1
|
Nadeem SA, Comellas AP, Chan K, Hoffman EA, Fain SB, Saha PK. Automated CT-based measurements of radial and longitudinal expansion of airways due to breathing-related lung volume change. Med Phys 2025; 52:2316-2329. [PMID: 39704489 PMCID: PMC11972036 DOI: 10.1002/mp.17592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/04/2024] [Accepted: 12/07/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Respiratory function is impaired in chronic obstructive pulmonary disease (COPD). Automation of multi-volume CT-based measurements of different components of breathing-related airway deformations will help understand multi-pathway impairments in respiratory mechanics in COPD. PURPOSE To develop and evaluate multi-volume chest CT-based automated measurements of breathing-related radial and longitudinal expansion of individual airways between inspiratory and expiratory lung volumes. METHODS We developed a method to compute breathing-related airway deformation metrics and applied it to total lung capacity (TLC) and functional residual capacity (FRC) chest CT scans. The computational pipeline involves: (1) segmentation of airways; (2) skeletonization of airways; (3) labeling of anatomical airway segments at TLC and FRC; and (4) computation of radial and longitudinal expansion metrics of individual airways across lung volumes. Radial expansion (∆CSA) of an airway is computed as the percent change of its cross-sectional area (CSA) between two lung volumes. Longitudinal expansion (∆L) of an airway is computed as the percent change in its airway path-length from the carina between lung volumes. These measures are summarized at different airway anatomic generations. Agreement of automated measures with their manually derived values was examined in terms of concordance correlation coefficient (CCC) of automated measures with those derived using manual outlining. Intra-class correlation coefficient (ICC) of automated measures from repeat CT scans (n = 37) was computed to assess repeatability. The method was also applied to a set of participants from the Genetic Epidemiology of COPD (COPDGene) Iowa cohort, distributed across COPD severity groups (n = 4 × 60). RESULTS The CCC values for the automated ∆CSA measure with manually derived values were 0.930 at the trachea, 0.898 at primary bronchi, and greater than 0.95 at pre-segmental and segmental airways; these CCC values were consistently greater than 0.95 for ∆L at all airway generations. ICC values for repeatability of ∆CSA were 0.974, 0.950, 0.943, and 0.901 at trachea, primary bronchi, pre-segmental, and segmental airways, respectively; these ICC values for ∆L were 0.973, 0.954, and 0.952 at primary bronchi, pre-segmental, and segmental airways, respectively. ∆CSA values were significantly reduced (p < 0.001) with increasing COPD severity at each of primary bronchi, pre-segmental, and segmental airways. Significantly lower ∆L values were observed for moderate (p = 0.042 at pre-segmental and p = 0.037 at segmental) and severe (p = 0.019 at pre-segmental and p < 0.001 at segmental) COPD groups as compared to the preserved lung function group. Body mass index (BMI) and smoking status were found to significantly associate with ∆CSA at segmental airways (r = 0.17 and -0.19, respectively; significance threshold = 0.13), while age and sex were significantly associated with ∆L (r = -0.21 and -0.17, respectively); COPD severity was significantly associated with both ∆CSA and ∆L (r = -0.35 and -0.22, respectively). CONCLUSION Our CT-based automated measures of breathing-related radial and longitudinal expansion of airways are repeatable and in agreement with manually derived values. Automation of different airway mechanical biomarkers and their observed significant associations with age, sex, BMI, smoking, and COPD severity establish an effective tool to investigate multi-pathway impairments of respiratory mechanics in COPD and other lung diseases.
Collapse
Affiliation(s)
- Syed Ahmed Nadeem
- Department of Radiology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Alejandro P. Comellas
- Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
| | - Kung‐Sik Chan
- Department of Statistics and Actuarial Science, College of Liberal Arts and SciencesUniversity of IowaIowa CityIowaUSA
| | - Eric A. Hoffman
- Department of Radiology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Department of Internal Medicine, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Department of Biomedical Engineering, College of EngineeringUniversity of IowaIowa CityIowaUSA
| | - Sean B. Fain
- Department of Radiology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Department of Biomedical Engineering, College of EngineeringUniversity of IowaIowa CityIowaUSA
- Department of Electrical and Computer Engineering, College of EngineeringUniversity of IowaIowa CityIowaUSA
| | - Punam K. Saha
- Department of Radiology, Carver College of MedicineUniversity of IowaIowa CityIowaUSA
- Department of Electrical and Computer Engineering, College of EngineeringUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
2
|
Adami A, Duan F, Calmelat RA, Chen Z, Casaburi R, Rossiter HB. SEVERITY OF LUNG OBSTRUCTION AND OLDER AGE, BUT NOT PHYSICAL ACTIVITY, PREDICT LOCOMOTOR MUSCLE OXIDATIVE IMPAIRMENT IN COPD. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.28.25321301. [PMID: 39974145 PMCID: PMC11838953 DOI: 10.1101/2025.01.28.25321301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Low muscle oxidative capacity is an extrapulmonary manifestation of chronic obstructive pulmonary disease (COPD) with unclear etiology. We sought to determine clinical and behavioral features associated with muscle oxidative capacity in smokers with and without COPD and never smoker individuals. Methods 243 adults enrolled in the Muscle Health Study, an observational study ancillary to COPDGene. Gastrocnemius oxidative capacity was measured by near-infrared spectroscopy from muscle oxygen uptake recovery rate constant (k). Physical activity by accelerometry (vector magnitude units, VMU/min). Pulmonary assessments included spirometry (FEV1%predicted), diffusing capacity (DLCO), and quantitative chest computed tomography (CT). Eighty-seven variables related to COPD features were considered. Variables selected by univariate analysis of log-transformed k with p≤0.20, and filtered by machine learning, were entered into multivariable linear regression to determine association with k. Results 241(99%) participants were allocated to analysis. FEV1%predicted, DLCO, CT, pack-years, age and VMU/min were among 24 variables selected by univariate analysis. After machine learning filtering on 161(66%) cases with complete data, 11 variables were included in multivariable analysis. Only FEV1%predicted, age and race were significantly associated with k (R2=0.26). Model coefficients equate a 10% lower FEV1%predicted to a 4.4% lower k, or 10-years of aging to a 9.7% lower k. In 118 cases with CT available, FEV1%predicted and age remained associated with k (R2=0.24). Physical activity was not retained in any model. Conclusions Locomotor muscle oxidative capacity was positively associated with FEV1%predicted and negatively associated with age. Physical activity or radiographic COPD manifestations were not significantly associated with muscle oxidative impairment.
Collapse
Affiliation(s)
- Alessandra Adami
- Department of Kinesiology, College of Health Sciences, University of Rhode Island, Kingston, RI, USA
| | - Fenghai Duan
- Department of Biostatistics and Center for Statistical Sciences, Brown University School of Public Health, Providence, RI, USA
| | - Robert A. Calmelat
- Institute of Respiratory Medicine and Exercise Physiology, Division of Pulmonary and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor–UCLA Medical Center, Torrance, CA, USA
| | - Zeyu Chen
- Department of Biostatistics and Center for Statistical Sciences, Brown University School of Public Health, Providence, RI, USA
| | - Richard Casaburi
- Institute of Respiratory Medicine and Exercise Physiology, Division of Pulmonary and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor–UCLA Medical Center, Torrance, CA, USA
| | - Harry B. Rossiter
- Institute of Respiratory Medicine and Exercise Physiology, Division of Pulmonary and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor–UCLA Medical Center, Torrance, CA, USA
| |
Collapse
|
3
|
Sun M, Gao M, Luo M, Wang T, Ruan X, Chen Q, Qin J. Causal relationship between air pollution and chronic obstructive pulmonary disease in European and East Asian populations: a Mendelian randomization study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:3944-3959. [PMID: 38563461 DOI: 10.1080/09603123.2024.2334781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Epidemiologic studies have suggested a possible association between air pollution and chronic obstructive pulmonary disease (COPD), but it is controversial and difficult to draw causal inferences. Five methods were adopted to evaluate the causal relationship between air pollution and COPD in European and East Asian populations by using MR Analysis. A statistically significant causal relationship between PM2.5 and COPD was observed in the European population (OR: 2.34; 95% CI: 1.06-5.05; p = 0.033). Statistical significance remained after adjustment for confounding factors (adjusted OR: 2.28; 95% CI: 1.01-5.20; p = 0.048). In East Asian populations, PM2.5 absorbance, a proxy for black carbon, was statistically associated with COPD (OR: 1.41; 95% CI: 1.09-1.81; p = 0.007). We did not adjust for confounders in East Asian populations, as the association was independent of known confounders (e.g. smoking, respiratory tract infections, etc.). In conclusion, increased concentrations of PM2.5 and PM2.5 absorbance were associated with an increased risk of COPD.
Collapse
Affiliation(s)
- Mengting Sun
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Ming Gao
- Department of Geriatric Medicine, Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manjun Luo
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Tingting Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Xiaorui Ruan
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Qian Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jiabi Qin
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China
| |
Collapse
|
4
|
Nakamura S, Wakahara K, Majima S, Yokoi E, Fukutani E, Otsuki R, Iwano S, Chen-Yoshikawa TF, Kinoshita F, Abe T, Sashio T, Kimura T, Izuhara K, Hashimoto N, Ishii M, Hasegawa Y. Blood eosinophil count correlates with alveolar damage in emphysema-predominant COPD. BMC Pulm Med 2024; 24:510. [PMID: 39396940 PMCID: PMC11472558 DOI: 10.1186/s12890-024-03320-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Although blood eosinophil count is recognized as a useful biomarker for the management of chronic obstructive pulmonary disease (COPD), the impact of eosinophils in COPD has not been fully elucidated. Here we aimed to investigate the relationships between the blood eosinophil count and various clinical parameters including lung structural changes. METHODS Ninety-three COPD patients without concomitant asthma were prospectively enrolled in this study. Blood eosinophil count, serum IgE level, serum periostin level, and chest computed tomography (CT) scans were evaluated. Eosinophilic COPD was defined as COPD with a blood eosinophil count ≧ 300/µL. We examined the correlation between the blood eosinophil count and structural changes graded by chest CT, focusing specifically on thin airway wall (WT thin) and thick airway wall (WT thick) groups. In a separate cohort, the number of eosinophils in the peripheral lungs of COPD patients with low attenuation area (LAA) on chest CT was assessed using lung resection specimens. RESULTS The mean blood eosinophil count was 212.1/µL, and 18 patients (19.3%) were categorized as having eosinophilic COPD. In the whole group analysis, the blood eosinophil count correlated only with blood white blood cells, blood basophils, C-reactive protein level, and sputum eosinophils. However, the blood eosinophil count positively correlated with the percentage of LAA and negatively correlated with the diffusing capacity for carbon monoxide in the WT thin group. Lung specimen data showed an increased number of eosinophils in the peripheral lungs of COPD patients with LAA on chest CT scans compared to normal controls. CONCLUSIONS Some COPD patients without concomitant asthma showed a phenotype of high blood eosinophils. Alveolar damage may be related to eosinophilic inflammation in patients with COPD without asthma and thickening of the central airway wall.
Collapse
Affiliation(s)
- Saya Nakamura
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Keiko Wakahara
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Suguru Majima
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Eito Yokoi
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Eriko Fukutani
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Ryo Otsuki
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shingo Iwano
- Department of Radiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Fumie Kinoshita
- Data Coordinating Center, Department of Advanced Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Takashi Abe
- Department of Respiratory Medicine, Ogaki Municipal Hospital, Ogaki, Japan
| | - Toyokazu Sashio
- Department of Respiratory Medicine, Meijo Hospital, Nagoya, Japan
| | - Tomoki Kimura
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Kenji Izuhara
- Division of Medicine Biochemistry, Department of Biomolecular Sciences, Saga Medical School, Saga, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
- National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
5
|
Kwee AKAL, Andrinopoulou ER, van der Veer T, Gallardo-Estrella L, Charbonnier JP, Humphries SM, Lynch DA, Tiddens HAWM, de Jong PA, Pompe E. Higher small pulmonary artery and vein volume on computed tomography is associated with mortality in current and former smokers. EBioMedicine 2024; 108:105366. [PMID: 39353280 PMCID: PMC11464249 DOI: 10.1016/j.ebiom.2024.105366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND In chronic obstructive pulmonary disease (COPD), vascular alterations have been shown to contribute to hypoxia and pulmonary hypertension, but the independent contribution of small vessel abnormalities to mortality remains unclear. METHODS We quantified artery and vein dimensions on computed tomography (CT) down to 0.2 mm. Small vessel volumes (<1 mmᴓ) were normalized by body surface area. In 7903 current and former smokers of the COPDGene study (53.2% male) the independent contribution of small artery and small vein volume to all-cause mortality was tested in multivariable Cox models. Additionally, we calculated the 95th percentile of small arteries and veins in 374 never smokers to create two groups: normal and high small artery or vein volume. We describe clinical, physiological and imaging characteristics of subjects with a high small artery and high small vein volume. FINDINGS Both high small artery and high small vein volumes were independently associated with mortality with an adjusted hazard ratio of 1.07 [1.01, 1.14] and 1.34 [1.21, 1.49] per mL/m2 increase, respectively. In COPDGene, 447 (5.7%) had high small artery volume and 519 (9.1%) subjects had high small vein volume and both had more emphysema, more air trapping and more severe coronary calcium. INTERPRETATION In smokers, abnormally high volumes in small arteries and veins are both relevant for mortality, which urges investigations into the aetiology of small pulmonary vessels and cardiac function in smokers. FUNDING Award Number U01-HL089897 and U01-HL089856 from the NHLBI. COPD Foundation with contributions from AstraZeneca, Boehringer Ingelheim, Genentech, GlaxoSmithKline, Novartis, Pfizer, Siemens, and Sunovion.
Collapse
Affiliation(s)
- Anastasia K A L Kwee
- University Medical Centre Utrecht and Utrecht University, Dept. Radiology, Utrecht, Netherlands.
| | - Eleni-Rosalina Andrinopoulou
- Erasmus Medical Centre, Dept. Biostatistics, Rotterdam, Netherlands; Erasmus Medical Centre, Dept. Epidemiology, Rotterdam, Netherlands
| | - Tjeerd van der Veer
- Leiden University Medical Centre, Dept. Pulmonology, Leiden, Netherlands; Erasmus Medical Centre, Dept. Pulmonology, Rotterdam, Netherlands
| | | | | | | | - David A Lynch
- National Jewish Health, Dept. Radiology, Denver, USA
| | - Harm A W M Tiddens
- Erasmus Medical Centre, Dept. Pulmonology, Rotterdam, Netherlands; Thirona B.V., Nijmegen, Netherlands; Erasmus MC-Sophia Children's Hospital, Dept. Paediatric Pulmonology and Allergology, Rotterdam, Netherlands
| | - Pim A de Jong
- University Medical Centre Utrecht and Utrecht University, Dept. Radiology, Utrecht, Netherlands
| | - Esther Pompe
- University Medical Centre Utrecht and Utrecht University, Dept. Radiology, Utrecht, Netherlands; Meander Medical Centre, Dept. Radiology, Amersfoort, Netherlands
| |
Collapse
|
6
|
Ogoshi T, Yatera K, Mukae H, Tsutsui M. Role of Nitric Oxide Synthases in Respiratory Health and Disease: Insights from Triple Nitric Oxide Synthases Knockout Mice. Int J Mol Sci 2024; 25:9317. [PMID: 39273265 PMCID: PMC11395504 DOI: 10.3390/ijms25179317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The system of nitric oxide synthases (NOSs) is comprised of three isoforms: nNOS, iNOS, and eNOS. The roles of NOSs in respiratory diseases in vivo have been studied by using inhibitors of NOSs and NOS-knockout mice. Their exact roles remain uncertain, however, because of the non-specificity of inhibitors of NOSs and compensatory up-regulation of other NOSs in NOS-KO mice. We addressed this point in our triple-n/i/eNOSs-KO mice. Triple-n/i/eNOSs-KO mice spontaneously developed pulmonary emphysema and displayed exacerbation of bleomycin-induced pulmonary fibrosis as compared with wild-type (WT) mice. Triple-n/i/eNOSs-KO mice exhibited worsening of hypoxic pulmonary hypertension (PH), which was reversed by treatment with sodium nitrate, and WT mice that underwent triple-n/i/eNOSs-KO bone marrow transplantation (BMT) also showed aggravation of hypoxic PH compared with those that underwent WT BMT. Conversely, ovalbumin-evoked asthma was milder in triple-n/i/eNOSs-KO than WT mice. These results suggest that the roles of NOSs are different in different pathologic states, even in the same respiratory diseases, indicating the diversity of the roles of NOSs. In this review, we describe these previous studies and discuss the roles of NOSs in respiratory health and disease. We also explain the current state of development of inorganic nitrate as a new drug for respiratory diseases.
