1
|
Seyfinejad B, Nemutlu E, Taghizadieh A, Khoubnasabjafari M, Ozkan SA, Jouyban A. Biomarkers in exhaled breath condensate as fingerprints of asthma, chronic obstructive pulmonary disease and asthma-chronic obstructive pulmonary disease overlap: a critical review. Biomark Med 2023; 17:811-837. [PMID: 38179966 DOI: 10.2217/bmm-2023-0420] [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: 01/06/2024] Open
Abstract
Asthma, chronic obstructive pulmonary disease (COPD) and asthma-COPD overlap are the third leading cause of mortality around the world. They share some common features, which can lead to misdiagnosis. To properly manage these conditions, reliable markers for early and accurate diagnosis are needed. Over the past 20 years, many molecules have been investigated in the exhaled breath condensate to better understand inflammation pathways and mechanisms related to these disorders. Recently, more advanced techniques, such as sensitive metabolomic and proteomic profiling, have been used to obtain a more comprehensive understanding. This article reviews the use of targeted and untargeted metabolomic methodology to study asthma, COPD and asthma-COPD overlap.
Collapse
Affiliation(s)
- Behrouz Seyfinejad
- Pharmaceutical Analysis Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, 06100, Turkiye
| | - Ali Taghizadieh
- Tuberculosis & Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Internal Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Khoubnasabjafari
- Tuberculosis & Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anesthesiology & Intensive Care, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sibel A Ozkan
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, Ankara, 06560, Turkiye
| | - Abolghasem Jouyban
- Pharmaceutical Analysis Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Near East University, PO Box 99138 Nicosia, North Cyprus, Mersin 10, Turkiye
| |
Collapse
|
2
|
Merikallio H, Pincikova T, Kotortsi I, Karimi R, Li CX, Forsslund H, Mikko M, Nyrén S, Lappi-Blanco E, Wheelock ÅM, Kaarteenaho R, Sköld MC. Mucins 3A and 3B Are Expressed in the Epithelium of Human Large Airway. Int J Mol Sci 2023; 24:13546. [PMID: 37686350 PMCID: PMC10487631 DOI: 10.3390/ijms241713546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Aberrant mucus secretion is a hallmark of chronic obstructive pulmonary disease (COPD). Expression of the membrane-tethered mucins 3A and 3B (MUC3A, MUC3B) in human lung is largely unknown. In this observational cross-sectional study, we recruited subjects 45-65 years old from the general population of Stockholm, Sweden, during the years 2007-2011. Bronchial mucosal biopsies, bronchial brushings, and bronchoalveolar lavage fluid (BALF) were retrieved from COPD patients (n = 38), healthy never-smokers (n = 40), and smokers with normal lung function (n = 40). Protein expression of MUC3A and MUC3B in bronchial mucosal biopsies was assessed by immunohistochemical staining. In a subgroup of subjects (n = 28), MUC3A and MUC3B mRNAs were quantified in bronchial brushings using microarray. Non-parametric tests were used to perform correlation and group comparison analyses. A value of p < 0.05 was considered statistically significant. MUC3A and MUC3B immunohistochemical expression was localized to ciliated cells. MUC3B was also expressed in basal cells. MUC3A and MUC3B immunohistochemical expression was equal in all study groups but subjects with emphysema had higher MUC3A expression, compared to those without emphysema. Smokers had higher mRNA levels of MUC3A and MUC3B than non-smokers. MUC3A and MUC3B mRNA were higher in male subjects and correlated negatively with expiratory air flows. MUC3B mRNA correlated positively with total cell concentration and macrophage percentage, and negatively with CD4/CD8 T cell ratio in BALF. We concluded that MUC3A and MUC3B in large airways may be a marker of disease or may play a role in the pathophysiology of airway obstruction.
Collapse
Affiliation(s)
- Heta Merikallio
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, 90570 Oulu, Finland; (H.M.)
- Center of Internal Medicine and Respiratory Medicine, Medical Research Center Oulu, University Hospital of Oulu, 90220 Oulu, Finland
| | - Terezia Pincikova
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Stockholm CF-Center, Albatross, K56, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Ioanna Kotortsi
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Reza Karimi
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Chuan-Xing Li
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Helena Forsslund
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mikael Mikko
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Sven Nyrén
- Department of Molecular Medicine and Surgery, Division of Radiology, Karolinska Institutet, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden
| | - Elisa Lappi-Blanco
- Cancer and Translational Medicine Research Unit, Department of Pathology, University Hospital of Oulu, Oulu University, 90220 Oulu, Finland
| | - Åsa M. Wheelock
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Riitta Kaarteenaho
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, 90570 Oulu, Finland; (H.M.)
- Center of Internal Medicine and Respiratory Medicine, Medical Research Center Oulu, University Hospital of Oulu, 90220 Oulu, Finland
| | - Magnus C. Sköld
- Respiratory Medicine Unit, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 77 Stockholm, Sweden
| |
Collapse
|
3
|
Sun W, Cao Z, Ma Y, Wang J, Zhang L, Luo Z. Fibrinogen, a Promising Marker to Evaluate Severity and Prognosis of Acute Exacerbation of Chronic Obstructive Pulmonary Disease: A Retrospective Observational Study. Int J Chron Obstruct Pulmon Dis 2022; 17:1299-1310. [PMID: 35686213 PMCID: PMC9172736 DOI: 10.2147/copd.s361929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/19/2022] [Indexed: 11/23/2022] Open
Abstract
Background Fibrinogen is increasingly being studied as an inflammatory biomarker in chronic obstructive pulmonary disease (COPD), but there are limited data on the role of fibrinogen in assessing the severity of acute exacerbation of COPD (AECOPD). This study aimed to explore whether circulating fibrinogen could be used as a surrogate to measure the severity and predict the prognosis of AECOPD. Methods A total of 535 AECOPD patients diagnosed at our center from January 2016 to June 2021 were retrospectively enrolled in this study. The electronic medical record of each patient was retrieved to collect data on baseline characteristics and laboratory parameters, as well as the use of noninvasive positive-pressure ventilation (NPPV) and prognosis. Multiple linear regression analysis was used to identify independent factors associated with circulating fibrinogen values. Receiver-operating characteristic curve and multivariate logistic regression analysis were applied to further verify the use of fibrinogen to predict NPPV failure. Results Compared to patients with fibrinogen <4 g/L, patients with increased fibrinogen levels (>4 g/L) tended to have elevated inflammatory response and higher incidence of DVT/PTE, emphysema, pneumonia, and atherosclerosis. In addition, fibrinogen levels in NPPV-failure patients were significantly higher than non-NPPV patients and NPPV-success ones. The presence of emphysema, pneumonia, and history of long-term oxygen therapy and higher CRP levels and leukocyte counts were independent risk factors associated with increased fibrinogen levels in AECOPD. Furthermore, our data indicated that fibrinogen could be considered as a reliable biomarker to predict NPPV failure (AUC, 0.899, 95% CI 0.846–0.952), with an OR of 7.702 (95% CI 2.984–19.875; P<0.001). Conclusion The level of circulating fibrinogen can be used to measure severity of AECOPD, and among AECOPD patients managed with NPPV, fibrinogen >3.55 g/L can independently predict NPPV failure.
Collapse
Affiliation(s)
- Wei Sun
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhixin Cao
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Wang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Liming Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zujin Luo
- Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China
- Correspondence: Zujin Luo, Department of Respiratory and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, People’s Republic of China, Email
| |
Collapse
|
4
|
Rajabi H, Konyalilar N, Erkan S, Mortazavi D, Korkunc SK, Kayalar O, Bayram H, Rahbarghazi R. Emerging role of exosomes in the pathology of chronic obstructive pulmonary diseases; destructive and therapeutic properties. Stem Cell Res Ther 2022; 13:144. [PMID: 35379335 PMCID: PMC8978512 DOI: 10.1186/s13287-022-02820-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 03/18/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is known as the third leading cause of human death globally. Enhanced chronic inflammation and pathological remodeling are the main consequences of COPD, leading to decreased life span. Histological and molecular investigations revealed that prominent immune cell infiltration and release of several cytokines contribute to progressive chronic remodeling. Recent investigations have revealed that exosomes belonging to extracellular vesicles are involved in the pathogenesis of COPD. It has been elucidated that exosomes secreted from immune cells are eligible to carry numerous pro-inflammatory factors exacerbating the pathological conditions. Here, in this review article, we have summarized various and reliable information about the negative role of immune cell-derived exosomes in the remodeling of pulmonary tissue and airways destruction in COPD patients.
Collapse
Affiliation(s)
- Hadi Rajabi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Nur Konyalilar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Sinem Erkan
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Deniz Mortazavi
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Seval Kubra Korkunc
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
| | - Ozgecan Kayalar
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey
- Department of Pulmonary Medicine, School of Medicine, Koç University, Istanbul, Turkey
| | - Hasan Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), Koç University School of Medicine, Istanbul, Turkey.
- Department of Pulmonary Medicine, School of Medicine, Koç University, Istanbul, Turkey.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Huber MB, Kurz C, Kirsch F, Schwarzkopf L, Schramm A, Leidl R. The relationship between body mass index and health-related quality of life in COPD: real-world evidence based on claims and survey data. Respir Res 2020; 21:291. [PMID: 33143706 PMCID: PMC7607880 DOI: 10.1186/s12931-020-01556-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Body mass index (BMI) is an important parameter associated with mortality and health-related quality of life (HRQoL) in chronic obstructive pulmonary disease (COPD). However, informed guidance on stratified weight recommendations for COPD is still lacking. This study aims to determine the association between BMI and HRQoL across different severity grades of COPD to support patient management. METHODS We use conjunct analysis of claims and survey data based on a German COPD disease management program from 2016 to 2017. The EQ-5D-5L visual analog scale (VAS) and COPD Assessment Test (CAT) are used to measure generic and disease-specific HRQoL. Generalized additive models with smooth functions are implemented to evaluate the relationship between BMI and HRQoL, stratified by COPD severity. RESULTS 11,577 patients were included in this study. Mean age was 69.4 years and 59% of patients were male. In GOLD grades 1-3, patients with BMI of around 25 had the best generic and disease-specific HRQoL, whereas in GOLD grade 4, obese patients had the best HRQoL using both instruments when controlled for several variables including smoking status, income, COPD severity, comorbidities, emphysema, corticosteroid use, and days spent in hospital. CONCLUSION This real-world analysis shows the non-linear relationship between BMI and HRQoL in COPD. HRQoL of obese patients with mild to severe COPD might improve following weight reduction. For very severe COPD, a negative association of obesity and HRQoL could not be confirmed. The results hint at the need to stratify COPD patients by disease stage for optimal BMI management.
