1
|
Jebaraj P, Paul B, Isaac R, Reddy SR, Kumar R, Vikas B, Das D, Norrie J, Weller D, Pinnock H. Optimising participation in a pulmonary rehabilitation programme for people living with chronic respiratory diseases in rural India: a feasibility study. J Glob Health 2025; 15:04143. [PMID: 40340958 DOI: 10.7189/jogh.15.04143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025] Open
Abstract
Background Pulmonary rehabilitation (PR) plays a vital role in managing chronic obstructive pulmonary disease and other chronic respiratory diseases (CRDs). However, implementation of this multidisciplinary approach in resource-poor settings may not be sufficient because of referrers' uncertainty regarding the effectiveness of PR, inconvenient timing, travel issues, patients' lack of motivation, and poor family support. The aim was to test the feasibility of a peer-led, professionally assisted community-based PR programme for CRD patients in a rural, low literacy setting. Methods We conducted a single-centre, pre-post feasibility study. Participants with a confirmed diagnosis of CRD were recruited and treatment was optimised. After completing baseline assessments, the participants underwent eight weeks of PR training (16 sessions) in six groups at five local facilities led by peers selected by the participants and assisted by professionals. Exercise capacity was assessed with 6-Minute Walk Test. Other outcomes were: International Physical Activity Questionnaire; Hospital Anxiety and Depression Scale; London Chest Activity of Daily Living scale. Upper and lower limb strength were assessed using a handheld dynamometer and cycle ergometer, respectively. Descriptive analysis was performed, and pre-and post-outcomes were compared using parametric tests. Results Thirty participants (20 chronic obstructive pulmonary disease and 10 asthma; 15 female; median age 57.5 years) completed baseline and endline assessments. Seventy percent completed at least 12/16 sessions. After eight weeks of training, the 6-Minute Walk Test had improved from 263.3 (standard deviation (SD) = 72.3) to 319.6 (SD = 84.7) metres (P < 0.001) with significant improvement in modified Medical Research Council (P = 0.022), London Chest Activity of Daily Living scale (P < 0.001) and dominant handgrip strength (P < 0.001) but no significant change in physical activity (P = 0.791). Conclusions The community-based PR led by peer volunteers and supported by professionals proved to be feasible in our low-resource setting and was associated with improved exercise tolerance and other outcomes. Registration The study was registered at the Clinical Trials Registry - India (CTRI/2020/09/027818).
Collapse
Affiliation(s)
- Paul Jebaraj
- Christian Medical College, Vellore, Rural Unit for Health and Social Affairs Department, Vellore, India
| | - Biswajit Paul
- Christian Medical College, Vellore, Rural Unit for Health and Social Affairs Department, Vellore, India
| | - Rita Isaac
- Karkinos Healthcare Private limited, Ernakulam, India
| | - Shadrack Ravindra Reddy
- Christian Medical College, Vellore, Rural Unit for Health and Social Affairs Department, Vellore, India
| | - Rakesh Kumar
- Christian Medical College, Vellore, Rural Unit for Health and Social Affairs Department, Vellore, India
| | - Bochu Vikas
- Christian Medical College, Vellore, Rural Unit for Health and Social Affairs Department, Vellore, India
| | - Deepa Das
- Bangalore Baptist Hospital, Bangalore, India
| | - John Norrie
- Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - David Weller
- Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Hilary Pinnock
- Usher Institute, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
2
|
Tzeng IS, Hu WC, Wu CW, Wu MY, Yiang GT, Hsieh PC, Su WL. Age, period and cohort processes in chronic obstructive pulmonary disease related emergency department visit rate in Taiwan, 2001-2015. J Infect Public Health 2025; 18:102658. [PMID: 39842192 DOI: 10.1016/j.jiph.2025.102658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 01/24/2025] Open
Abstract
PURPOSE Emergency room (ER) physicians must deal with patients with clinically suspected symptoms, such as dyspnea, cough, and increased sputum production, on the frontlines of medical care if patients present with severe chronic obstructive pulmonary disease (COPD). This study aims to investigate the longitudinal tendencies of COPD-related ER visits. PATIENTS AND METHODS A total of 360,313 patients were included in this study. The COPD-related ER visit rates between 2001 and 2015 were categorized using the International Classification of Disease (ICD) codes (496). The effects of age, period, and cohort on COPD-related ER visit rates were determined using an age-period-cohort (APC) model. RESULTS Age was associated with a high risk of COPD in the pediatric and older populations. A significant increase was observed in the period effect, from 2001 to 2015. The cohort effect tended to oscillate from 1918 to 1973, and was reversed in the latest cohort. Furthermore, the COPD-related ER visit rate increased between 2001 and 2015 in both men and women. CONCLUSION Age, period, and cohort were observed to increase COPD visit rates. The APC model can be used to determine trends in COPD-related ER visits.
Collapse
Affiliation(s)
- I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Wan-Chung Hu
- Department of Clinical Pathology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Chih-Wei Wu
- Department of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Po-Chun Hsieh
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Wen-Lin Su
- Department of Pulmonary Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| |
Collapse
|
3
|
Goeminne LJE, Vladimirova A, Eames A, Tyshkovskiy A, Argentieri MA, Ying K, Moqri M, Gladyshev VN. Plasma protein-based organ-specific aging and mortality models unveil diseases as accelerated aging of organismal systems. Cell Metab 2025; 37:205-222.e6. [PMID: 39488213 DOI: 10.1016/j.cmet.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/04/2024] [Accepted: 10/04/2024] [Indexed: 11/04/2024]
Abstract
Aging is a complex process manifesting at molecular, cellular, organ, and organismal levels. It leads to functional decline, disease, and ultimately death, but the relationship between these fundamental biomedical features remains elusive. By applying elastic net regularization to plasma proteome data of over 50,000 human subjects in the UK Biobank and other cohorts, we report interpretable organ-specific and conventional aging models trained on chronological age, mortality, and longitudinal proteome data. These models predict organ/system-specific disease and indicate that men age faster than women in most organs. Accelerated organ aging leads to diseases in these organs, and specific diets, lifestyles, professions, and medications influence organ aging rates. We then identify proteins driving these associations with organ-specific aging. Our analyses reveal that age-related chronic diseases epitomize accelerated organ- and system-specific aging, modifiable through environmental factors, advocating for both universal whole-organism and personalized organ/system-specific anti-aging interventions.
Collapse
Affiliation(s)
- Ludger J E Goeminne
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anastasiya Vladimirova
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alec Eames
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - M Austin Argentieri
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kejun Ying
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mahdi Moqri
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
4
|
Bouchard A, Mun J, Vazquez F, Tang A, Delsole E, Strom R, Chen T. Radiographic Robustness of Lumbar Interbody Fusion Techniques. Global Spine J 2025; 15:59-65. [PMID: 38197369 PMCID: PMC11572118 DOI: 10.1177/21925682241226659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
STUDY DESIGN Retrospective chart review. OBJECTIVES Lumbar interbody fusion (LIF) can be achieved with various techniques. Evidence supporting the long-term clinical advantages of one technique over another are inconclusive. The purpose of this study was to (1) determine the changes in sagittal parameters in the preoperative, intraoperative, and post-operative phase, (2) evaluate the radiographic maintenance of these parameters over time, and (3) compare the demographics and patient reported outcomes of patients undergoing various LIF techniques. METHODS We performed a retrospective chart review of patients with degenerative spine disease undergoing single level anterior (ALIF), lateral (LLIF), posterior (PLIF), or transforaminal (TLIF) lumbar interbody fusion. Data collected included patient demographics and diagnosis at time of surgery. Upright lumbar radiographs taken pre-operatively, intra-operatively, and post-operatively were measured for lumbar lordosis (LL), segmental lordosis (SL), posterior disc height (PDH), and foraminal height (FH). RESULTS 194 patients in a single center were included. PDH and FH increased intra-operatively following ALIF (P < .0001), PLIF (P < .0001), LLIF (P < .0001), and TLIF (P < .0001). SL also increased intra-operatively for ALIF (P = .002) and LLIF (P = .0007). Compared to intra-operative radiographs, PDH and FH decreased at latest post-operative phase for ALIF (P < .03), LLIF (P < .003), TLIF (P < .001), and PLIF (P < .005). SL decreased for ALIF (P = .0008), and TLIF (P = .02). LL did not change postoperatively across techniques. Patient reported outcomes improved post-surgically and disability index decreased, but neither differed between techniques. CONCLUSION LIF, regardless of technique, was shown to provide significant radiographic changes in PDH and FH. Techniques utilizing larger intervertebral cage sizes (ALIF/LLIF) improved SL. Single level LIF did not affect overall LL. No single technique displayed superior radiographic robustness over time.
