CAT) questionnaires. RESULTS Among 136 COPD patients, the frequency of Non-Exacerbator (NE), Exacerbator Emphysema (EEM), Exacerbator Chronic Bronchitis (ECB), and Asthma COPD overlap (ACO) phenotypes was 79(58.1 %), 16(11.8 %), 31(22.8 %), and 10(7.4 %) respectively. Based on the SGRQ-c score, the ECB and EEM phenotypes had a significantly poorer Quality of life (QoL) when compared with NE(P<0.0001), ACO(P=0.011), phenotypes. Similarly, ECB and EEM phenotypes had significantly poorer QoL when compared to NE(P<0.0001), and ACO(P=0.015), based on the CAT score. ECB and EEM patients also had the worst scores in all individual CAT items and SGRQ-c components. CONCLUSION NE was the most common followed by ECB phenotype. ECB and EEM phenotypes recorded the poorest quality of life without any significant differences among them. Further research is needed in the future to determine whether phenotype-specific therapies can produce better clinical outcomes.
Collapse
Affiliation(s)
- Jeevanandham Anandan
- Department of Pulmonary Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India.
| | - Dharm Prakash Dwivedi
- Department of Pulmonary Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India.
| | - Vishnukanth Govindaraj
- Department of Pulmonary Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India.
| |
Collapse
|
27
|
Bell AJ, Pal R, Labaki WW, Hoff BA, Wang JM, Murray S, Kazerooni EA, Galban S, Lynch DA, Humphries SM, Martinez FJ, Hatt CR, Han MK, Ram S, Galban CJ. Quantitative CT of Normal Lung Parenchyma and Small Airways Disease Topologies are Associated With COPD Severity and Progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.26.23290532. [PMID: 37333382 PMCID: PMC10274970 DOI: 10.1101/2023.05.26.23290532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Objectives Small airways disease (SAD) is a major cause of airflow obstruction in COPD patients, and has been identified as a precursor to emphysema. Although the amount of SAD in the lungs can be quantified using our Parametric Response Mapping (PRM) approach, the full breadth of this readout as a measure of emphysema and COPD progression has yet to be explored. We evaluated topological features of PRM-derived normal parenchyma and SAD as surrogates of emphysema and predictors of spirometric decline. Materials and Methods PRM metrics of normal lung (PRMNorm) and functional SAD (PRMfSAD) were generated from CT scans collected as part of the COPDGene study (n=8956). Volume density (V) and Euler-Poincaré Characteristic (χ) image maps, measures of the extent and coalescence of pocket formations (i.e., topologies), respectively, were determined for both PRMNorm and PRMfSAD. Association with COPD severity, emphysema, and spirometric measures were assessed via multivariable regression models. Readouts were evaluated as inputs for predicting FEV1 decline using a machine learning model. Results Multivariable cross-sectional analysis of COPD subjects showed that V and χ measures for PRMfSAD and PRMNorm were independently associated with the amount of emphysema. Readouts χfSAD (β of 0.106, p<0.001) and VfSAD (β of 0.065, p=0.004) were also independently associated with FEV1% predicted. The machine learning model using PRM topologies as inputs predicted FEV1 decline over five years with an AUC of 0.69. Conclusions We demonstrated that V and χ of fSAD and Norm have independent value when associated with lung function and emphysema. In addition, we demonstrated that these readouts are predictive of spirometric decline when used as inputs in a ML model. Our topological PRM approach using PRMfSAD and PRMNorm may show promise as an early indicator of emphysema onset and COPD progression.
Collapse
Affiliation(s)
- Alexander J. Bell
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Ravi Pal
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Wassim W. Labaki
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Benjamin A. Hoff
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer M. Wang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Susan Murray
- School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Ella A. Kazerooni
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Stefanie Galban
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - David A. Lynch
- Department of Radiology, National Jewish Health, Denver, CO, United States
| | | | | | | | - MeiLan K. Han
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Sundaresh Ram
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Craig J. Galban
- Department of Radiology, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
28
|
Koblizek V, Milenkovic B, Svoboda M, Kocianova J, Holub S, Zindr V, Ilic M, Jankovic J, Cupurdija V, Jarkovsky J, Popov B, Valipour A. RETRO-POPE: A Retrospective, Multicenter, Real-World Study of All-Cause Mortality in COPD. Int J Chron Obstruct Pulmon Dis 2023; 18:2661-2672. [PMID: 38022829 PMCID: PMC10661906 DOI: 10.2147/copd.s426919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose The Phenotypes of COPD in Central and Eastern Europe (POPE) study assessed the prevalence and clinical characteristics of four clinical COPD phenotypes, but not mortality. This retrospective analysis of the POPE study (RETRO-POPE) investigated the relationship between all-cause mortality and patient characteristics using two grouping methods: clinical phenotyping (as in POPE) and Burgel clustering, to better identify high-risk patients. Patients and Methods The two largest POPE study patient cohorts (Czech Republic and Serbia) were categorized into one of four clinical phenotypes (acute exacerbators [with/without chronic bronchitis], non-exacerbators, asthma-COPD overlap), and one of five Burgel clusters based on comorbidities, lung function, age, body mass index (BMI) and dyspnea (very severe comorbid, very severe respiratory, moderate-to-severe respiratory, moderate-to-severe comorbid/obese, and mild respiratory). Patients were followed-up for approximately 7 years for survival status. Results Overall, 801 of 1,003 screened patients had sufficient data for analysis. Of these, 440 patients (54.9%) were alive and 361 (45.1%) had died at the end of follow-up. Analysis of survival by clinical phenotype showed no significant differences between the phenotypes (P=0.211). However, Burgel clustering demonstrated significant differences in survival between clusters (P<0.001), with patients in the "very severe comorbid" and "very severe respiratory" clusters most likely to die. Overall survival was not significantly different between Serbia and the Czech Republic after adjustment for age, BMI, comorbidities and forced expiratory volume in 1 second (hazard ratio [HR] 0.80, 95% confidence interval [CI] 0.65-0.99; P=0.036 [unadjusted]; HR 0.88, 95% CI 0.7-1.1; P=0.257 [adjusted]). The most common causes of death were respiratory-related (36.8%), followed by cardiovascular (25.2%) then neoplasm (15.2%). Conclusion Patient clusters based on comorbidities, lung function, age, BMI and dyspnea were more likely to show differences in COPD mortality risk than phenotypes defined by exacerbation history and presence/absence of chronic bronchitis and/or asthmatic features.