Collapse
Affiliation(s)
- Manuel B Huber
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| | - Christoph Kurz
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Florian Kirsch
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Munich School of Management and Munich Center of Health Sciences, Ludwig-Maximilians-Universität, Munich, Germany
| | - Larissa Schwarzkopf
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Neuherberg, Germany
- IFT-Institute Fuer Therapieforschung, Working Group Therapy and Health Services Research, Munich, Germany
| | - Anja Schramm
- AOK Bayern, Service Center of Health Care Management, Regensburg, Germany
| | - Reiner Leidl
- Institute of Health Economics and Health Care Management, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Munich School of Management and Munich Center of Health Sciences, Ludwig-Maximilians-Universität, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| |
Collapse
|
6
|
Aisanov Z, Khaltaev N. Management of cardiovascular comorbidities in chronic obstructive pulmonary disease patients. J Thorac Dis 2020; 12:2791-2802. [PMID: 32642187 PMCID: PMC7330365 DOI: 10.21037/jtd.2020.03.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is а highly prevalent, complex and heterogeneous clinical condition which is associated with significant concomitant diseases. COPD and cardiovascular diseases (CVDs) often coexist due to the high prevalence of each of these pathological conditions separately as well as the common risk factors (particularly smoking), mechanisms of interaction and influence of systemic inflammation. In addition, decreased pulmonary function in COPD is closely associated with an increased risk of congestive CVDs. One of the most important pathophysiological markers of COPD—lung hyperinflation—plays a significant role in the appearance of functional limitations of the pumping function of the heart, creating unfavorable conditions by exerting a compression effect on the heart muscle. The latter was confirmed by significant correlation between the COPD severity according to GOLD classification and the basic dimensions of the heart chambers. Several decades ago, the term “microcardia” was commonly used and indicated a radiological sign of emphysema. Some studies demonstrated a close relationship between the chance of occurrence of CVD and the severity of pulmonary dysfunction. Such an association has been demonstrated for the whole spectrum of CVD—including cerebrovascular disease, congestive heart failure (CHF) and rhythm disturbances—and was detected in the early stages of the disease. A large proportion of patients with mild and moderate COPD die due to CVD, which is much more likely than deaths in the same group due to respiratory insufficiency. COPD patients have a higher rate of hospitalization and death, the cause of which are coronary heart disease (CHD), stroke and CHF. Treatment of COPD today is mainly determined by national and international clinical guidelines, which should pay more attention to the problems of the treatment of COPD patients with comorbid conditions.
Collapse
Affiliation(s)
- Zaurbek Aisanov
- Pulmonology Department, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Nikolai Khaltaev
- WHO Global Coordination Mechanism for NCD Prevention and Control, WHO, Geneva, Switzerland
| |
Collapse
|
7
|
Kazeminasab S, Emamalizadeh B, Jouyban A, Shoja MM, Khoubnasabjafari M. Macromolecular biomarkers of chronic obstructive pulmonary disease in exhaled breath condensate. Biomark Med 2020; 14:1047-1063. [PMID: 32940079 DOI: 10.2217/bmm-2020-0121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Biomarkers provide important diagnostic and prognostic information on heterogeneous diseases such as chronic obstructive pulmonary disease (COPD). However, finding a suitable specimen for clinical analysis of biomarkers for COPD is challenging. Exhaled breath condensate (EBC) sampling is noninvasive, rapid, cost-effective and easily repeatable. EBC sampling has also provided recent progress in the identification of biological macromolecules, such as lipids, proteins and DNA in EBC samples, which has increased its utility for clinical scientists. In this article, we review applications involving EBC sampling for the analysis of COPD biomarkers and discuss its future potential.
Collapse
Affiliation(s)
- Somayeh Kazeminasab
- Pharmaceutical Analysis Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Liver & Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14756, Iran
| | - Babak Emamalizadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences,Tabriz, Iran
| | - Abolghasem Jouyban
- Pharmaceutical Analysis Research Center & Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran 14117-13135, Iran
| | - Mohammadali M Shoja
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Maryam Khoubnasabjafari
- Tuberculosis & Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz 51666-14756, Iran
| |
Collapse
|
8
|
ADAM17 Activity and IL-6 Trans-Signaling in Inflammation and Cancer. Cancers (Basel) 2019; 11:cancers11111736. [PMID: 31694340 PMCID: PMC6895846 DOI: 10.3390/cancers11111736] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/02/2019] [Indexed: 02/08/2023] Open
Abstract
All ligands of the epidermal growth factor receptor (EGF-R) are transmembrane proteins, which need to be proteolytically cleaved in order to be systemically active. The major protease responsible for this cleavage is the membrane metalloprotease ADAM17, which also has been implicated in cleavage of TNFα and interleukin-6 (IL-6) receptor. It has been recently shown that in the absence of ADAM17, the main protease for EGF-R ligand processing, colon cancer formation is largely abrogated. Intriguingly, colon cancer formation depends on EGF-R activity on myeloid cells rather than on intestinal epithelial cells. A major activity of EGF-R on myeloid cells is the stimulation of IL-6 synthesis. Subsequently, IL-6 together with the ADAM17 shed soluble IL-6 receptor acts on intestinal epithelial cells via IL-6 trans-signaling to induce colon cancer formation, which can be blocked by the inhibitor of IL-6 trans-signaling, sgp130Fc. Blockade of IL-6 trans-signaling therefore offers a new therapeutic window downstream of the EGF-R for the treatment of colon cancer and possibly of other EGF-R related neoplastic diseases.
Collapse
|
9
|
Aisanov ZR, Chuchalin AG, Kalmanova EN. [Chronic obstructive pulmonary disease and cardiovascular comorbidity]. ACTA ACUST UNITED AC 2019; 59:24-36. [PMID: 31526359 DOI: 10.18087/cardio.2572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/16/2019] [Indexed: 11/18/2022]
Abstract
In recent years, a greater understanding of the heterogeneity and complexity of chronic obstructive pulmonary disease (COPD) has come from the point of view of an integrated clinical assessment of severity, pathophysiology, and the relationship with other pathologies. A typical COPD patient suffers on average 4 or more concomitant diseases and every day about a third of patients take from 5 to 10 different drugs. The mechanisms of the interaction of COPD and cardiovascular disease (CVD) include the effects of systemic inflammation, hyperinflation (hyperinflation) of the lungs and bronchial obstruction. The risk of developing CVD in patients with COPD is on average 2-3 times higher than in people of a comparable age in the general population, even taking into account the risk of smoking. The prevalence of coronary heart disease, heart failure, and rhythm disturbances among COPD patients is significantly higher than in the general population. The article discusses in detail the safety of prescribing various groups of drugs for the treatment of CVD in patients with COPD. Achieving success in understanding and managing patients with COPD and CVD is possible using an integrated multidisciplinary approach.
Collapse
Affiliation(s)
- Z R Aisanov
- Pirogov Russian National Research Medical University
| | - A G Chuchalin
- Pirogov Russian National Research Medical University
| | - E N Kalmanova
- Pirogov Russian National Research Medical University
| |
Collapse
|
10
|
Dong W, Zhu Y, Du Y, Ma S. Association between features of COPD and risk of venous thromboembolism. CLINICAL RESPIRATORY JOURNAL 2019; 13:499-504. [PMID: 31172658 DOI: 10.1111/crj.13051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 05/03/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is associated with risk of venous thromboembolism (VTE) events. A detailed understanding of which clinical features of COPD increase risk of VTE events is needed. OBJECTIVES To investigate the association between features of COPD and risk of venous thromboembolism. METHODS A retrospective observational clinical study was conducted on 551 consecutive COPD patients visiting the Department of Respiratory and Critical Care Medicine of Tianjin Chest Hospital between February 2014 and April 2018. Finally, 151 patients were eligible for inclusion. Of these, 29 patients had COPD with VTE and 121 patients had COPD without VTE. Patient informations regarding age, gender, BMI, smoking history, smoking status (package/year), COPD-related symptoms, lung function, number of acute exacerbations and imaging visual emphysema were gathered. RESULTS Among the 29 VTE patients, 18 patients had PE and five had DVT, while 6 patients had simultaneous PE and DVT. There were statistically significant differences in GOLD grade, Imaging visual emphysema, and frequent acute exacerbations between the two groups. Multivariate logistic regression analysis showed that after adjustment for gender, age, BMI and smoking history, there were statistically significant for visible emphysema (OR = 3.54, 95% CI: 1.13-11.08; P = 0.03) and GOLD grade (OR = 1.77, 95% CI: 1.04-3.01; P = 0.035), but not for frequent acute exacerbations (OR = 1.65, 95% CI: 0.62-4.38; P = 0.31). CONCLUSIONS Visual emphysema is an independent risk factor for VTE events and the risk of VTE in COPD patients increases with the degree of airway obstruction. However, there is no evidence of an association between exacerbation frequency and VTE events.
Collapse
Affiliation(s)
- Weigang Dong
- Department of respiratory and CriticalCare Medicine, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Yaqian Zhu
- Department of respiratory and CriticalCare Medicine, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Yan Du
- Department of respiratory and CriticalCare Medicine, Tianjin Chest Hospital, Tianjin, People's Republic of China
| | - Shuping Ma
- Department of respiratory and CriticalCare Medicine, Tianjin Chest Hospital, Tianjin, People's Republic of China
| |
Collapse
|
11
|
Tarigan AP, Ananda FR, Pandia P, Sinaga BYM, Maryaningsih M, Anggriani A. The Impact of Upper Limb Training with Breathing Maneuver in Lung Function, Functional Capacity, Dyspnea Scale, and Quality of Life in Patient with Stable Chronic Obstructive of Lung Disease. Open Access Maced J Med Sci 2019; 7:567-572. [PMID: 30894913 PMCID: PMC6420952 DOI: 10.3889/oamjms.2019.113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/25/2019] [Accepted: 01/26/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Exercise tolerance is one of the main impacts of COPD. COPD patients often experience dyspnea and fatigue after doing daily activities using their limb parts, even in simple thing such as lifting or grooming. Nowadays, many pulmonologists concerned in pulmonary rehabilitation to modify some limb training with breathing manoeuvre to get positive impact in stable COPD patient. AIM The purpose of this study is to examine the impact of this modified upper limb training in lung function, functional capacity, dyspnea scale, and quality of life in patients with stable COPD. METHOD This was a quasi-experimental study held in 2017 on 22 stable COPD patients (based on GOLD 2018 criteria). Patients were given modified upper limb training with breathing manoeuvre that leads and monitored by a physiotherapist and physician in 10-20 minutes twice a week for 8 weeks. Before and after completed all sessions of training, we measured pulmonary functions test include FEV1 and FVC, functional capacity by 6 MWT, dyspnea scale by mMRC, and quality of life by CAT assessment. Statistical analysis was performed by Wilcoxon and paired t-test. RESULTS There was an improvement of lung function, both FEV1 (40.7 ± 13.8 to 47.3 ± 14.2; p-value 0.001) and FVC (50.7 ± 14.1 to 54.1 ± 14.7; p-value: 0.207) after training. There was a significant change of functional capacity in 6 MWT mean (277.3 ± 80.8 to 319.1 ± 78.3; p-value: 0.001). There was an improved quality of life after training, measured by decreasing in CAT score (23.9 ± 5.5 to 18.3 ± 5.2; p-value: 0.000). There was no significant change in the mMRC scale (p-value: 0.429). CONCLUSION There was an improvement of lung function, functional capacity, and quality of life in stable COPD after upper limb training with breathing manoeuvre in stable COPD patients.