Collapse
Affiliation(s)
- Alice Bouchard
- Department of Orthopedic Surgery, Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Jeffrey Mun
- Department of Orthopedic Surgery, Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Frank Vazquez
- Department of Orthopedic Surgery, Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Alex Tang
- Northeast Orthopaedic Surgery Residency, Geisinger, Wilkes-Barren, PA, USA
| | - Edward Delsole
- Department of Orthopaedic Surgery, Geisinger Medical Center, Danville, PA, USA
| | - Russell Strom
- Department of Neurosurgery, Geisinger Medical Center, Danville, PA, USA
| | - Tan Chen
- Department of Orthopaedic Surgery, Geisinger Medical Center, Danville, PA, USA
| |
Collapse
|
5
|
Dong Y, Cromer P, Layman D, Altvater M, Dong Y, Zhu H. The prevalence of small airways disease and association with handgrip strength in young Hispanic farmworkers. BMC Pulm Med 2024; 24:636. [PMID: 39734202 DOI: 10.1186/s12890-024-03382-2] [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: 06/14/2023] [Accepted: 11/05/2024] [Indexed: 12/31/2024] Open
Abstract
BACKGROUND Small airways disease (SAD) is a key risk in developing obstructive lung diseases (OLD). Handgrip strength (HGS) is found to be associated with pulmonary function in populations with lung conditions. Hispanics remain the main workforce in farming industry, but their prevalence of lung conditions remain understudied. Likewise, HGS also remains understudied in Hispanic and farmworker populations. Our study investigated the prevalence of SAD and OLD as well as their associations with HGS among Hispanic farmworkers. METHODS A cross-sectional study analyzed 113 Hispanic farmworkers (54% female) who were screened using pulmonary function tests during annual health fairs in rural Southeastern US from 2013 to 2017. Smoking status was self-reported. SAD was defined as forced expiratory flow at 25-75% predicted of vital capacity (FEF25-75%predicted) ≤ 60% per literature and OLD defined as forced expiratory volume in 1 s/ forced vital capacity (FEV1/FVC) ratio < 70% per Global Initiative for Chronic Obstructive Lung Disease criteria. Seated isometric absolute (the sum of both hands) and relative (absolute handgrip strength divided by body mass index) handgrip strengths were collected. RESULTS 26.5% of subjects had SAD and 15.9% had OLD. 50% of subjects with SAD had OLD while 83% of subjects with OLD had SAD. 13% of overall population smoked. Lower absolute and relative HGS groups had higher prevalence of SAD and OLD. Multivariate linear regression showed that lower absolute and relative HGS were associated with worsened small airway function. Age and FEF25-75%predicted were associated with FEV1/FVC. Smoking, body mass index, blood pressures, hemoglobin A1C and lipids were not predictors in either model. CONCLUSIONS This is one of the first studies reporting prevalence of pulmonary function in Hispanic farmworkers. Although this population was relatively young and healthy, there was high prevalence of SAD and OLD, which was higher than the overall prevalence in Hispanic population. There were more females subjects with SAD. Most of the subjects with OLD had SAD but not vice versa. Lower HGS levels were associated with worsened pulmonary function, and HGS was a significant predictor of FEF25-75%predicted, a potential marker for small airway physiology.
Collapse
Affiliation(s)
- Yutong Dong
- Pulmonary-Critical Care Fellowship, Mount Sinai Hospital, 1468 Madison Ave, New York, NY, 10029, USA.
| | - Pam Cromer
- College of Nursing, Augusta University, Augusta, GA, USA
| | - Debbie Layman
- Community Liaison Between Augusta University and Costa-Layman Farm, Augusta, GA, USA
| | - Michelle Altvater
- Department of Medicine, Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15 Street, Augusta, GA, 30912, USA
| | - Yanbin Dong
- Department of Medicine, Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15 Street, Augusta, GA, 30912, USA
| | - Haidong Zhu
- Department of Medicine, Georgia Prevention Institute, Medical College of Georgia, Augusta University, 1120 15 Street, Augusta, GA, 30912, USA
| |
Collapse
|
6
|
Bochenek B, Jankowski M, Wieczorek J, Gruszczynska M, Jaczewski A, Goryński P, Figurski M, Pinkas J. Changes in Hospital Admissions of Patients with COPD in Poland: A 14-Year Nationwide Analysis (2006-2019). COPD 2024; 21:2387114. [PMID: 39129331 DOI: 10.1080/15412555.2024.2387114] [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: 03/14/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024]
Abstract
Poland is a country with a high burden of COPD and its risk factors. This population-wide and geospatial study on COPD in Poland, aimed to analyze changes in hospitalizations of patients with a diagnosis of COPD between 2006 and 2019 as well as to identify changes in the demographic characteristics of patients hospitalized with COPD. This is a retrospective analysis of hospital discharge records of patients with a diagnosis of COPD (ICD-10 code J44 and age 40 and over), hospitalized in Poland between 2006 and 2019. Data were analyzed separately for all patients hospitalized with COPD (primary and secondary diagnosis), for patients admitted due to COPD (primary diagnosis) and patients hospitalized with COPD as a comorbidity (secondary diagnosis). Between 2006 and 2019, a total of 1,663,420 hospital admissions of patients with COPD were reported. Between 2006 and 2019, the annual number of patients hospitalized due to COPD decreased by 53.8%, including 57.3% among men and 46.6% among women. The number of patients hospitalized with COPD as comorbidity increased by 35.8%. The lowest hospital admissions rate was in north-western Poland and the highest in north-eastern Poland. Among patients admitted due to COPD, the percentage of women increased from 32.6% to 37.7%. The percentage of patients admitted due to COPD and living in rural areas decreased from 51.2% in 2006 to 40.8%in 2019. This study provided data on changes in sociodemographic characteristics of hospitalizations of patients with a diagnosis of COPD, including overall reduction in COPD admissions but an increase in COPD burden among women.