Collapse
Affiliation(s)
- Takaaki Ogoshi
- Department of Respiratory Medicine, Kokura Memorial Hospital, 1-1 Asano, Kokura-kita-ku, Kitakyushu 803-0802, Japan;
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan;
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan;
| | - Hiroshi Mukae
- Department of Respiratory Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan;
| | - Masato Tsutsui
- Department of Pharmacology, Graduate School of Medicine, University of the Ryukyus, 207 Uehara, Nishihara, Okinawa 903-0215, Japan
| |
Collapse
|
7
|
Kaji S, Tanabe N, Maetani T, Shiraishi Y, Sakamoto R, Oguma T, Suzuki K, Terada K, Fukui M, Muro S, Sato S, Hirai T. Quantification of Airway Structures by Persistent Homology. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:2758-2768. [PMID: 38478453 DOI: 10.1109/tmi.2024.3376683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
We propose two types of novel morphological metrics for quantifying the geometry of tubular structures on computed tomography (CT) images. We apply our metrics to identify irregularities in the airway of patients with chronic obstructive pulmonary disease (COPD) and demonstrate that they provide complementary information to the conventional metrics used to assess COPD, such as the tissue density distribution in lung parenchyma and the wall area ratio of the segmented airway. The three-dimensional shape of the airway and its abstraction as a rooted tree with the root at the trachea carina are automatically extracted from a lung CT volume, and the two metrics are computed based on a mathematical tool called persistent homology; treeH0 quantifies the distribution of branch lengths to assess the complexity of the tree-like structure and radialH0 quantifies the irregularities in the luminal radius along the airway. We show our metrics are associated with clinical outcomes.
Collapse
|
8
|
Elbehairy AF, Marshall H, Naish JH, Wild JM, Parraga G, Horsley A, Vestbo J. Advances in COPD imaging using CT and MRI: linkage with lung physiology and clinical outcomes. Eur Respir J 2024; 63:2301010. [PMID: 38548292 DOI: 10.1183/13993003.01010-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 03/16/2024] [Indexed: 05/04/2024]
Abstract
Recent years have witnessed major advances in lung imaging in patients with COPD. These include significant refinements in images obtained by computed tomography (CT) scans together with the introduction of new techniques and software that aim for obtaining the best image whilst using the lowest possible radiation dose. Magnetic resonance imaging (MRI) has also emerged as a useful radiation-free tool in assessing structural and more importantly functional derangements in patients with well-established COPD and smokers without COPD, even before the existence of overt changes in resting physiological lung function tests. Together, CT and MRI now allow objective quantification and assessment of structural changes within the airways, lung parenchyma and pulmonary vessels. Furthermore, CT and MRI can now provide objective assessments of regional lung ventilation and perfusion, and multinuclear MRI provides further insight into gas exchange; this can help in structured decisions regarding treatment plans. These advances in chest imaging techniques have brought new insights into our understanding of disease pathophysiology and characterising different disease phenotypes. The present review discusses, in detail, the advances in lung imaging in patients with COPD and how structural and functional imaging are linked with common resting physiological tests and important clinical outcomes.
Collapse
Affiliation(s)
- Amany F Elbehairy
- Department of Chest Diseases, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester and Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Helen Marshall
- POLARIS, Imaging, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Josephine H Naish
- MCMR, Manchester University NHS Foundation Trust, Manchester, UK
- Bioxydyn Limited, Manchester, UK
| | - Jim M Wild
- POLARIS, Imaging, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Insigneo Institute for in silico Medicine, Sheffield, UK
| | - Grace Parraga
- Robarts Research Institute, Western University, London, ON, Canada
- Department of Medical Biophysics, Western University, London, ON, Canada
- Division of Respirology, Western University, London, ON, Canada
| | - Alexander Horsley
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester and Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, The University of Manchester and Manchester University NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| |
Collapse
|
9
|
Ji Y, Chen L, Yang J, Yang X, Yang F. Quantitative assessment of airway wall thickness in COPD patients with interstitial lung abnormalities. Front Med (Lausanne) 2023; 10:1280651. [PMID: 38146423 PMCID: PMC10749311 DOI: 10.3389/fmed.2023.1280651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/24/2023] [Indexed: 12/27/2023] Open
Abstract
Background Whether the airway is involved in the pathogenesis of interstitial lung abnormalities (ILA) is not well understood. Also the impact of ILA on lung function in COPD patients remains controversial. We aimed to assess the quantitative CT measurements of airway wall thickness (AWT) and lung function according to ILA status in COPD patients. Methods 157 COPD patients discharged from our hospital from August 1, 2019 through August 31, 2022 who underwent chest CT imagings and pulmonary function tests were retrospectively enrolled. Linear regression analysis and multiple models were used to analyze associations between quantitative assessment of airway wall changes and the presence of ILA. Results In 157 COPD patients, 23 patients (14.6%) had equivocal ILA, 42 patients (26.8%) had definite ILA. The definite ILA group had the highest measurements of Pi10 (square root of theoretical airway wall area with a lumen perimeter of 10 mm), segmental AWT and segmental WA% (percentage of wall area), whereas the no ILA group had the lowest measurements of Pi10, segmental AWT and segmental WA%. In the adjusted analyses (adjusted by age, sex, body mass index, smoking intensity, COPD GOLD stage, lung function, slice thickness and scanner type), compared to COPD patients without ILA, the measurements of Pi10, segmental AWT and segmental WA% were higher in definite ILA group with differences of 0.225 mm (p = 0.012), 0.152 mm (p < 0.001), 4.8% (p < 0.001) respectively. COPD patients with definite ILA tended to have higher FEV1% predicted, FVC% predicted and lower MMEF75/25% predicted, but there were no statistically differences among the three groups. Conclusion Our study demonstrates the higher AWT measures in COPD patients with ILA compared to the patients without ILA. These findings suggest that the airway may be involved in the pathogenesis of ILA.
Collapse
Affiliation(s)
- Yingying Ji
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Leqing Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jinrong Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xiangying Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Fan Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
10
|
Lim WH, Jeong S, Park CM. Cigarette smoking and disproportionate changes of thoracic skeletal muscles in low-dose chest computed tomography. Sci Rep 2023; 13:20110. [PMID: 37978301 PMCID: PMC10656498 DOI: 10.1038/s41598-023-46360-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023] Open
Abstract
Association between smoking intensity and the quantity and quality of thoracic skeletal muscles (TSMs) remains unexplored. Skeletal muscle index (SMI; skeletal muscle area/height2) and percentage of normal attenuation muscle area (NAMA%) were measured to represent the quantity and quality of the skeletal muscles, respectively, and quantification was performed in pectoralis muscle at aortic arch (AA-PM), TSM at carina (C-TSM), erector spinae muscle at T12 (T12-ESM), and skeletal muscle at L1 (L1-SM). Among the 258 men (median age, 62 years [IQR: 58-69]), 183 were current smokers (median smoking intensity, 40 pack-years [IQR: 30-46]). SMI and NAMA% of AA-PM significantly decreased with pack-year (β = - 0.028 and - 0.076; P < 0.001 and P = 0.021, respectively). Smoking intensity was inversely associated with NAMA% of C-TSM (β = - 0.063; P = 0.001), whereas smoking intensity showed a borderline association with SMI of C-TSM (β = - 0.023; P = 0.057). Smoking intensity was associated with the change in NAMA% of L1-SM (β = - 0.040; P = 0.027), but was not associated with SMI of L1-SM (P > 0.05). Neither NAMA% nor SMI of T12-ESM was affected by smoking intensity (P > 0.05). In conclusion, smoking intensity was associated with the change of TSMs. Its association varied according to the location of TSMs, with the most associated parts being the upper (AA-PM) and middle TSMs (C-TSM).
Collapse
Affiliation(s)
- Woo Hyeon Lim
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Suhyun Jeong
- Department of Radiology, Namwon Medical Center, 365 Chungjeong-no, Namwon, Jeollabuk-do, 55726, Republic of Korea
| | - Chang Min Park
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Institute of Radiation Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea.
- Institute of Medical and Biological Engineering, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Zhuan B, Ma HH, Zhang BC, Li P, Wang X, Yuan Q, Yang Z, Xie J. Identification of non-small cell lung cancer with chronic obstructive pulmonary disease using clinical symptoms and routine examination: a retrospective study. Front Oncol 2023; 13:1158948. [PMID: 37576878 PMCID: PMC10419203 DOI: 10.3389/fonc.2023.1158948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/03/2023] [Indexed: 08/15/2023] Open
Abstract
Background Patients with non-small cell lung cancer (NSCLC) and patients with NSCLC combined with chronic obstructive pulmonary disease (COPD) have similar physiological conditions in early stages, and the latter have shorter survival times and higher mortality rates. The purpose of this study was to develop and compare machine learning models to identify future diagnoses of COPD combined with NSCLC patients based on the patient's disease and routine clinical data. Methods Data were obtained from 237 patients with COPD combined with NSCLC as well as NSCLC admitted to Ningxia Hui Autonomous Region People's Hospital from October 2013 to July 2022. Six machine learning algorithms (K-nearest neighbor, logistic regression, eXtreme gradient boosting, support vector machine, naïve Bayes, and artificial neural network) were used to develop prediction models for NSCLC combined with COPD. Sensitivity, specificity, positive predictive value, negative predictive value, accuracy, F1 score, Mathews correlation coefficient (MCC), Kappa, area under the receiver operating characteristic curve (AUROC)and area under the precision-recall curve (AUPRC) were used as performance indicators to evaluate the performance of the models. Results 135 patients with NSCLC combined with COPD, 102 patients with NSCLC were included in the study. The results showed that pulmonary function and emphysema were important risk factors and that the support vector machine-based identification model showed optimal performance with accuracy:0.946, recall:0.940, specificity:0.955, precision:0.972, npv:0.920, F1 score:0.954, MCC:0.893, Kappa:0.888, AUROC:0.975, AUPRC:0.987. Conclusion The use of machine learning tools combining clinical symptoms and routine examination data features is suitable for identifying the risk of concurrent NSCLC in COPD patients.
Collapse
Affiliation(s)
- Bing Zhuan
- Department of Respiratory Medicine, Ningxia Hui Autonomous Region People’s Hospital, Yinchuan, Ningxia, China
- Department of Respiratory Medicine, Ningxia Hui Autonomous Region People’s Hospital Affiliated to Ningxia Medical University, Yinchuan, Ningxia, China
| | - Hong-Hong Ma
- Department of Respiratory Medicine, Ningxia Hui Autonomous Region People’s Hospital, Yinchuan, Ningxia, China
- Department of Respiratory Medicine, Ningxia Hui Autonomous Region People’s Hospital Affiliated to Ningxia Medical University, Yinchuan, Ningxia, China
| | - Bo-Chao Zhang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou, Jiangsu, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Ping Li
- Department of Respiratory Medicine, Ningxia Hui Autonomous Region People’s Hospital, Yinchuan, Ningxia, China
- Department of Respiratory Medicine, Ningxia Hui Autonomous Region People’s Hospital Affiliated to Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xi Wang
- Department of Respiratory Medicine, Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Qun Yuan
- Department of Respiratory Medicine, Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Zhao Yang
- Department of Respiratory Medicine, Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jun Xie
- Department of Thoracic Surgery, Affiliated Suzhou Science and Technology Town Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
12
|
Byon JH, Jin GY, Han YM, Choi EJ, Chae KJ, Park EH. Quantitative CT Analysis Based on Smoking Habits and Chronic Obstructive Pulmonary Disease in Patients with Normal Chest CT. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2023; 84:900-910. [PMID: 37559818 PMCID: PMC10407071 DOI: 10.3348/jksr.2022.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/26/2022] [Accepted: 11/13/2022] [Indexed: 08/11/2023]
Abstract
PURPOSE To assess normal CT scans with quantitative CT (QCT) analysis based on smoking habits and chronic obstructive pulmonary disease (COPD). MATERIALS AND METHODS From January 2013 to December 2014, 90 male patients with normal chest CT and quantification analysis results were enrolled in our study [non-COPD never-smokers (n = 38) and smokers (n = 45), COPD smokers (n = 7)]. In addition, an age-matched cohort study was performed for seven smokers with COPD. The square root of the wall area of a hypothetical bronchus of internal perimeter 10 mm (Pi10), skewness, kurtosis, mean lung attenuation (MLA), and percentage of low attenuation area (%LAA) were evaluated. RESULTS Among patients without COPD, the Pi10 of smokers (4.176 ± 0.282) was about 0.1 mm thicker than that of never-smokers (4.070 ± 0.191, p = 0.047), and skewness and kurtosis of smokers (2.628 ± 0.484 and 6.448 ± 3.427) were lower than never-smokers (2.884 ± 0.624, p = 0.038 and 8.594 ± 4.944, p = 0.02). The Pi10 of COPD smokers (4.429 ± 0.435, n = 7) was about 0.4 mm thicker than never-smokers without COPD (3.996 ± 0.115, n = 14, p = 0.005). There were no significant differences in MLA and %LAA between groups (p > 0.05). CONCLUSION Even on normal CT scans, QCT showed that the airway walls of smokers are thicker than never-smokers regardless of COPD and it preceded lung parenchymal changes.
Collapse
|
13
|
Abstract
Understanding how biases originate in medical technologies and developing safeguards to identify, mitigate, and remove their harms are essential to ensuring equal performance in all individuals. Drawing upon examples from pulmonary medicine, this article describes how bias can be introduced in the physical aspects of the technology design, via unrepresentative data, or by conflation of biological with social determinants of health. It then can be perpetuated by inadequate evaluation and regulatory standards. Research demonstrates that pulse oximeters perform differently depending on patient race and ethnicity. Pulmonary function testing and algorithms used to predict healthcare needs are two additional examples of medical technologies with racial and ethnic biases that may perpetuate health disparities.