Collapse
Affiliation(s)
- Amira Permatasari Tarigan
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | | | - Pandiaman Pandia
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Bintang YM Sinaga
- Department of Pulmonology and Respiratory Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | | | | |
Collapse
|
12
|
Bhatt SP, Nath HP, Kim YI, Ramachandran R, Watts JR, Terry NLJ, Sonavane S, Deshmane SP, Woodruff PG, Oelsner EC, Bodduluri S, Han MK, Labaki WW, Michael Wells J, Martinez FJ, Barr RG, Dransfield MT. Centrilobular emphysema and coronary artery calcification: mediation analysis in the SPIROMICS cohort. Respir Res 2018; 19:257. [PMID: 30563576 PMCID: PMC6299495 DOI: 10.1186/s12931-018-0946-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/20/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is associated with a two-to-five fold increase in the risk of coronary artery disease independent of shared risk factors. This association is hypothesized to be mediated by systemic inflammation but this link has not been established. METHODS We included 300 participants enrolled in the SPIROMICS cohort, 75 each of lifetime non-smokers, smokers without airflow obstruction, mild-moderate COPD, and severe-very severe COPD. We quantified emphysema and airway disease on computed tomography, characterized visual emphysema subtypes (centrilobular and paraseptal) and airway disease, and used the Weston visual score to quantify coronary artery calcification (CAC). We used the Sobel test to determine whether markers of systemic inflammation mediated a link between spirometric and radiographic features of COPD and CAC. RESULTS FEV1/FVC but not quantitative emphysema or airway wall thickening was associated with CAC (p = 0.036), after adjustment for demographics, diabetes mellitus, hypertension, statin use, and CT scanner type. To explain this discordance, we examined visual subtypes of emphysema and airway disease, and found that centrilobular emphysema but not paraseptal emphysema or bronchial thickening was independently associated with CAC (p = 0.019). MMP3, VCAM1, CXCL5 and CXCL9 mediated 8, 8, 7 and 16% of the association between FEV1/FVC and CAC, respectively. Similar biomarkers partially mediated the association between centrilobular emphysema and CAC. CONCLUSIONS The association between airflow obstruction and coronary calcification is driven primarily by the centrilobular subtype of emphysema, and is linked through bioactive molecules implicated in the pathogenesis of atherosclerosis. TRIAL REGISTRATION ClinicalTrials.gov: Identifier: NCT01969344 .
Collapse
Affiliation(s)
- Surya P Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA.
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Hrudaya P Nath
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Young-Il Kim
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Rekha Ramachandran
- Department of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jubal R Watts
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nina L J Terry
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sushil Sonavane
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Swati P Deshmane
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University California San Francisco, San Francisco, CA, 94143, USA
| | - Elizabeth C Oelsner
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Sandeep Bodduluri
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Wassim W Labaki
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - J Michael Wells
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Birmingham Veterans Affairs Hospital, Birmingham, AL, 35294, USA
| | - Fernando J Martinez
- Division of Pulmonary and Critical Care Medicine, Weill Cornell School of Medicine, New York, NY, 10065, USA
| | - R Graham Barr
- Division of Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mark T Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine and Lung Health Center, University of Alabama at Birmingham, THT 422, 1720, 2nd Avenue South, Birmingham, AL, 35294, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Birmingham Veterans Affairs Hospital, Birmingham, AL, 35294, USA
| |
Collapse
|
13
|
Kohli P, Staziaki PV, Janjua SA, Addison DA, Hallett TR, Hennessy O, Takx RAP, Lu MT, Fintelmann FJ, Semigran M, Harris RS, Celli BR, Hoffmann U, Neilan TG. The effect of emphysema on readmission and survival among smokers with heart failure. PLoS One 2018; 13:e0201376. [PMID: 30059544 PMCID: PMC6066229 DOI: 10.1371/journal.pone.0201376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/13/2018] [Indexed: 12/22/2022] Open
Abstract
Heart Failure (HF) and chronic obstructive pulmonary disease (COPD) are morbid diseases that often coexist. In patients with coexisting disease, COPD is an independent risk factor for readmission and mortality. However, spirometry is often inaccurate in those with active heart failure. Therefore, we investigated the association between the presence of emphysema on computed tomography (CT) and readmission rates in smokers admitted with heart failure (HF). The cohort included a consecutive group of smokers discharged with HF from a tertiary center between January 1, 2014 and April 1, 2014 who also had a CT of the chest for dyspnea. The primary endpoint was any readmission for HF before April 1, 2016; secondary endpoints were 30-day readmission for HF, length of stay and all-cause mortality. Over the study period, there were 225 inpatient smokers with HF who had a concurrent chest CT (155 [69%] males, age 69±11 years, ejection fraction [EF] 46±18%, 107 [48%] LVEF of < 50%). Emphysema on CT was present in 103 (46%) and these were older, had a lower BMI, more pack-years, less diabetes and an increased afterload. During a follow-up of 2.1 years, there were 110 (49%) HF readmissions and 55 (24%) deaths. When separated by emphysema on CT, any readmission, 30-day readmission, length of stay and mortality were higher among HF patients with emphysema. In multivariable regression, emphysema by CT was associated with a two-fold higher (adjusted HR 2.11, 95% CI 1.41–3.15, p < 0.001) risk of readmission and a trend toward increased mortality (adjusted HR 1.70 95% CI 0.86–3.34, p = 0.12). In conclusion, emphysema by CT is a frequent finding in smokers hospitalized with HF and is associated with adverse outcomes in HF. This under recognized group of patients with both emphysema and heart failure may benefit from improved recognition and characterization of their co-morbid disease processes and optimization of therapies for their lung disease.
Collapse
Affiliation(s)
- Puja Kohli
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| | - Pedro V. Staziaki
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Sumbal A. Janjua
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Daniel A. Addison
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Travis R. Hallett
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Orla Hennessy
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Richard A. P. Takx
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Michael T. Lu
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Florian J. Fintelmann
- Division of Thoracic Imaging and Intervention, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Marc Semigran
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Robert S. Harris
- Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Bartolome R. Celli
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
| | - Udo Hoffmann
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Tomas G. Neilan
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
14
|
Ostridge K, Williams NP, Kim V, Harden S, Bourne S, Clarke SC, Aris E, Mesia-Vela S, Devaster JM, Tuck A, Williams A, Wootton S, Staples KJ, Wilkinson TMA. Relationship of CT-quantified emphysema, small airways disease and bronchial wall dimensions with physiological, inflammatory and infective measures in COPD. Respir Res 2018; 19:31. [PMID: 29458372 PMCID: PMC5819274 DOI: 10.1186/s12931-018-0734-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/01/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND COPD is a complex, heterogeneous disease characterised by progressive development of airflow limitation. Spirometry provides little information about key aspects of pathology and is poorly related to clinical outcome, so other tools are required to investigate the disease. We sought to explore the relationships between quantitative CT analysis with functional, inflammatory and infective assessments of disease to identify the utility of imaging to stratify disease to better predict outcomes and disease response. METHODS Patients from the AERIS study with moderate-very severe COPD underwent HRCT, with image analysis determining the quantity of emphysema (%LAA<- 950), small airways disease (E/I MLD) and bronchial wall thickening (Pi10). At enrolment subjects underwent lung function testing, six-minute walk testing (6MWT), blood sampling for inflammatory markers and sputum sampling for white cell differential and microbiological culture and PCR. RESULTS 122 subjects were included in this analysis. Emphysema and small airways disease had independent associations with airflow obstruction (β = - 0.34, p < 0.001 and β = - 0.56, p < 0.001). %LAA<- 950 had independent associations with gas transfer (β = - 0.37, p < 0.001) and E/I MLD with RV/TLC (β = 0.30, p =0.003). The distance walked during the 6MWT was not associated with CT parameters, but exertional desaturation was independently associated with emphysema (β = 0.73, p < 0.001). Pi10 did not show any independent associations with lung function or functional parameters. No CT parameters had any associations with sputum inflammatory cells. Greater emphysema was associated with lower levels of systemic inflammation (CRP β = - 0.34, p < 0.001 and fibrinogen β = - 0.28, p =0.003). There was no significant difference in any of the CT parameters between subjects where potentially pathogenic bacteria were detected in sputum and those where it was not. CONCLUSIONS This study provides further validation for the use of quantitative CT measures of emphysema and small airways disease in COPD as they showed strong associations with pulmonary physiology and functional status. In contrast to this quantitative CT measures showed few convincing associations with biological measures of disease, suggesting it is not an effective tool at measuring disease activity.
Collapse
Affiliation(s)
- Kristoffer Ostridge
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK. .,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Nicholas P Williams
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Viktoriya Kim
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Stephen Harden
- Department of Radiology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Simon Bourne
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Portsmouth Hospitals NHS Trust, Queen Alexandra Hospital, Portsmouth, UK
| | - Stuart C Clarke
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | | | | | | | - Andrew Tuck
- Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Anthony Williams
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Stephen Wootton
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Karl J Staples
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Wessex Investigational Sciences Hub, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | | |
Collapse
|
15
|
Papaioannou AI, Kostikas K, Papaporfyriou A, Angelakis L, Papathanasiou E, Hillas G, Mazioti A, Bakakos P, Koulouris N, Papiris S, Loukides S. Emphysematous Phenotype is Characterized by Low Blood Eosinophils: A Cross-Sectional Study. COPD 2017; 14:635-640. [DOI: 10.1080/15412555.2017.1386644] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, University of Athens, Attikon Hospital, Athens, Greece
| | | | - Leonidas Angelakis
- 1st Respiratory Medicine Department, University of Athens, Sotiria Hospital, Athens Greece
| | - Evgenia Papathanasiou
- 2nd Respiratory Medicine Department, University of Athens, Attikon Hospital, Athens, Greece
| | - Georgios Hillas
- 1st Respiratory Medicine Department, University of Athens, Sotiria Hospital, Athens Greece
| | - Argyro Mazioti
- Department of Radiology, University of Athens “Attikon” University Hospital, Athens Greece
| | - Petros Bakakos
- 1st Respiratory Medicine Department, University of Athens, Sotiria Hospital, Athens Greece
| | - Nikolaos Koulouris
- 1st Respiratory Medicine Department, University of Athens, Sotiria Hospital, Athens Greece
| | - Spyros Papiris
- 2nd Respiratory Medicine Department, University of Athens, Attikon Hospital, Athens, Greece
| | - Stelios Loukides
- 1st Respiratory Medicine Department, University of Athens, Sotiria Hospital, Athens Greece
| |
Collapse
|
16
|
Tufvesson E, Markstad H, Bozovic G, Ekberg M, Bjermer L. Inflammation and chronic colonization of Haemophilus influenzae in sputum in COPD patients related to the degree of emphysema and bronchiectasis in high-resolution computed tomography. Int J Chron Obstruct Pulmon Dis 2017; 12:3211-3219. [PMID: 29138549 PMCID: PMC5677300 DOI: 10.2147/copd.s137578] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The presence of bacteria in the lower airways in COPD results in inflammation, further airway structural damage, and might lead to repeated exacerbations. We have previously shown that chronic colonization of Haemophilus influenzae during stable disease is related to increased inflammation, and we now aimed to relate previous findings of bacterial colonization and inflammation to the degree of radiological findings of bronchiectasis and emphysema. Thirty-nine patients with COPD were included in their stable state, and a high-resolution computed tomography of the lung was performed. They were followed-up monthly for up to a maximum of 6 months or until exacerbation, and they answered questionnaires, performed spirometry, and induced sputum at every visit. Thirty-five patients had emphysema with an emphysema degree of median 20% (interquartile range 10–50), and five patients had bronchiectasis, of which only four could expectorate sputum. The degree of emphysema correlated with several inflammatory mediators in sputum, such as interleukin-8 concentration, myeloperoxidase activity, and Leukotriene B4 concentration. Ten patients were chronically colonized with H. influenzae (ie, had a positive culture for H. influenzae at all visits). The four sputum patients with bronchiectasis were chronically colonized with H. influenzae and showed higher degree of H. influenzae growth compared to patients without bronchiectasis. During exacerbation, there was no longer any correlation between emphysema degree and inflammation, but patients with bronchiectasis showed higher sputum purulence score than patients without bronchiectasis. Emphysema and bronchiectasis in COPD patients show different clinical features. The presence of emphysema is more related to inflammation, while bronchiectasis is associated with bacterial colonization. We believe that both emphysema and bronchiectasis are therefore COPD phenotypes of highest impact and need evaluation to prevent further disease progression.