Collapse
Affiliation(s)
- Bogdan Bochenek
- Centre of Numerical Weather Prediction, Institute of Meteorology and Water Management - National Research Institute, Warsaw, Poland
| | - Mateusz Jankowski
- School of Public Health, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Joanna Wieczorek
- Centre of Numerical Weather Prediction, Institute of Meteorology and Water Management - National Research Institute, Warsaw, Poland
| | - Marta Gruszczynska
- Centre of Numerical Weather Prediction, Institute of Meteorology and Water Management - National Research Institute, Warsaw, Poland
| | - Adam Jaczewski
- Centre of Numerical Weather Prediction, Institute of Meteorology and Water Management - National Research Institute, Warsaw, Poland
| | - Paweł Goryński
- School of Public Health, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Mariusz Figurski
- Centre of Numerical Weather Prediction, Institute of Meteorology and Water Management - National Research Institute, Warsaw, Poland
| | - Jarosław Pinkas
- School of Public Health, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
7
|
Akhtari M, Jalalvand M, Sadr M, Sharifi H. Autophagy in the Cellular Consequences of Tobacco Smoking: Insights into Senescence. J Biochem Mol Toxicol 2024; 38:e70065. [PMID: 39588771 DOI: 10.1002/jbt.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/27/2024]
Abstract
Smoking is a significant contributing factor to the development of many complex diseases. One of the most important stimuli for aging in the human body is constant exposure to environmental factors such as cigarette smoke. Free radicals in cigarette smoke cause reactive oxygen species production at the cellular level and induce inflammatory responses. The respiratory system of smokers exhibits age-related characteristics, such as enhanced oxidative stress, accumulated damaged proteins, and increased inflammation. Autophagy is triggered by tobacco smoke as a protective mechanism to prevent and reduce molecular stress. However, smoking can interfere with the normal functioning of autophagy in various ways. Smoking-induced impairment of autophagy leads to irreversible cellular damage accumulation, causing cells to undergo cellular aging or senescence. Senescent cells lose their ability to divide and display a distinct secretory phenotype called the senescence-associated secretory phenotype (SASP) and produce numerous growth factors, immune modulators, and inflammatory cytokines. This review discusses the effects of tobacco smoke exposure on autophagy alteration, cellular aging, and senescence induction in exposed animal models, as well as in exposed epithelial and immune cells in the body.
Collapse
Affiliation(s)
- Maryam Akhtari
- Tobacco Prevention and Control Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Jalalvand
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Makan Sadr
- Virology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hooman Sharifi
- Tobacco Prevention and Control Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Rao Q, Li H, Zhou Q, Zhang M, Zhao X, Shi L, Xie J, Fan L, Han Y, Guo F, Liu S, Zhou X. Assessment of pulmonary physiological changes caused by aging, cigarette smoking, and COPD with hyperpolarized 129Xe magnetic resonance. Eur Radiol 2024; 34:7450-7459. [PMID: 38748243 DOI: 10.1007/s00330-024-10800-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/14/2024] [Accepted: 04/05/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE To comprehensively assess the impact of aging, cigarette smoking, and chronic obstructive pulmonary disease (COPD) on pulmonary physiology using 129Xe MR. METHODS A total of 90 subjects were categorized into four groups, including healthy young (HY, n = 20), age-matched control (AMC, n = 20), asymptomatic smokers (AS, n = 28), and COPD patients (n = 22). 129Xe MR was utilized to obtain pulmonary physiological parameters, including ventilation defect percent (VDP), alveolar sleeve depth (h), apparent diffusion coefficient (ADC), total septal wall thickness (d), and ratio of xenon signal from red blood cells and interstitial tissue/plasma (RBC/TP). RESULTS Significant differences were found in the measured VDP (p = 0.035), h (p = 0.003), and RBC/TP (p = 0.003) between the HY and AMC groups. Compared with the AMC group, higher VDP (p = 0.020) and d (p = 0.048) were found in the AS group; higher VDP (p < 0.001), d (p < 0.001) and ADC (p < 0.001), and lower h (p < 0.001) and RBC/TP (p < 0.001) were found in the COPD group. Moreover, significant differences were also found in the measured VDP (p < 0.001), h (p < 0.001), ADC (p < 0.001), d (p = 0.008), and RBC/TP (p = 0.032) between the AS and COPD groups. CONCLUSION Our findings indicate that pulmonary structure and functional changes caused by aging, cigarette smoking, and COPD are various, and show a progressive deterioration with the accumulation of these risk factors, including cigarette smoking and COPD. CLINICAL RELEVANCE STATEMENT Pathophysiological changes can be difficult to comprehensively understand due to limitations in common techniques and multifactorial etiologies. 129Xe MRI can demonstrate structural and functional changes caused by several common factors and can be used to better understand patients' underlying pathology. KEY POINTS Standard techniques for assessing pathophysiological lung function changes, spirometry, and chest CT come with limitations. 129Xe MR demonstrated progressive deterioration with accumulation of the investigated risk factors, without these limitations. 129Xe MR can assess lung changes related to these risk factors to stage and evaluate the etiology of the disease.
Collapse
Affiliation(s)
- Qiuchen Rao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
| | - Haidong Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
| | - Ming Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiuchao Zhao
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Shi
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junshuai Xie
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Fan
- Department of Radiology, Changzheng Hospital of the Second Military Medical University, Shanghai, 200003, China
| | - Yeqing Han
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fumin Guo
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China
| | - Shiyuan Liu
- Department of Radiology, Changzheng Hospital of the Second Military Medical University, Shanghai, 200003, China
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430071, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Biomedical Engineering, Hainan University, Haikou, 570228, China.
| |
Collapse
|
9
|
Shigematsu M, Kawamura T, Deshpande DA, Kirino Y. Immunoactive signatures of circulating tRNA- and rRNA-derived RNAs in chronic obstructive pulmonary disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102285. [PMID: 39220268 PMCID: PMC11364045 DOI: 10.1016/j.omtn.2024.102285] [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: 01/07/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is the most prevalent lung disease, and macrophages play a central role in the inflammatory response in COPD. We here report a comprehensive characterization of circulating short non-coding RNAs (sncRNAs) in plasma from patients with COPD. While circulating sncRNAs are increasingly recognized for their regulatory roles and biomarker potential in various diseases, the conventional RNA sequencing (RNA-seq) method cannot fully capture these circulating sncRNAs due to their heterogeneous terminal structures. By pre-treating the plasma RNAs with T4 polynucleotide kinase, which converts all RNAs to those with RNA-seq susceptible ends (5'-phosphate and 3'-hydroxyl), we comprehensively sequenced a wide variety of non-microRNA sncRNAs, such as 5'-tRNA halves containing a 2',3'-cyclic phosphate. We discovered a remarkable accumulation of the 5'-half derived from tRNAValCAC in plasma from COPD patients, whereas the 5'-tRNAGlyGCC half is predominant in healthy donors. Further, the 5'-tRNAValCAC half activates human macrophages via Toll-like receptor 7 and induces cytokine production. Additionally, we identified circulating rRNA-derived fragments that were upregulated in COPD patients and demonstrated their ability to induce cytokine production in macrophages. Our findings provide evidence of circulating, immune-active sncRNAs in patients with COPD, suggesting that they serve as inflammatory mediators in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Megumi Shigematsu
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Takuya Kawamura
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Deepak A. Deshpande
- Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Yohei Kirino
- Computational Medicine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
10
|
Liu C, Zhu J, Zhao L, Li G, Sun J, Zhang S, Liang X. Blood MALT1 reflects acute exacerbation risk and inflammation in elderly chronic obstructive pulmonary disease patients. Biomark Med 2024; 18:513-521. [PMID: 39136445 PMCID: PMC11364060 DOI: 10.1080/17520363.2024.2347199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 03/01/2024] [Indexed: 08/30/2024] Open
Abstract
Aim: This study intended to investigate the ability of blood MALT1 to estimate acute exacerbation risk in elderly chronic obstructive pulmonary disease (COPD) patients.Methods: Blood MALT1 was detected in 176 elderly COPD patients (aged more than 60 years).Results: MALT1 was elevated in patients with COPD acute exacerbation versus patients with stable COPD (p < 0.001). In patients with COPD acute exacerbation, MALT1 was negatively related to forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) (p = 0.024) and FEV1% predicted (p = 0.002), but positively linked with global initiative for chronic obstructive lung disease stage (p = 0.005).Conclusion: Blood MALT1 reflects increased acute exacerbation risk and inflammation in elderly COPD patients.