Collapse
Affiliation(s)
- Vladimir Koblizek
- Department of Pneumology, University Hospital, Hradec Kralove, Czech Republic
- Faculty of Medicine Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Branislava Milenkovic
- Clinic for Pulmonary Diseases, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Michal Svoboda
- Institute of Biostatistics and Analyses Ltd., Brno, Czech Republic
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jana Kocianova
- Outpatient Department of Pneumology Alveolus, APRO MED, Ostrava, Czech Republic
| | - Stanislav Holub
- Outpatient Chest Clinic, Plicni Stredisko Teplice Ltd., Teplice, Czech Republic
| | - Vladimir Zindr
- Outpatient Chest Clinic, PNEUMO KV Ltd., Karlovy Vary, Czech Republic
| | - Miroslav Ilic
- Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
- Clinic for Tuberculosis and Interstitial Lung Diseases, PolyClinic Department, Institute for Pulmonary Diseases of Vojvodina, Sremska Kamenica, Serbia
| | - Jelena Jankovic
- Clinic for Pulmonary Diseases, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vojislav Cupurdija
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Clinic for Pulmonology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Jiri Jarkovsky
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Boris Popov
- Medicine Department, Boehringer Ingelheim Serbia d.o.o. Beograd, Belgrade, Serbia
| | - Arschang Valipour
- Karl Landsteiner Institute for Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna Health Care Group, Vienna, Austria
| |
Collapse
|
29
|
Liu G, Haw TJ, Starkey MR, Philp AM, Pavlidis S, Nalkurthi C, Nair PM, Gomez HM, Hanish I, Hsu AC, Hortle E, Pickles S, Rojas-Quintero J, Estepar RSJ, Marshall JE, Kim RY, Collison AM, Mattes J, Idrees S, Faiz A, Hansbro NG, Fukui R, Murakami Y, Cheng HS, Tan NS, Chotirmall SH, Horvat JC, Foster PS, Oliver BG, Polverino F, Ieni A, Monaco F, Caramori G, Sohal SS, Bracke KR, Wark PA, Adcock IM, Miyake K, Sin DD, Hansbro PM. TLR7 promotes smoke-induced experimental lung damage through the activity of mast cell tryptase. Nat Commun 2023; 14:7349. [PMID: 37963864 PMCID: PMC10646046 DOI: 10.1038/s41467-023-42913-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/25/2023] [Indexed: 11/16/2023] Open
Abstract
Toll-like receptor 7 (TLR7) is known for eliciting immunity against single-stranded RNA viruses, and is increased in both human and cigarette smoke (CS)-induced, experimental chronic obstructive pulmonary disease (COPD). Here we show that the severity of CS-induced emphysema and COPD is reduced in TLR7-deficient mice, while inhalation of imiquimod, a TLR7-agonist, induces emphysema without CS exposure. This imiquimod-induced emphysema is reduced in mice deficient in mast cell protease-6, or when wild-type mice are treated with the mast cell stabilizer, cromolyn. Furthermore, therapeutic treatment with anti-TLR7 monoclonal antibody suppresses CS-induced emphysema, experimental COPD and accumulation of pulmonary mast cells in mice. Lastly, TLR7 mRNA is increased in pre-existing datasets from patients with COPD, while TLR7+ mast cells are increased in COPD lungs and associated with severity of COPD. Our results thus support roles for TLR7 in mediating emphysema and COPD through mast cell activity, and may implicate TLR7 as a potential therapeutic target.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Tatt Jhong Haw
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Malcolm R Starkey
- Depatrment of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Stelios Pavlidis
- The Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Christina Nalkurthi
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Prema M Nair
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Henry M Gomez
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Irwan Hanish
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Alan Cy Hsu
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Elinor Hortle
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Sophie Pickles
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | | | - Raul San Jose Estepar
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Richard Y Kim
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Adam M Collison
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Joerg Mattes
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Sobia Idrees
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Alen Faiz
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Musashino University, Nishitokyo-shi, Tokyo, Japan
| | - Hong Sheng Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sanjay H Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Respiratory and Critical Care Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jay C Horvat
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Paul S Foster
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Brian Gg Oliver
- Woolcock Institute of Medical Research, University of Sydney & School of Life Sciences, University of Technology, Sydney, Australia
| | | | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Section of Anatomic Pathology, Università di Messina, Messina, Italy
| | - Francesco Monaco
- Thoracic Surgery, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento BIOMORF and Dipartimento di Medicina e Chirurgia, Universities of Messina and Parma, Messina, Italy
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Peter A Wark
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia
| | - Ian M Adcock
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare clinical campus, UNSW, Sydney, Australia
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minatoku, Tokyo, Japan
| | - Don D Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital & Respiratory Division, Dept of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, and Faculty of Science, University of Technology Sydney, Camperdown, New South Wales, Australia.
- Immune Healthy &/or Grow Up Well, Hunter Medical Research Institute & University of Newcastle, Callaghan, New South Wales, Australia.
| |
Collapse
|
30
|
Wang J, Xia B, Ma R, Ye Q. Comprehensive Analysis of a Competing Endogenous RNA Co-Expression Network in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:2417-2429. [PMID: 37955025 PMCID: PMC10637225 DOI: 10.2147/copd.s431041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is the main cause of mortality world widely. Non-coding RNAs (lncRNAs) and associated competitive endogenous RNAs (ceRNAs) networks were recently proved to lead to mRNA gene expression downregulation but were still unclear in COPD. This study aims to investigate and elucidate the mechanisms underlying the involvement of ceRNA co-expression networks in COPD pathogenesis. Methods Obtained expression signature of data from the Gene Expression Omnibus database and compared the differentially expression of mRNAs and miRNAs between COPD patients and healthy smokers. Predicted the miRNA-lncRNA and miRNA-mRNA interaction using online library and employed CIBERSORT to measure the proportions of the 22 immune cells in the COPD and control groups. Results Established a ceRNA-network comprising 11 lncRNAs, 5 miRNAs, and 16 mRNAs. Using the weighted correlation network analysis method, we identified hub genes and hub miRNAs and obtained one core sub-network, XIST, FGD5-AS1, KCNQ1OT1, HOXA11-AS, LINC00667, H19, PRKCQ-AS1, NUTM2A-AS1/has-mir-454-3p/ZNF678, PRRG4. COPD patients had different proportions of immune cells than controls, and these variations were associated with the magnitude of pulmonary function parameters. Conclusion The ceRNA-network, particularly the core sub-network, may be a putative goal for COPD, in which specific immune cells were involved.
Collapse
Affiliation(s)
- Jingwei Wang
- Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Bowen Xia
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Ruimin Ma
- Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Qiao Ye
- Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
- Department of Occupational Medicine and Toxicology, Clinical Center for Interstitial Lung Diseases, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
31
|
Singh G, Acharya S, Shukla S, Jain D. Muco-Obstructive Lung Disease: A Systematic Review. Cureus 2023; 15:e46866. [PMID: 37954759 PMCID: PMC10637992 DOI: 10.7759/cureus.46866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Muco-obstructive lung disease is a new classification under the diseases of respiratory tract. A lot of discussion is still going on regarding this new group of diseases. It is characterised by obstruction of the respiratory tract with a thick mucin layer. Usually in normal individuals, the mucus is swept out of the respiratory system while coughing in the form of sputum or phlegm, but if the consistency of the mucus is thick, or the amount is heavy or there is a certain defect in the ciliary function of the respiratory tract, the mucus is not cleared and it gets accumulated in the lungs alveoli, therefore blocking it. The mucus trapped in the distal airways cannot be cleared by coughing therefore forming a layer in the alveoli and bronchioles. Long-standing condition causes inflammation and infection. This new group of diseases specifically includes chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), primary ciliary dyskinesia (PCD) and non-cystic fibrosis bronchiectasis (NCFB). Asthma, although an obstructive disease of the lung, is not particularly included under muco-obstructive lung disease. The major symptoms with which these diseases present are sputum production, chronic cough and acute exacerbations of the condition. The mucus adheres to the lung parenchyma causing airway obstruction and hyperinflation. In this article, we will see how muco-obstructive lung diseases affect the normal physiology of the respiratory system and how is it different from other obstructive and restrictive lung diseases. We will individually look into all the four conditions that come under the category of muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Garima Singh
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sourya Acharya
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Samarth Shukla
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Dhriti Jain
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
32
|
Arnaert A, Ahmed A, Debe Z, Charbonneau S, Paul S. Telehealth nursing interventions for phenotypes of older adults with COPD: an exploratory study. Front Digit Health 2023; 5:1144075. [PMID: 37808916 PMCID: PMC10558261 DOI: 10.3389/fdgth.2023.1144075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/01/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Inconclusive results exist around the effectiveness of telemonitoring for patients with COPD, and studies recommended conducting subgroup analyses to identify patient phenotypes that could benefit from these services. This exploratory study investigated what type of COPD patients were receiving which type of telenursing interventions more frequently using the telemonitoring platform. Methods A sample of 36 older adults with COPD were receiving telenursing services for 12 months and were asked to answer five COPD-symptom related questions and submit their vital signs daily. Results Findings revealed two phenotypes of older adults for whom the frequency of telenursing calls and related interventions differed. Although no statistically significant differences were observed in participants' GOLD grades and hospitalizations, cluster one participants used their COPD action plan significantly more frequently, and were in frequent contact with the telenurse. Discussion It is paramount that further research is needed on the development of patient phenotypes who may benefit from telemonitoring.