Collapse
Affiliation(s)
| | - Hanna Markstad
- Radiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | - Gracijela Bozovic
- Radiology, Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Lund, Sweden
| | | | | |
Collapse
|
17
|
Bradford E, Jacobson S, Varasteh J, Comellas AP, Woodruff P, O’Neal W, DeMeo DL, Li X, Kim V, Cho M, Castaldi PJ, Hersh C, Silverman EK, Crapo JD, Kechris K, Bowler RP. The value of blood cytokines and chemokines in assessing COPD. Respir Res 2017; 18:180. [PMID: 29065892 PMCID: PMC5655820 DOI: 10.1186/s12931-017-0662-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 10/09/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Blood biomarkers are increasingly used to stratify high risk chronic obstructive pulmonary disease (COPD) patients; however, there are fewer studies that have investigated multiple biomarkers and replicated in multiple large well-characterized cohorts of susceptible current and former smokers. METHODS We used two MSD multiplex panels to measure 9 cytokines and chemokines in 2123 subjects from COPDGene and 1117 subjects from SPIROMICS. These biomarkers included: interleukin (IL)-2, IL-6, IL-8, IL-10, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, eotaxin/CCL-11, eotaxin-3/CCL-26, and thymus and activation-regulated chemokine (TARC)/CCL-17. Regression models adjusted for clinical covariates were used to determine which biomarkers were associated with the following COPD phenotypes: airflow obstruction (forced expiratory flow at 1 s (FEV1%) and FEV1/forced vital capacity (FEV1/FVC), chronic bronchitis, COPD exacerbations, and emphysema. Biomarker-genotype associations were assessed by genome-wide association of single nucleotide polymorphisms (SNPs). RESULTS Eotaxin and IL-6 were strongly associated with airflow obstruction and accounted for 3-5% of the measurement variance on top of clinical variables. IL-6 was associated with progressive airflow obstruction over 5 years and both IL-6 and IL-8 were associated with progressive emphysema over 5 years. None of the biomarkers were consistently associated with chronic bronchitis or COPD exacerbations. We identified one novel SNP (rs9302690 SNP) that was associated with CCL17 plasma measurements. CONCLUSION When assessing smoking related pulmonary disease, biomarkers of inflammation such as IL-2, IL-6, IL-8, and eotaxin may add additional modest predictive value on top of clinical variables alone. TRIAL REGISTRATION COPDGene (ClinicalTrials.gov Identifier: NCT02445183 ). Subpopulations and Intermediate Outcomes Measures in COPD Study (SPIROMICS) ( ClinicalTrials.gov Identifier: NCT 01969344 ).
Collapse
Affiliation(s)
- Eric Bradford
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Sean Jacobson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Jason Varasteh
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Alejandro P. Comellas
- University of Iowa, Internal Medicine, 200 Hawkins Dr C331-GH, Iowa City, IA 52242 USA
| | - Prescott Woodruff
- UCSF, Division of Pulmonary and Critical Care Medicine and Cardiovascular Research Institute, Box 0130, Rm HSE 1305, 513 Parnassus Ave, San Francisco, CA 94143 USA
| | - Wanda O’Neal
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Dawn L. DeMeo
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts USA
| | - Xingnan Li
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ USA
| | - Victor Kim
- Temple University School of Medicine, Pulmonary and Critical Care Medicine, 785 Parkinson Pavilion, 3401 North Broad Street, Philadelphia, PA 19140 USA
| | - Michael Cho
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Peter J. Castaldi
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts USA
- Tufts Medical Center, ICRHPS, 800 Washington St, Box 63, Boston, MA 02111 USA
| | - Craig Hersh
- Division of General Internal Medicine and Primary Care, Brigham and Women’s Hospital, Boston, Massachusetts USA
| | - Edwin K. Silverman
- Channing Division of Network Medicine and the Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - James D. Crapo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, University of Colorado Denver, Colorado School of Public Health, Mail Stop B119, 13001 E. 17th Place, Aurora, CO 80045 USA
| | - Russell P. Bowler
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., K715, Denver, CO 80206 USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, University of Colorado Anschutz Medical Campus, Research Building 2, 9th Floor, 12700 E. 19th Ave, Aurora, CO USA
| |
Collapse
|
18
|
Skronska-Wasek W, Mutze K, Baarsma HA, Bracke KR, Alsafadi HN, Lehmann M, Costa R, Stornaiuolo M, Novellino E, Brusselle GG, Wagner DE, Yildirim AÖ, Königshoff M. Reduced Frizzled Receptor 4 Expression Prevents WNT/β-Catenin-driven Alveolar Lung Repair in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2017; 196:172-185. [PMID: 28245136 DOI: 10.1164/rccm.201605-0904oc] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD), in particular emphysema, is characterized by loss of parenchymal alveolar tissue and impaired tissue repair. Wingless and INT-1 (WNT)/β-catenin signaling is reduced in COPD; however, the mechanisms thereof, specifically the role of the frizzled (FZD) family of WNT receptors, remain unexplored. OBJECTIVES To identify and functionally characterize specific FZD receptors that control downstream WNT signaling in impaired lung repair in COPD. METHODS FZD expression was analyzed in lung homogenates and alveolar epithelial type II (ATII) cells of never-smokers, smokers, patients with COPD, and two experimental COPD models by quantitative reverse transcriptase-polymerase chain reaction, immunoblotting, and immunofluorescence. The functional effects of cigarette smoke on FZD4, WNT/β-catenin signaling, and elastogenic components were investigated in primary ATII cells in vitro and in three-dimensional lung tissue cultures ex vivo. Gain- and loss-of-function approaches were applied to determine the effects of FZD4 signaling on alveolar epithelial cell wound healing and repair, as well as on expression of elastogenic components. MEASUREMENTS AND MAIN RESULTS FZD4 expression was reduced in human and experimental COPD lung tissues as well as in primary human ATII cells from patients with COPD. Cigarette smoke exposure down-regulated FZD4 expression in vitro and in vivo, along with reduced WNT/β-catenin activity. Inhibition of FZD4 decreased WNT/β-catenin-driven epithelial cell proliferation and wound closure, and it interfered with ATII-to-ATI cell transdifferentiation and organoid formation, which were augmented by FZD4 overexpression. Moreover, FZD4 restoration by overexpression or pharmacological induction led to induction of WNT/β-catenin signaling and expression of elastogenic components in three-dimensional lung tissue cultures ex vivo. CONCLUSIONS Reduced FZD4 expression in COPD contributes to impaired alveolar repair capacity.
Collapse
Affiliation(s)
- Wioletta Skronska-Wasek
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Kathrin Mutze
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Hoeke A Baarsma
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Ken R Bracke
- 2 Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Hani N Alsafadi
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Mareike Lehmann
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Rita Costa
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Mariano Stornaiuolo
- 3 Department of Pharmacy, University of Naples Federico II, Naples, Italy; and
| | - Ettore Novellino
- 3 Department of Pharmacy, University of Naples Federico II, Naples, Italy; and
| | - Guy G Brusselle
- 2 Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Darcy E Wagner
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Ali Ö Yildirim
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany
| | - Melanie Königshoff
- 1 Helmholtz Zentrum Munich, Comprehensive Pneumology Center, Member of the German Center for Lung Research, Munich, Germany.,4 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, Aurora, Colorado
| |
Collapse
|
19
|
Onishi K. Total management of chronic obstructive pulmonary disease (COPD) as an independent risk factor for cardiovascular disease. J Cardiol 2017; 70:128-134. [PMID: 28325523 DOI: 10.1016/j.jjcc.2017.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Patients with cardiovascular disease (CVD) often have multiple comorbid conditions that may interact with each other, confound the choice of treatments, and reduce mortality. Chronic obstructive pulmonary disease (COPD) is one of the most important comorbidities of CVD, which causes serious consequences in patients with ischemic heart disease, stroke, arrhythmia, and heart failure. COPD shares common risk factors such as tobacco smoking and aging with CVD, is associated with less physical activity, and produces systemic inflammation and oxidative stress. Overall, patients with COPD have a 2-3-fold increased risk of CVD as compared to age-matched controls when adjusted for tobacco smoking. Chronic heart failure (HF) is a frequent and important comorbidity which has a significant impact on prognosis in COPD, and vice versa. HF overlaps in symptoms and signs and has a common comorbidity with COPD, so that diagnosis of COPD is difficult in patients with HF. The combination of HF and COPD presents many therapeutic challenges including beta-blockers (BBs) and beta-agonists. Inhaled long-acting bronchodilators including beta2-agonists and anticholinergics for COPD would not worsen HF. Diuretics are relatively safe, and angiotensin-converting enzyme inhibitors are preferred to treat HF accompanied with COPD. BBs are only relatively contraindicated in asthma, but not in COPD. Low doses of cardioselective BBs should be aggressively initiated in clinically stable patients with HF accompanied with COPD combined with close monitoring for signs of airway obstruction and gradually up-titrated to the maximum tolerated dose. Encouraging appropriate and aggressive treatment for both HF and COPD should be recommended to improve quality of life and mortality in HF patients with COPD.