Collapse
Affiliation(s)
- Cui Liu
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| | - Jinsong Zhu
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| | - Lei Zhao
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| | - Guanying Li
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| | - Jiawei Sun
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| | - Shengli Zhang
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| | - Xijun Liang
- Department of Respiratory Medicine, Cangzhou People's Hospital, Cangzhou061000, China
| |
Collapse
|
11
|
Liu X, Zhang X, Yao C, Liang J, Noble PW, Jiang D. Transcriptomics Analysis Identifies the Decline in the Alveolar Type II Stem Cell Niche in Aged Human Lungs. Am J Respir Cell Mol Biol 2024; 71:229-241. [PMID: 38635761 PMCID: PMC11299088 DOI: 10.1165/rcmb.2023-0363oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 04/18/2024] [Indexed: 04/20/2024] Open
Abstract
Aging poses a global public health challenge, which is linked to the rise of age-related lung diseases. The precise understanding of the molecular and genetic changes in the aging lung that elevate the risk of acute and chronic lung diseases remains incomplete. Alveolar type II (AT2) cells are stem cells that maintain epithelial homeostasis and repair the lung after injury. AT2 progenitor function decreases with aging. The maintenance of AT2 function requires niche support from other cell types, but little has been done to characterize alveolar alterations with aging in the AT2 niche. To systematically profile the genetic changes associated with age, we present a single-cell transcriptional atlas comprising nearly half a million cells from the healthy lungs of human subjects spanning various ages, sexes, and smoking statuses. Most annotated cell lineages in aged lungs exhibit dysregulated genetic programs. Specifically, the aged AT2 cells demonstrate loss of epithelial identities, heightened inflammaging characterized by increased expression of AP-1 (Activator Protein-1) transcription factor and chemokine genes, and significantly increased cellular senescence. Furthermore, the aged mesenchymal cells display a remarkable decrease in collagen and elastin transcription and a loss of support to epithelial cell stemness. The decline of the AT2 niche is further exacerbated by a dysregulated genetic program in macrophages and dysregulated communications between AT2 and macrophages in aged human lungs. These findings highlight the dysregulations observed in both AT2 stem cells and their supportive niche cells, potentially contributing to the increased susceptibility of aged populations to lung diseases.
Collapse
Affiliation(s)
- Xue Liu
- Department of Medicine and Women’s Guild Lung Institute and
| | - Xuexi Zhang
- Department of Medicine and Women’s Guild Lung Institute and
| | - Changfu Yao
- Department of Medicine and Women’s Guild Lung Institute and
| | - Jiurong Liang
- Department of Medicine and Women’s Guild Lung Institute and
| | - Paul W. Noble
- Department of Medicine and Women’s Guild Lung Institute and
| | - Dianhua Jiang
- Department of Medicine and Women’s Guild Lung Institute and
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
12
|
Chung C, Lee KN, Shin DW, Lee SW, Han K. Low household income increases risks for chronic obstructive pulmonary disease in young population: a nationwide retrospective cohort study in South Korea. BMJ Open Respir Res 2024; 11:e002444. [PMID: 39074962 PMCID: PMC11288150 DOI: 10.1136/bmjresp-2024-002444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Low socioeconomic status is a risk factor for chronic obstructive pulmonary disease (COPD); however, the association between low household income and COPD in young populations remains unclear. METHODS We screened individuals aged 20-39 years who underwent the national health examination between 2009 and 2012 using the Korean National Health Information Database, which was searched until December 2019. We identified 5 965 366 eligible individuals, and 13 296 had newly developed COPD based on health insurance claims. We evaluated household income levels based on the health insurance premiums, categorised them into quartiles and 'Medical aid' (the lowest 3% income group), and assessed the annual income status from the preceding 4 years. Multivariate Cox proportional hazard models were used to estimate the adjusted HR (aHR) of risk factors for COPD. RESULTS In the Medical aid group, the incidence rate for developing COPD was 0.56/1000 person-years, with an aHR of 2.45 (95% CI 1.91 to 3.13) compared with that of the highest income quartile group. This association was prominent in consecutive recipients of Medical aid (aHR 2.37, 95% CI 1.80 to 3.11) compared with those who had never been Medical aid beneficiaries. Those who experienced a decline in household income between the previous (preceding 4 years) and baseline time points had an increased risk of developing COPD, regardless of previous income status. CONCLUSION Low household income was associated with an increased risk of developing COPD in the young population. This risk was augmented by sustained low income and declining income status.
Collapse
Affiliation(s)
- Chiwook Chung
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
- Department of Pulmonary and Critical Care Medicine, GangNeung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea (the Republic of)
| | - Kyu Na Lee
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea (the Republic of)
| | - Dong Wook Shin
- Department of Family Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea (the Republic of)
- Department of Clinical Research Design & Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Korea (the Republic of)
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea (the Republic of)
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea (the Republic of)
| |
Collapse
|
13
|
Yan D. Association between α-klotho levels and adults with COPD in the United States. Front Med (Lausanne) 2024; 11:1361922. [PMID: 39091285 PMCID: PMC11291460 DOI: 10.3389/fmed.2024.1361922] [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: 12/27/2023] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is accompanied by increased inflammation, persistent lung function decline, and extensive lung injury. Klotho, a well-known antiaging protein, has anti-inflammatory and antioxidative effects. However, the effects of klotho on COPD have yet to be thoroughly elucidated. This study examined the association among COPD adults and their α-klotho level. Patients and methods Data were collected from the 2007 to 2012 National Health and Nutrition Examination Survey (NHANES). A total of 676 participants were analyzed and divided into COPD (n = 403) and non-COPD (n = 273) groups. The two groups were compared with respect to clinical characteristics. Logistic regression analysis and a generalized additive model were used to estimate the association between COPD incidence and serum α-klotho concentration. All COPD participants were stratified according to the levels of α-klotho (Q1: <687 pg./mL; Q2: 687-900 pg./mL; Q3: ≥900 pg./mL), and clinical characteristics were compared. Results Non-COPD individuals had higher α-klotho levels than did COPD individuals (863.09 ± 267.13 vs. 817.51 ± 302.20, p < 0.05). Logistic regression analysis revealed that the Q2 and Q3 layers had a lower risk of COPD than did the Q1 layer, with odds ratios (ORs) of 0.73 (0.50, 0.99) for Q2 and 0.58 (0.41, 0.86) for Q3 (p < 0.001). The generalized additive model showed that the risk of COPD gradually decreased with increasing α-klotho concentration when the α-klotho concentration < 1,500 pg./mL, while the risk of COPD increased as the α-klotho concentration increased to ≥1,500 pg./mL. Compared with individuals in the Q2 or Q3 groups, individuals with COPD in the Q1 group were more likely to be current smokers, have lower levels of erythrocytes, and have higher levels of creatinine and leukocytes. Conclusion Increased α-klotho levels were negatively correlated with the risk of COPD in participants over 40 years old with α-klotho <1,500 pg./mL. When α-klotho was ≥1,500 pg./mL, the risk of COPD increased as α-klotho levels increased. Pulmonary ventilation function and the number of hemocytes differed among COPD patients with different levels of α-klotho.
Collapse
Affiliation(s)
- Dan Yan
- Department of Pulmonary and Critical Care Medicine, Jinhua Municipal Central Hospital, The Affiliated Jinhua Hospital, College of Medicine, Zhejiang University, Jinhua, China
| |
Collapse
|
14
|
Jha SK, De Rubis G, Devkota SR, Zhang Y, Adhikari R, Jha LA, Bhattacharya K, Mehndiratta S, Gupta G, Singh SK, Panth N, Dua K, Hansbro PM, Paudel KR. Cellular senescence in lung cancer: Molecular mechanisms and therapeutic interventions. Ageing Res Rev 2024; 97:102315. [PMID: 38679394 DOI: 10.1016/j.arr.2024.102315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/03/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Lung cancer stands as the primary contributor to cancer-related fatalities worldwide, affecting both genders. Two primary types exist where non-small cell lung cancer (NSCLC), accounts for 80-85% and SCLC accounts for 10-15% of cases. NSCLC subtypes include adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Smoking, second-hand smoke, radon gas, asbestos, and other pollutants, genetic predisposition, and COPD are lung cancer risk factors. On the other hand, stresses such as DNA damage, telomere shortening, and oncogene activation cause a prolonged cell cycle halt, known as senescence. Despite its initial role as a tumor-suppressing mechanism that slows cell growth, excessive or improper control of this process can cause age-related diseases, including cancer. Cellular senescence has two purposes in lung cancer. Researchers report that senescence slows tumor growth by constraining multiplication of impaired cells. However, senescent cells also demonstrate the pro-inflammatory senescence-associated secretory phenotype (SASP), which is widely reported to promote cancer. This review will look at the role of cellular senescence in lung cancer, describe its diagnostic markers, ask about current treatments to control it, look at case studies and clinical trials that show how senescence-targeting therapies can be used in lung cancer, and talk about problems currently being faced, and possible solutions for the same in the future.