Collapse
Affiliation(s)
- A. Arnaert
- Ingram School of Nursing, McGill University, Montreal, QC, Canada
| | - A.M.I. Ahmed
- Ingram School of Nursing, McGill University, Montreal, QC, Canada
| | - Z. Debe
- Ingram School of Nursing, McGill University, Montreal, QC, Canada
| | - S. Charbonneau
- Montreal West Island Integrated University Health and Social Service Centre, Montreal, QC, Canada
| | - S. Paul
- Ingram School of Nursing, McGill University, Montreal, QC, Canada
| |
Collapse
|
33
|
Lenoir A, Whittaker H, Gayle A, Jarvis D, Quint JK. Mortality in non-exacerbating COPD: a longitudinal analysis of UK primary care data. Thorax 2023; 78:904-911. [PMID: 36423926 DOI: 10.1136/thorax-2022-218724] [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: 01/17/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Non-exacerbating patients with chronic obstructive pulmonary disease (COPD) are a less studied phenotype. We investigated clinical characteristics, mortality rates and causes of death among non-exacerbating compared with exacerbating patients with COPD. METHODS We used data from the Clinical Practice Research Datalink, Hospital Episode Statistics and Office for National Statistics between 1 January 2004 and 31 December 2018. Ever smokers with a COPD diagnosis with minimum 3 years of baseline information were included. We compared overall using Cox regression and cause-specific mortality rates using competing risk analysis, adjusted for age, sex, deprivation, smoking status, body mass index, GOLD stage and comorbidities. Causes of death were identified using International Classification of Diseases-10 codes. RESULTS Among 67 516 patients, 17.3% did not exacerbate during the 3-year baseline period. Mean follow-up was 4 years. Non-exacerbators were more likely to be male (63.3% vs 52.4%, p<0.001) and less often had a history of asthma (33.9% vs 43.6%, p<0.001) or FEV1<50% predicted (23.7 vs 31.8%) compared with exacerbators. Adjusted HR for overall mortality in non-exacerbators compared with exacerbators was 0.62 (95% CI 0.56 to 0.70) in the first year of follow-up and 0.87 (95% CI 0.83 to 0.91) thereafter. Non-exacerbating patients with COPD died less of respiratory causes than exacerbators (29.2% vs 40.3%) and more of malignancies (29.4% vs 23.4%) and cardiovascular diseases (26.2% vs 22.9%). HRs for malignant and circulatory causes of death were increased after the first year of follow-up. DISCUSSION In this primary care cohort, non-exacerbators showed distinct clinical characteristics and lower mortality rates. Non-exacerbators were equally likely to die of respiratory, malignant or cardiovascular diseases.
Collapse
Affiliation(s)
- Alexandra Lenoir
- Department of Respiratory Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Gesundheitsamt Fürstenfeldbruck, Fürstenfeldbruck, Germany
| | - Hannah Whittaker
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Alicia Gayle
- National Heart and Lung Institute, Imperial College London, London, UK
- Epidemiology Department, AstraZeneca, Cambridge, UK
| | - Debbie Jarvis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jennifer K Quint
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
34
|
Xie W, Jacobs C, Charbonnier JP, Slebos DJ, van Ginneken B. Emphysema subtyping on thoracic computed tomography scans using deep neural networks. Sci Rep 2023; 13:14147. [PMID: 37644032 PMCID: PMC10465555 DOI: 10.1038/s41598-023-40116-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/04/2023] [Indexed: 08/31/2023] Open
Abstract
Accurate identification of emphysema subtypes and severity is crucial for effective management of COPD and the study of disease heterogeneity. Manual analysis of emphysema subtypes and severity is laborious and subjective. To address this challenge, we present a deep learning-based approach for automating the Fleischner Society's visual score system for emphysema subtyping and severity analysis. We trained and evaluated our algorithm using 9650 subjects from the COPDGene study. Our algorithm achieved the predictive accuracy at 52%, outperforming a previously published method's accuracy of 45%. In addition, the agreement between the predicted scores of our method and the visual scores was good, where the previous method obtained only moderate agreement. Our approach employs a regression training strategy to generate categorical labels while simultaneously producing high-resolution localized activation maps for visualizing the network predictions. By leveraging these dense activation maps, our method possesses the capability to compute the percentage of emphysema involvement per lung in addition to categorical severity scores. Furthermore, the proposed method extends its predictive capabilities beyond centrilobular emphysema to include paraseptal emphysema subtypes.
Collapse
Affiliation(s)
- Weiyi Xie
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Colin Jacobs
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Dirk Jan Slebos
- Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bram van Ginneken
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
35
|
Çiftçi R, Kurtoğlu A, Eken Ö, Durmaz D, Eler S, Eler N, Aldhahi MI. Investigation of Factors Affecting Shuttle Walking Performance at Increased Speed for Patients with Chronic Obstructive Pulmonary Disease. J Clin Med 2023; 12:4752. [PMID: 37510866 PMCID: PMC10381562 DOI: 10.3390/jcm12144752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The aim of this study was to examine the factors affecting the shuttle walking test (SWT) in patients with chronic obstructive pulmonary disease (COPD). A total of 29 patients with COPD (the COPD group) and a healthy group (HG) of 34 women aged between 55 and 74 years were included in the study. After the pulmonary function profiles of the participants were assessed, and the SWT was performed. Walking distances, walking speeds, and SWT levels (SWT-L) were determined with the SWT. Before and after the SWT, the heart rate (HR), oxygen saturation level (SPO2), and Borg scale (perceived exertion (BSe) and dyspnea (BSd)) results were analyzed with a paired sample t-test. The dyspnea levels during activity of daily living were determined with the Medical Research Council (MRC) dyspnea scale, and the relationship between MRC dyspnea (MRCD) and walking distance, speed, and SWT-L was tested using multiple linear regression and Pearson correlation analysis. The walking distance, speed, and SWT-L were lower in the COPD group (p < 0.001) than in the HG. The HR values before and after the SWT changed significantly in the COPD group and the HG (p< 0.001), and the effect size was higher in the COPD group. Although the BSe and BSd results before and after the SWT in the COPD group increased significantly (p < 0.001), they did not change in the HG. There was a highly negative correlation between MRCD and walking distance, speed, and SWT-L in the COPD group (p = 0.002, p = 0.000, and p = 0.001, respectively), but no correlation was found in the HG. The results showed that the HR, perceived exertion, and dyspnea levels of women with COPD whose respiratory functions were lower than the HG were significantly affected on the SWT.
Collapse
Affiliation(s)
- Rukiye Çiftçi
- Department of Anatomy, Medical Faculty, Gaziantep Islamic Science and Technology University, Gaziantep 27260, Turkey;
| | - Ahmet Kurtoğlu
- Department of Coaching Education, Sport Science Faculty, Bandirma Onyedi Eylul University, Balikesir 10250, Turkey;
| | - Özgür Eken
- Department of Physical Education and Sport Teaching, Inonu University, Malatya 44000, Turkey;
| | - Dilber Durmaz
- Department of Thoracic Diseases, Balikesir, Medical Faculty, Bandirma Onyedi Eylul University, Bandırma 10250, Turkey;
| | - Serdar Eler
- Department of Coaching Education, Faculty of Sport Sciences, Gazi University, Ankara 06560, Turkey;
| | - Nebahat Eler
- Department of Coaching Education, School of Physical Education and Sports, Zonguldak Bulent Ecevit University, Zonguldak 67100, Turkey;
| | - Monira I. Aldhahi
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| |
Collapse
|
36
|
Curtis JL. Understanding COPD Etiology, Pathophysiology, and Definition. Respir Care 2023; 68:859-870. [PMID: 37353333 PMCID: PMC10289621 DOI: 10.4187/respcare.10873] [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] [Indexed: 06/25/2023]
Abstract
COPD, one of the leading worldwide health problems, currently lacks truly disease-modifying medical therapies applicable to most patients. Developing such novel therapies has been hampered by the marked heterogeneity of phenotypes between individuals with COPD. Such heterogeneity suggests that, rather than a single cause (particularly just direct inhalation of tobacco products), development and progression of COPD likely involve both complex gene-by-environment interactions to multiple inhalational exposures and a variety of molecular pathways. However, there has been considerable recent progress toward understanding how specific pathological processes can lead to discrete COPD phenotypes, particularly that of small airways disease. Advances in imaging techniques that correlate to specific types of histological damage, and in the immunological mechanisms of lung damage in COPD, hold promise for development of personalized therapies. At the same time, there is growing recognition that the current diagnostic criteria for COPD, based solely on spirometry, exclude large numbers of individuals with very similar disease manifestations. This concise review summarizes current understanding of the etiology and pathophysiology of COPD and provides background explaining the increasing calls to expand the diagnostic criteria used to diagnose COPD and some challenges in doing so.