Collapse
|
20
|
Hawkins NM, Khosla A, Virani SA, McMurray JJV, FitzGerald JM. B-type natriuretic peptides in chronic obstructive pulmonary disease: a systematic review. BMC Pulm Med 2017; 17:11. [PMID: 28073350 PMCID: PMC5223538 DOI: 10.1186/s12890-016-0345-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/09/2016] [Indexed: 12/29/2022] Open
Abstract
Background Patients with chronic obstructive pulmonary disease (COPD) have increased cardiovascular risk. Natriuretic peptides (NP) in other populations are useful in identifying cardiovascular disease, stratifying risk, and guiding therapy. Methods We performed a systematic literature review to examine NP in COPD, utilising Medline, EMBASE, and the Cochrane Library. Results Fifty one studies were identified. NP levels were lower in stable compared to exacerbation of COPD, and significantly increased with concomitant left ventricular systolic dysfunction or cor pulmonale. Elevation occurred in 16 to 60% of exacerbations and persisted in approximately one half of patients at discharge. Cardiovascular comorbidities were associated with increased levels. Levels consistently correlated with pulmonary artery pressure and left ventricular ejection fraction, but not pulmonary function or oxygen saturation. NP demonstrated high negative predictive values (0.80 to 0.98) to exclude left ventricular dysfunction in both stable and exacerbation of COPD, but relatively low positive predictive values. NP elevation predicted early adverse outcomes, but the association with long term mortality was inconsistent. Conclusion NP reflect diverse aspects of the cardiopulmonary continuum which limits utility when applied in isolation. Strategies integrating NP with additional variables, biomarkers and imaging require further investigation.
Collapse
Affiliation(s)
- Nathaniel M Hawkins
- Division of Cardiology, University of British Columbia, BC Centre for Improved Cardiovascular Health, St. Paul's Hospital, 1081 Burrard Street, Vancouver, V6Z 1Y6, BC, Canada.
| | - Amit Khosla
- Division of Cardiology, University of British Columbia, BC Centre for Improved Cardiovascular Health, St. Paul's Hospital, 1081 Burrard Street, Vancouver, V6Z 1Y6, BC, Canada
| | - Sean A Virani
- Division of Cardiology, University of British Columbia, BC Centre for Improved Cardiovascular Health, St. Paul's Hospital, 1081 Burrard Street, Vancouver, V6Z 1Y6, BC, Canada
| | - John J V McMurray
- Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - J Mark FitzGerald
- Division of Respiratory Medicine, University of British Columbia and Institute for Heart and Lung Health, Vancouver, Canada
| |
Collapse
|
21
|
Henriques I, Lopes-Pacheco M, Padilha GA, Marques PS, Magalhães RF, Antunes MA, Morales MM, Rocha NN, Silva PL, Xisto DG, Rocco PRM. Moderate Aerobic Training Improves Cardiorespiratory Parameters in Elastase-Induced Emphysema. Front Physiol 2016; 7:329. [PMID: 27536247 PMCID: PMC4971418 DOI: 10.3389/fphys.2016.00329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
Aim: We investigated the therapeutic effects of aerobic training on lung mechanics, inflammation, morphometry and biological markers associated with inflammation, and endothelial cell damage, as well as cardiac function in a model of elastase-induced emphysema. Methods: Eighty-four BALB/c mice were randomly allocated to receive saline (control, C) or 0.1 IU porcine pancreatic elastase (emphysema, ELA) intratracheally once weekly for 4 weeks. After the end of administration period, once cardiorespiratory impairment associated with emphysema was confirmed, each group was further randomized into sedentary (S) and trained (T) subgroups. Trained mice ran on a motorized treadmill, at moderate intensity, 30 min/day, 3 times/week for 4 weeks. Results: Four weeks after the first instillation, ELA animals, compared to C, showed: (1) reduced static lung elastance (Est,L) and levels of vascular endothelial growth factor (VEGF) in lung tissue, (2) increased elastic and collagen fiber content, dynamic elastance (E, in vitro), alveolar hyperinflation, and levels of interleukin-1β and tumor necrosis factor (TNF)-α, and (3) increased right ventricular diastolic area (RVA). Four weeks after aerobic training, ELA-T group, compared to ELA-S, was associated with reduced lung hyperinflation, elastic and collagen fiber content, TNF-α levels, and RVA, as well as increased Est,L, E, and levels of VEGF. Conclusion: Four weeks of regular and moderate intensity aerobic training modulated lung inflammation and remodeling, thus improving pulmonary function, and reduced RVA and pulmonary arterial hypertension in this animal model of elastase-induced emphysema.
Collapse
Affiliation(s)
- Isabela Henriques
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Gisele A Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Patrícia S Marques
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Raquel F Magalhães
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Nazareth N Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Department of Physiology, Fluminense Federal UniversityNiterói, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Débora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Papaioannou AI, Kostikas K, Manali ED, Papadaki G, Roussou A, Spathis A, Mazioti A, Tomos I, Papanikolaou I, Loukides S, Chainis K, Karakitsos P, Griese M, Papiris S. Serum Levels of Surfactant Proteins in Patients with Combined Pulmonary Fibrosis and Emphysema (CPFE). PLoS One 2016; 11:e0157789. [PMID: 27337142 PMCID: PMC4919090 DOI: 10.1371/journal.pone.0157789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/03/2016] [Indexed: 01/05/2023] Open
Abstract
Introduction Emphysema and idiopathic pulmonary fibrosis (IPF) present either per se or coexist in combined pulmonary fibrosis and emphysema (CPFE). Serum surfactant proteins (SPs) A, B, C and D levels may reflect lung damage. We evaluated serum SP levels in healthy controls, emphysema, IPF, and CPFE patients and their associations to disease severity and survival. Methods 122 consecutive patients (31 emphysema, 62 IPF, and 29 CPFE) and 25 healthy controls underwent PFTs, ABG-measurements, 6MWT and chest HRCT. Serum levels of SPs were measured. Patients were followed-up for 1-year. Results SP-A and SP-D levels differed between groups (p = 0.006 and p<0.001 respectively). In post-hoc analysis, SP-A levels differed only between controls and CPFE (p<0.05) and CPFE and emphysema (p<0.05). SP-D differed between controls and IPF or CPFE (p<0.001 for both comparisons). In IPF SP-B correlated to pulmonary function while SP-A, correlated to the Composite Physiological Index (CPI). Controls current smokers had higher SP-A and SP-D levels compared to non-smokers (p = 0.026 and p = 0.023 respectively). SP-D levels were higher in CPFE patients with extended emphysema (p = 0.042). In patients with IPF, SP-B levels at the upper quartile of its range (≥26 ng/mL) presented a weak association with reduced survival (p = 0.05). Conclusion In conclusion, serum SP-A and SP-D levels were higher where fibrosis exists or coexists and related to disease severity, suggesting that serum SPs relate to alveolar damage in fibrotic lungs and may reflect either local overproduction or overleakage. The weak association between high levels of SP-B and survival needs further validation in clinical trials.
Collapse
Affiliation(s)
- Andriana I. Papaioannou
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
- * E-mail:
| | - Konstantinos Kostikas
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Effrosyni D. Manali
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Papadaki
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aneza Roussou
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris Spathis
- Department of Cytopathology, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Argyro Mazioti
- Department of Radiology, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Tomos
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ilias Papanikolaou
- Respiratory Medicine Department, “Corfu General Hospital”, Corfu, Greece
| | - Stelios Loukides
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriakos Chainis
- Respiratory Medicine Department, “Corfu General Hospital”, Corfu, Greece
| | - Petros Karakitsos
- Department of Cytopathology, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Matthias Griese
- Hauner Children’s University Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Lindwurmstrasse 4, 80337, Munich, Germany
| | - Spyros Papiris
- 2nd Respiratory Medicine Department, “Attikon” University Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
23
|
Ostridge K, Wilkinson TMA. Present and future utility of computed tomography scanning in the assessment and management of COPD. Eur Respir J 2016; 48:216-28. [PMID: 27230448 DOI: 10.1183/13993003.00041-2016] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/21/2016] [Indexed: 01/08/2023]
Abstract
Computed tomography (CT) is the modality of choice for imaging the thorax and lung structure. In chronic obstructive pulmonary disease (COPD), it used to recognise the key morphological features of emphysema, bronchial wall thickening and gas trapping. Despite this, its place in the investigation and management of COPD is yet to be determined, and it is not routinely recommended. However, lung CT already has important clinical applications where it can be used to diagnose concomitant pathology and determine which patients with severe emphysema are appropriate for lung volume reduction procedures. Furthermore, novel quantitative analysis techniques permit objective measurements of pulmonary and extrapulmonary manifestations of the disease. These techniques can give important insights into COPD, and help explore the heterogeneity and underlying mechanisms of the condition. In time, it is hoped that these techniques can be used in clinical trials to help develop disease-specific therapy and, ultimately, as a clinical tool in identifying patients who would benefit most from new and existing treatments. This review discusses the current clinical applications for CT imaging in COPD and quantification techniques, and its potential future role in stratifying disease for optimal outcome.
Collapse
Affiliation(s)
- Kristoffer Ostridge
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| | - Tom M A Wilkinson
- Southampton NIHR Respiratory Biomedical Research Unit, Southampton General Hospital, Southampton, UK Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton General Hospital, Southampton, UK
| |
Collapse
|
24
|
Takahashi Y, Matsuda M, Aoki S, Dejima H, Nakayama T, Matsutani N, Kawamura M. Qualitative Analysis of Preoperative High-Resolution Computed Tomography: Risk Factors for Pulmonary Complications After Major Lung Resection. Ann Thorac Surg 2016; 101:1068-74. [DOI: 10.1016/j.athoracsur.2015.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/21/2015] [Accepted: 09/08/2015] [Indexed: 11/24/2022]
|
25
|
Padilha GA, Henriques I, Lopes-Pacheco M, Abreu SC, Oliveira MV, Morales MM, Lima LM, Barreiro EJ, Silva PL, Xisto DG, Rocco PRM. Therapeutic effects of LASSBio-596 in an elastase-induced mouse model of emphysema. Front Physiol 2015; 6:267. [PMID: 26483698 PMCID: PMC4588117 DOI: 10.3389/fphys.2015.00267] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/11/2015] [Indexed: 11/13/2022] Open
Abstract
Emphysema is an intractable pulmonary disease characterized by an inflammatory process of the airways and lung parenchyma and ongoing remodeling process in an attempt to restore lung structure. There is no effective drug therapy that regenerates lung tissue or prevents the progression of emphysema; current treatment is aimed at symptomatic relief. We hypothesized that LASSBio-596, a molecule with potent anti-inflammatory and immunomodulatory effects, might reduce pulmonary inflammation and remodeling and thus improve lung function in experimental emphysema. Emphysema was induced in BALB/c mice by intratracheal administration of porcine pancreatic elastase (0.1 IU) once weekly during 4 weeks. A control group received saline using the same protocol. After the last instillation of saline or elastase, dimethyl sulfoxide, or LASSBio-596 were administered intraperitoneally, once daily for 8 days. After 24 h, in elastase-induced emphysema animals, LASSBio-596 yielded: (1) decreased mean linear intercept, hyperinflation and collagen fiber content, (2) increased elastic fiber content, (3) reduced number of M1 macrophages, (4) decreased tumor necrosis factor-α, interleukin-1β, interleukin-6, and transforming growth factor-β protein levels in lung tissue, and increased vascular endothelial growth factor. These changes resulted in increased static lung elastance. In conclusion, LASSBio-596 therapy reduced lung inflammation, airspace enlargement, and small airway wall remodeling, thus improving lung function, in this animal model of elastase-induced emphysema.