Collapse
Affiliation(s)
- Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur, Uttar Pradesh 208016, India
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Shankar Raj Devkota
- Monash Biomedicine Discovery Institute, and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Yali Zhang
- School of Chemical Engineering, University of Adelaide, Adelaide 5005, Australia
| | - Radhika Adhikari
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Laxmi Akhileshwar Jha
- Naraina Vidya Peeth Group of Institutions, Faculty of Pharmacy, Dr. A. P. J. Abdul Kalam Technical University, Lucknow, Uttar Pradesh 0208020, India
| | - Kunal Bhattacharya
- Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam 781026, India; Royal School of Pharmacy, The Assam Royal Global University, Guwahati, Assam 781035, India
| | - Samir Mehndiratta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sachin Kumar Singh
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Nisha Panth
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW 2007, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia.
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia.
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, NSW 2007, Australia.
| |
Collapse
|
15
|
Xue H, Lan X, Xue T, Tang X, Yang H, Hu Z, Xu N, Xie B. PD-1 + T lymphocyte proportions and hospitalized exacerbation of COPD: a prospective cohort study. Respir Res 2024; 25:218. [PMID: 38789950 PMCID: PMC11127417 DOI: 10.1186/s12931-024-02847-6] [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: 01/12/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
OBJECTIVE To evaluate the predictive value of PD-1 expression in T lymphocytes for rehospitalization due to acute exacerbations of COPD (AECOPD) in discharged patients. METHODS 115 participants hospitalized with COPD (average age 71.8 ± 6.0 years) were recruited at Fujian Provincial Hospital. PD1+T lymphocytes proportions (PD1+T%), baseline demographics and clinical data were recorded at hospital discharge. AECOPD re-admission were collected at 1-year follow-up. Kaplan-Meier analysis compared the time to AECOPD readmissions among groups stratified by PD1+T%. Multivariable Cox proportional hazards regression and stratified analysis determined the correlation between PD1+T%, potential confounders, and AECOPD re-admission. ROC and DCA evaluated PD1+T% in enhancing the clinical predictive values of Cox models, BODE and CODEX. RESULTS 68 participants (59.1%) were AECOPD readmitted, those with AECOPD readmission exhibited significantly elevated baseline PD-1+CD4+T/CD4+T% and PD-1+CD8 + T/CD8 + T% compared to non-readmitted counterparts. PD1+ T lymphocyte levels statistically correlated with BODE and CODEX indices. Kaplan-Meier analysis demonstrated that those in Higher PD1+ T lymphocyte proportions had reduced time to AECOPD readmission (logRank p < 0.05). Cox analysis identified high PD1+CD4+T and PD1+CD8+T ratios as risk factors of AECOPD readmission, with hazard ratios of 1.384(95%CI [1.043-1.725]) and 1.401(95%CI [1.013-1.789]), respectively. Notably, in patients aged < 70 years and with fewer than twice AECOPD episodes in the previous year, high PD1+T lymphocyte counts significantly increased risk for AECOPD readmission(p < 0.05). The AECOPD readmission predictive model, incorporating PD1+T% exhibited superior discrimination to the Cox model, BODE index and CODEX index, AUC of ROC were 0.763(95%CI [0.633-0.893]) and 0.734(95%CI [0.570-0.899]) (DeLong's test p < 0.05).The DCA illustrates that integrating PD1+T% into models significantly enhances the utility in aiding clinical decision-making. CONCLUSION Evaluation of PD1+ lymphocyte proportions offer a novel perspective for identifying high-risk COPD patients, potentially providing insights for COPD management. TRIAL REGISTRATION Chinese Clinical Trial Registry (ChiCTR, URL: www.chictr.org.cn/ ), Registration number: ChiCTR2200055611 Date of Registration: 2022-01-14.
Collapse
Affiliation(s)
- Hong Xue
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China
| | - Xiuyan Lan
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China
| | - Ting Xue
- Center of Health Management, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Xuwei Tang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xuefu north Road, Fuzhou, 350122, China
| | - Haitao Yang
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, 1 Xuefu north Road, Fuzhou, 350122, China
| | - Nengluan Xu
- Department of Infectious Diseases, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, No.516 Jinrong South Street, Fuzhou, Fujian, 350001, China.
| | - Baosong Xie
- Department of Respiratory and Critical Care Medicine, Provincial School of Clinical Medicine, Fujian Provincial Hospital, Fujian Medical University, No.134 East Street, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
16
|
Jehloh L, Songwathana P, Kitrungrote L, Bourbonnais A. Perspectives of family caregivers and nurses on hospital discharge transitional care for Muslim older adults living with COPD: a qualitative study. BMC Nurs 2024; 23:273. [PMID: 38659051 PMCID: PMC11044287 DOI: 10.1186/s12912-024-01943-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND The increased number of emergency department visits among older adults living with chronic obstructive pulmonary disease reflects the challenges of hospital discharge transition, especially in those from a cultural minority. The barriers and facilitators of this discharge from the perspective of formal and informal care providers, such as nurses and family caregivers, are important to identify to provide effective symptom management and quality of care. The purpose of this study was to describe the barriers and facilitators in caring for Muslim older adults with chronic obstructive pulmonary disease (COPD) during hospital discharge transitional care. METHODS A descriptive qualitative study was conducted in a hospital of Thailand where Muslim people are a cultural minority. Thirteen family caregivers of Muslim older adults living with COPD and seven nurses were purposively recruited and participated in semi-structured interviews and focus group discussions. Content analysis was used to analyze the data. RESULTS Five barriers and three facilitating factors of transitional care for Muslim older adults living with COPD were outlined. Barriers included: (1) lack of knowledge about the causes and management of dyspnea, (2) inadequate discharge preparation, (3) language barrier, (4) discontinuity of care, and (5) COVID-19 epidemic. Facilitators included: (1) the ability to understand Malayu language, (2) the presence of healthcare professionals of the same gender, and (3) the presence of Muslim healthcare providers. CONCLUSION Family caregivers require more supportive care to meet the care needs of Muslim older adults living with COPD. Alternative nurse-based transitional care programs for these older adult caregivers should be developed.