Collapse
Affiliation(s)
- Jeffrey L Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan; Division of Pulmonary and Critical Care Medicine, Michigan Medicine, Ann Arbor, Michigan; and Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
37
|
Asai N, Ethridge AD, Fonseca W, Yagi K, Rasky AJ, Morris SB, Falkowski NR, Huang YJ, Huffnagle GB, Lukacs NW. A steroid-resistant cockroach allergen model is associated with lung and cecal microbiome changes. Physiol Rep 2023; 11:e15761. [PMID: 37403414 PMCID: PMC10320043 DOI: 10.14814/phy2.15761] [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: 03/23/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 07/06/2023] Open
Abstract
The pathogenesis of asthma has been partially linked to lung and gut microbiome. We utilized a steroid-resistant chronic model of cockroach antigen-induced (CRA) asthma with corticosteroid (fluticasone) treatment to examine lung and gut microbiome during disease. The pathophysiology assessment demonstrated that mucus and airway hyperresponsiveness were increased in the chronic CRA with no alteration in the fluticasone (Flut)-treated group, demonstrating steroid resistance. Analysis of mRNA from lungs showed no decrease of MUC5AC or Gob5 in the Flut-treated group. Furthermore, flow-cytometry in lung tissue showed eosinophils and neutrophils were not significantly reduced in the Flut-treated group compared to the chronic CRA group. When the microbiome profiles were assessed, data showed that only the Flut-treated animals were significantly different in the gut microbiome. Finally, a functional analysis of cecal microbiome metabolites using PiCRUSt showed several biosynthetic pathways were significantly enriched in the Flut-treated group, with tryptophan pathway verified by ELISA with increased kynurenine in homogenized cecum samples. While the implications of these data are unclear, they may suggest a significant impact of steroid treatment on future disease pathogenesis through microbiome and associated metabolite pathway changes.
Collapse
Affiliation(s)
- Nobuhiro Asai
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - Alexander D. Ethridge
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
- Immunology Graduate ProgramUniversity of MichiganAnn ArborMichiganUSA
| | - Wendy Fonseca
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - Kazuma Yagi
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - Andrew J. Rasky
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - Susan B. Morris
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
| | - Nicole R. Falkowski
- Division of Pulmonary and Critical Medicine, Department of MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Yvonne J. Huang
- Division of Pulmonary and Critical Medicine, Department of MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Gary B. Huffnagle
- Immunology Graduate ProgramUniversity of MichiganAnn ArborMichiganUSA
- Division of Pulmonary and Critical Medicine, Department of MedicineUniversity of MichiganAnn ArborMichiganUSA
- Mary H. Weiser Food Allergy CenterUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular, Cellular and Developmental BiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Nicholas W. Lukacs
- Department of PathologyUniversity of MichiganAnn ArborMichiganUSA
- Immunology Graduate ProgramUniversity of MichiganAnn ArborMichiganUSA
- Mary H. Weiser Food Allergy CenterUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
38
|
Zhang YH, Cho MH, Morrow JD, Castaldi PJ, Hersh CP, Midha MK, Hoopmann MR, Lutz SM, Moritz RL, Silverman EK. Integrating Genetics, Transcriptomics, and Proteomics in Lung Tissue to Investigate Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2023; 68:651-663. [PMID: 36780661 PMCID: PMC10257075 DOI: 10.1165/rcmb.2022-0302oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/13/2023] [Indexed: 02/15/2023] Open
Abstract
The integration of transcriptomic and proteomic data from lung tissue with chronic obstructive pulmonary disease (COPD)-associated genetic variants could provide insight into the biological mechanisms of COPD. Here, we assessed associations between lung transcriptomics and proteomics with COPD in 98 subjects from the Lung Tissue Research Consortium. Low correlations between transcriptomics and proteomics were generally observed, but higher correlations were found for COPD-associated proteins. We integrated COPD risk SNPs or SNPs near COPD-associated proteins with lung transcripts and proteins to identify regulatory cis-quantitative trait loci (QTLs). Significant expression QTLs (eQTLs) and protein QTLs (pQTLs) were found regulating multiple COPD-associated biomarkers. We investigated mediated associations from significant pQTLs through transcripts to protein levels of COPD-associated proteins. We also attempted to identify colocalized effects between COPD genome-wide association studies and eQTL and pQTL signals. Evidence was found for colocalization between COPD genome-wide association study signals and a pQTL for RHOB and an eQTL for DSP. We applied weighted gene co-expression network analysis to find consensus COPD-associated network modules. Two network modules generated by consensus weighted gene co-expression network analysis were associated with COPD with a false discovery rate lower than 0.05. One network module is related to the catenin complex, and the other module is related to plasma membrane components. In summary, multiple cis-acting determinants of transcripts and proteins associated with COPD were identified. Colocalization analysis, mediation analysis, and correlation-based network analysis of multiple omics data may identify key genes and proteins that work together to influence COPD pathogenesis.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- Channing Division of Network Medicine, Harvard Medical School, and
| | - Michael H. Cho
- Channing Division of Network Medicine, Harvard Medical School, and
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | | | | | - Craig P. Hersh
- Channing Division of Network Medicine, Harvard Medical School, and
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | | | | | - Sharon M. Lutz
- Channing Division of Network Medicine, Harvard Medical School, and
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| | | | - Edwin K. Silverman
- Channing Division of Network Medicine, Harvard Medical School, and
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts; and
| |
Collapse
|
39
|
Chen D, Zeng Q, Liu L, Zhou Z, Qi W, Yu S, Zhao L. Global Research Trends on the Link Between the Microbiome and COPD: A Bibliometric Analysis. Int J Chron Obstruct Pulmon Dis 2023; 18:765-783. [PMID: 37180751 PMCID: PMC10167978 DOI: 10.2147/copd.s405310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023] Open
Abstract
Background The pathogenesis of chronic obstructive pulmonary disease (COPD) has been studied in relation to the microbiome, providing space for more targeted interventions and new treatments. Numerous papers on the COPD microbiome have been reported in the last 10 years, yet few publications have used bibliometric methods to evaluate this area. Methods We searched the Web of Science Core Collection for all original research articles in the field of COPD microbiome from January 2011 to August 2022 and used CiteSpace for visual analysis. Results A total of 505 relevant publications were obtained, and the number of global publications in this field is steadily increasing every year, with China and the USA occupying the first two spots in international publications. Imperial College London and the University of Leicester produced the most publications. Brightling C from the UK was the most prolific writer, while Huang Y and Sze M from the USA were first and second among the authors cited. The American Journal of Respiratory and Critical Care Medicine had the highest frequency of citations. The top 10 institutions, cited authors and journals are mostly from the UK and the US. In the ranking of citations, the first article was a paper published by Sze M on changes in the lung tissue's microbiota in COPD patients. The keywords "exacerbation", "gut microbiota", "lung microbiome", "airway microbiome", "bacterial colonization", and "inflammation" were identified as cutting-edge research projects for 2011-2022. Conclusion Based on the visualization results, in the future, we can use the gut-lung axis as the starting point to explore the immunoinflammatory mechanism of COPD, and study how to predict the effects of different treatments of COPD by identifying the microbiota, and how to achieve the optimal enrichment of beneficial bacteria and the optimal consumption of harmful bacteria to improve COPD.