Collapse
Affiliation(s)
- Gisele A. Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Isabela Henriques
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Soraia C. Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Milena V. Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Marcelo M. Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Lidia M. Lima
- Laboratory of Evaluation and Synthesis of Bioactive Substances, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Eliezer J. Barreiro
- Laboratory of Evaluation and Synthesis of Bioactive Substances, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Pedro L. Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Debora G. Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
26
|
Miłkowska-Dymanowska J, Białas AJ, Zalewska-Janowska A, Górski P, Piotrowski WJ. Underrecognized comorbidities of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2015. [PMID: 26203239 PMCID: PMC4507790 DOI: 10.2147/copd.s82420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
COPD is associated with different comorbid diseases, and their frequency increases with age. Comorbidities severely impact costs of health care, intensity of symptoms, quality of life and, most importantly, may contribute to life span shortening. Some comorbidities are well acknowledged and established in doctors’ awareness. However, both everyday practice and literature searches provide evidence of other, less recognized diseases, which are frequently associated with COPD. We call them underrecognized comorbidities, and the reason why this is so may be related to their relatively low clinical significance, inefficient literature data, or data ambiguity. In this review, we describe rhinosinusitis, skin abnormalities, eye diseases, different endocrinological disorders, and gastroesophageal reflux disease. Possible links to COPD pathogenesis have been discussed, if the data were available.
Collapse
Affiliation(s)
- Joanna Miłkowska-Dymanowska
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland ; Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| | - Adam J Białas
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland ; Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| | - Anna Zalewska-Janowska
- Unit of Psychodermatology, Chair of Clinical Immunology and Microbiology, Medical University of Lodz, Łódź, Poland
| | - Paweł Górski
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland ; Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| | - Wojciech J Piotrowski
- Department of Pneumology and Allergy, 1st Chair of Internal Medicine, Medical University of Lodz, Łódź, Poland ; Healthy Aging Research Centre (HARC), Medical University of Lodz, Łódź, Poland
| |
Collapse
|
27
|
Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther 2015; 18:381-8. [PMID: 24781789 DOI: 10.1007/s40291-014-0100-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The diagnosis of chronic obstructive pulmonary disease (COPD) is based on a post-bronchodilator fixed forced expiratory volume in 1 second (FEV1)/forced vital capacity (FVC) <70 % ratio and the presence of symptoms such as shortness of breath and productive cough. Despite the simplicity in making a diagnosis of COPD, this morbid condition is very heterogeneous, and at least three different phenotypes can be recognized: the exacerbator, the emphysema-hyperinflation and the overlap COPD-asthma. These subgroups show different clinical and radiological features. It has been speculated that there is an enormous variability in the response to drugs among the COPD phenotypes, and it is expected that subjects with the same phenotype will have a similar response to each specific treatment. We believe that phenotyping COPD patients would be very useful to predict the response to a treatment and the progression of the disease. This personalized approach allows identification of the right treatment for each COPD patient, and at the same time, leads to improvement in the effectiveness of therapies, avoidance of treatments not indicated, and reduction in the onset of adverse effects. The objective of the present review is to report the current knowledge about different COPD phenotypes, focusing on specific treatments for each subgroup. However, at present, COPD phenotypes have not been studied by randomized clinical trials and therefore we hope that well designed studies will focus on this topic.
Collapse
Affiliation(s)
- Andrea Segreti
- Unit of Respiratory Medicine, Department of System Medicine, University of Rome Tor Vergata, via Montpellier 1, 00131, Rome, Italy
| | | | | | | |
Collapse
|
28
|
Phenotyping provides potential for a personalized approach in patients with chronic obstructive pulmonary disease (COPD). DRUGS & THERAPY PERSPECTIVES 2015. [DOI: 10.1007/s40267-015-0183-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Lock-Johansson S, Vestbo J, Sorensen GL. Surfactant protein D, Club cell protein 16, Pulmonary and activation-regulated chemokine, C-reactive protein, and Fibrinogen biomarker variation in chronic obstructive lung disease. Respir Res 2014; 15:147. [PMID: 25425298 PMCID: PMC4256818 DOI: 10.1186/s12931-014-0147-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 11/07/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a multifaceted condition that cannot be fully described by the severity of airway obstruction. The limitations of spirometry and clinical history have prompted researchers to investigate a multitude of surrogate biomarkers of disease for the assessment of patients, prediction of risk, and guidance of treatment. The aim of this review is to provide a comprehensive summary of observations for a selection of recently investigated pulmonary inflammatory biomarkers (Surfactant protein D (SP-D), Club cell protein 16 (CC-16), and Pulmonary and activation-regulated chemokine (PARC/CCL-18)) and systemic inflammatory biomarkers (C-reactive protein (CRP) and fibrinogen) with COPD. The relevance of these biomarkers for COPD is discussed in terms of their biological plausibility, their independent association to disease and hard clinical outcomes, their modification by interventions, and whether changes in clinical outcomes are reflected by changes in the biomarker.
Collapse
Affiliation(s)
- Sofie Lock-Johansson
- Institute of Molecular Medicine, University of Southern Denmark, JB Winsloews Vej 25.3, Odense, 5000, Denmark.
| | - Jørgen Vestbo
- Department of Respiratory Medicine, Gentofte Hospital, Hellerup, Denmark.
- Respiratory Research Group, Manchester Academic Science Centre University Hospital South Manchester NHS Foundation Trust Manchester, Manchester, UK.
| | - Grith Lykke Sorensen
- Institute of Molecular Medicine, University of Southern Denmark, JB Winsloews Vej 25.3, Odense, 5000, Denmark.
| |
Collapse
|
30
|
Kheir F, Salerno DA. Brain natriuretic peptide measurement in patients with COPD and cardiovascular disease. Chest 2014; 144:1081. [PMID: 24008964 DOI: 10.1378/chest.13-0948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Fayez Kheir
- Department of Pulmonary Diseases, Critical Care, and Environmental Medicine, Tulane University Health Sciences Center, New Orleans, LA
| | - Daniel A Salerno
- Department of Pulmonary Diseases, Critical Care, and Environmental Medicine, Tulane University Health Sciences Center, New Orleans, LA.
| |
Collapse
|
31
|
de-Torres JP, Blanco D, Alcaide AB, Seijo LM, Bastarrika G, Pajares MJ, Muñoz-Barrutia A, Ortiz-de-Solorzano C, Pio R, Campo A, Montes U, Segura V, Pueyo J, Montuenga LM, Zulueta JJ. Smokers with CT detected emphysema and no airway obstruction have decreased plasma levels of EGF, IL-15, IL-8 and IL-1ra. PLoS One 2013; 8:e60260. [PMID: 23577098 PMCID: PMC3618450 DOI: 10.1371/journal.pone.0060260] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 02/24/2013] [Indexed: 01/03/2023] Open
Abstract
RATIONALE Low-grade inflammation and emphysema have been shown to be associated with an increased risk of lung cancer. However, the systemic inflammatory response in patients with emphysema is still unknown. OBJECTIVE TO COMPARE THE PLASMA CYTOKINE PROFILES IN TWO GROUPS OF CURRENT OR FORMER SMOKERS WITHOUT AIRWAY OBSTRUCTION: a control group of individuals without computed tomography (CT) detected emphysema vs. a study group of individuals with CT detected emphysema. METHODS Subjects underwent a chest CT, spirometry, and determination of EGF, IL-15, IL-1ra, IL-8, MCP-1, MIP-1β, TGFα, TNFα, and VEGF levels in plasma. Cytokine levels in each group were compared adjusting for confounding factors. RESULTS 160 current smokers and former smokers without airway obstruction participated in the study: 80 without emphysema and 80 subjects with emphysema. Adjusted group comparisons revealed significant reductions in EGF (-0.317, p = 0.01), IL-15 (-0.21, p = 0.01), IL-8 (-0.180, p = 0.02) and IL-1ra (-0.220, p = 0.03) in subjects with emphysema and normal spirometry. CONCLUSIONS Current or former smokers expressing a well-defined disease characteristic such as emphysema, has a specific plasma cytokine profile. This includes a decrease of cytokines mainly implicated in activation of apoptosis or decrease of immunosurveillance. This information should be taken into account when evaluated patients with tobacco respiratory diseases.
Collapse
Affiliation(s)
- Juan P de-Torres
- Pulmonary Department, Clínica Universidad de Navarra, University of Navarra, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
OBJECTIVE Studies of fractional exhaled NO (FeNO) or induced sputum are now well standardized and the exponential increase in publications about exhaled breath condensate reflects growing interest in a noninvasive diagnosis of pulmonary diseases in occupational medicine. METHODS This review describes current techniques (FeNO, induced sputum, and exhaled breath condensate) for the study of inflammation and oxidative stress biomarkers. RESULTS These biomarkers are FeNO, cytokines, H2O2, 8-isoprostane, malondialdehyde, and nitrogen oxides. These techniques also include the study of markers of the toxic burden in the lungs (heavy metals and mineral compounds) that are important in occupational health exposure assessment. CONCLUSIONS In occupational medicine, the study of both volatile and nonvolatile respiratory biomarkers can be useful in medical surveillance of exposed workers, the early identification of respiratory diseases, or the monitoring of their development.
Collapse
|
33
|
Abstract
COPD is a worldwide public health problem that reduces the quality of life. The exact pathways by which CS and other environmental toxins produce COPD are not known. Currently, the leading candidates are (1) the protease-antiprotease hypothesis, (2) the Dutch hypothesis, (3) the British hypothesis, and the (4) autoimmunity hypothesis. Given the heterogeneity of the disease (and phenotypes), it is probably unrealistic that one pathway will fully explain COPD pathophysiology.