Collapse
Affiliation(s)
- Michael W. Sjoding
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA,Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan, USA,Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan, USA
| | - Sardar Ansari
- Weil Institute for Critical Care Research and Innovation, Ann Arbor, Michigan, USA; .,Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Thomas S. Valley
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA,Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan, USA,Center for Bioethics and Social Sciences in Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
14
|
Lahmar Z, Ahmed E, Fort A, Vachier I, Bourdin A, Bergougnoux A. Hedgehog pathway and its inhibitors in chronic obstructive pulmonary disease (COPD). Pharmacol Ther 2022; 240:108295. [PMID: 36191777 DOI: 10.1016/j.pharmthera.2022.108295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022]
Abstract
COPD affects millions of people and is now ranked as the third leading cause of death worldwide. This largely untreatable chronic airway disease results in irreversible destruction of lung architecture. The small lung hypothesis is now supported by epidemiological, physiological and clinical studies. Accordingly, the early and severe COPD phenotype carries the most dreadful prognosis and finds its roots during lung growth. Pathophysiological mechanisms remain poorly understood and implicate individual susceptibility (genetics), a large part of environmental factors (viral infections, tobacco consumption, air pollution) and the combined effects of those triggers on gene expression. Genetic susceptibility is most likely involved as the disease is severe and starts early in life. The latter observation led to the identification of Mendelian inheritance via disease-causing variants of SERPINA1 - known as the basis for alpha-1 anti-trypsin deficiency, and TERT. In the last two decades multiple genome wide association studies (GWAS) identified many single nucleotide polymorphisms (SNPs) associated with COPD. High significance SNPs are located in 4q31 near HHIP which encodes an evolutionarily highly conserved physiological inhibitor of the Hedgehog signaling pathway (HH). HHIP is critical to several in utero developmental lung processes. It is also implicated in homeostasis, injury response, epithelial-mesenchymal transition and tumor resistance to apoptosis. A few studies have reported decreased HHIP RNA and protein levels in human adult COPD lungs. HHIP+/- murine models led to emphysema. HH pathway inhibitors, such as vismodegib and sonidegib, are already validated in oncology, whereas other drugs have evidenced in vitro effects. Targeting the Hedgehog pathway could lead to a new therapeutic avenue in COPD. In this review, we focused on the early and severe COPD phenotype and the small lung hypothesis by exploring genetic susceptibility traits that are potentially treatable, thus summarizing promising therapeutics for the future.
Collapse
Affiliation(s)
- Z Lahmar
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France
| | - E Ahmed
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Fort
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - I Vachier
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bourdin
- Department of Respiratory Diseases, CHU de Montpellier, Montpellier, France; PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France
| | - A Bergougnoux
- PhyMedExp, Univ Montpellier, Inserm U1046, CNRS UMR 9214, Montpellier, France; Laboratoire de Génétique Moléculaire et de Cytogénomique, CHU de Montpellier, Montpellier, France.
| |
Collapse
|
15
|
Bhatt SP, Bodduluri S, Nakhmani A, Kim YI, Reinhardt JM, Hoffman EA, Motahari A, Wilson CG, Humphries SM, Regan EA, DeMeo DL. Sex Differences in Airways at Chest CT: Results from the COPDGene Cohort. Radiology 2022; 305:699-708. [PMID: 35916677 PMCID: PMC9713451 DOI: 10.1148/radiol.212985] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 05/10/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
Background The prevalence of chronic obstructive pulmonary disease (COPD) in women is fast approaching that in men, and women experience greater symptom burden. Although sex differences in emphysema have been reported, differences in airways have not been systematically characterized. Purpose To evaluate whether structural differences in airways may underlie some of the sex differences in COPD prevalence and clinical outcomes. Materials and Methods In a secondary analyses of a multicenter study of never-, current-, and former-smokers enrolled from January 2008 to June 2011 and followed up longitudinally until November 2020, airway disease on CT images was quantified using seven metrics: airway wall thickness, wall area percent, and square root of the wall thickness of a hypothetical airway with internal perimeter of 10 mm (referred to as Pi10) for airway wall; and lumen diameter, airway volume, total airway count, and airway fractal dimension for airway lumen. Least-squares mean values for each airway metric were calculated and adjusted for age, height, ethnicity, body mass index, pack-years of smoking, current smoking status, total lung capacity, display field of view, and scanner type. In ever-smokers, associations were tested between each airway metric and postbronchodilator forced expiratory volume in 1 second (FEV1)-to-forced vital capacity (FVC) ratio, modified Medical Research Council dyspnea scale, St George's Respiratory Questionnaire score, and 6-minute walk distance. Multivariable Cox proportional hazards models were created to evaluate the sex-specific association between each airway metric and mortality. Results In never-smokers (n = 420), men had thicker airway walls than women as quantified on CT images for segmental airway wall area percentage (least-squares mean, 47.68 ± 0.61 [standard error] vs 45.78 ± 0.55; difference, -1.90; P = .02), whereas airway lumen dimensions were lower in women than men after accounting for height and total lung capacity (segmental lumen diameter, 8.05 mm ± 0.14 vs 9.05 mm ± 0.16; difference, -1.00 mm; P < .001). In ever-smokers (n = 9363), men had greater segmental airway wall area percentage (least-squares mean, 52.19 ± 0.16 vs 48.89 ± 0.18; difference, -3.30; P < .001), whereas women had narrower segmental lumen diameter (7.80 mm ± 0.05 vs 8.69 mm ± 0.04; difference, -0.89; P < .001). A unit change in each of the airway metrics (higher wall or lower lumen measure) resulted in lower FEV1-to-FVC ratio, more dyspnea, poorer respiratory quality of life, lower 6-minute walk distance, and worse survival in women compared with men (all P < .01). Conclusion Airway lumen sizes quantified at chest CT were smaller in women than in men after accounting for height and lung size, and these lower baseline values in women conferred lower reserves against respiratory morbidity and mortality for equivalent changes compared with men. © RSNA, 2022 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Surya P. Bhatt
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Sandeep Bodduluri
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Arie Nakhmani
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Young-il Kim
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Joseph M. Reinhardt
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Eric A. Hoffman
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Amin Motahari
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Carla G. Wilson
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Stephen M. Humphries
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Elizabeth A. Regan
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| | - Dawn L. DeMeo
- From the UAB Lung Imaging Lab (S.P.B., S.B., A.N.), UAB Lung Health
Center (S.P.B., S.B.), Division of Pulmonary, Allergy and Critical Care Medicine
(S.P.B., S.B.), Department of Electrical and Computer Engineering (A.N.), and
Division of Preventive Medicine (Y.I.K.), University of Alabama at Birmingham,
1720 2nd Ave S, THT 422, Birmingham, AL 35294; Roy J. Carver Department
of Biomedical Engineering (J.M.R.) and Department of Radiology (E.A.H., A.M.),
University of Iowa, Iowa City, Iowa; Departments of Biostatistics and
Bioinformatics (C.G.W.), Radiology (S.M.H.), and Medicine (E.A.R.), National
Jewish Health, Denver, Colo; and Channing Division of Network Medicine and the
Division of Pulmonary and Critical Care Medicine, Brigham and Women's
Hospital, Harvard Medical School, Boston, Mass (D.L.D.)
| |
Collapse
|
16
|
Kaza N, Lin VY, Stanford D, Hussain SS, Falk Libby E, Kim H, Borgonovi M, Conrath K, Mutyam V, Byzek SA, Tang LP, Trombley JE, Rasmussen L, Schoeb T, Leung HM, Tearney GJ, Raju SV, Rowe SM. Evaluation of a novel CFTR potentiator in COPD ferrets with acquired CFTR dysfunction. Eur Respir J 2022; 60:13993003.01581-2021. [PMID: 34916262 PMCID: PMC10079430 DOI: 10.1183/13993003.01581-2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 11/21/2021] [Indexed: 11/05/2022]
Abstract
RATIONALE The majority of chronic obstructive pulmonary disease (COPD) patients have chronic bronchitis, for which specific therapies are unavailable. Acquired cystic fibrosis transmembrane conductance regulator (CFTR) dysfunction is observed in chronic bronchitis, but has not been proven in a controlled animal model with airway disease. Furthermore, the potential of CFTR as a therapeutic target has not been tested in vivo, given limitations to rodent models of COPD. Ferrets exhibit cystic fibrosis-related lung pathology when CFTR is absent and COPD with bronchitis following cigarette smoke exposure. OBJECTIVES To evaluate CFTR dysfunction induced by smoking and test its pharmacological reversal by a novel CFTR potentiator, GLPG2196, in a ferret model of COPD with chronic bronchitis. METHODS Ferrets were exposed for 6 months to cigarette smoke to induce COPD and chronic bronchitis and then treated with enteral GLPG2196 once daily for 1 month. Electrophysiological measurements of ion transport and CFTR function, assessment of mucociliary function by one-micron optical coherence tomography imaging and particle-tracking microrheology, microcomputed tomography imaging, histopathological analysis and quantification of CFTR protein and mRNA expression were used to evaluate mechanistic and pathophysiological changes. MEASUREMENTS AND MAIN RESULTS Following cigarette smoke exposure, ferrets exhibited CFTR dysfunction, increased mucus viscosity, delayed mucociliary clearance, airway wall thickening and airway epithelial hypertrophy. In COPD ferrets, GLPG2196 treatment reversed CFTR dysfunction, increased mucus transport by decreasing mucus viscosity, and reduced bronchial wall thickening and airway epithelial hypertrophy. CONCLUSIONS The pharmacologic reversal of acquired CFTR dysfunction is beneficial against pathological features of chronic bronchitis in a COPD ferret model.
Collapse
Affiliation(s)
- Niroop Kaza
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,Equal contributions
| | - Vivian Y Lin
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,Equal contributions
| | - Denise Stanford
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,Equal contributions
| | - Shah S Hussain
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily Falk Libby
- The Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Harrison Kim
- Dept of Radiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Venkateshwar Mutyam
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephen A Byzek
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Li Ping Tang
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - John E Trombley
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lawrence Rasmussen
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Trenton Schoeb
- Dept of Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA.,Animal Resources Program, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui Min Leung
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Guillermo J Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Dept of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - S Vamsee Raju
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.,The Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, USA.,Dept of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL, USA.,Co-senior authors
| | - Steven M Rowe
- Dept of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA .,The Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, USA.,Dept of Cell, Developmental, and Integrative Biology, The University of Alabama at Birmingham, Birmingham, AL, USA.,Co-senior authors
| |
Collapse
|
17
|
Zhang L, Jiang B, Wisselink HJ, Vliegenthart R, Xie X. COPD identification and grading based on deep learning of lung parenchyma and bronchial wall in chest CT images. Br J Radiol 2022; 95:20210637. [PMID: 35143286 PMCID: PMC10993953 DOI: 10.1259/bjr.20210637] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 01/20/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Chest CT can display the main pathogenic factors of chronic obstructive pulmonary disease (COPD), emphysema and airway wall remodeling. This study aims to establish deep convolutional neural network (CNN) models using these two imaging markers to diagnose and grade COPD. METHODS Subjects who underwent chest CT and pulmonary function test (PFT) from one hospital (n = 373) were retrospectively included as the training cohort, and subjects from another hospital (n = 226) were used as the external test cohort. According to the PFT results, all subjects were labeled as Global Initiative for Chronic Obstructive Lung Disease (GOLD) Grade 1, 2, 3, 4 or normal. Two DenseNet-201 CNNs were trained using CT images of lung parenchyma and bronchial wall to generate two corresponding confidence levels to indicate the possibility of COPD, then combined with logistic regression analysis. Quantitative CT was used for comparison. RESULTS In the test cohort, CNN achieved an area under the curve of 0.899 (95%CI: 0.853-0.935) to determine the existence of COPD, and an accuracy of 81.7% (76.2-86.7%), which was significantly higher than the accuracy 68.1% (61.6%-74.2%) using quantitative CT method (p < 0.05). For three-way (normal, GOLD 1-2, and GOLD 3-4) and five-way (normal, GOLD 1, 2, 3, and 4) classifications, CNN reached accuracies of 77.4 and 67.9%, respectively. CONCLUSION CNN can identify emphysema and airway wall remodeling on CT images to infer lung function and determine the existence and severity of COPD. It provides an alternative way to detect COPD using the extensively available chest CT. ADVANCES IN KNOWLEDGE CNN can identify the main pathological changes of COPD (emphysema and airway wall remodeling) based on CT images, to infer lung function and determine the existence and severity of COPD. CNN reached an area under the curve of 0.853 to determine the existence of COPD in the external test cohort. The CNN approach provides an alternative and effective way for early detection of COPD using extensively used chest CT, as an important alternative to pulmonary function test.
Collapse
Affiliation(s)
- Lin Zhang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao
Tong University School of Medicine,
Shanghai, China
| | - Beibei Jiang
- Radiology Department, Shanghai General Hospital, Shanghai Jiao
Tong University School of Medicine,
Shanghai, China
| | - Hendrik Joost Wisselink
- Radiology Department, University of Groningen, University
Medical Center Groningen,
Groningen, The Netherlands
| | - Rozemarijn Vliegenthart
- Radiology Department, University of Groningen, University
Medical Center Groningen,
Groningen, The Netherlands
| | - Xueqian Xie
- Radiology Department, Shanghai General Hospital, Shanghai Jiao
Tong University School of Medicine,
Shanghai, China
| |
Collapse
|
18
|
Dudurych I, Muiser S, McVeigh N, Kerstjens HAM, van den Berge M, de Bruijne M, Vliegenthart R. Bronchial wall parameters on CT in healthy never-smoking, smoking, COPD, and asthma populations: a systematic review and meta-analysis. Eur Radiol 2022; 32:5308-5318. [PMID: 35192013 PMCID: PMC9279249 DOI: 10.1007/s00330-022-08600-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/14/2021] [Accepted: 01/29/2022] [Indexed: 11/25/2022]
Abstract
Objective Research on computed tomography (CT) bronchial parameter measurements shows that there are conflicting results on the values for bronchial parameters in the never-smoking, smoking, asthma, and chronic obstructive pulmonary disease (COPD) populations. This review assesses the current CT methods for obtaining bronchial wall parameters and their comparison between populations. Methods A systematic review of MEDLINE and Embase was conducted following PRISMA guidelines (last search date 25th October 2021). Methodology data was collected and summarised. Values of percentage wall area (WA%), wall thickness (WT), summary airway measure (Pi10), and luminal area (Ai) were pooled and compared between populations. Results A total of 169 articles were included for methodologic review; 66 of these were included for meta-analysis. Most measurements were obtained from multiplanar reconstructions of segmented airways (93 of 169 articles), using various tools and algorithms; third generation airways in the upper and lower lobes were most frequently studied. COPD (12,746) and smoking (15,092) populations were largest across studies and mostly consisted of men (median 64.4%, IQR 61.5 – 66.1%). There were significant differences between populations; the largest WA% was found in COPD (mean SD 62.93 ± 7.41%, n = 6,045), and the asthma population had the largest Pi10 (4.03 ± 0.27 mm, n = 442). Ai normalised to body surface area (Ai/BSA) (12.46 ± 4 mm2, n = 134) was largest in the never-smoking population. Conclusions Studies on CT-derived bronchial parameter measurements are heterogenous in methodology and population, resulting in challenges to compare outcomes between studies. Significant differences between populations exist for several parameters, most notably in the wall area percentage; however, there is a large overlap in their ranges. Key Points • Diverse methodology in measuring airways contributes to overlap in ranges of bronchial parameters among the never-smoking, smoking, COPD, and asthma populations. • The combined number of never-smoking participants in studies is low, limiting insight into this population and the impact of participant characteristics on bronchial parameters. • Wall area percent of the right upper lobe apical segment is the most studied (87 articles) and differentiates all except smoking vs asthma populations. Supplementary Information The online version contains supplementary material available at 10.1007/s00330-022-08600-1.