Collapse
Affiliation(s)
- Daohong Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Qian Zeng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Lu Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Ziyang Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wenchuan Qi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Shuguang Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
40
|
Abadi E, Jadick G, Lynch DA, Segars WP, Samei E. Emphysema Quantifications With CT Scan: Assessing the Effects of Acquisition Protocols and Imaging Parameters Using Virtual Imaging Trials. Chest 2023; 163:1084-1100. [PMID: 36462532 PMCID: PMC10206513 DOI: 10.1016/j.chest.2022.11.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 11/01/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND CT scan has notable potential to quantify the severity and progression of emphysema in patients. Such quantification should ideally reflect the true attributes and pathologic conditions of subjects, not scanner parameters. To achieve such an objective, the effects of the scanner conditions need to be understood so the influence can be mitigated. RESEARCH QUESTION How do CT scan imaging parameters affect the accuracy of emphysema-based quantifications and biomarkers? STUDY DESIGN AND METHODS Twenty anthropomorphic digital phantoms were developed with diverse anatomic attributes and emphysema abnormalities informed by a real COPD cohort. The phantoms were input to a validated CT scan simulator (DukeSim), modeling a commercial scanner (Siemens Flash). Virtual images were acquired under various clinical conditions of dose levels, tube current modulations (TCM), and reconstruction techniques and kernels. The images were analyzed to evaluate the effects of imaging parameters on the accuracy of density-based quantifications (percent of lung voxels with HU < -950 [LAA-950] and 15th percentile of lung histogram HU [Perc15]) across varied subjects. Paired t tests were performed to explore statistical differences between any two imaging conditions. RESULTS The most accurate imaging condition corresponded to the highest acquired dose (100 mAs) and iterative reconstruction (SAFIRE) with the smooth kernel of I31, where the measurement errors (difference between measurement and ground truth) were 35 ± 3 Hounsfield Units (HU), -4% ± 5%, and 26 ± 10 HU (average ± SD), for the mean lung HU, LAA-950, and Perc15, respectively. Without TCM and at the I31 kernel, increase of dose (20 to 100 mAs) improved the lung mean absolute error (MAE) by 4.2 ± 2.3 HU (average ± SD). TCM did not contribute to a systematic improvement of lung MAE. INTERPRETATION The results highlight that although CT scan quantification is possible, its reliability is impacted by the choice of imaging parameters. The developed virtual imaging trial platform in this study enables comprehensive evaluation of CT scan methods in reliable quantifications, an effort that cannot be readily made with patient images or simplistic physical phantoms.
Collapse
Affiliation(s)
- Ehsan Abadi
- Center for Virtual Imaging Trials, Carl E. Ravin Advanced Imaging Laboratories, Department of Radiology, Duke University School of Medicine, Durham, NC; Department of Electrical & Computer Engineering, Duke University, Durham, NC; Medical Physics Graduate Program, Duke University, Durham, NC.
| | - Giavanna Jadick
- Center for Virtual Imaging Trials, Carl E. Ravin Advanced Imaging Laboratories, Department of Radiology, Duke University School of Medicine, Durham, NC
| | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, CO
| | - W Paul Segars
- Center for Virtual Imaging Trials, Carl E. Ravin Advanced Imaging Laboratories, Department of Radiology, Duke University School of Medicine, Durham, NC; Medical Physics Graduate Program, Duke University, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC
| | - Ehsan Samei
- Center for Virtual Imaging Trials, Carl E. Ravin Advanced Imaging Laboratories, Department of Radiology, Duke University School of Medicine, Durham, NC; Department of Electrical & Computer Engineering, Duke University, Durham, NC; Medical Physics Graduate Program, Duke University, Durham, NC; Department of Biomedical Engineering, Duke University, Durham, NC; Department of Physics, Duke University, Durham, NC
| |
Collapse
|
41
|
Hobbs BD, Morrow JD, Wang XW, Liu YY, DeMeo DL, Hersh CP, Celli BR, Bueno R, Criner GJ, Silverman EK, Cho MH. Identifying chronic obstructive pulmonary disease from integrative omics and clustering in lung tissue. BMC Pulm Med 2023; 23:115. [PMID: 37041558 PMCID: PMC10091624 DOI: 10.1186/s12890-023-02389-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 03/15/2023] [Indexed: 04/13/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a highly morbid and heterogenous disease. While COPD is defined by spirometry, many COPD characteristics are seen in cigarette smokers with normal spirometry. The extent to which COPD and COPD heterogeneity is captured in omics of lung tissue is not known. METHODS We clustered gene expression and methylation data in 78 lung tissue samples from former smokers with normal lung function or severe COPD. We applied two integrative omics clustering methods: (1) Similarity Network Fusion (SNF) and (2) Entropy-Based Consensus Clustering (ECC). RESULTS SNF clusters were not significantly different by the percentage of COPD cases (48.8% vs. 68.6%, p = 0.13), though were different according to median forced expiratory volume in one second (FEV1) % predicted (82 vs. 31, p = 0.017). In contrast, the ECC clusters showed stronger evidence of separation by COPD case status (48.2% vs. 81.8%, p = 0.013) and similar stratification by median FEV1% predicted (82 vs. 30.5, p = 0.0059). ECC clusters using both gene expression and methylation were identical to the ECC clustering solution generated using methylation data alone. Both methods selected clusters with differentially expressed transcripts enriched for interleukin signaling and immunoregulatory interactions between lymphoid and non-lymphoid cells. CONCLUSIONS Unsupervised clustering analysis from integrated gene expression and methylation data in lung tissue resulted in clusters with modest concordance with COPD, though were enriched in pathways potentially contributing to COPD-related pathology and heterogeneity.
Collapse
Affiliation(s)
- Brian D Hobbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA.
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Jarrett D Morrow
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
| | - Xu-Wen Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bartolome R Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raphael Bueno
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Gerard J Criner
- Division of Pulmonary and Critical Care Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Ave, Rm 460, Boston, MA, 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Yang Y, Ge H, Lu J, Huang X, Wang K, Jin L, Qi L, Li M. Structural features on quantitative chest computed tomography of patients with maximal mid-expiratory flow impairment in a normal lung function population. BMC Pulm Med 2023; 23:86. [PMID: 36922831 PMCID: PMC10015933 DOI: 10.1186/s12890-023-02380-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 03/03/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND Maximal mid-expiratory flow (MMEF) is an earlier predictor of chronic obstructive pulmonary disease (COPD) development than forced expiratory volume in 1 s (FEV1). Changes of lung structure in patients with MMEF impairment only is still not clear. Therefore, this study aimed to investigate the structural features of patients with decreased MMEF by quantitative computed tomography (QCT) and develop a predictive model for predicting patients with reduced MMEF in normal lung function population. METHODS In this study, 131 patients with normal spirometry results and available volumetric chest CT images were enrolled and divided into the reduced MMEF group (FEV1/forced expiratory vital capacity (FEV1/FVC) > 0.7, FEV1% predictive values (FEV1%pred) > 80%, MMEF%pred < 80%, n = 52) and the normal MMEF group (FEV1/FVC > 0.7, FEV1%pred > 80%, MMEF%pred ≥ 80%, n = 79). The emphysema, small airway disease and medium-size airway parameters were measured by a commercial software. The differences were investigated in clinical features, spirometrical parameters and QCT parameters between the two groups. A nomogram model was constructed based on the results of the multivariable logistic regression model. Spearman's correlation coefficients were calculated between QCT measurements and spirometrical parameters. RESULTS There were more males in reduced MMEF group than normal group (P < 0.05). Lung parenchyma parameter (PRMEmph) and airway-related parameters (functional small airway disease (PRMfSAD), luminal area of fifth- and sixth- generation airway (LA5, LA6) were significantly different between the reduced MMEF group and the normal group (20.2 ± 17.4 vs 9.4 ± 6.7, 3.4 ± 3.5 vs 1.9 ± 2.0, 12.2 ± 2.5 vs 13.7 ± 3.4, 7.7 ± 2.4 vs 8.9 ± 2.8, respectively, all P < 0.01). After multivariable logistical regression, only sex (odds ratio [OR]: 2.777; 95% confidence interval [CI]:1.123-3.867), PRMfSAD (OR:1.102, 95%CI:1.045-1.162) and LA6 (OR:0.650, 95%CI:0.528-0.799) had significant differences between the two groups (P < 0.05) and a model incorporating with the three indicators was constructed (area under curve, 0.836). Correlation analysis showed MMEF%pred had mild to moderate correlation with airway-related measurements. CONCLUSION In normal lung function population, patients with reduced MMEF have potential medium-size and small airway changes, and MMEF%pred is significantly associated with airway-related CT parameters. The nomogram incorporating with sex, PRMfSAD and LA6 has good predictive value and offers more objective evidences in a group with reduced MMEF.
Collapse
Affiliation(s)
- Yuling Yang
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China
| | - Haiyan Ge
- Department of Respiratory Medicine, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China
| | - Jinjuan Lu
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China
| | - Xuemei Huang
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China
| | - Kun Wang
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China
| | - Liang Jin
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China
| | - Lin Qi
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China.