Collapse
Affiliation(s)
- Anthony Tam
- Department of Medicine, The UBC James Hogg Research Centre, Providence Heart and Lung Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | | |
Collapse
|
34
|
Duvoix A, Dickens J, Haq I, Mannino D, Miller B, Tal-Singer R, Lomas DA. Blood fibrinogen as a biomarker of chronic obstructive pulmonary disease. Thorax 2012; 68:670-6. [PMID: 22744884 PMCID: PMC3711372 DOI: 10.1136/thoraxjnl-2012-201871] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a multicomponent condition that is characterised by airflow obstruction that is not fully reversible and is a major global cause of morbidity and mortality. The most widely used marker of disease severity and progression is FEV1. However, FEV1 correlates poorly with both symptoms and other measures of disease progression and thus there is an urgent need for other biological markers to better characterise individuals with COPD. Fibrinogen is an acute phase plasma protein that has emerged as a promising biomarker in COPD. Here we review the current clinical evidence linking fibrinogen with COPD and its associated co-morbidities and discuss its potential utility as a biomarker. Methods Searches for appropriate studies were undertaken on PubMed using search terms fibrinogen, COPD, emphysema, chronic bronchitis, FEV1, cardiovascular disease, exacerbation and mortality. Results There is strong evidence of an association between fibrinogen and the presence of COPD, the presence and frequency of exacerbations and with mortality. Fibrinogen is associated with disease severity but does not predict lung function decline, a measure used as a surrogate for disease activity. The role of fibrinogen in identifying inflammatory co morbidities, particularly cardiovascular disease, remains unclear. Fibrinogen is reduced by p38 mitogen-activated protein kinase inhibitors in individuals with stable disease and by oral corticosteroids during exacerbations. Conclusions Fibrinogen is likely to be a useful biomarker to stratify individuals with COPD into those with a high or low risk of future exacerbations and may identify those with a higher risk of mortality.
Collapse
Affiliation(s)
- Annelyse Duvoix
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/MRC Building, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
35
|
Silva DR, Coelho AC, Gazzana MB, Menna Barreto SS, Knorst MM. D-dimer Levels in Stable COPD Patients: A Case-control Study. COPD 2012; 9:426-31. [DOI: 10.3109/15412555.2012.683840] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Denise Rossato Silva
- Universidade Federal do Rio Grande do Sul; Hospital de Clínicas de Porto Alegre, Brazil
| | - Ana Cláudia Coelho
- Universidade Federal do Rio Grande do Sul; Hospital de Clínicas de Porto Alegre, Brazil
| | - Marcelo Basso Gazzana
- Universidade Federal do Rio Grande do Sul; Hospital de Clínicas de Porto Alegre, Brazil
| | | | - Marli Maria Knorst
- Universidade Federal do Rio Grande do Sul; Hospital de Clínicas de Porto Alegre, Brazil
| |
Collapse
|
36
|
Gaki E, Kontogianni K, Papaioannou AI, Bakakos P, Gourgoulianis KI, Kostikas K, Alchanatis M, Papiris S, Loukides S. Associations between BODE index and systemic inflammatory biomarkers in COPD. COPD 2012; 8:408-13. [PMID: 22149400 DOI: 10.3109/15412555.2011.619599] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND COPD is a multicomponent disease and systemic inflammation represents one of the possible mechanisms responsible for its systemic manifestations, including skeletal muscle weakness and cachexia. Fat-free mass index (FFMI) that reflects the skeletal muscle mass, has been shown to be associated with both dyspnoea and exercise capacity. We hypothesized that the multidimensional BODE index, that reflects the multicomponent nature of COPD, might be related to biomarkers of systemic inflammation. We further evaluated associations between FFMI and systemic inflammation. METHODS BODE index and FFMI were calculated in 222 stable COPD patients and 132 smokers or ex-smokers with normal lung function. Systemic inflammation was evaluated with the measurement of leptin, adiponectin, CRP, IL-6, and TNF-α in serum samples of COPD patients. RESULTS In patients with COPD, both BODE index and FFMI presented significant positive and negative associations respectively with leptin levels (R(2) 0.61 and 0.65, respectively), whereas FFMI presented an additional negative association with the levels of TNF-α (R(2) 0.38). No significant associations were observed in smokers or ex-smokers with normal lung function. CONCLUSIONS Both BODE index and FFMI, are related to the circulating levels of leptin in patients with COPD, suggesting a possible role for leptin in the systemic component of COPD. The additional association of FFMI with TNF-α may further support a role of systemic inflammation in muscle wasting in COPD.
Collapse
Affiliation(s)
- Eleni Gaki
- 2nd Respiratory Medicine Department, University of Athens Medical School, Attikon Hospital, Rimini 1, Athens, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Minas M, Kostikas K, Papaioannou AI, Mystridou P, Karetsi E, Georgoulias P, Liakos N, Pournaras S, Gourgoulianis KI. The association of metabolic syndrome with adipose tissue hormones and insulin resistance in patients with COPD without co-morbidities. COPD 2012; 8:414-20. [PMID: 22149401 DOI: 10.3109/15412555.2011.619600] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and metabolic syndrome represent common causes of morbidity and mortality in ageing populations. The effect of the co-existence of COPD and metabolic syndrome on adipose tissue hormones and insulin resistance as well as the differences between COPD patients with and without metabolic syndrome have not been adequately studied. The prevalence of metabolic syndrome, based on Adult Treatment Panel III (ATP III) criteria, was evaluated in 114 male patients with COPD without significant co-morbidities. Pulmonary functions tests (PFTs), arterial blood gases, quality of life and BODE index were assessed. Blood samples were obtained for the assessment of adipose tissue hormones and insulin resistance. The overall prevalence of metabolic syndrome was 21%, being more prevalent in earlier stages of COPD. Patients with COPD and metabolic syndrome were younger with higher body-mass index (BMI), had better pulmonary function, less static hyperinflation and air-trapping, better diffusing capacity for carbon monoxide and BODE index. These patients had higher levels of leptin, lower levels of adiponectin and increased insulin resistance, as expressed by HOMA index, compared with patients without metabolic syndrome. Metabolic syndrome was more prevalent in younger patients with less severe COPD. These patients may constitute a specific COPD phenotype with greater leptin to adiponectin imbalance and insulin resistance, despite smaller impairment in PFTs. The prognosis and differences of these patients compared with other COPD phenotypes needs to be determined in prospective studies.
Collapse
Affiliation(s)
- Markos Minas
- Respiratory Medicine Department, University of Thessaly Medical School, Larissa, Greece.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fountoulis GA, Minas M, Georgoulias P, Fezoulidis IV, Gourgoulianis KI, Vlychou M. Association of bone mineral density, parameters of bone turnover, and body composition in patients with chronic obstructive pulmonary disease. J Clin Densitom 2012; 15:217-23. [PMID: 22154434 DOI: 10.1016/j.jocd.2011.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 10/16/2011] [Accepted: 10/21/2011] [Indexed: 01/06/2023]
Abstract
Patients with chronic obstructive pulmonary disease (COPD) often develop osteoporosis. Many hormones regulate bone metabolism and body composition, and some of them are affected in COPD patients vs controls. In 46 COPD patients, we measured hip neck, total hip, lumbar spine, and whole-body T-score with dual-energy X-ray absorptiometry, parameters of body composition (body mass index [BMI], fat mass index [FMI], and fat-free mass index [FFMI]), and adiponectin, leptin, parathormone, osteocalcin, calcitonin, and insulin-like growth factor I (IGF-I) serum levels and correlated them with COPD stage. Our results suggest that total hip bone mineral density (BMD) is affected by FFMI and COPD stage; lumbar spine BMD is affected by FMI and COPD stage; and whole-body BMD is affected by BMI, COPD stage, and leptin. Adiponectin, parathormone, osteocalcin, calcitonin, and IGF-I levels were not significantly correlated to BMD at any of the measured sites. Our findings are in agreement with the current literature in that a decline in lung function is correlated to a decline in BMD.
Collapse
Affiliation(s)
- Georgios A Fountoulis
- Department of Diagnostic Radiology, School of Health Sciences, University of Thessaly, University Hospital of Larissa, Biopolis, Larissa, Greece
| | | | | | | | | | | |
Collapse
|
39
|
Harting JR, Gleason A, Romberger DJ, Von Essen SG, Qiu F, Alexis N, Poole JA. Chronic obstructive pulmonary disease patients have greater systemic responsiveness to ex vivo stimulation with swine dust extract and its components versus healthy volunteers. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2012; 75:1456-70. [PMID: 23116451 PMCID: PMC4001714 DOI: 10.1080/15287394.2012.722186] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by an airway and systemic inflammatory response. Bioaerosols/organic dusts are important agricultural pollutants that may lead to COPD. These environments are complex, containing a rich source of various microbial components. The objective of this study was to determine whether individuals with COPD have enhanced systemic responsiveness to settled swine facility organic dust extract (ODE) or its main pathogenic components (peptidoglycan [PGN], lipopolysaccharide [LPS]) versus healthy volunteers. A modified whole blood assay (WBA) that included occupational levels of ODE and concentrations of LPS and PGN found in ODE was used to determine systemic responsiveness (mediator release), and sputum inflammatory markers were measured to explore for systemic and airway associations. Sputum samples were evaluated for cell counts, and tumor necrosis factor (TNF)-α, interleukin (IL)-8/CXCL8, IL-6, and IL-10. Ex vivo whole blood stimulation with ODE, LPS, and PGN each resulted in significant mediator release in all subjects, with the highest occurring with ODE; PGN resulted in significantly enhanced TNF-α and IL-8 as compared to LPS. COPD subjects demonstrated greater systemic responsiveness using the modified WBA versus healthy controls. Within COPD subjects, blood baseline TNF-α, IL-8, and IL-10 and ODE-, PGN-, and LPS-stimulated IL-8 levels significantly correlated with lung function. In conclusion, dust-induced mediator release was robust, and PGN, in part, resembled dust-induced mediator release. Subjects with COPD demonstrated increased mediator release following ex vivo whole blood stimulation with bioaerosol components, suggesting that circulating blood cells in COPD subjects may be primed to respond greater to microbial/inflammatory insult.