Collapse
Affiliation(s)
- Ivan Dudurych
- Department of Radiology, EB49, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9700RB, Groningen, The Netherlands
| | - Susan Muiser
- Department of Pulmonology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Niall McVeigh
- Department of Cardiothoracic Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - Huib A M Kerstjens
- Department of Pulmonology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Department of Pulmonology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Marleen de Bruijne
- Department of Radiology and Nuclear Medicine, Biomedical Imaging Group Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Rozemarijn Vliegenthart
- Department of Radiology, EB49, University Medical Centre Groningen, University of Groningen, Hanzeplein 1, 9700RB, Groningen, The Netherlands.
| |
Collapse
|
19
|
Kooner HK, McIntosh MJ, Desaigoudar V, Rayment JH, Eddy RL, Driehuys B, Parraga G. Pulmonary functional MRI: Detecting the structure-function pathologies that drive asthma symptoms and quality of life. Respirology 2022; 27:114-133. [PMID: 35008127 PMCID: PMC10025897 DOI: 10.1111/resp.14197] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/09/2021] [Accepted: 12/12/2021] [Indexed: 12/21/2022]
Abstract
Pulmonary functional MRI (PfMRI) using inhaled hyperpolarized, radiation-free gases (such as 3 He and 129 Xe) provides a way to directly visualize inhaled gas distribution and ventilation defects (or ventilation heterogeneity) in real time with high spatial (~mm3 ) resolution. Both gases enable quantitative measurement of terminal airway morphology, while 129 Xe uniquely enables imaging the transfer of inhaled gas across the alveolar-capillary tissue barrier to the red blood cells. In patients with asthma, PfMRI abnormalities have been shown to reflect airway smooth muscle dysfunction, airway inflammation and remodelling, luminal occlusions and airway pruning. The method is rapid (8-15 s), cost-effective (~$300/scan) and very well tolerated in patients, even in those who are very young or very ill, because unlike computed tomography (CT), positron emission tomography and single-photon emission CT, there is no ionizing radiation and the examination takes only a few seconds. However, PfMRI is not without limitations, which include the requirement of complex image analysis, specialized equipment and additional training and quality control. We provide an overview of the three main applications of hyperpolarized noble gas MRI in asthma research including: (1) inhaled gas distribution or ventilation imaging, (2) alveolar microstructure and finally (3) gas transfer into the alveolar-capillary tissue space and from the tissue barrier into red blood cells in the pulmonary microvasculature. We highlight the evidence that supports a deeper understanding of the mechanisms of asthma worsening over time and the pathologies responsible for symptoms and disease control. We conclude with a summary of approaches that have the potential for integration into clinical workflows and that may be used to guide personalized treatment planning.
Collapse
Affiliation(s)
- Harkiran K Kooner
- Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Marrissa J McIntosh
- Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Vedanth Desaigoudar
- Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Jonathan H Rayment
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rachel L Eddy
- Centre of Heart Lung Innovation, Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bastiaan Driehuys
- Center for In Vivo Microscopy, Duke University Medical Centre, Durham, North Carolina, USA
| | - Grace Parraga
- Robarts Research Institute, Western University, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Division of Respirology, Department of Medicine, Western University, London, Ontario, Canada
- School of Biomedical Engineering, Western University, London, Ontario, Canada
| |
Collapse
|
20
|
Nishiki K, Nojiri M, Kato R, Shinomiya S, Oikawa T, Ishizaki T, Toga H, Mizuno S. Serum Creatinine/Cystatin C Ratio Associated with Cross-Sectional Area of Erector Spinae Muscles and Pulmonary Function in Patients with Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2022; 16:3513-3524. [PMID: 34992359 PMCID: PMC8713710 DOI: 10.2147/copd.s339243] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/06/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose Muscle atrophy is a major clinical feature of chronic obstructive pulmonary disease (COPD) and is considered a predictor of mortality in COPD patients. Recently, the cross-sectional area (CSA) of the erector spinae muscles measured by chest computed tomography (CT) scans (ESMCSA) has been reported as a clinical parameter reflecting disease severity and future prognosis in patients with COPD. In addition, the serum creatinine (Cr)/cystatin C (CysC) ratio has been considered a quantitative marker of residual muscle mass, because serum Cr levels are affected by muscle mass, and correction by CysC counteracts the effect of renal function on serum Cr levels. The purpose of this study was to assess whether the serum Cr level corrected by serum CysC can be used as a predictive marker of pulmonary function and disease severity in patients with COPD. Patients and Methods A total of 99 patients without COPD and 201 patients with COPD, with a smoking history of more than 10 pack-years were enrolled in this study, and serum Cr and CysC levels were measured. On chest high-resolution CT images, %low attenuation area (LAA%) (≤960 Hounsfield units (HU)) and ESMCSA at the Th12 level were identified. Results There was a significant correlation between the ESMCSA and the Cr/CysC ratio. The Cr/CysC ratio was significantly associated with forced vital capacity (FVC) and forced expiratory volume in 1 second (FEV1) values, especially in former smokers. Conclusion The serum Cr/CysC ratio could be a convenient substitute for the measurement of muscle atrophy and pulmonary function testing in patients with COPD.
Collapse
Affiliation(s)
- Kazuaki Nishiki
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Masafumi Nojiri
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Ryo Kato
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Shohei Shinomiya
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Taku Oikawa
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Takeshi Ishizaki
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Hirohisa Toga
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| | - Shiro Mizuno
- Department of Respiratory Medicine, Kanazawa Medical University, Ishikawa, Japan
| |
Collapse
|
21
|
Zhao D, Abbasi A, Casaburi R, Adami A, Tiller NB, Yuan W, Yee C, Jendzjowsky NG, MacDonald DM, Kunisaki KM, Stringer WW, Porszasz J, Make BJ, Bowler RP, Rossiter HB, On behalf of the COPDGene Investigators. Identifying a Heart Rate Recovery Criterion After a 6-Minute Walk Test in COPD. Int J Chron Obstruct Pulmon Dis 2021; 16:2545-2560. [PMID: 34511898 PMCID: PMC8427685 DOI: 10.2147/copd.s311572] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Slow heart rate recovery (HRR) after exercise is associated with autonomic dysfunction and increased mortality. What HRR criterion at 1-minute after a 6-minute walk test (6MWT) best defines pulmonary impairment?. STUDY DESIGN AND METHODS A total of 5008 phase 2 COPDGene (NCT00608764) participants with smoking history were included. A total of 2127 had COPD and, of these, 385 were followed-up 5-years later. Lung surgery, transplant, bronchiectasis, atrial fibrillation, heart failure and pacemakers were exclusionary. HR was measured from pulse oximetry at end-walk and after 1-min seated recovery. A receiver operator characteristic (ROC) identified optimal HRR cut-off. Generalized linear regression determined HRR association with spirometry, chest CT, symptoms and exacerbations. RESULTS HRR after 6MWT (bt/min) was categorized in quintiles: ≤5 (23.0% of participants), 6-10 (20.7%), 11-15 (18.9%), 16-22 (18.5%) and ≥23 (18.9%). Compared to HRR≤5, HRR≥11 was associated with (p<0.001): lower pre-walk HR and 1-min post HR; greater end-walk HR; greater 6MWD; greater FEV1%pred; lower airway wall area and wall thickness. HRR was positively associated with FEV1%pred and negatively associated with airway wall thickness. An optimal HRR ≤10 bt/min yielded an area under the ROC curve of 0.62 (95% CI 0.58-0.66) for identifying FEV1<30%pred. HRR≥11 bt/min was the lowest HRR associated with consistently less impairment in 6MWT, spirometry and CT variables. In COPD, HRR≤10 bt/min was associated with (p<0.001): ≥2 exacerbations in the previous year (OR=1.76[1.33-2.34]); CAT≥10 (OR=1.42[1.18-1.71]); mMRC≥2 (OR=1.42[1.19-1.69]); GOLD 4 (OR=1.98[1.44-2.73]) and GOLD D (OR=1.51[1.18-1.95]). HRR≤10 bt/min was predicted COPD exacerbations at 5-year follow-up (RR=1.83[1.07-3.12], P=0.027). CONCLUSION HRR≤10 bt/min after 6MWT in COPD is associated with more severe expiratory flow limitation, airway wall thickening, worse dyspnoea and quality of life, and future exacerbations, suggesting that an abnormal HRR≤10 bt/min after a 6MWT may be used in a comprehensive assessment in COPD for risk of severity, symptoms and future exacerbations.
Collapse
Affiliation(s)
- Dongxing Zhao
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, People’s Republic of China
| | - Asghar Abbasi
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Richard Casaburi
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Alessandra Adami
- Department of Kinesiology, University of Rhode Island, Kingston, RI, USA
| | - Nicholas B Tiller
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Wei Yuan
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- Respiratory Medicine Department, Beijing Friendship Hospital Affiliated of Capital Medical University, Beijing, 100050, People’s Republic of China
| | | | - Nicholas G Jendzjowsky
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David M MacDonald
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Ken M Kunisaki
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - William W Stringer
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Janos Porszasz
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | | | - Harry B Rossiter
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - On behalf of the COPDGene Investigators
- Rehabilitation Clinical Trials Center, Division of Respiratory and Critical Care Physiology and Medicine, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, People’s Republic of China
- Department of Kinesiology, University of Rhode Island, Kingston, RI, USA
- Respiratory Medicine Department, Beijing Friendship Hospital Affiliated of Capital Medical University, Beijing, 100050, People’s Republic of China
- MemorialCare Long Beach Medical Center, Long Beach, CA, USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
- National Jewish Health, Denver, CO, USA
| |
Collapse
|
22
|
Krings JG, Goss CW, Lew D, Samant M, McGregor MC, Boomer J, Bacharier LB, Sheshadri A, Hall C, Brownell J, Schechtman KB, Peterson S, McEleney S, Mauger DT, Fahy JV, Fain SB, Denlinger LC, Israel E, Washko G, Hoffman E, Wenzel SE, Castro M. Quantitative CT metrics are associated with longitudinal lung function decline and future asthma exacerbations: Results from SARP-3. J Allergy Clin Immunol 2021; 148:752-762. [PMID: 33577895 PMCID: PMC8349941 DOI: 10.1016/j.jaci.2021.01.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/02/2020] [Accepted: 01/08/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND Currently, there is limited knowledge regarding which imaging assessments of asthma are associated with accelerated longitudinal decline in lung function. OBJECTIVES We aimed to assess whether quantitative computed tomography (qCT) metrics are associated with longitudinal decline in lung function and morbidity in asthma. METHODS We analyzed 205 qCT scans of adult patients with asthma and calculated baseline markers of airway remodeling, lung density, and pointwise regional change in lung volume (Jacobian measures) for each participant. Using multivariable regression models, we then assessed the association of qCT measurements with the outcomes of future change in lung function, future exacerbation rate, and changes in validated measurements of morbidity. RESULTS Greater baseline wall area percent (β = -0.15 [95% CI = -0.26 to -0.05]; P < .01), hyperinflation percent (β = -0.25 [95% CI = -0.41 to -0.09]; P < .01), and Jacobian gradient measurements (cranial-caudal β = 10.64 [95% CI = 3.79-17.49]; P < .01; posterior-anterior β = -9.14, [95% CI = -15.49 to -2.78]; P < .01) were associated with more severe future lung function decline. Additionally, greater wall area percent (rate ratio = 1.06 [95% CI = 1.01-1.10]; P = .02) and air trapping percent (rate ratio =1.01 [95% CI = 1.00-1.02]; P = .03), as well as lower decline in the Jacobian determinant mean (rate ratio = 0.58 [95% CI = 0.41-0.82]; P < .01) and Jacobian determinant standard deviation (rate ratio = 0.52 [95% CI = 0.32-0.85]; P = .01), were associated with a greater rate of future exacerbations. However, imaging metrics were not associated with clinically meaningful changes in scores on validated asthma morbidity questionnaires. CONCLUSIONS Baseline qCT measures of more severe airway remodeling, more small airway disease and hyperinflation, and less pointwise regional change in lung volumes were associated with future lung function decline and asthma exacerbations.
Collapse
Affiliation(s)
- James G Krings
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, St Louis, Mo
| | - Charles W Goss
- Division of Biostatistics, Washington University in St Louis School of Medicine, St Louis, Mo
| | - Daphne Lew
- Division of Biostatistics, Washington University in St Louis School of Medicine, St Louis, Mo
| | - Maanasi Samant
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, St Louis, Mo
| | - Mary Clare McGregor
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, St Louis, Mo
| | - Jonathan Boomer
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Kansas School of Medicine, Kansas City, Kan
| | - Leonard B Bacharier
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Ajay Sheshadri
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Tex
| | - Chase Hall
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Kansas School of Medicine, Kansas City, Kan
| | - Joshua Brownell
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wis
| | - Ken B Schechtman
- Division of Biostatistics, Washington University in St Louis School of Medicine, St Louis, Mo
| | | | | | - David T Mauger
- Division of Statistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University, Hershey, Pa
| | - John V Fahy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, the University of California San Francisco, San Francisco, Calif
| | - Sean B Fain
- Department of Radiology and Biomedical Engineering, University of Wisconsin, Madison, Wis
| | - Loren C Denlinger
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin, Madison, Wis
| | - Elliot Israel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Mass
| | - George Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Eric Hoffman
- Department of Radiology, Biomedical Engineering, and Medicine, University of Iowa, Iowa City, IA
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, the University of Pittsburgh, Pittsburgh, Pa
| | - Mario Castro
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Kansas School of Medicine, Kansas City, Kan.
| |
Collapse
|
23
|
Sin S, Choi HM, Lim J, Kim J, Bak SH, Choi SS, Park J, Lee JH, Oh YM, Lee MK, Hobbs BD, Cho MH, Silverman EK, Kim WJ. A genome-wide association study of quantitative computed tomographic emphysema in Korean populations. Sci Rep 2021; 11:16692. [PMID: 34404834 PMCID: PMC8371078 DOI: 10.1038/s41598-021-95887-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 06/28/2021] [Indexed: 11/18/2022] Open
Abstract
Emphysema is an important feature of chronic obstructive pulmonary disease (COPD). Genetic factors likely affect emphysema pathogenesis, but this question has predominantly been studied in those of European ancestry. In this study, we sought to determine genetic components of emphysema severity and characterize the potential function of the associated loci in Korean population. We performed a genome-wide association study (GWAS) on quantitative emphysema in subjects with or without COPD from two Korean COPD cohorts. We investigated the functional consequences of the loci using epigenetic annotation and gene expression data. We also compared our GWAS results with an epigenome-wide association study and previous differential gene expression analysis. In total, 548 subjects (476 [86.9%] male) including 514 COPD patients were evaluated. We identified one genome-wide significant SNP (P < 5.0 × 10-8), rs117084279, near PIBF1. We identified an additional 57 SNPs (P < 5.0 × 10-6) associated with emphysema in all subjects, and 106 SNPs (P < 5.0 × 10-6) in COPD patients. Of these candidate SNPs, 2 (rs12459249, rs11667314) near CYP2A6 were expression quantitative trait loci in lung tissue and a SNP (rs11214944) near NNMT was an expression quantitative trait locus in whole blood. Of note, rs11214944 was in linkage disequilibrium with variants in enhancer histone marks in lung tissue. Several genes near additional SNPs were identified in our previous EWAS study with nominal level of significance. We identified a novel SNP associated with quantitative emphysema on CT. Including the novel SNP, several candidate SNPs in our study may provide clues to the genetic etiology of emphysema in Asian populations. Further research and validation of the loci will help determine the genetic factors for the development of emphysema.