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated With Fudan University, No. 221 West Yanan Road, Shanghai, 200040, China.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW With the improvement in device technology and delivery methods of inhaled medications, along with development of novel compounds and recognition of the importance of personalized approach in the management of chronic airway diseases, nebulizers have not only maintained their place in the treatment hierarchy of airway disease but have also proven a vital platform for the development of new classes of drugs. RECENT FINDINGS This short review explores recent advances in nebulized drug delivery in chronic obstructive pulmonary disease and other chronic airway diseases, emphasizing the progress in nebulizer technology, physiologic advantages of nebulized drug delivery and the high versatility of currently available and developing nebulizer-delivered pharmacotherapies. SUMMARY Versatility and efficiency of nebulizers allows for a broad spectrum of existing and novel therapies to be clinically studied, facilitating the progress in phenotype-targeted pharmacotherapies in the management of chronic airway diseases.
Collapse
|
44
|
LeMaster WB, Quibrera PM, Couper D, Tashkin DP, Bleecker ER, Doerschuk CM, Ortega VE, Cooper C, Han MK, Woodruff PG, O'Neal WK, Anderson WH, Alexis NE, Bowler RP, Barr RG, Kaner RJ, Dransfield MT, Paine R, Kim V, Curtis JL, Martinez FJ, Hastie AT, Barjaktarevic I. Clinical Implications of Low Absolute Blood Eosinophil Count in the SPIROMICS COPD Cohort. Chest 2023; 163:515-528. [PMID: 36343688 PMCID: PMC10083128 DOI: 10.1016/j.chest.2022.10.029] [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: 05/12/2022] [Revised: 10/17/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The Global Initiative for Chronic Obstructive Lung Disease (GOLD) considers blood eosinophil counts < 100 cells/μL (BEC≤100) in people with COPD to predict poor inhaled corticosteroid (ICS) responsiveness. However, the BEC≤100 phenotype is inadequately characterized, especially in advanced COPD. RESEARCH QUESTION Are there differences between GOLD group D patients with high BEC and those with low BEC regarding baseline characteristics and longitudinal outcomes? STUDY DESIGN AND METHODS We used multivariable mixed models and logistic regression to contrast clinical characteristics and outcomes of BEC≤100 vs BEC > 100 (BEC100+) in all subjects with COPD (n = 1,414) and GOLD group D subjects (n = 185) not receiving ICS. RESULTS We identified n = 485 with BEC≤100 (n = 61 GOLD group D) and n = 929 people with BEC100+ (n = 124 GOLD group D). BEC≤100 status was stable at 6 weeks and approximately 52 weeks (intraclass correlations of 0.78 and 0.71, respectively). Compared with BEC100+, BEC≤100 comprised more women, with greater current smoking, and less frequent childhood asthma. Among all analyzed participants, the two BEC-defined subsets showed similar rates of lung function decline (mean slope, BEC≤100 vs BEC100+, -50 vs -39 mL/y; P = .140), exacerbations (0.40 vs 0.36/y; P = .098), subsequent ICS initiation (2.5% vs 4.4%; P = .071), and mortality (7.8% vs 8.4%; P = .715). However, in GOLD group D, people with BEC≤100 showed higher exacerbation rates within 365 days of enrollment (0.62 vs 0.33/y; P = .002) and total follow-up (1.16 vs 0.83/y; P = .014). They also had greater lung function decline (mean slope of -68 mL/y vs -23 mL/y; P = .036) and had greater emphysema at baseline (voxels < 950 Hounsfield units at total lung capacity of 7.46% vs 4.61%; P = .029). INTERPRETATION In non-ICS-treated GOLD group D COPD, people with BEC≤100 had more baseline emphysema, prospective exacerbations, and lung function decline. Our analysis has identified a particularly vulnerable subpopulation of people with COPD, suggesting the need for studies focused specifically on their therapeutic treatment. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; No.: NCT01969344; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- W Blake LeMaster
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | | | | | - Donald P Tashkin
- Division of Pulmonary and Critical Care Medicine, UCLA, Los Angeles, CA
| | | | | | - Victor E Ortega
- Division of Respiratory Medicine, Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ
| | | | - MeiLan K Han
- University of Michigan School of Medicine, Ann Arbor, MI
| | - Prescott G Woodruff
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | | | | | | | | | - R Graham Barr
- Presbyterian Hospital, Columbia University Medical Center, New York, NY
| | | | - Mark T Dransfield
- University of Alabama Birmingham and Birmingham VA Medical Center, Birmingham, AL
| | | | - Victor Kim
- Department of Thoracic Medicine and Surgery, Temple Lung Center, Philadelphia, PA
| | - Jeffrey L Curtis
- University of Michigan School of Medicine, Ann Arbor, MI; Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, MI
| | | | - Annette T Hastie
- Atrium Health Wake Forest Baptist, School of Medicine, Winston Salem, NC
| | - Igor Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, UCLA, Los Angeles, CA.
| |
Collapse
|
45
|
Valera-Novella E, Bernabeu-Mora R, Montilla-Herrador J, Escolar-Reina P, García-Vidal JA, Medina-Mirapeix F. Development of the ESEx index: a tool for predicting risk of recurrent severe COPD exacerbations. Ther Adv Chronic Dis 2023; 14:20406223231155115. [PMID: 38405221 PMCID: PMC10893840 DOI: 10.1177/20406223231155115] [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: 06/22/2022] [Accepted: 01/18/2023] [Indexed: 02/27/2024] Open
Abstract
Background In chronic obstructive pulmonary disease (COPD), multiple recurrent severe exacerbations that require hospitalization can occur. These events are strongly associated with death and other clinical complications. Objectives We aimed to develop a prognostic model that could identify patients with COPD that are at risk of multiple recurrent severe exacerbations within 3 years. Design Prospective cohort. Methods The derivation cohort comprised patients with stable, moderate-to-severe COPD. Multivariable logistic regression analyses were performed to develop the final model. Based on regression coefficients, a simplified index (ESEx) was established. Both, model and index, were assessed for predictive performance by measuring discrimination and calibration. Results Over 3 years, 16.4% of patients with COPD experienced at least three severe recurrent exacerbations. The prognostic model showed good discrimination of high-risk patients, based on three characteristics: the number of severe exacerbations in the previous year, performance in the five-repetition sit-to-stand test, and in the 6-minute-walk test. The ESEx index provided good level of discrimination [areas under the receiver operating characteristic curve (AUCs): 0.913]. Conclusions The ESEx index showed good internal validation for the identification of patients at risk of three recurrent severe COPD exacerbations within 3 years. These tools could be used to identify patients who require early interventions and motivate patients to improve physical performance to prevent recurrent exacerbations.