Collapse
Affiliation(s)
- Latifah Jehloh
- Faculty of Nursing, Prince of Narathiwas University, Muang, Narathiwat, Thailand
| | - Praneed Songwathana
- Faculty of Nursing, Prince of Songkla University, Hatyai, Songkhla, Thailand.
| | - Luppana Kitrungrote
- Faculty of Nursing, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Anne Bourbonnais
- Faculty of Nursing, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
17
|
Zhu Z, Zhao S, Li J, Wang Y, Xu L, Jia Y, Li Z, Li W, Chen G, Wu X. Development and application of a deep learning-based comprehensive early diagnostic model for chronic obstructive pulmonary disease. Respir Res 2024; 25:167. [PMID: 38637823 PMCID: PMC11027407 DOI: 10.1186/s12931-024-02793-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/28/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a frequently diagnosed yet treatable condition, provided it is identified early and managed effectively. This study aims to develop an advanced COPD diagnostic model by integrating deep learning and radiomics features. METHODS We utilized a dataset comprising CT images from 2,983 participants, of which 2,317 participants also provided epidemiological data through questionnaires. Deep learning features were extracted using a Variational Autoencoder, and radiomics features were obtained using the PyRadiomics package. Multi-Layer Perceptrons were used to construct models based on deep learning and radiomics features independently, as well as a fusion model integrating both. Subsequently, epidemiological questionnaire data were incorporated to establish a more comprehensive model. The diagnostic performance of standalone models, the fusion model and the comprehensive model was evaluated and compared using metrics including accuracy, precision, recall, F1-score, Brier score, receiver operating characteristic curves, and area under the curve (AUC). RESULTS The fusion model exhibited outstanding performance with an AUC of 0.952, surpassing the standalone models based solely on deep learning features (AUC = 0.844) or radiomics features (AUC = 0.944). Notably, the comprehensive model, incorporating deep learning features, radiomics features, and questionnaire variables demonstrated the highest diagnostic performance among all models, yielding an AUC of 0.971. CONCLUSION We developed and implemented a data fusion strategy to construct a state-of-the-art COPD diagnostic model integrating deep learning features, radiomics features, and questionnaire variables. Our data fusion strategy proved effective, and the model can be easily deployed in clinical settings. TRIAL REGISTRATION Not applicable. This study is NOT a clinical trial, it does not report the results of a health care intervention on human participants.
Collapse
Affiliation(s)
- Zecheng Zhu
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shunjin Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Lanxi Branch (Lanxi People's Hospital), Hangzhou, Zhejiang, China
| | - Jiahui Li
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuting Wang
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Luopiao Xu
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yubing Jia
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zihan Li
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenyuan Li
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Gang Chen
- College of Computer Science and Technology, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xifeng Wu
- Center of Clinical Big Data and Analytics of The Second Affiliated Hospital and Department of Big Data in Health Science School of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Li CL, Liu JF, Liu SF. Mitochondrial Dysfunction in Chronic Obstructive Pulmonary Disease: Unraveling the Molecular Nexus. Biomedicines 2024; 12:814. [PMID: 38672169 PMCID: PMC11048013 DOI: 10.3390/biomedicines12040814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent and debilitating respiratory disorder characterized by persistent airflow limitation and chronic inflammation. In recent years, the role of mitochondrial dysfunction in COPD pathogenesis has emerged as a focal point of investigation. This review endeavors to unravel the molecular nexus between mitochondrial dysfunction and COPD, delving into the intricate interplay of oxidative stress, bioenergetic impairment, mitochondrial genetics, and downstream cellular consequences. Oxidative stress, a consequence of mitochondrial dysfunction, is explored as a driving force behind inflammation, exacerbating the intricate cascade of events leading to COPD progression. Bioenergetic impairment sheds light on the systemic consequences of mitochondrial dysfunction, impacting cellular functions and contributing to the overall energy imbalance observed in COPD patients. This review navigates through the genetic landscape, elucidating the role of mitochondrial DNA mutations, variations, and haplogroups in COPD susceptibility and severity. Cellular consequences, including apoptosis, autophagy, and cellular senescence, are examined, providing insights into the intricate mechanisms by which mitochondrial dysfunction influences COPD pathology. Therapeutic implications, spanning antioxidant strategies, mitochondria-targeted compounds, and lifestyle modifications, are discussed in the context of translational research. Important future directions include identifying novel biomarkers, advancing mitochondria-targeted therapies, and embracing patient-centric approaches to redefine COPD management. This abstract provides a comprehensive overview of our review, offering a roadmap for understanding and addressing the molecular nexus between mitochondrial dysfunction and COPD, with potential implications for precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- Chin-Ling Li
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Jui-Fang Liu
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
| | - Shih-Feng Liu
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Respiratory Care, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
- Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Chiayi 600, Taiwan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
19
|
Polman R, Hurst JR, Uysal OF, Mandal S, Linz D, Simons S. Cardiovascular disease and risk in COPD: a state of the art review. Expert Rev Cardiovasc Ther 2024; 22:177-191. [PMID: 38529639 DOI: 10.1080/14779072.2024.2333786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/19/2024] [Indexed: 03/27/2024]
Abstract
INTRODUCTION Chronic Obstructive Pulmonary Disease (COPD) and cardiovascular diseases (CVD) commonly co-exist. Outcomes of people living with both conditions are poor in terms of symptom burden, receiving evidence-based treatment and mortality. Increased understanding of the underlying mechanisms may help to identify treatments to relieve this disease burden. This narrative review covers the overlap of COPD and CVD with a focus on clinical presentation, mechanisms, and interventions. Literature up to December 2023 are cited. AREAS COVERED 1. What is COPD 2. The co-existence of COPD and cardiovascular disease 3. Mechanisms of cardiovascular disease in COPD. 4. Populations with COPD are at risk of CVD 5. Complexity in the co-diagnosis of COPD in those with cardiovascular disease. 6. Therapy for COPD and implications for cardiovascular events and risk. 7. Cardiovascular risk and exacerbations of COPD. 8. Pro-active identification and management of CV risk in COPD. EXPERT OPINION The prospective identification of co-morbid COPD in CVD patients and of CVD and CV risk in people with COPD is crucial for optimizing clinical outcomes. This includes the identification of novel treatment targets and the design of clinical trials specifically designed to reduce the cardiovascular burden and mortality associated with COPD. Databases searched: Pubmed, 2006-2023.
Collapse
Affiliation(s)
- Ricardo Polman
- Department of Respiratory Medicine, Maastricht UMC+, Maastricht, the Netherlands
| | - John R Hurst
- UCL Respiratory, University College London, London, UK
| | | | - Swapna Mandal
- UCL Respiratory, University College London, London, UK
| | - Dominik Linz
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
- Centre for Heart Rhythm Disorders, South Australian Health and Medical Research Institute, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Sami Simons
- Department of Respiratory Medicine, Maastricht UMC+, Maastricht, the Netherlands
- Department of Respiratory Medicine, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
20
|
Jesus FR, Moraes ACS, da Silva ILN, Passos FC, Salles C, Neves MCLC, Baccan GC. Analysis of Endocrine and Inflammatory Markers in Preserved Ratio Impaired Spirometry. Med Sci (Basel) 2024; 12:18. [PMID: 38651412 PMCID: PMC11036252 DOI: 10.3390/medsci12020018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a disease of the lungs characterized by chronic airflow obstruction. Individuals with preserved ratio impaired spirometry (PRISm) may be at risk for developing COPD. This study aimed to characterize PRISm and COPD patients in terms of their immune response and endocrine profile to identify differences extending beyond lung function. The participants performed the clinical assessment, pulmonary function test, and blood collection to determine serum hormone levels and concentrations of cytokine. Differences were observed in the nutritional status, lung function, and comorbidity. There were no differences in IL-6, IL-8, IL-10, IL-12, and TNF levels between PRISm and COPD groups. Both PRISm and COPD patients have lower dehydroepiandrosterone (DHEA) and dehydroepiandrosterone sulfate (DHEA-S) levels than controls. Correlation analysis of PRISm and COPD patients revealed positive correlations between serum levels of DHEA-S and DHEA, with forced expiratory volume in 1 second (FEV1) and forced vital capacity (FVC), which negatively correlated with IL-8 levels. The results indicated that despite differences in lung function parameters, the PRISm and COPD groups exhibited similarities in endocrine profile alterations. This study represents the first attempt to link endocrine with immune markers and lung function in individuals with PRISm.