Collapse
Affiliation(s)
- Sooim Sin
- grid.412010.60000 0001 0707 9039Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - Hye-Mi Choi
- grid.412010.60000 0001 0707 9039Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jiwon Lim
- grid.412010.60000 0001 0707 9039Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jeeyoung Kim
- grid.412010.60000 0001 0707 9039Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea
| | - So Hyeon Bak
- grid.412010.60000 0001 0707 9039Department of Radiology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, Republic of Korea
| | - Sun Shim Choi
- grid.412010.60000 0001 0707 9039Division of Biomedical Convergence, College of Biomedical Science, and Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Jinkyeong Park
- grid.470090.a0000 0004 1792 3864Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| | - Jin Hwa Lee
- grid.255649.90000 0001 2171 7754Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Yeon-Mok Oh
- grid.267370.70000 0004 0533 4667Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Kyeong Lee
- grid.280664.e0000 0001 2110 5790Epidemiology Branch, Division of Intramural Research, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC USA
| | - Brian D. Hobbs
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Michael H. Cho
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Edwin K. Silverman
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA ,grid.38142.3c000000041936754XDivision of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Republic of Korea.
| |
Collapse
|
24
|
Zhang J, DeMeo DL, Silverman EK, Make BJ, Wade RC, Wells JM, Cho MH, Hobbs BD. Secondary polycythemia in chronic obstructive pulmonary disease: prevalence and risk factors. BMC Pulm Med 2021; 21:235. [PMID: 34261472 PMCID: PMC8278596 DOI: 10.1186/s12890-021-01585-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 07/06/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Secondary polycythemia is associated with cigarette smoking and chronic obstructive pulmonary disease (COPD). However, the prevalence of polycythemia in COPD and the contributing risk factors for polycythemia in COPD have not been extensively studied. METHODS We analyzed the presence of secondary polycythemia in current and former smokers with moderate to very severe COPD at the five-year follow-up visit in the observational COPDGene study. We used logistic regression to evaluate the association of polycythemia with age, sex, race, altitude, current smoking status, spirometry, diffusing capacity for carbon monoxide (DLCO), quantitative chest CT measurements (including emphysema, airway wall thickness, and pulmonary artery to aorta diameter ratio), resting hypoxemia, exercise-induced hypoxemia, and long-term oxygen therapy. RESULTS In a total of 1928 COPDGene participants with moderate to very severe COPD, secondary polycythemia was found in 97 (9.2%) male and 31 (3.5%) female participants. In a multivariable logistic model, severe resting hypoxemia (OR 3.50, 95% CI 1.41-8.66), impaired DLCO (OR 1.28 for each 10-percent decrease in DLCO % predicted, CI 1.09-1.49), male sex (OR 3.60, CI 2.20-5.90), non-Hispanic white race (OR 3.33, CI 1.71-6.50), current smoking (OR 2.55, CI 1.49-4.38), and enrollment in the Denver clinical center (OR 4.42, CI 2.38-8.21) were associated with higher risk for polycythemia. In addition, continuous (OR 0.13, CI 0.05-0.35) and nocturnal (OR 0.46, CI 0.21-0.97) supplemental oxygen were associated with lower risk for polycythemia. Results were similar after excluding participants with anemia and participants enrolled at the Denver clinical center. CONCLUSIONS In a large cohort of individuals with moderate to very severe COPD, male sex, current smoking, enrollment at the Denver clinical center, impaired DLCO, and severe hypoxemia were associated with increased risk for secondary polycythemia. Continuous or nocturnal supplemental oxygen use were associated with decreased risk for polycythemia.
Collapse
Affiliation(s)
- Jingzhou Zhang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Barry J Make
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - R Chad Wade
- Lung Health Center and the Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - J Michael Wells
- Lung Health Center and the Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Birmingham VA Medical Center, Birmingham, AL, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian D Hobbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Moll M, Jackson VE, Yu B, Grove ML, London SJ, Gharib SA, Bartz TM, Sitlani CM, Dupuis J, O'Connor GT, Xu H, Cassano PA, Patchen BK, Kim WJ, Park J, Kim KH, Han B, Barr RG, Manichaikul A, Nguyen JN, Rich SS, Lahousse L, Terzikhan N, Brusselle G, Sakornsakolpat P, Liu J, Benway CJ, Hall IP, Tobin MD, Wain LV, Silverman EK, Cho MH, Hobbs BD. A systematic analysis of protein-altering exonic variants in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2021; 321:L130-L143. [PMID: 33909500 PMCID: PMC8321852 DOI: 10.1152/ajplung.00009.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/15/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified regions associated with chronic obstructive pulmonary disease (COPD). GWASs of other diseases have shown an approximately 10-fold overrepresentation of nonsynonymous variants, despite limited exonic coverage on genotyping arrays. We hypothesized that a large-scale analysis of coding variants could discover novel genetic associations with COPD, including rare variants with large effect sizes. We performed a meta-analysis of exome arrays from 218,399 controls and 33,851 moderate-to-severe COPD cases. All exome-wide significant associations were present in regions previously identified by GWAS. We did not identify any novel rare coding variants with large effect sizes. Within GWAS regions on chromosomes 5q, 6p, and 15q, four coding variants were conditionally significant (P < 0.00015) when adjusting for lead GWAS single-nucleotide polymorphisms A common gasdermin B (GSDMB) splice variant (rs11078928) previously associated with a decreased risk for asthma was nominally associated with a decreased risk for COPD [minor allele frequency (MAF) = 0.46, P = 1.8e-4]. Two stop variants in coiled-coil α-helical rod protein 1 (CCHCR1), a gene involved in regulating cell proliferation, were associated with COPD (both P < 0.0001). The SERPINA1 Z allele was associated with a random-effects odds ratio of 1.43 for COPD (95% confidence interval = 1.17-1.74), though with marked heterogeneity across studies. Overall, COPD-associated exonic variants were identified in genes involved in DNA methylation, cell-matrix interactions, cell proliferation, and cell death. In conclusion, we performed the largest exome array meta-analysis of COPD to date and identified potential functional coding variants. Future studies are needed to identify rarer variants and further define the role of coding variants in COPD pathogenesis.
Collapse
Affiliation(s)
- Matthew Moll
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Victoria E Jackson
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Bing Yu
- School of Public Health, University of Texas Health Science Center, Houston, Texas
| | - Megan L Grove
- School of Public Health, University of Texas Health Science Center, Houston, Texas
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services Research, Research Triangle Park, Durham, North Carolina
| | - Sina A Gharib
- Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - George T O'Connor
- Division of Pulmonary, Allergy, Sleep, and Critical Care Medicine, Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Patricia A Cassano
- Division of Nutritional Sciences, Cornell University, Ithaca, New York
- Division of Epidemiology, Department of Population Health Sciences, Weill Cornell Medicine, New York, New York
| | | | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jinkyeong Park
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Department of Internal Medicine, Dongguk University Ilsan Hospital, Goyang-Si, Gyeonggi-do, South Korea
| | - Kun Hee Kim
- Department of Convergence Medicine and Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Buhm Han
- Department of Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - R Graham Barr
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Jennifer N Nguyen
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Lies Lahousse
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Natalie Terzikhan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Guy Brusselle
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Phuwanat Sakornsakolpat
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jiangyuan Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Christopher J Benway
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ian P Hall
- NIHR Nottingham Biomedical Research Centre, Queen's Medical Centre, Nottingham, United Kingdom
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, United Kingdom
| | - Edwin K Silverman
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Michael H Cho
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Brian D Hobbs
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
26
|
Bhatt SP, Bodduluri S, Kizhakke Puliyakote AS, Oelsner EC, Nakhmani A, Lynch DA, Wilson CG, Fortis S, Kim V. Structural airway imaging metrics are differentially associated with persistent chronic bronchitis. Thorax 2021; 76:343-349. [PMID: 33408194 PMCID: PMC8225550 DOI: 10.1136/thoraxjnl-2020-215853] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/09/2020] [Accepted: 11/23/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Chronic bronchitis (CB) is strongly associated with cigarette smoking, but not all smokers develop CB. We aimed to evaluate whether measures of structural airway disease on CT are differentially associated with CB. METHODS In smokers between ages 45 and 80 years, and with Global Initiative for Obstructive Lung Disease stages 0-4, CB was defined by the classic definition. Airway disease on CT was quantified by (i) wall area percent (WA%) of segmental airways; (ii) Pi10, the square root of the wall area of a hypothetical airway with 10 mm internal perimeter; (iii) total airway count (TAC) and (iv) airway fractal dimension (AFD), a measure of the complex branching pattern and remodelling of airways. CB was also assessed at the 5-year follow-up visit. MEASUREMENTS AND MAIN RESULTS Of 8917 participants, 1734 (19.4%) had CB at baseline. Airway measures were significantly worse in those with CB compared with those without CB: WA% 54.5 (8.8) versus 49.8 (8.3); Pi10 2.58 (0.67) versus 2.28 (0.59) mm; TAC 156.7 (81.6) versus 177.8 (91.1); AFD 1.477 (0.091) versus 1.497 (0.092) (all p<0.001). On follow-up of 5517 participants at 5 years, 399 (7.2%) had persistent CB. With adjustment for between-visits changes in smoking status and lung function, greater WA% and Pi10 were associated with significantly associated with persistent CB, adjusted OR per SD change 1.75, 95% CI 1.56 to 1.97; p<0.001 and 1.66, 95% CI 1.42 to 1.86; p<0.001, respectively. Higher AFD and TAC were associated with significantly lower odds of persistent CB, adjusted OR per SD change 0.76, 95% CI 0.67 to 0.86; p<0.001 and 0.69, 95% CI 0.60 to 0.80; p<0.001, respectively. CONCLUSIONS Higher baseline AFD and TAC are associated with a lower risk of persistent CB, irrespective of changes in smoking status, suggesting preserved airway structure can confer a reserve against CB.
Collapse
Affiliation(s)
- Surya P Bhatt
- Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sandeep Bodduluri
- Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Arie Nakhmani
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, AL, USA
- Electrical Engineering, University of Alabama At Birmingham, Birmingham, Alabama, USA
| | - David A Lynch
- Radiology, National Jewish Health, Denver, Colorado, USA
| | - Carla G Wilson
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA
| | - Spyridon Fortis
- Pulmonary, Critical Care and Occupation Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Victor Kim
- Division of Pulmonary and Critical Care Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
27
|
Shin HJ, Kim TO, Kim YI, Kim SH, Kim HK, Kim YH, Byun MK, Jung KS, Yoo KH, Lee JS, Lim SC. The paradoxical response to short-acting bronchodilator administration in patients with chronic obstructive pulmonary disease. J Thorac Dis 2021; 13:511-520. [PMID: 33717524 PMCID: PMC7947541 DOI: 10.21037/jtd-20-985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background There are a few studies about paradoxical bronchodilator response (BDR), which means a decrease in forced expiratory volume in 1 second (FEV1) or forced vital capacity (FVC) after short-acting bronchodilator administration in patients with chronic obstructive pulmonary disease (COPD). We evaluated the effect of paradoxical BDR on the clinical outcomes of COPD patients in South Korea. Methods We analyzed the KOrea COpd Subgroup Study team (KOCOSS) cohort data in South Korea between January 2012 and December 2017. BDR was defined as at least a 12% and 200-mL reduction in FEV1 or FVC after bronchodilator administration. Results A total of 1,991 patients were included in this study. A paradoxical BDR was noted in 57 (2.9%) patients and was independently associated with worse dyspnea and poor quality of life. High C-reactive protein (CRP) levels were associated with a paradoxical BDR (OR, 1.05; 95% CI, 1.01-1.09; P=0.003). However, paradoxical BDR was not associated with severe acute exacerbations. Pre-bronchodilator FEV1 (L) showed a higher area under the curve (AUC) for predicting severe acute exacerbations than the post-bronchodilator FEV1 (L) in the paradoxical BDR group (0.788 vs. 0.752). Conclusion A paradoxical reduction of FEV1 or FVC after bronchodilator administration may be associated with chronic inflammation in the airway and independently associated with worse respiratory symptoms and poor quality of life.
Collapse
Affiliation(s)
- Hong-Joon Shin
- Division of Pulmonary Medicine, Department of Internal Medicine, Chonnam National University Hospital, Kwangju, South Korea
| | - Tae-Ok Kim
- Division of Pulmonary Medicine, Department of Internal Medicine, Chonnam National University Hospital, Kwangju, South Korea
| | - Yu-Il Kim
- Division of Pulmonary Medicine, Department of Internal Medicine, Chonnam National University Hospital, Kwangju, South Korea
| | - Sang-Hoon Kim
- Department of Internal Medicine, Eulji General Hospital, Eulji University School of Medicine, Seoul, South Korea
| | - Hyun Kuk Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Inje University Haeundae Paik Hospital, Busan, South Korea
| | - Yong-Hyun Kim
- Division of Allergy and Pulmonology, Department of Internal Medicine, Bucheon St Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon, South Korea
| | - Min Kwang Byun
- Division of Pulmonology, Department of Internal Medicine, Gangnam Severance Hospital, South Korea
| | - Ki-Suck Jung
- Division of Pulmonary Medicine, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Hallym University Medical School, Anyang, South Korea
| | - Kwang-Ha Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, South Korea
| | - Jae Seung Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, South Korea
| | - Sung-Chul Lim
- Division of Pulmonary Medicine, Department of Internal Medicine, Chonnam National University Hospital, Kwangju, South Korea
| | | |
Collapse
|
28
|
Xiong M, Guo M, Huang D, Li J, Zhou Y. TRPV1 genetic polymorphisms and risk of COPD or COPD combined with PH in the Han Chinese population. Cell Cycle 2020; 19:3066-3073. [PMID: 33103544 PMCID: PMC7714492 DOI: 10.1080/15384101.2020.1831246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
COPD is a common chronic disease with genetic predisposition. TRPV1 is mainly expressed in peripheral neuron which widely exists in entire respiratory tract. In present study, we aimed to study the relationship between single nucleotide polymorphisms (SNPs) of transient receptor potential vanilloid-1 (TRPV1) and the risk of chronic obstructive pulmonary disease (COPD) or COPD combined with pulmonary hypertension (PH) in Chinese Han population. A total of 1019 individuals, including 506 healthy volunteers and 513 COPD patients (150 patients combined with PH among them) were recruited in this study. Genomic DNA were extracted and sequenced. Genotype and allele frequencies of the TRPV1 SNPs among COPD, COPD combined with PH and control groups were compared. Then, the association of TRPV1 SNPs and smoking status were analyzed. Genotype frequencies of SNP rs3744683 had a significant difference in COPD patients with PH patients compared with control (p = 0.006) or COPD patients without PH patients (p = 0.016). Likewise, SNP rs3744683 was remarkedly associated with the risk of COPD (p = 0.004) in current-smoker groups which phenomenon was not observed in nonsmoker or former-smoker groups. Compared with the control group, there was a significant difference for the distribution of SNP rs4790521 alleles in the COPD group (p = 0.041). For further, logical regression analysis showed that SNP rs3744683 genotype of “TC” was a protective factor for PH in COPD patients compared with the genotype of “TT” (OR = 0.364, 95%CI = 0.159–0.829, p = 0.016). Our findings firstly revealed the relevance between TRPV1 SNPs and the risk for COPD/COPD combined with PH.