Collapse
Affiliation(s)
- Elisa Valera-Novella
- Department of Physical Therapy, University of Murcia, Murcia, Spain
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria Virgen de La Arrixaca (IMIB), Murcia, Spain
| | - Roberto Bernabeu-Mora
- Department of Pneumology, Hospital General Universitario Morales Meseguer, Adva. Marqués de los Vélez s/n, Murcia 30008, Spain
- Department of Internal Medicine, University of Murcia, Murcia, Spain
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Joaquina Montilla-Herrador
- Department of Physical Therapy, University of Murcia, Murcia, Spain
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria Virgen de La Arrixaca (IMIB), Murcia, Spain
| | - Pilar Escolar-Reina
- Department of Physical Therapy, University of Murcia, Murcia, Spain
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria Virgen de La Arrixaca (IMIB), Murcia, Spain
| | - José Antonio García-Vidal
- University of Murcia, Murcia, Spain
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria Virgen de La Arrixaca (IMIB), Murcia, Spain
| | - Francesc Medina-Mirapeix
- Department of Physical Therapy, University of Murcia, Murcia, Spain
- Research Group Fisioterapia y Discapacidad, Instituto Murciano de Investigación Biosanitaria Virgen de La Arrixaca (IMIB), Murcia, Spain
| |
Collapse
|
46
|
Agusti A, Ambrosino N, Blackstock F, Bourbeau J, Casaburi R, Celli B, Crouch R, Negro RD, Dreher M, Garvey C, Gerardi D, Goldstein R, Hanania N, Holland AE, Kaur A, Lareau S, Lindenauer PK, Mannino D, Make B, Maltais F, Marciniuk JD, Meek P, Morgan M, Pepin JL, Reardon JZ, Rochester C, Singh S, Spruit MA, Steiner MC, Troosters T, Vitacca M, Clini E, Jardim J, Nici L, Raskin J, ZuWallack R. COPD: Providing the right treatment for the right patient at the right time. Respir Med 2023; 207:107041. [PMID: 36610384 DOI: 10.1016/j.rmed.2022.107041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a common disease associated with significant morbidity and mortality that is both preventable and treatable. However, a major challenge in recognizing, preventing, and treating COPD is understanding its complexity. While COPD has historically been characterized as a disease defined by airflow limitation, we now understand it as a multi-component disease with many clinical phenotypes, systemic manifestations, and associated co-morbidities. Evidence is rapidly emerging in our understanding of the many factors that contribute to the pathogenesis of COPD and the identification of "early" or "pre-COPD" which should provide exciting opportunities for early treatment and disease modification. In addition to breakthroughs in our understanding of the origins of COPD, we are optimizing treatment strategies and delivery of care that are showing impressive benefits in patient-centered outcomes and healthcare utilization. This special issue of Respiratory Medicine, "COPD: Providing the Right Treatment for the Right Patient at the Right Time" is a summary of the proceedings of a conference held in Stresa, Italy in April 2022 that brought together international experts to discuss emerging evidence in COPD and Pulmonary Rehabilitation in honor of a distinguished friend and colleague, Claudio Ferdinando Donor (1948-2021). Claudio was a true pioneer in the field of pulmonary rehabilitation and the comprehensive care of individuals with COPD. He held numerous leadership roles in in the field, provide editorial stewardship of several respiratory journals, authored numerous papers, statement and guidelines in COPD and Pulmonary Rehabilitation, and provided mentorship to many in our field. Claudio's most impressive talent was his ability to organize spectacular conferences and symposia that highlighted cutting edge science and clinical medicine. It is in this spirit that this conference was conceived and planned. These proceedings are divided into 4 sections which highlight crucial areas in the field of COPD: (1) New concepts in COPD pathogenesis; (2) Enhancing outcomes in COPD; (3) Non-pharmacologic management of COPD; and (4) Optimizing delivery of care for COPD. These presentations summarize the newest evidence in the field and capture lively discussion on the exciting future of treating this prevalent and impactful disease. We thank each of the authors for their participation and applaud their efforts toward pushing the envelope in our understanding of COPD and optimizing care for these patients. We believe that this edition is a most fitting tribute to a dear colleague and friend and will prove useful to students, clinicians, and researchers as they continually strive to provide the right treatment for the right patient at the right time. It has been our pleasure and a distinct honor to serve as editors and oversee such wonderful scholarly work.
Collapse
Affiliation(s)
- Alvar Agusti
- Clinic Barcelona Hospital University, Barcelona, Spain.
| | | | | | - Jean Bourbeau
- Department of Medicine, Division of Experimental Medicine, McGill University Health Centre, Montreal, QC, CA, USA.
| | | | | | | | - Roberto Dal Negro
- National Centre for Pharmacoeconomics and Pharmacoepidemiology (CESFAR), Verona, Italy.
| | - Michael Dreher
- Clinic of Cardiology, Angiology, Pneumology and Intensive Medicine, University Hospital Aachen, Aachen, 52074, DE, USA.
| | | | | | - Roger Goldstein
- Respiratory Rehabilitation Service, West Park Health Care Centre, Toronto, Ontario, CA, USA.
| | | | - Anne E Holland
- Departments of Physiotherapy and Respiratory Medicine, Alfred Health, Melbourne, Australia; Central Clinical School, Monash University, Melbourne, Australia; Institute for Breathing and Sleep, Melbourne, Australia.
| | - Antarpreet Kaur
- Section of Pulmonary, Critical Care, and Sleep Medicine, Trinity Health of New England, Hartford, CT, USA; University of Colorado School of Nursing, Aurora, CO, USA.
| | - Suzanne Lareau
- University of Colorado School of Nursing, Aurora, CO, USA.
| | - Peter K Lindenauer
- Department of Healthcare Delivery and Population Sciences, University of Massachusetts Chan Medical School - Baystate, Springfield, MA, USA.
| | | | - Barry Make
- National Jewish Health, Denver, CO, USA.
| | - François Maltais
- Institut Universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec, CA, USA.
| | - Jeffrey D Marciniuk
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, CA, USA.
| | - Paula Meek
- University of Utah College of Nursing, Salt Lake City, UT, USA.
| | - Mike Morgan
- Dept of Respiratory Medicine, University Hospitals of Leicester, UK.
| | - Jean-Louis Pepin
- CHU de Grenoble - Clin Univ. de physiologie, sommeil et exercice, Grenoble, France.
| | - Jane Z Reardon
- Section of Pulmonary, Critical Care, and Sleep Medicine, Trinity Health of New England, Hartford, CT, USA.
| | | | - Sally Singh
- Department of Respiratory Diseases, University of Leicester, UK.
| | | | - Michael C Steiner
- Department of Respiratory Sciences, Leicester NIHR Biomedical Research Centre, Professor, University of Leicester, UK.
| | - Thierry Troosters
- Laboratory of Respiratory Diseases and Thoracic Surgery, KU Leuven: Leuven, Vlaanderen, Belgium.
| | - Michele Vitacca
- Department of Respiratory Rehabilitation, ICS S. Maugeri Care and Research Institutes, IRCCS Pavia, Italy.
| | - Enico Clini
- University of Modena and Reggio Emilia, Italy.
| | - Jose Jardim
- Federal University of Sao Paulo Paulista, Brazil.
| | - Linda Nici
- nBrown University School of Medicine, USA.
| | | | - Richard ZuWallack
- Section of Pulmonary, Critical Care, and Sleep Medicine, Saint Francis Hospital and Medical Center, 114 Woodland Street, Hartford, CT, 06105, USA.
| |
Collapse
|
47
|
Sonar M, Jayadeva BT, Shashibhushan B. Comparative Study of Systemic Inflammatory Markers in Clinical Phenotypes of Chronic Obstructive Pulmonary Disease. TANAFFOS 2023; 22:215-220. [PMID: 38628879 PMCID: PMC11016923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 02/01/2023] [Indexed: 04/19/2024]
Abstract
Background Chronic Obstructive Pulmonary Disease (COPD) is an inflammatory pulmonary disorder with systemic inflammatory manifestations. This study aims to identify the profile of systemic inflammatory markers in the different phenotypes of COPD to help predict the disease and identify suitable treatment options. Materials and Methods A prospective observational study was conducted on 92 patients with COPD admitted to Victoria Hospital, Bangalore between August 2021 to December 2021. Levels of C-reactive protein (CRP), Serum Creatinine, Erythrocyte Sedimentation Rate (ESR), Absolute Lymphocyte Count (ALC), Absolute Eosinophil Count (AEC), and Lactate Dehydrogenase (LDH) were measured within 48 hours of presentation. Results Significantly higher levels of CRP were found in frequent exacerbator emphysema and chronic bronchitis phenotypes (p=0.001). The frequent exacerbator emphysema phenotype had significantly higher levels of LDH (p=0.001) and serum creatinine (p=0.001). Not surprisingly, absolute eosinophil counts were significantly raised in the overlap COPD-Asthma phenotype (p=0.001). Conclusion Raised serum CRP levels in the frequent exacerbator phenotypes of emphysema and chronic bronchitis suggest a possible inflammatory response to an infective etiology. Raised LDH levels in frequent exacerbator emphysema phenotype could signify underlying lung parenchymal destruction. Systemic inflammation and oxidative stress can lead to skeletal muscle injury and atrophy in COPD patients. This may explain the raised serum creatinine levels in frequent exacerbator emphysema phenotype. Eosinophilia seen in Overlap COPD-Asthma phenotype is suggestive of type 2 inflammation of the airways with better response to steroids.