Collapse
Affiliation(s)
- Fabíola Ramos Jesus
- Maternidade Climério de Oliveira (MCO/EBSERH), Universidade Federal da Bahia, Salvador 40055-150, Bahia, Brazil;
| | - Anna Clara Santiago Moraes
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40110-110, Bahia, Brazil
| | - Ingrid Lorena Neves da Silva
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40110-110, Bahia, Brazil
| | - Fabine Correia Passos
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40110-110, Bahia, Brazil
| | - Cristina Salles
- Unidade do Sistema Respiratório, Ambulatório Professor Francisco Magalhães Neto-Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador 40110-200, Bahia, Brazil
| | - Margarida Célia Lima Costa Neves
- Unidade do Sistema Respiratório, Ambulatório Professor Francisco Magalhães Neto-Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador 40110-200, Bahia, Brazil
| | - Gyselle Chrystina Baccan
- Departamento de Bioquímica e Biofísica, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40110-110, Bahia, Brazil
| |
Collapse
|
21
|
Steinhardt M, Marka AW, Ziegelmayer S, Makowski M, Braren R, Graf M, Gawlitza J. Comparison of Virtual Non-Contrast and True Non-Contrast CT Images Obtained by Dual-Layer Spectral CT in COPD Patients. Bioengineering (Basel) 2024; 11:301. [PMID: 38671723 PMCID: PMC11047621 DOI: 10.3390/bioengineering11040301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/10/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the leading causes of death. Recent studies have underlined the importance of non-contrast-enhanced chest CT scans not only for emphysema progression quantification, but for correlation with clinical outcomes as well. As about 40 percent of the 300 million CT scans per year are contrast-enhanced, no proper emphysema quantification is available in a one-stop-shop approach for patients with known or newly diagnosed COPD. Since the introduction of spectral imaging (e.g., dual-energy CT scanners), it has been possible to create virtual non-contrast-enhanced images (VNC) from contrast-enhanced images, making it theoretically possible to offer proper COPD imaging despite contrast enhancing. This study is aimed towards investigating whether these VNC images are comparable to true non-contrast-enhanced images (TNC), thereby reducing the radiation exposure of patients and usage of resources in hospitals. In total, 100 COPD patients with two scans, one with (VNC) and one without contrast media (TNC), within 8 weeks or less obtained by a spectral CT using dual-layer technology, were included in this retrospective study. TNC and VNC were compared according to their voxel-density histograms. While the comparison showed significant differences in the low attenuated volumes (LAVs) of TNC and VNC regarding the emphysema threshold of -950 Houndsfield Units (HU), the 15th and 10th percentiles of the LAVs used as a proxy for pre-emphysema were comparable. Upon further investigation, the threshold-based LAVs (-950 HU) of TNC and VNC were comparable in patients with a water equivalent diameter (DW) below 270 mm. The study concludes that VNC imaging may be a viable option for assessing emphysema progression in COPD patients, particularly those with a normal body mass index (BMI). Further, pre-emphysema was generally comparable between TNC and VNC. This approach could potentially reduce radiation exposure and hospital resources by making additional TNC scans obsolete.
Collapse
Affiliation(s)
- Manuel Steinhardt
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (A.W.M.); (S.Z.); (M.M.); (R.B.); (M.G.)
| | | | | | | | | | | | - Joshua Gawlitza
- Correspondence: (M.S.); (J.G.); Tel.: +49-176-24498226 (M.S.); +49-89-4140-8834 (J.G.)
| |
Collapse
|
22
|
Wang Q, Goracci C, Sundar IK, Rahman I. Environmental tobacco smoke exposure exaggerates bleomycin-induced collagen overexpression during pulmonary fibrogenesis. J Inflamm (Lond) 2024; 21:9. [PMID: 38509574 PMCID: PMC10956237 DOI: 10.1186/s12950-024-00377-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 02/12/2024] [Indexed: 03/22/2024] Open
Abstract
Environmental tobacco smoke (ETS) is known to cause lung inflammatory and injurious responses. Smoke exposure is associated with the pathobiology related to lung fibrosis, whereas the mechanism that ETS exposure augments pulmonary fibrogenesis is unclear. We hypothesized that ETS exposure could exacerbate fibrotic responses via collagen dynamic dysregulation and complement activation. C57BL/6J and p16-3MR mice were exposed to ETS followed by bleomycin administration. ETS exposure exacerbated bleomycin-induced collagen and lysyl oxidase overexpression in the fibrotic lesion. ETS exposure also led to augmented bleomycin-induced upregulation of C3 and C3AR, which are pro-fibrotic markers. Moreover, overexpressed collagens and C3 levels were highly significant in males than females. The old mice (17 months old) were exposed to ETS and treated with bleomycin to induce fibrogenesis which is considered as an aging-associated disease. Fewer gene and protein dysregulations trends were identified between ETS exposure with the bleomycin group and the bleomycin alone group in old mice. Based on our findings, we suggested that ETS exposure increases the risk of developing severe lung fibrotic responses via collagen overexpression and lysyl oxidase-mediated collagen stabilization in the fibrotic lesion, and potentially affected the complement system activation induced by bleomycin. Further, male mice were more susceptible than females during fibrogenesis exacerbation. Thus ETS and bleomycin induced lung fibrotic changes via collagen-lysyl oxidase in an age-dependent mechanism.
Collapse
Affiliation(s)
- Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Chiara Goracci
- Department of Environmental Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Isaac Kirubakaran Sundar
- Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.
| |
Collapse
|
23
|
Banturaki A, Munyambalu DK, Kajoba D, Onchoke VB, Peris A, Ryamugwiza P, Amandua J, Akaba K. Chronic obstructive pulmonary disease burden, grades and erythrocytosis at a tertiary hospital in western Uganda. BMC Pulm Med 2024; 24:119. [PMID: 38448860 PMCID: PMC10918867 DOI: 10.1186/s12890-024-02944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide among people over 40 years of age, and erythrocytosis is one of the major complications associated with increased mortality among COPD patients. The study aimed to determine the proportion of COPD, associated factors, and the burden of erythrocytosis among COPD participants. METHODS AND MATERIALS A descriptive cross-sectional study design was used. A consecutive sampling technique was used to obtain study participants at the Fort Portal Regional Referral Hospital outpatient clinic. Focused history and physical examination were carried out to select eligible participants. Participants were screened using the COPD population screener for spirometry after consenting to participate. The study enrolled all adults at risk of having COPD based on the COPD population screener and able to undergo spirometry. Spirometry was carried out according to the Global Chronic Obstructive Lung Disease and European Respiratory Society guidelines, and haemoglobin concentration was measured. RESULTS One hundred eighty participants were enrolled in the study, most of whom were females. The modal and mean age of participants was 60 years with 139 (77.2%) females and primary as the highest education level 149(82.8%). The proportion of COPD was 25% (45) [95% CI 18.9 - 32] and highest among females (68.9%) and those aged 60 years and above (70%). The combined COPD assessment tool groups had a proportion of 55.6%, 37.8%, 4.4%, and 2.2% for groups A, B, C, and D, respectively. Age < 50 years was protective against COPD, while for every additional year of smoking, there was an associated 6.5% increased risk compared to the general population. Additionally, the proportion of erythrocytosis among COPD participants was 6.7%. CONCLUSIONS AND RECOMMENDATIONS There was a high proportion of COPD among study participants (25%), with a 6.7% proportion of erythrocytosis. We recommend a complete blood count for every patient in groups C and D of the ABCD COPD GOLD groups.