Collapse
Affiliation(s)
- Mingmei Xiong
- Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Meihua Guo
- Department of Respiration, Guangzhou Chest Hospital , Guangzhou, China
| | - Dongjian Huang
- Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Jing Li
- Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Yan Zhou
- Department of Critical Care Medicine, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| |
Collapse
|
29
|
Nikolaou V, Massaro S, Fakhimi M, Stergioulas L, Price D. COPD phenotypes and machine learning cluster analysis: A systematic review and future research agenda. Respir Med 2020; 171:106093. [PMID: 32745966 DOI: 10.1016/j.rmed.2020.106093] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/19/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a highly heterogeneous condition projected to become the third leading cause of death worldwide by 2030. To better characterize this condition, clinicians have classified patients sharing certain symptomatic characteristics, such as symptom intensity and history of exacerbations, into distinct phenotypes. In recent years, the growing use of machine learning algorithms, and cluster analysis in particular, has promised to advance this classification through the integration of additional patient characteristics, including comorbidities, biomarkers, and genomic information. This combination would allow researchers to more reliably identify new COPD phenotypes, as well as better characterize existing ones, with the aim of improving diagnosis and developing novel treatments. Here, we systematically review the last decade of research progress, which uses cluster analysis to identify COPD phenotypes. Collectively, we provide a systematized account of the extant evidence, describe the strengths and weaknesses of the main methods used, identify gaps in the literature, and suggest recommendations for future research.
Collapse
Affiliation(s)
- Vasilis Nikolaou
- Surrey Business School, University of Surrey, Guildford, GU2 7HX, UK.
| | - Sebastiano Massaro
- Surrey Business School, University of Surrey, Guildford, GU2 7HX, UK; The Organizational Neuroscience Laboratory, London, WC1N 3AX, UK
| | - Masoud Fakhimi
- Surrey Business School, University of Surrey, Guildford, GU2 7HX, UK
| | | | - David Price
- Observational and Pragmatic Research Institute, Singapore, Singapore; Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
30
|
Stanford D, Kim H, Bodduluri S, LaFontaine J, Byzek SA, Schoeb TR, Harris ES, Nath HP, Bhatt SP, Raju SV, Rowe SM. Airway remodeling in ferrets with cigarette smoke-induced COPD using µCT imaging. Am J Physiol Lung Cell Mol Physiol 2020; 319:L11-L20. [PMID: 32374671 PMCID: PMC7468842 DOI: 10.1152/ajplung.00328.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 11/22/2022] Open
Abstract
Structural changes to airway morphology, such as increased bronchial wall thickness (BWT) and airway wall area, are cardinal features of chronic obstructive pulmonary disease (COPD). Ferrets are a recently established animal model uniquely exhibiting similar clinical and pathological characteristics of COPD as humans, including chronic bronchitis. Our objective was to develop a microcomputed tomography (µCT) method for evaluating structural changes to the airways in ferrets and assess whether the effects of smoking induce changes consistent with chronic bronchitis in humans. Ferrets were exposed to mainstream cigarette smoke or air control twice daily for 6 mo. µCT was conducted in vivo at 6 mo; a longitudinal cohort was imaged monthly. Manual measurements of BWT, luminal diameter (LD), and BWT-to-LD ratio (BWT/LD) were conducted and confirmed by a semiautomated algorithm. The square root of bronchial wall area (√WA) versus luminal perimeter was determined on an individual ferret basis. Smoke-exposed ferrets reproducibly demonstrated 34% increased BWT (P < 0.001) along with increased LD and BWT/LD versus air controls. Regression indicated that the effect of smoking on BWT persisted despite controlling for covariates. Semiautomated measurements replicated findings. √WA for the theoretical median airway luminal perimeter of 4 mm (Pi4) was elevated 4.4% in smoke-exposed ferrets (P = 0.015). Increased BWT and Pi4 developed steadily over time. µCT-based airway measurements in ferrets are feasible and reproducible. Smoke-exposed ferrets develop increased BWT and Pi4, changes similar to humans with chronic bronchitis. µCT can be used as a significant translational platform to measure dynamic airway morphological changes.
Collapse
Affiliation(s)
- Denise Stanford
- Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Harrison Kim
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sandeep Bodduluri
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer LaFontaine
- Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephen A Byzek
- Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Trenton R Schoeb
- Genetics and Animal Resources Program, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elex S Harris
- Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hrudaya P Nath
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Surya P Bhatt
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- UAB Lung Imaging Core, University of Alabama at Birmingham, Birmingham, Alabama
| | - S Vamsee Raju
- Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Steven M Rowe
- Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
31
|
Moll M, Sakornsakolpat P, Shrine N, Hobbs BD, DeMeo DL, John C, Guyatt AL, McGeachie MJ, Gharib SA, Obeidat M, Lahousse L, Wijnant SRA, Brusselle G, Meyers DA, Bleecker ER, Li X, Tal-Singer R, Manichaikul A, Rich SS, Won S, Kim WJ, Do AR, Washko GR, Barr RG, Psaty BM, Bartz TM, Hansel NN, Barnes K, Hokanson JE, Crapo JD, Lynch D, Bakke P, Gulsvik A, Hall IP, Wain L, Weiss ST, Silverman EK, Dudbridge F, Tobin MD, Cho MH. Chronic obstructive pulmonary disease and related phenotypes: polygenic risk scores in population-based and case-control cohorts. THE LANCET. RESPIRATORY MEDICINE 2020; 8:696-708. [PMID: 32649918 PMCID: PMC7429152 DOI: 10.1016/s2213-2600(20)30101-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/24/2020] [Accepted: 02/17/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Genetic factors influence chronic obstructive pulmonary disease (COPD) risk, but the individual variants that have been identified have small effects. We hypothesised that a polygenic risk score using additional variants would predict COPD and associated phenotypes. METHODS We constructed a polygenic risk score using a genome-wide association study of lung function (FEV1 and FEV1/forced vital capacity [FVC]) from the UK Biobank and SpiroMeta. We tested this polygenic risk score in nine cohorts of multiple ethnicities for an association with moderate-to-severe COPD (defined as FEV1/FVC <0·7 and FEV1 <80% of predicted). Associations were tested using logistic regression models, adjusting for age, sex, height, smoking pack-years, and principal components of genetic ancestry. We assessed predictive performance of models by area under the curve. In a subset of studies, we also studied quantitative and qualitative CT imaging phenotypes that reflect parenchymal and airway pathology, and patterns of reduced lung growth. FINDINGS The polygenic risk score was associated with COPD in European (odds ratio [OR] per SD 1·81 [95% CI 1·74-1·88] and non-European (1·42 [1·34-1·51]) populations. Compared with the first decile, the tenth decile of the polygenic risk score was associated with COPD, with an OR of 7·99 (6·56-9·72) in European ancestry and 4·83 (3·45-6·77) in non-European ancestry cohorts. The polygenic risk score was superior to previously described genetic risk scores and, when combined with clinical risk factors (ie, age, sex, and smoking pack-years), showed improved prediction for COPD compared with a model comprising clinical risk factors alone (AUC 0·80 [0·79-0·81] vs 0·76 [0·75-0·76]). The polygenic risk score was associated with CT imaging phenotypes, including wall area percent, quantitative and qualitative measures of emphysema, local histogram emphysema patterns, and destructive emphysema subtypes. The polygenic risk score was associated with a reduced lung growth pattern. INTERPRETATION A risk score comprised of genetic variants can identify a small subset of individuals at markedly increased risk for moderate-to-severe COPD, emphysema subtypes associated with cigarette smoking, and patterns of reduced lung growth. FUNDING US National Institutes of Health, Wellcome Trust.
Collapse
Affiliation(s)
- Matthew Moll
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Phuwanat Sakornsakolpat
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nick Shrine
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Brian D Hobbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Catherine John
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Anna L Guyatt
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Michael J McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA, USA; Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ma'en Obeidat
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; University of British Columbia Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Lies Lahousse
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, Netherlands; Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Sara R A Wijnant
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, Netherlands; Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Guy Brusselle
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, Netherlands; Department of Respiratory Medicine, Erasmus Medical Centre, Rotterdam, Netherlands; Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | | | | | - Xingnan Li
- Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Ruth Tal-Singer
- GlaxoSmithKline Research and Development, Collegeville, PA, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA; Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Sungho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, South Korea; Interdisciplinary Program of Bioinformatics, College of National Sciences, Seoul National University, Seoul, South Korea; Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Woo Jin Kim
- Department of Internal Medicine, Kangwon National University, Chuncheon, South Korea
| | - Ah Ra Do
- Interdisciplinary Program of Bioinformatics, College of National Sciences, Seoul National University, Seoul, South Korea
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - R Graham Barr
- Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York, NY, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA; Kaiser Permanente Washington Health Research Institute, Seattle, WA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Nadia N Hansel
- School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kathleen Barnes
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - James D Crapo
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - David Lynch
- Department of Radiology, National Jewish Health, Denver, CO, USA
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Amund Gulsvik
- Division of Respiratory Medicine, Queen's Medical Centre, Nottingham, UK
| | - Ian P Hall
- National Institute for Health Research Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Louise Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK; National Institute for Health Research Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Frank Dudbridge
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Martin D Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK; National Institute for Health Research Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK.
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
32
|
Increased Airway Wall Thickness in Interstitial Lung Abnormalities and Idiopathic Pulmonary Fibrosis. Ann Am Thorac Soc 2020; 16:447-454. [PMID: 30543456 DOI: 10.1513/annalsats.201806-424oc] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
RATIONALE There is increasing evidence that aberrant processes occurring in the airways may precede the development of idiopathic pulmonary fibrosis (IPF); however, there has been no prior confirmatory data derived from imaging studies. OBJECTIVES To assess quantitative measures of airway wall thickness (AWT) in populations characterized for interstitial lung abnormalities (ILA) and for IPF. METHODS Computed tomographic imaging of the chest and measures of AWT were available for 6,073, 615, 1,167, and 38 participants from COPDGene (Genetic Epidemiology of COPD study), ECLIPSE (Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints study), and the Framingham Heart Study (FHS) and in patients with IPF from the Brigham and Women's Hospital Herlihy Registry, respectively. To evaluate these associations, we used multivariable linear regression to compare a standardized measure of AWT (the square root of AWT for airways with an internal perimeter of 10 mm [Pi10]) and characterizations of ILA and IPF by computed tomographic imaging of the chest. RESULTS In COPDGene, ECLIPSE, and FHS, research participants with ILA had increased measures of Pi10 compared with those without ILA. Patients with IPF had mean measures of Pi10 that were even greater than those noted in research participants with ILA. After adjustment for important covariates (e.g., age, sex, race, body mass index, smoking behavior, and chronic obstructive pulmonary disease severity when appropriate), research participants with ILA had increased measures of Pi10 compared with those without ILA (0.03 mm in COPDGene, 95% confidence interval [CI], 0.02-0.03; P < 0.001; 0.02 mm in ECLIPSE, 95% CI, 0.005-0.04; P = 0.01; 0.07 mm in FHS, 95% CI, 0.01-0.1; P = 0.01). Compared with COPDGene participants without ILA older than 60 years of age, patients with IPF were also noted to have increased measures of Pi10 (2.0 mm, 95% CI, 2.0-2.1; P < 0.001). Among research participants with ILA, increases in Pi10 were correlated with reductions in lung volumes in some but not all populations. CONCLUSIONS These results demonstrate that measurable increases in AWT are consistently noted in research participants with ILA and in patients with IPF. These findings suggest that abnormalities of the airways may play a role in, or be correlated with, early pathogenesis of pulmonary fibrosis.
Collapse
|
33
|
Combined Forced Expiratory Volume in 1 Second and Forced Vital Capacity Bronchodilator Response, Exacerbations, and Mortality in Chronic Obstructive Pulmonary Disease. Ann Am Thorac Soc 2020; 16:826-835. [PMID: 30908927 DOI: 10.1513/annalsats.201809-601oc] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rationale: The American Thoracic Society (ATS)/European Respiratory Society defines a positive bronchodilator response (BDR) by a composite of BDR in either forced expiratory volume in 1 second (FEV1) and/or forced vital capacity (FVC) greater than or equal to 12% and 200 ml (ATS-BDR). We hypothesized that ATS-BDR components would be differentially associated with important chronic obstructive pulmonary disease (COPD) outcomes. Objectives: To examine whether ATS-BDR components are differentially associated with clinical, functional, and radiographic features in COPD. Methods: We included subjects with COPD enrolled in the COPDGene study. In the main analysis, we excluded subjects with self-reported asthma. We categorized BDR into the following: 1) No-BDR, no BDR in either FEV1 or FVC; 2) FEV1-BDR, BDR in FEV1 but no BDR in FVC; 3) FVC-BDR, BDR in FVC but no BDR in FEV1; and 4) Combined-BDR, BDR in both FEV1 and FVC. We constructed multivariable logistic, linear, zero-inflated negative binomial, and Cox hazards models to examine the association of BDR categories with symptoms, computed tomography findings, change in FEV1 over time, respiratory exacerbations, and mortality. We also created models using the ATS BDR definition (ATS-BDR) as the main independent variable. Results: Of 3,340 COPD subjects included in the analysis, 1,083 (32.43%) had ATS-BDR, 182 (5.45%) had FEV1-BDR, 522 (15.63%) had FVC-BDR, and 379 (11.34%) had Combined-BDR. All BDR categories were associated with FEV1 decline compared with No-BDR. Compared with No-BDR, both ATS-BDR and Combined-BDR were associated with higher functional residual capacity %predicted, greater internal perimeter of 10 mm, and greater 6-minute-walk distance. In contrast to ATS-BDR, Combined-BDR was independently associated with less emphysema (adjusted beta regression coefficient, -1.67; 95% confidence interval [CI], -2.68 to -0.65; P = 0.001), more frequent respiratory exacerbations (incidence rate ratio, 1.25; 95% CI, 1.03-1.50; P = 0.02) and severe exacerbations (incidence rate ratio, 1.34; 95% CI, 1.05-1.71; P = 0.02), and lower mortality (adjusted hazards ratio, 0.76; 95% CI, 0.58-0.99; P = 0.046). Sensitivity analysis that included subjects with self-reported history of asthma showed similar findings. Conclusions: BDR in both FEV1 and FVC indicates a COPD phenotype with asthma-like characteristics, and provides clinically more meaningful information than current definitions of BDR.