Collapse
Affiliation(s)
- Janel R. Harting
- Omaha Veterans Administration Medical Center, Omaha, NE 68105
- Pulmonary, Critical Care, Sleep & Allergy Division; Department of Medicine, University of Nebraska Medical Center, 985300 The Nebraska Medical Center, Omaha, NE 68198-5300
| | - Angela Gleason
- Omaha Veterans Administration Medical Center, Omaha, NE 68105
- Pulmonary, Critical Care, Sleep & Allergy Division; Department of Medicine, University of Nebraska Medical Center, 985300 The Nebraska Medical Center, Omaha, NE 68198-5300
| | - Debra J. Romberger
- Omaha Veterans Administration Medical Center, Omaha, NE 68105
- Pulmonary, Critical Care, Sleep & Allergy Division; Department of Medicine, University of Nebraska Medical Center, 985300 The Nebraska Medical Center, Omaha, NE 68198-5300
| | | | - Fang Qiu
- College of Public Health, University of Nebraska Medical Center, 985300 The Nebraska Medical Center, Omaha, NE 68198-5300
| | - Neil Alexis
- University of North Carolina School of Medicine, Center for Environmental Medicine, Asthma & Lung Biology, Chapel Hill, NC 27599-7310
| | - Jill A. Poole
- Omaha Veterans Administration Medical Center, Omaha, NE 68105
- Pulmonary, Critical Care, Sleep & Allergy Division; Department of Medicine, University of Nebraska Medical Center, 985300 The Nebraska Medical Center, Omaha, NE 68198-5300
| |
Collapse
|
40
|
García-Rio F, Romero D, Lores V, Casitas R, Hernanz A, Galera R, Alvarez-Sala R, Torres I. Dynamic Hyperinflation, Arterial Blood Oxygen, and Airway Oxidative Stress in Stable Patients With COPD. Chest 2011; 140:961-969. [DOI: 10.1378/chest.10-2788] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
41
|
Bazeghi N, Gerds TA, Budtz-Jørgensen E, Hove J, Vestbo J. Exhaled nitric oxide measure using multiple flows in clinically relevant subgroups of COPD. Respir Med 2011; 105:1338-44. [DOI: 10.1016/j.rmed.2011.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/17/2011] [Accepted: 03/21/2011] [Indexed: 10/18/2022]
|
42
|
Todd NW, Jeudy J, Lavania S, Franks TJ, Galvin JR, Deepak J, Britt EJ, Atamas SP. Centrilobular emphysema combined with pulmonary fibrosis results in improved survival. FIBROGENESIS & TISSUE REPAIR 2011; 4:6. [PMID: 21324139 PMCID: PMC3055815 DOI: 10.1186/1755-1536-4-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 02/15/2011] [Indexed: 12/21/2022]
Abstract
Background We hypothesized that, in patients with pulmonary fibrosis combined with emphysema, clinical characteristics and outcomes may differ from patients with pulmonary fibrosis without emphysema. We identified 102 patients who met established criteria for pulmonary fibrosis. The amount of emphysema (numerical score) and type of emphysema (centrilobular, paraseptal, or mixed) were characterized in each patient. Clinical characteristics, pulmonary function tests and patient survival were analysed. Results Based on the numerical emphysema score, patients were classified into those having no emphysema (n = 48), trivial emphysema (n = 26) or advanced emphysema (n = 28). Patients with advanced emphysema had a significantly higher amount of smoking in pack/years than patients with no emphysema or trivial emphysema (P < 0.0001). Median survival [1st, 3rd quartiles] of patients with advanced emphysema was 63 [36, 82] months compared to 29 [18, 49] months in patients without emphysema and 32 [19, 48] months in patients with trivial emphysema (P < 0.001). Median forced vital capacity (FVC) and total lung capacity (TLC) were higher in the advanced emphysema group compared to patients with no emphysema (P < 0.01 and P < 0.001, respectively), whereas median DLCO did not differ among groups and was overall low. Within the advanced emphysema group (n = 28), further characterization of the type of emphysema was performed and, within these subgroups of patients, survival was 75 [58, 85] months for patients with centrilobular emphysema, 75 [48, 85] months for patients with mixed centrilobular/paraseptal emphysema, and 24 [22, 35] months for patients with paraseptal emphysema (P < 0.01). Patients with advanced paraseptal emphysema had similar survival times to patients without emphysema. Conclusions Patients with pulmonary fibrosis combined with advanced centrilobular or mixed emphysema have an improved survival compared with patients with pulmonary fibrosis without emphysema, with trivial emphysema or with advanced paraseptal emphysema.
Collapse
Affiliation(s)
- Nevins W Todd
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Ruwanpura SM, McLeod L, Miller A, Jones J, Bozinovski S, Vlahos R, Ernst M, Armes J, Bardin PG, Anderson GP, Jenkins BJ. Interleukin-6 promotes pulmonary emphysema associated with apoptosis in mice. Am J Respir Cell Mol Biol 2011; 45:720-30. [PMID: 21297079 DOI: 10.1165/rcmb.2010-0462oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The IL-6 cytokine family, which signals via the shared gp130 coreceptor, is linked with the pathogenesis of emphysema. However, the definitive mechanisms by which these cytokines cause emphysema remain ill-defined. We took an in vivo genetic complementation approach to identify the specific IL-6 cytokine family members and gp130-regulated cellular processes that cause emphysema. We used gp130(F/F) mice homozygous for a subtle knock-in mutation in gp130 that deregulates intracellular signaling by the IL-6 cytokine family. The gp130(F/F) mice spontaneously develop emphysema by age 6 months. Within the IL-6 cytokine family, only IL-6 was significantly up-regulated in the lungs of gp130(F/F) mice, and the genetic targeting of IL-6 in gp130(F/F) mice (gp130(F/F):IL-6(-/-)) prevented emphysema. By contrast, the genetic ablation of receptor signaling via IL-11, which like IL-6 signals via a gp130 homodimer and uses the same signaling machinery, failed to ameliorate emphysema in gp130(F/F) mice. Among the disease-associated processes examined, emphysema strongly correlated with elevated alveolar cell apoptosis. Acute (4-day) exposure to cigarette smoke (CS) further augmented the expression of IL-6 in lungs of gp130(F/F) mice, and subchronic (6-week) exposure to CS exacerbated emphysematous and apoptotic changes in the lungs of gp130(F/F) but not gp130(F/F): IL-6(-/-) mice. IL-6 is the main causative agent of IL-6 cytokine family-induced emphysema, and operates to induce apoptosis in the lung. We propose that the discrete targeting of IL-6 signaling may provide an effective therapeutic strategy against human lung disease.
Collapse
Affiliation(s)
- Saleela M Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton 3168, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Costa D, Cancelliero KM, Ike D, Laranjeira TL, Pantoni CBF, Borghi-Silva A. Strategy for respiratory exercise pattern associated with upper limb movements in COPD patients. Clinics (Sao Paulo) 2011; 66:299-305. [PMID: 21484050 PMCID: PMC3059857 DOI: 10.1590/s1807-59322011000200020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Accepted: 11/26/2010] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Upper limb exercises are frequently used in respiratory physiotherapy, with UL elevation and controlled inspiratory timing. However, the use of expiration during upper limb elevation appears to be a strategy that could minimize the action of accessory muscles in patients with chronic obstructive pulmonary disease. In this context, little is known about the synchrony of upper limb (UL) movements associated with breathing. The aim of this study was to investigate the respiratory pattern of chronic obstructive pulmonary disease patients during different UL exercises associated with respiratory exercises. METHODS Fifteen chronic obstructive pulmonary disease patients participated in this study. Respiratory pattern analysis by inductance plethysmography was performed during four types of upper limb exercises, two shoulder flexion-extension (one associated with inspiratory time during the concentric phase and the other associated with expiratory time) and two shoulder abduction-adduction (same timing as above). Statistical analysis was performed by the Kolmogorov-Smirnov test and ANOVA with Tukey tests (p< 0.05). RESULTS The thoracoabdominal coordination measurements increased in the two exercises using both inspiration during shoulder flexion (PhRIB: 172%; PhREB: 131%; PhRTB: 142% and PhAng: 238%) as well as in shoulder horizontal abduction (PhRIB: 145%; PhREB: 109%; PhRTB: 130% and PhAng: 229%), differing from the exercises with expiration at the time of shoulder flexion and horizontal abduction. CONCLUSION The exercises performed with inverted respiratory time produced less asynchrony and can be used as important strategies during physical exercise programs in these patients.
Collapse
Affiliation(s)
- Dirceu Costa
- Respiratory Physiotherapy Laboratory, Federal University of Sao Carlos, Sao Paulo, Brazil.
| | | | | | | | | | | |
Collapse
|
45
|
The effects of sleep hypoxia on coagulant factors and hepatic inflammation in emphysematous rats. PLoS One 2010; 5:e13201. [PMID: 20949089 PMCID: PMC2950855 DOI: 10.1371/journal.pone.0013201] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 09/13/2010] [Indexed: 01/24/2023] Open
Abstract
Objectives To develop a sleep hypoxia (SH) in emphysema (SHE) rat model and to explore whether SHE results in more severe hepatic inflammation than emphysema alone and whether the inflammation changes levels of coagulant/anticoagulant factors synthesized in the liver. Methods Seventy-five rats were put into 5 groups: SH control (SHCtrl), treated with sham smoke exposure (16 weeks) and SH exposure (12.5% O2, 3 h/d, latter 8 weeks); emphysema control (ECtrl), smoke exposure and sham SH exposure (21% O2); short SHE (SHEShort), smoke exposure and short SH exposure (1.5 h/d); mild SHE (SHEMild), smoke exposure and mild SH exposure (15% O2); standard SHE (SHEStand), smoke exposure and SH exposure. Therefore, ECtrl, SHEShort, SHEMild and SHEStand group were among emphysematous groups. Arterial blood gas (ABG) data was obtained during preliminary tests. After exposure, hepatic inflammation (interleukin -6 [IL-6] mRNA and protein, tumor necrosis factor α [TNFα] mRNA and protein) and liver coagulant/anticoagulant factors (antithrombin [AT], fibrinogen [FIB] and Factor VIII [F VIII]) were evaluated. SPSS 11.5 software was used for statistical analysis. Results Characteristics of emphysema were obvious in emphysematous groups and ABGs reached SH criteria on hypoxia exposure. Hepatic inflammation parameters and coagulant factors are the lowest in SHCtrl and the highest in SHEStand while AT is the highest in SHCtrl and the lowest in SHEStand. Inflammatory cytokines of liver correlate well with coagulant factors positively and with AT negatively. Conclusions When SH is combined with emphysema, hepatic inflammation and coagulability enhance each other synergistically and produce a more significant liver-derivative inflammatory and prothrombotic status.
Collapse
|
46
|
de Torres JP, Bastarrika G, Zagaceta J, Sáiz-Mendiguren R, Alcaide AB, Seijo LM, Montes U, Campo A, Zulueta JJ. Emphysema presence, severity, and distribution has little impact on the clinical presentation of a cohort of patients with mild to moderate COPD. Chest 2010; 139:36-42. [PMID: 20705800 DOI: 10.1378/chest.10-0984] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Phenotypic characterization of patients with COPD may have potential prognostic and therapeutic implications. Available information on the relationship between emphysema and the clinical presentation in patients with COPD is limited to advanced stages of the disease. The objective of this study was to describe emphysema presence, severity, and distribution and its impact on clinical presentation of patients with mild to moderate COPD. METHODS One hundred fifteen patients with COPD underwent clinical and chest CT scan evaluation for the presence, severity, and distribution of emphysema. Patients with and without emphysema and with different forms of emphysema distribution (upper/lower/core/peel) were compared. The impact of emphysema severity and distribution on clinical presentation was determined. RESULTS Fifty percent of the patients had mild homogeneously distributed emphysema (1.84; 0.76%-4.77%). Upper and core zones had the more severe degree of emphysema. Patients with emphysema were older, more frequently men, and had lower FEV(1)%, higher total lung capacity percentage, and lower diffusing capacity of the lung for carbon monoxide. No differences were found between the clinical or physiologic parameters of the different emphysema distributions. CONCLUSIONS In patients with mild to moderate COPD, although the presence of emphysema has an impact on physiologic presentation, its severity and distribution seem to have little impact on clinical presentation.
Collapse
Affiliation(s)
- Juan P de Torres
- Department of Pulmonology, University Clinic of Navarra, Pamplona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|