Collapse
Affiliation(s)
- Manjushree Sonar
- Department of Pulmonary Medicine, Bangalore Medical College and Research Institute, Bangalore, Karnataka, India
| | - Basavaraju Tejur Jayadeva
- Department of Pulmonary Medicine, Bangalore Medical College and Research Institute, Bangalore, Karnataka, India
| | - B.L. Shashibhushan
- Department of Pulmonary Medicine, Bangalore Medical College and Research Institute, Bangalore, Karnataka, India
| |
Collapse
|
48
|
Buschur KL, Riley C, Saferali A, Castaldi P, Zhang G, Aguet F, Ardlie KG, Durda P, Craig Johnson W, Kasela S, Liu Y, Manichaikul A, Rich SS, Rotter JI, Smith J, Taylor KD, Tracy RP, Lappalainen T, Graham Barr R, Sciurba F, Hersh CP, Benos PV. Distinct COPD subtypes in former smokers revealed by gene network perturbation analysis. Respir Res 2023; 24:30. [PMID: 36698131 PMCID: PMC9875487 DOI: 10.1186/s12931-023-02316-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/05/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) varies significantly in symptomatic and physiologic presentation. Identifying disease subtypes from molecular data, collected from easily accessible blood samples, can help stratify patients and guide disease management and treatment. METHODS Blood gene expression measured by RNA-sequencing in the COPDGene Study was analyzed using a network perturbation analysis method. Each COPD sample was compared against a learned reference gene network to determine the part that is deregulated. Gene deregulation values were used to cluster the disease samples. RESULTS The discovery set included 617 former smokers from COPDGene. Four distinct gene network subtypes are identified with significant differences in symptoms, exercise capacity and mortality. These clusters do not necessarily correspond with the levels of lung function impairment and are independently validated in two external cohorts: 769 former smokers from COPDGene and 431 former smokers in the Multi-Ethnic Study of Atherosclerosis (MESA). Additionally, we identify several genes that are significantly deregulated across these subtypes, including DSP and GSTM1, which have been previously associated with COPD through genome-wide association study (GWAS). CONCLUSIONS The identified subtypes differ in mortality and in their clinical and functional characteristics, underlining the need for multi-dimensional assessment potentially supplemented by selected markers of gene expression. The subtypes were consistent across cohorts and could be used for new patient stratification and disease prognosis.
Collapse
Affiliation(s)
- Kristina L Buschur
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Joint CMU-Pitt PhD Program in Computational Biology, Pittsburgh, PA, USA
- Division of General Medicine, Columbia University Medical Center, New York, NY, USA
- New York Genome Center, New York, NY, USA
| | - Craig Riley
- Division of Pulmonary Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Grace Zhang
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Francois Aguet
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - W Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Silva Kasela
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Josh Smith
- Northwest Genome Center, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Tuuli Lappalainen
- New York Genome Center, New York, NY, USA
- Department of Systems Biology, Columbia University, New York, NY, USA
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - R Graham Barr
- Division of General Medicine, Columbia University Medical Center, New York, NY, USA
| | - Frank Sciurba
- Division of Pulmonary Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Joint CMU-Pitt PhD Program in Computational Biology, Pittsburgh, PA, USA.
- Department of Epidemiology, University of Florida, 2004 Mowry Rd, Gainesville, FL, 32603, USA.
| |
Collapse
|
49
|
Hizawa N. The understanding of asthma pathogenesis in the era of precision medicine. Allergol Int 2023; 72:3-10. [PMID: 36195530 DOI: 10.1016/j.alit.2022.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 01/25/2023] Open
Abstract
Asthma is a syndrome with extremely diverse clinical phenotypes in which the onset, severity, and response to treatment are defined by the complex interplay of many genetic and environmental factors. Environmental factors epigenetically affect gene expression, and the disease is driven by a multidimensional dynamic network involving RNA and protein molecules derived from gene expression, as well as various metabolic products. In other words, specific pathophysiological mechanisms or endotypes are dynamic networks that arise in response to individual genotypes and the various environmental factors to which individuals have been exposed since before birth, such as diet, infection, air pollution, smoking, antibiotic use, and the bacterial flora of the intestinal tract, skin, and lungs. A key feature of asthma genome scans is their potential to reveal the molecular pathways that lead to pathogenesis. Endotypes that drive the disease have a significant impact on the phenotypes of asthma patients, including their drug responsiveness. Understanding endotypes will lead to not only the implementation of therapies that are tailored to the specific molecular network(s) underlying the patient's condition, but also to the development of therapeutic strategies that target individual endotypes, as well as to precision health, which will enable the prediction of disease onset with high accuracy from an early stage and the implementation of preventive strategies based on endotypes. Understanding of endotypes will pave the way for the practice of precision medicine in asthma care, moving away from 'one-size-fits-all' medicine and population-based prevention approaches that do not take individuals' susceptibility into account.
Collapse
Affiliation(s)
- Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
50
|
Nelson KE, Finlay M, Huang E, Chakravarti V, Feinstein JA, Diskin C, Thomson J, Mahant S, Widger K, Feudtner C, Cohen E. Clinical characteristics of children with severe neurologic impairment: A scoping review. J Hosp Med 2023; 18:65-77. [PMID: 36484088 PMCID: PMC9829450 DOI: 10.1002/jhm.13019] [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: 07/11/2022] [Revised: 10/21/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The aim of this study is to extrapolate the clinical features of children with severe neurologic impairment (SNI) based on the functional characteristics and comorbidities described in published studies. METHODS Four databases were searched. We included studies that describe clinical features of a group of children with SNI (≥20 subjects <19 years of age with >1 neurologic diagnosis and severe functional limitation) using data from caregivers, medical charts, or prospective collection. Studies that were not written in English were excluded. We extracted data about functional characteristics, comorbidities, and study topics. RESULTS We included 102 studies, spanning 5 continents over 43 years, using 41 distinct terms for SNI. The terms SNI and neurologic impairment (NI) were used in 59 studies (58%). Most studies (n = 81, 79%) described ≥3 types of functional characteristics, such as technology assistance and motor impairment. Studies noted 59 comorbidities and surgeries across 10 categories. The most common comorbidities were related to feeding, nutrition, and the gastrointestinal system, which were described in 79 studies (77%). Most comorbidities (76%) were noted in <10 studies. Studies investigated seven clinical topics, with "Gastrointestinal reflux and feeding tubes" as the most common research focus (n = 57, 56%). The next most common topic, "Aspiration and respiratory issues," included 13 studies (13%). Most studies (n = 54, 53%) were retrospective cohorts or case series; there were no clinical trials. CONCLUSIONS Despite the breadth of described comorbidities, studies focused on a narrow set of clinical topics. Further research is required to understand the prevalence, clinical impact, and interaction of the multiple comorbidities that are common in children with SNI.
Collapse
Affiliation(s)
- Katherine E Nelson
- Pediatric Advanced Care Team, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, Division of Paediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Institute for Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Melissa Finlay
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
| | - Emma Huang
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
| | - Vishakha Chakravarti
- Pediatric Advanced Care Team, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
| | - James A Feinstein
- Adult and Child Consortium for Health Outcomes Research and Delivery Science, University of Colorado and Children's Hospital Colorado, Aurora, Colorado, USA
| | - Catherine Diskin
- Department of Paediatrics, Division of Paediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joanna Thomson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Hospital Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sanjay Mahant
- Department of Paediatrics, Division of Paediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Institute for Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- CanChild Centre for Childhood Disability Research, McMaster University, Hamilton, Ontario, Canada
| | - Kimberley Widger
- Pediatric Advanced Care Team, Hospital for Sick Children, Toronto, Ontario, Canada
- Lawrence S. Bloomberg Faculty of Nursing, University of Toronto, Toronto, Ontario, Canada
| | - Chris Feudtner
- The Justin Michael Ingerman Center for Palliative Care, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Departments of Pediatrics and Medical Ethics and Health Policy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eyal Cohen
- Department of Paediatrics, Division of Paediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Ontario, Canada
- Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada
- Institute for Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Adult and Child Consortium for Health Outcomes Research and Delivery Science, University of Colorado and Children's Hospital Colorado, Aurora, Colorado, USA
- Edwin S.H. Leong Centre for Healthy Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|