Collapse
|
34
|
Gao J, Törölä T, Li CX, Ohlmeier S, Toljamo T, Nieminen P, Hattori N, Pulkkinen V, Iwamoto H, Mazur W. Sputum Vitamin D Binding Protein (VDBP) GC1S/1S Genotype Predicts Airway Obstruction: A Prospective Study in Smokers with COPD. Int J Chron Obstruct Pulmon Dis 2020; 15:1049-1059. [PMID: 32546996 PMCID: PMC7237691 DOI: 10.2147/copd.s234464] [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: 10/12/2019] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction The vitamin D binding protein (VDBP, also known as GC-globulin) and vitamin D deficiency have been associated with chronic obstructive pulmonary disease (COPD). rs7041 and rs4588 are two single nucleotide polymorphisms of the VDBP gene, including three common allelic variants (GC1S, GC1F and GC2). Previous studies primarily assessed the serum levels of vitamin D and VDBP in COPD. However, less is known regarding the impact of the local release of VDBP on COPD lung function. Thus, we examined the association of sputum and plasma VDBP with lung function at baseline and at four years, and examined potential genetic polymorphism interactions. Methods The baseline levels of sputum VDBP, plasma VDBP and plasma 25-OH vitamin D, as well as the GC rs4588 and rs7041 genotypes, were assessed in a 4-year Finnish follow-up cohort (n = 233) of non-smokers, and smokers with and without COPD. The associations between the VDBP levels and the longitudinal decline of lung function were further analysed. Results High frequencies of the haplotypes in rs7041/rs4588 were homozygous GC1S/1S (42.5%). Higher sputum VDBP levels in stage I and stage II COPD were observed only in carriers with GC1S/1S genotype when compared with non-smokers (p = 0.034 and p = 0.002, respectively). Genotype multivariate regression analysis indicated that the baseline sputum VDBP and FEV1/FVC ratio at baseline independently predicted FEV1% at follow-up. Discussion and Conclusion The baseline sputum VDBP expression was elevated in smokers with COPD among individuals with the GC1S/1S genotype, and predicted follow-up airway obstruction. Our results suggest that the GC polymorphism should be considered when exploring the potential of VDBP as a biomarker for COPD.
Collapse
Affiliation(s)
- Jing Gao
- Heart and Lung Centre, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tanja Törölä
- Heart and Lung Centre, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Chuan-Xing Li
- Pulmonomics Group, Respiratory Medicine Unit, Department of Medicine & Centre for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Steffen Ohlmeier
- Proteomics Core Facility, Biocentre Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Tuula Toljamo
- Department of Pulmonary Medicine, Lapland Central Hospital, Rovaniemi, Finland
| | - Pentti Nieminen
- Medical Informatics and Statistics Group, University of Oulu, Oulu, Finland
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Ville Pulkkinen
- Heart and Lung Centre, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Witold Mazur
- Heart and Lung Centre, Department of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
35
|
Kinose D, Ogawa E, Kawashima S, Matsuo-Kashiwagi Y, Yukimura-Seto R, Yamazaki A, Yoshihashi S, Hirayama Y, Nakano Y. An index of the fractal characteristic of an airway tree is associated with airflow limitations and future body mass index reduction in COPD patients. J Appl Physiol (1985) 2020; 128:1280-1286. [PMID: 32240020 DOI: 10.1152/japplphysiol.00461.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Disorders of the fractality of an airway tree and a vessel tree have been studied in pulmonary diseases. Here we successfully applied Mishima's D to the bronchial minimal inner cross-sectional area (iCSA) measured in multidetector computed tomography (MDCT) images of chronic obstructive pulmonary disease (COPD) and non-COPD smokers (n = 162), by defining D in the following formula: logN(≥X) = -D × logX + c, where X is a certain iCSA value, N(≥X) is the number of airway branches having iCSA greater than or equal to X, and c is a constant. Mathematically, this D of iCSA was associated with the expected reduction ratio of iCSA at bifurcations, which can be estimated by 2-1/D. This D of iCSA also correlated weakly with the box-counting fractal dimension and Weibel's reduction ratio over airway generations, which indicated that the airway tree was not a perfect fractal object and that the branch bifurcation was asymmetric. The D of iCSA showed positive correlations with lung function measurements of airflow limitation in study participants. In addition, D of iCSA representing the periphery showed an association with future body mass index reduction, most likely as an indicator of energy efficacy for breathing as predicted by Hess-Murray's law. D of iCSA may be helpful to understanding the pathogenesis of obstructive lung diseases.NEW & NOTEWORTHY An airway tree is a fractal object. We showed that the distribution of minimal inner cross-sectional area (iCSA) of airway branches can be expressed by a fractal index, D, of minimal iCSA. This D was correlated with airflow limitation and future body mass index reduction in chronic obstructive pulmonary disease patients, as predicted by Hess-Murray's law.
Collapse
Affiliation(s)
- Daisuke Kinose
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Emiko Ogawa
- Health Administration Center, Shiga University of Medical Science, Otsu, Japan
| | - Satoru Kawashima
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Yumiko Matsuo-Kashiwagi
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Ruriko Yukimura-Seto
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Akio Yamazaki
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Saiko Yoshihashi
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Yoko Hirayama
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Yasutaka Nakano
- Division of Respiratory Medicine, Department of Internal Medicine, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
36
|
Vasilescu DM, Phillion AB, Kinose D, Verleden SE, Vanaudenaerde BM, Verleden GM, Van Raemdonck D, Stevenson CS, Hague CJ, Han MK, Cooper JD, Hackett TL, Hogg JC. Comprehensive stereological assessment of the human lung using multiresolution computed tomography. J Appl Physiol (1985) 2020; 128:1604-1616. [PMID: 32298211 DOI: 10.1152/japplphysiol.00803.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The application of stereology to lung casts and two-dimensional microscopy images is the gold standard for quantification of the human lung anatomy. However, these techniques are labor intensive, involving fixation, embedding, and histological sectioning of samples and thus have prevented comprehensive studies. Our objective was to demonstrate the application of stereology to volumetric multiresolution computed tomography (CT) to efficiently and extensively quantify the human lung anatomy. Nontransplantable donor lungs from individuals with no evidence of respiratory disease (n = 13) were air inflated, frozen at 10 cmH2O, and scanned using CT. Systematic uniform random samples were taken, scanned using micro-CT, and assessed using stereology. The application of stereology to volumetric CT imaging enabled comprehensive quantification of total lung volume, volume fractions of alveolar, alveolar duct, and tissue, mean linear intercept, alveolar surface area, alveolar surface area density, septal wall thickness, alveolar number, number-weighted mean alveolar volume, and the number and morphometry of terminal and transitional bronchioles. With the use of this data set, we found that women and men have the same number of terminal bronchioles (last generation of conducting airways), but men have longer terminal bronchioles, a smaller wall area percentage, and larger lungs due to a greater number of alveoli per acinus. The application of stereology to multiresolution CT imaging enables comprehensive analysis of the human lung parenchyma that identifies differences between men and women. The reported data set of normal donor lungs aged 25-77 yr provides reference data for future studies of chronic lung disease to determine exact changes in tissue pathology.NEW & NOTEWORTHY Stereology has been the gold standard to quantify the three-dimensional lung anatomy using two-dimensional microscopy images. However, such techniques are labor intensive. This study provides a method that applies stereology to volumetric computed tomography images of frozen whole human lungs and systematic uniform random samples. The method yielded a comprehensive data set on the small airways and parenchymal lung structures, highlighting morphometric sex differences and providing a reference data set for future pathological studies.
Collapse
Affiliation(s)
- Dragoş M Vasilescu
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - André B Phillion
- Department of Materials Science and Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Daisuke Kinose
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stijn E Verleden
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven-Gasthuisberg, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven-Gasthuisberg, Leuven, Belgium
| | - Geert M Verleden
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven-Gasthuisberg, Leuven, Belgium
| | - Dirk Van Raemdonck
- Leuven Lung Transplant Unit, Katholieke Universiteit Leuven and Universitair Ziekenhuis Leuven-Gasthuisberg, Leuven, Belgium
| | | | - Cameron J Hague
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Joel D Cooper
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - James C Hogg
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
37
|
Cairncross A, Jones RL, Elliot JG, McFawn PK, James AL, Noble PB. Airway narrowing and response to simulated deep inspiration in bronchial segments from subjects with fixed airflow obstruction. J Appl Physiol (1985) 2020; 128:757-767. [PMID: 32105523 DOI: 10.1152/japplphysiol.00439.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The volume fraction of extracellular matrix (ECM) within the layer of airway smooth muscle (ASM) is increased in subjects with fixed airflow obstruction. We postulated that changes in ECM within the ASM layer will impact force transmission during induced contraction and/or in response to externally applied stresses like a deep inspiration (DI). Subjects were patients undergoing lung resection surgery who were categorized as unobstructed (n = 12) or "fixed" obstructed (n = 6) on the basis of preoperative spirometry. The response to a DI, assessed by the ratio of isovolumic flows from maximal and partial inspirations (M/P), was also measured preoperatively. M/P was reduced in the obstructed group (P = 0.02). Postoperatively, bronchial segments were obtained from resected tissue, and luminal narrowing to acetylcholine and bronchodilation to simulated DI were assessed in vitro. Airway wall dimensions and the volume fraction of ECM within the ASM were quantified. Maximal airway narrowing to acetylcholine (P = 0.01) and the volume fraction of ECM within the ASM layer (P = 0.02) were increased in the obstructed group, without a change in ASM thickness. Whereas bronchodilation to simulated DI in vitro was not different between obstructed and unobstructed groups, it was correlated with increased M/P (bronchodilation/less bronchoconstriction) in vivo (P = 0.03). The volume fraction of ECM was inversely related to forced expiratory volume in 1 s FEV1 %predicted (P = 0.04) and M/P (P = 0.01). Results show that in subjects with fixed airflow obstruction the mechanical behavior of the airway wall is altered and there is a contemporaneous shift in the structural composition of the ASM layer.NEW & NOTEWORTHY Cartilaginous airways from subjects with fixed airflow obstruction have an increase in the volume fraction of extracellular matrix within the airway smooth muscle layer. These airways are also intrinsically more reactive to a contractile stimulus, which is expected to contribute to airway hyperresponsiveness in this population, often attributed to geometric mechanisms. In view of these results, we speculate on how changes in extracellular matrix may impact airway mechanics.
Collapse
Affiliation(s)
- Alvenia Cairncross
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Robyn L Jones
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Peter K McFawn
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
38
|
Refaee T, Wu G, Ibrahim A, Halilaj I, Leijenaar RTH, Rogers W, Gietema HA, Hendriks LEL, Lambin P, Woodruff HC. The Emerging Role of Radiomics in COPD and Lung Cancer. Respiration 2020; 99:99-107. [PMID: 31991420 DOI: 10.1159/000505429] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/12/2019] [Indexed: 12/24/2022] Open
Abstract
Medical imaging plays a key role in evaluating and monitoring lung diseases such as chronic obstructive pulmonary disease (COPD) and lung cancer. The application of artificial intelligence in medical imaging has transformed medical images into mineable data, by extracting and correlating quantitative imaging features with patients' outcomes and tumor phenotype - a process termed radiomics. While this process has already been widely researched in lung oncology, the evaluation of COPD in this fashion remains in its infancy. Here we outline the main applications of radiomics in lung cancer and briefly review the workflow from image acquisition to the evaluation of model performance. Finally, we discuss the current assessments of COPD and the potential application of radiomics in COPD.
Collapse
Affiliation(s)
- Turkey Refaee
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands, .,Department of Diagnostic Radiology, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia,
| | - Guangyao Wu
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Abdallah Ibrahim
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands.,Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, Centre Hospitalier Universitaire de Liège, Liège, Belgium.,Department of Nuclear Medicine and Comprehensive Diagnostic Center Aachen (CDCA), University Hospital RWTH Aachen University, Aachen, Germany
| | - Iva Halilaj
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Ralph T H Leijenaar
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - William Rogers
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Thoracic Oncology, IRCCS Foundation National Cancer Institute, Milan, Italy
| | - Hester A Gietema
- Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.,Department of Radiology and Nuclear Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
39
|
Hansen JE, Dilektasli AG, Porszasz J, Stringer WW, Pak Y, Rossiter HB, Casaburi R. A New Bronchodilator Response Grading Strategy Identifies Distinct Patient Populations. Ann Am Thorac Soc 2019; 16:1504-1517. [PMID: 31404502 PMCID: PMC6956832 DOI: 10.1513/annalsats.201901-030oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/08/2019] [Indexed: 01/06/2023] Open
Abstract
Rationale: A positive bronchodilator response (BDR) according to American Thoracic Society/European Respiratory Society (ATS/ERS) guidelines require both 200 ml and 12% increase in forced expiratory volume in 1 second (FEV1) or forced vital capacity (FVC) after bronchodilator inhalation. This dual criterion is insensitive in those with high or low FEV1.Objectives: To establish BDR criteria with volume or percentage FEV1 change.Methods: The largest FEV1 and FVC were identified from three pre- and three post-bronchodilator maneuvers in COPDGene (Genetic Epidemiology of COPD) participants. A total of 7,741 individuals with coefficient of variation less than 15% for both FEV1 and FVC formed bronchodilator categories of FEV1 response: negative (≤0.00% or ≤0.00 L), minimal (>0.00% to ≤9.00% or >0.00 L to ≤0.09 L), mild (>9.00% to ≤16.00% or >0.09 L to ≤0.16 L), moderate (>16.00% to ≤26.00% or >0.16 L to ≤0.26 L), and marked (>26.00% or >0.26 L). These response size categories are based on empirical limits considering average FEV1 increase of approximately 160 ml and the clinically important difference for FEV1. To compare flow and volume response characteristics, BDR-FEV1 category assignments were applied for the BDR-FVC response.Results: Twenty percent met mild and 31% met moderate or marked BDR-FEV1 criteria, whereas 12% met mild and 33% met moderate or marked BDR-FVC criteria. In contrast, only 20.6% met ATS/ERS positive criteria. Compared with the negative BDR-FEV1 category, the minimal, mild, moderate, and marked BDR-FEV1 categories were associated with greater 6-minute-walk distance and lower St. George's Respiratory Questionnaire and modified Medical Research Council dyspnea scale scores. Compared with negative BDR, moderate and marked BDR-FEV1 categories were associated with fewer exacerbations, and minimal BDR was associated with lower computed tomography airway wall thickness. Compared with the negative category, all BDR-FVC categories were associated with increasing emphysema percentage and gas trapping percentage. Moderate and marked BDR-FVC categories were associated with higher St. George's Respiratory Questionnaire scores but fewer exacerbations and lower dyspnea scores.Conclusions: BDR grading by FEV1 volume or percentage response identified subjects otherwise missed by ATS/ERS criteria. BDR grades were associated with functional exercise performance, quality of life, exacerbation frequency, dyspnea, and radiological airway measures. BDR grades in FEV1 and FVC indicate different clinical and radiological characteristics.
Collapse
Affiliation(s)
| | - Asli G Dilektasli
- Rehabilitation Clinical Trials Center and
- Department of Pulmonary Medicine, Faculty of Medicine, Uludağ University, Bursa, Turkey; and
| | | | | | - Youngju Pak
- UCLA Clinical and Translational Science Institute, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Harry B Rossiter
- Rehabilitation Clinical Trials Center and
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | | |
Collapse
|
40
|
Goorsenberg A, Kalverda KA, Annema J, Bonta P. Advances in Optical Coherence Tomography and Confocal Laser Endomicroscopy in Pulmonary Diseases. Respiration 2019; 99:190-205. [PMID: 31593955 DOI: 10.1159/000503261] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Diagnosing and monitoring pulmonary diseases is highly dependent on imaging, physiological function tests and tissue sampling. Optical coherence tomography (OCT) and confocal laser endomicroscopy (CLE) are novel imaging techniques with near-microscopic resolution that can be easily and safely combined with conventional bronchoscopy. Disease-related pulmonary anatomical compartments can be visualized, real time, using these techniques. In obstructive lung diseases, airway wall layers and related structural remodelling can be identified and quantified. In malignant lung disease, normal and malignant areas of the central airways, lung parenchyma, lymph nodes and pleura can be discriminated. A growing number of interstitial lung diseases (ILDs) have been visualized using OCT or CLE. Several ILD-associated structural changes can be imaged: fibrosis, cellular infiltration, bronchi(ol)ectasis, cysts and microscopic honeycombing. Although not yet implemented in clinical practice, OCT and CLE have the potential to improve detection and monitoring pulmonary diseases and can contribute in unravelling the pathophysiology of disease and mechanism of action of novel treatments. Indeed, assessment of the airway wall layers with OCT might be helpful when evaluating treatments targeting airway remodelling. By visualizing individual malignant cells, CLE has the potential as a real-time lung cancer detection tool. In the future, both techniques could be combined with laser-enhanced fluorescent-labelled tracer detection. This review discusses the value of OCT and CLE in pulmonary medicine by summarizing the current evidence and elaborating on future perspectives.
Collapse
Affiliation(s)
- Annika Goorsenberg
- Department of Pulmonology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands,
| | - Kirsten A Kalverda
- Department of Pulmonology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Jouke Annema
- Department of Pulmonology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Peter Bonta
- Department of Pulmonology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
41
|
Washko GR. Rebuttal From Dr Washko. Chest 2019; 154:1281-1282. [PMID: 30526968 DOI: 10.1016/j.chest.2018.08.1054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 11/28/2022] Open
Affiliation(s)
- George R Washko
- Lung Center, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA.
| |
Collapse
|
42
|
Using Quantitative Computed Tomographic Imaging to Understand Chronic Obstructive Pulmonary Disease and Fibrotic Interstitial Lung Disease. J Thorac Imaging 2019; 35:246-254. [DOI: 10.1097/rti.0000000000000440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
43
|
Huang X, Mu X, Deng L, Fu A, Pu E, Tang T, Kong X. The etiologic origins for chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2019; 14:1139-1158. [PMID: 31213794 PMCID: PMC6549659 DOI: 10.2147/copd.s203215] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/18/2019] [Indexed: 12/27/2022] Open
Abstract
COPD, characterized by long-term poorly irreversible airway limitation and persistent respiratory symptoms, has resulted in enormous challenges to human health worldwide, with increasing rates of prevalence, death, and disability. Although its origin was thought to be in the interactions of genetic with environmental factors, the effects of environmental factors on the disease during different life stages remain little known. Without clear mechanisms and radical cure for it, early screening and prevention of COPD seem to be important. In this review, we will discuss the etiologic origins for poor lung function and COPD caused by specific adverse effects during corresponding life stages, as well as try to find new insights and potential prevention strategies for this disease.
Collapse
Affiliation(s)
- Xinwei Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming City, Yunnan Province, People's Republic of China.,Medical School, Kunming University of Science and Technology, Kunming City, Yunnan Province, People's Republic of China
| | - Xi Mu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming City, Yunnan Province, People's Republic of China
| | - Li Deng
- The Pathology Department, First People's Hospital of Yunnan Province, Kunming City, Yunnan Province, People's Republic of China
| | - Aili Fu
- Department of Oncology, Yunfeng Hospital, Xuanwei City, Yunnan Province, People's Republic of China
| | - Endong Pu
- Department of Thoracic Surgery, Yunfeng Hospital, Xuanwei City, Yunnan Province, People's Republic of China
| | - Tao Tang
- Medical School, Kunming University of Science and Technology, Kunming City, Yunnan Province, People's Republic of China
| | - Xiangyang Kong
- Medical School, Kunming University of Science and Technology, Kunming City, Yunnan Province, People's Republic of China
| |
Collapse
|
44
|
Quantitative computed tomography for predicting cardiopulmonary complications after lobectomy for lung cancer in patients with chronic obstructive pulmonary disease. Gen Thorac Cardiovasc Surg 2019; 67:697-703. [DOI: 10.1007/s11748-019-01080-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 01/20/2019] [Indexed: 01/21/2023]
|
45
|
Janssen R, Piscaer I, Franssen FME, Wouters EFM. Emphysema: looking beyond alpha-1 antitrypsin deficiency. Expert Rev Respir Med 2019; 13:381-397. [DOI: 10.1080/17476348.2019.1580575] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Rob Janssen
- Department of Pulmonary Medicine, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Ianthe Piscaer
- Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Frits M. E. Franssen
- Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
- CIRO, Center of Expertise for Chronic Organ Failure, Horn, The Netherlands
| | - Emiel F. M. Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
- CIRO, Center of Expertise for Chronic Organ Failure, Horn, The Netherlands
| |
Collapse
|
46
|
Parker MM, Lutz SM, Hobbs BD, Busch R, McDonald MN, Castaldi PJ, Beaty TH, Hokanson JE, Silverman EK, Cho MH. Assessing pleiotropy and mediation in genetic loci associated with chronic obstructive pulmonary disease. Genet Epidemiol 2019; 43:318-329. [PMID: 30740764 DOI: 10.1002/gepi.22192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/10/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022]
Abstract
Genetic association studies have increasingly recognized variant effects on multiple phenotypes. Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease with environmental and genetic causes. Multiple genetic variants have been associated with COPD, many of which show significant associations to additional phenotypes. However, it is unknown if these associations represent biological pleiotropy or if they exist through correlation of related phenotypes ("mediated pleiotropy"). Using 6,670 subjects from the COPDGene study, we describe the association of known COPD susceptibility loci with other COPD-related phenotypes and distinguish if these act directly on the phenotypes (i.e., biological pleiotropy) or if the association is due to correlation (i.e., mediated pleiotropy). We identified additional associated phenotypes for 13 of 25 known COPD loci. Tests for pleiotropy between genotype and associated outcomes were significant for all loci. In cases of significant pleiotropy, we performed mediation analysis to test if SNPs had a direct association to phenotype. Most loci showed a mediated effect through the hypothesized causal pathway. However, many loci also had direct associations, suggesting causal explanations (i.e., emphysema leading to reduced lung function) are incomplete. Our results highlight the high degree of pleiotropy in complex disease-associated loci and provide novel insights into the mechanisms underlying COPD.
Collapse
Affiliation(s)
- Margaret M Parker
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sharon M Lutz
- Department of Biostatistics and Informatics, University of Colorado, Anschutz Medical Campus, Denver, Colorado
| | - Brian D Hobbs
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Robert Busch
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - MerryLynn N McDonald
- Division of Pulmonary, Allergy, and Critical Care Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, Massachusetts
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - John E Hokanson
- Department of Epidemiology, University of Colorado, Denver, Aurora, Colorado
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
47
|
Young KA, Strand M, Ragland MF, Kinney GL, Austin EE, Regan EA, Lowe KE, Make BJ, Silverman EK, Crapo JD, Hokanson JE. Pulmonary Subtypes Exhibit Differential Global Initiative for Chronic Obstructive Lung Disease Spirometry Stage Progression: The COPDGene® Study. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2019; 6:414-429. [PMID: 31710796 DOI: 10.15326/jcopdf.6.5.2019.0155] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Rationale We classified individuals into pulmonary disease subtypes based on 2 underlying pathophysiologic disease axes (airway-predominant and emphysema-predominant) and their increased mortality risk. Our next objective was to determine whether some subcomponents of these subtypes are additionally associated with unique patterns of Global initiative for chronic Obstructive Lung Disease (GOLD) spirometry stage progression. Methods After accounting for intra-individual measurement variability in spirometry measures between baseline (Phase 1) and the 5-year follow up (Phase 2) of the COPD Genetic Epidemiology (COPDGene®) study, 4615 individuals had complete data that would characterize patterns of disease progression over 5 years (2033 non-Hispanic whites; 827 African Americans; 48% female). Individuals could express increased risk for mortality on one or both of the primary subtype axes (airway-predominant or emphysema-predominant) and thus they were further classified into 6 groups: high-risk airway-predominant disease only (APD-only), moderate-risk airway-predominant disease only (MR-APD-only), high-risk emphysema-predominant disease only (EPD-only), combined high-risk airway- and emphysema-predominant disease (combined APD-EPD), combined moderate-risk airway- and emphysema-predominant disease (combined MR-APD-EPD), and no high-risk pulmonary subtype. Outcomes were dichotomized for GOLD spirometry stage progression from Phase 1 to Phase 2. Logistic regression of the progression outcomes on the pulmonary subtypes were adjusted for age, sex, race, and change in smoking status. Results The MR-APD-only group was associated with conversion from GOLD 0 to preserved ratio-impaired spirometry (PRISm) status (odds ratio [OR] 11.3, 95% confidence interval [CI] 5.7-22.1) and GOLD 0 to GOLD 2-4 (OR 6.0, 95% CI 2.0-18.0). The EPD-only group was associated with conversion from GOLD 0 to GOLD 1 (OR 2.4, 95% CI 1.2-4.6), and GOLD 1 to GOLD 2-4 (OR 2.6, 95% CI 1.0-6.9). Conversion between PRISm and GOLD 2-4 (31%-38%) occurred in both the APD-only and the MR-APD-only groups. Conclusion Differential conversion occurs from GOLD 0 to PRISm and GOLD 0 to GOLD 1 based on groups expressing airway-predominant disease or emphysema-predominant disease independently or in combination. Airway-predominant and emphysema-predominant subtypes are highly important in determining patterns of early disease progression.
Collapse
Affiliation(s)
- Kendra A Young
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | - Mathew Strand
- Division of Biostatistics and Bioinformatics, Office of Academic Affairs, National Jewish Health, Denver, Colorado
| | - Margaret F Ragland
- Department of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora
| | - Gregory L Kinney
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | - Erin E Austin
- Department of Mathematical and Statistical Sciences, University of Colorado at Denver
| | | | - Katherine E Lowe
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | - Barry J Make
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - James D Crapo
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora
| | | |
Collapse
|
48
|
Charbonnier JP, Pompe E, Moore C, Humphries S, van Ginneken B, Make B, Regan E, Crapo JD, van Rikxoort EM, Lynch DA. Airway wall thickening on CT: Relation to smoking status and severity of COPD. Respir Med 2019; 146:36-41. [PMID: 30665516 PMCID: PMC6639031 DOI: 10.1016/j.rmed.2018.11.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/13/2018] [Accepted: 11/18/2018] [Indexed: 11/17/2022]
Abstract
Airway wall thickening in cigarette smokers is thought to be a result of inflammatory changes and airway remodeling. This study investigates if CT-derived airway wall thickening associates to disease severity in smokers with and without COPD and if airway wall thickening is reversible by smoking cessation. We examined 2000 smokers and 46 never-smokers who returned for a 5-year follow-up visit in the COPDGene-study. Multivariable regression analyses were performed at visit 1 to associate airway wall thickness (expressed as Pi10) with percent predicted forced expiratory volume in 1 s (FEV1%-predicted), 6-min walking distance (6MWD), and St. George Respiratory Questionnaire (SGRQ). Longitudinal analyses were performed to assess the effect of smoking cessation on Pi10 using linear mixed models. A higher Pi10 was significantly associated with worse FEV1%-predicted, 6MWD, and SGRQ in all GOLD-stages. Longitudinal analyses showed that subjects that quit smoking significantly decreased in Pi10 (ΔPi10 = -0.18 mm, p < 0.001). Subjects that started smoking had a significant increase in Pi10 (ΔPi10 = 0.14 mm, p < 0.001). Pi10 is a clinically relevant biomarker of smoking-related airway injury in smokers with and without COPD. The change in Pi10 with change in smoking status suggests that it can quantify a reversible component of smoking-related airway inflammation.
Collapse
Affiliation(s)
- Jean-Paul Charbonnier
- Thirona, Nijmegen, the Netherlands; Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Esther Pompe
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Camille Moore
- Department of Biostatistics and Bioinformatics, National Jewish Health, Denver, CO, USA
| | | | - Bram van Ginneken
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Barry Make
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Elizabeth Regan
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - James D Crapo
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Eva M van Rikxoort
- Thirona, Nijmegen, the Netherlands; Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - David A Lynch
- Department of Radiology, National Jewish Health, Denver, CO, USA
| |
Collapse
|
49
|
Occupational COPD and emphysema: two case histories. Br J Gen Pract 2018; 69:42-43. [PMID: 30591614 DOI: 10.3399/bjgp19x700625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 10/31/2022] Open
|
50
|
Bodduluri S, Puliyakote ASK, Gerard SE, Reinhardt JM, Hoffman EA, Newell JD, Nath HP, Han MK, Washko GR, San José Estépar R, Dransfield MT, Bhatt SP. Airway fractal dimension predicts respiratory morbidity and mortality in COPD. J Clin Invest 2018; 128:5374-5382. [PMID: 30256767 DOI: 10.1172/jci120693] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/11/2018] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is characterized by airway remodeling. Characterization of airway changes on computed tomography has been challenging due to the complexity of the recurring branching patterns, and this can be better measured using fractal dimensions. METHODS We analyzed segmented airway trees of 8,135 participants enrolled in the COPDGene cohort. The fractal complexity of the segmented airway tree was measured by the Airway Fractal Dimension (AFD) using the Minkowski-Bougliand box-counting dimension. We examined associations between AFD and lung function and respiratory morbidity using multivariable regression analyses. We further estimated the extent of peribronchial emphysema (%) within 5 mm of the airway tree, as this is likely to affect AFD. We classified participants into 4 groups based on median AFD, percentage of peribronchial emphysema, and estimated survival. RESULTS AFD was significantly associated with forced expiratory volume in one second (FEV1; P < 0.001) and FEV1/forced vital capacity (FEV1/FVC; P < 0.001) after adjusting for age, race, sex, smoking status, pack-years of smoking, BMI, CT emphysema, air trapping, airway thickness, and CT scanner type. On multivariable analysis, AFD was also associated with respiratory quality of life and 6-minute walk distance, as well as exacerbations, lung function decline, and mortality on longitudinal follow-up. We identified a subset of participants with AFD below the median and peribronchial emphysema above the median who had worse survival compared with participants with high AFD and low peribronchial emphysema (adjusted hazards ratio [HR]: 2.72; 95% CI: 2.20-3.35; P < 0.001), a substantial number of whom were not identified by traditional spirometry severity grades. CONCLUSION Airway fractal dimension as a measure of airway branching complexity and remodeling in smokers is associated with respiratory morbidity and lung function change, offers prognostic information additional to traditional CT measures of airway wall thickness, and can be used to estimate mortality risk. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00608764. FUNDING This study was supported by NIH K23 HL133438 (SPB) and the COPDGene study (NIH Grant Numbers R01 HL089897 and R01 HL089856). The COPDGene project is also supported by the COPD Foundation through contributions made to an Industry Advisory Board comprised of AstraZeneca, Boehringer Ingelheim, Novartis, Pfizer, Siemens, Sunovion and GlaxoSmithKline.
Collapse
Affiliation(s)
- Sandeep Bodduluri
- Division of Pulmonary, Allergy and Critical Care Medicine.,UAB Lung Imaging Core, and.,UAB Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Sarah E Gerard
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Joseph M Reinhardt
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Eric A Hoffman
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA.,Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - John D Newell
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA.,Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Hrudaya P Nath
- UAB Lung Imaging Core, and.,Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Raúl San José Estépar
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark T Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine.,UAB Lung Imaging Core, and.,UAB Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Surya P Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine.,UAB Lung Imaging Core, and.,UAB Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | -
- The COPDGene Investigators are detailed in the Supplemental Acknowledgments
| |
Collapse
|