Collapse
Affiliation(s)
- Amon Banturaki
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda.
| | - Dalton Kambale Munyambalu
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda
| | - Dickson Kajoba
- Department of Paediatrics and Child Health, Kampala International University-Teaching Hospital, Ishaka- Bushenyi, Uganda
| | - Verah Bella Onchoke
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda
| | - Alina Peris
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda
| | - Prosper Ryamugwiza
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda
| | - Jacinto Amandua
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda
| | - Kingsley Akaba
- Department of Internal Medicine, Kampala International University-Teaching Hospital, P.O. BOX 71, Ishaka- Bushenyi, Uganda
| |
Collapse
|
24
|
Gilad N, Mohanam MP, Darlyuk-Saadon I, Heng CKM, Plaschkes I, Benyamini H, Berezhnoy NV, Engelberg D. Asynchronous Pattern of MAPKs' Activity during Aging of Different Tissues and of Distinct Types of Skeletal Muscle. Int J Mol Sci 2024; 25:1713. [PMID: 38338990 PMCID: PMC10855984 DOI: 10.3390/ijms25031713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
The MAPK p38α was proposed to be a prominent promoter of skeletal muscle aging. The skeletal muscle tissue is composed of various muscle types, and it is not known if p38α is associated with aging in all of them. It is also not known if p38α is associated with aging of other tissues. JNK and ERK were also proposed to be associated with aging of several tissues. Nevertheless, the pattern of p38α, JNK, and ERK activity during aging was not documented. Here, we documented the levels of phosphorylated/active p38α, Erk1/2, and JNKs in several organs as well as the soleus, tibialis anterior, quadriceps, gastrocnemius, and EDL muscles of 1-, 3-, 6-, 13-, 18-, and 24-month-old mice. We report that in most tissues and skeletal muscles, the MAPKs' activity does not change in the course of aging. In most tissues and muscles, p38α is in fact active at younger ages. The quadriceps and the lungs are exceptions, where p38α is significantly active only in mice 13 months old or older. Curiously, levels of active JNK and ERKs are also elevated in aged lungs and quadriceps. RNA-seq analysis of the quadriceps during aging revealed downregulation of proteins related to the extra-cellular matrix (ECM) and ERK signaling. A panel of mRNAs encoding cell cycle inhibitors and senescence-associated proteins, considered to be aging markers, was not found to be elevated. It seems that the pattern of MAPKs' activation in aging, as well as expression of known 'aging' components, are tissue- and muscle type-specific, supporting a notion that the process of aging is tissue- and even cell-specific.
Collapse
Affiliation(s)
- Nechama Gilad
- Department of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
| | - Manju Payini Mohanam
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Ilona Darlyuk-Saadon
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
| | - C. K. Matthew Heng
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Inbar Plaschkes
- Info-CORE, Bioinformatics Unit of the I-CORE, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Hadar Benyamini
- Info-CORE, Bioinformatics Unit of the I-CORE, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Nikolay V. Berezhnoy
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - David Engelberg
- Department of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem 91904, Israel;
- Singapore-HUJ Alliance for Research and Enterprise, Mechanisms of Liver Inflammatory Diseases Program, National University of Singapore, Singapore 138602, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
25
|
Pandics T, Major D, Fazekas-Pongor V, Szarvas Z, Peterfi A, Mukli P, Gulej R, Ungvari A, Fekete M, Tompa A, Tarantini S, Yabluchanskiy A, Conley S, Csiszar A, Tabak AG, Benyo Z, Adany R, Ungvari Z. Exposome and unhealthy aging: environmental drivers from air pollution to occupational exposures. GeroScience 2023; 45:3381-3408. [PMID: 37688657 PMCID: PMC10643494 DOI: 10.1007/s11357-023-00913-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/14/2023] [Indexed: 09/11/2023] Open
Abstract
The aging population worldwide is facing a significant increase in age-related non-communicable diseases, including cardiovascular and brain pathologies. This comprehensive review paper delves into the impact of the exposome, which encompasses the totality of environmental exposures, on unhealthy aging. It explores how environmental factors contribute to the acceleration of aging processes, increase biological age, and facilitate the development and progression of a wide range of age-associated diseases. The impact of environmental factors on cognitive health and the development of chronic age-related diseases affecting the cardiovascular system and central nervous system is discussed, with a specific focus on Alzheimer's disease, Parkinson's disease, stroke, small vessel disease, and vascular cognitive impairment (VCI). Aging is a major risk factor for these diseases. Their pathogenesis involves cellular and molecular mechanisms of aging such as increased oxidative stress, impaired mitochondrial function, DNA damage, and inflammation and is influenced by environmental factors. Environmental toxicants, including ambient particulate matter, pesticides, heavy metals, and organic solvents, have been identified as significant contributors to cardiovascular and brain aging disorders. These toxicants can inflict both macro- and microvascular damage and many of them can also cross the blood-brain barrier, inducing neurotoxic effects, neuroinflammation, and neuronal dysfunction. In conclusion, environmental factors play a critical role in modulating cardiovascular and brain aging. A deeper understanding of how environmental toxicants exacerbate aging processes and contribute to the pathogenesis of neurodegenerative diseases, VCI, and dementia is crucial for the development of preventive strategies and interventions to promote cardiovascular, cerebrovascular, and brain health. By mitigating exposure to harmful environmental factors and promoting healthy aging, we can strive to reduce the burden of age-related cardiovascular and brain pathologies in the aging population.
Collapse
Affiliation(s)
- Tamas Pandics
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Department of Public Health Laboratory, National Public Health Centre, Budapest, Hungary
- Department of Public Health Siences, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - David Major
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Vince Fazekas-Pongor
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Peterfi
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Ungvari
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Monika Fekete
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Anna Tompa
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adam G Tabak
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- UCL Brain Sciences, University College London, London, UK
- Department of Internal Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zoltan Benyo
- Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University (ELKH-SE) Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, H-1052, Hungary
| | - Roza Adany
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- ELKH-DE Public Health Research Group, Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary
- Epidemiology and Surveillance Centre, Semmelweis University, 1085, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
26
|
Wang Q, Goracci C, Sundar IK, Rahman I. Environmental tobacco smoke exposure exaggerates bleomycin- induced collagen overexpression during pulmonary fibrogenesis. RESEARCH SQUARE 2023:rs.3.rs-3406872. [PMID: 37886473 PMCID: PMC10602094 DOI: 10.21203/rs.3.rs-3406872/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Environmental tobacco smoke (ETS) is known to cause lung inflammatory and injurious responses. Smoke exposure is associated with the pathobiology related to lung fibrosis, whereas the mechanism by which ETS exposure augments lung fibrogenesis is unclear. We hypothesized that ETS exposure could exacerbate fibrotic responses via collagen dynamic dysregulation and complement activation. C57BL/6J and p16-3MR mice were exposed to ETS followed by bleomycin administration. ETS exposure exacerbated bleomycin-induced collagen and lysyl oxidase overexpression in the fibrotic lesion. ETS exposure also led to augmented bleomycin-induced upregulation of C3 and C3AR, which are pro-fibrotic markers. Moreover, overexpressed collagens and C3 levels were highly significant in males than females. The old mice (17 months old) were exposed to ETS and treated with bleomycin to induce fibrogenesis, since fibrogenesis is an aging-associated disease. Fewer gene and protein dysregulations trends were identified between ETS exposure with the bleomycin group and the bleomycin alone group in old mice. Based on our findings, we suggested that ETS exposure increases the risk of developing severe lung fibrotic responses via collagen overexpression and lysyl oxidase-mediated collagen stabilization in the fibrotic lesion. ETS exposure also potentially affected the complement system activation induced by bleomycin. Further, male mice were more susceptible than females during fibrogenesis exacerbation.
Collapse
|
27
|
Song Y, Liu YS, Talarico F, Zhang Y, Hayward J, Wang M, Stroulia E, Dixon RA, Greiner R, Li X, Greenshaw A, Jie S, Cao B. Associations between Differential Aging and Lifestyle, Environment, Current, and Future Health Conditions: Findings from Canadian Longitudinal Study on Aging. Gerontology 2023; 69:1394-1403. [PMID: 37725932 DOI: 10.1159/000534015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 09/05/2023] [Indexed: 09/21/2023] Open
Abstract
INTRODUCTION An aging population will bring a pressing challenge for the healthcare system. Insights into promoting healthy longevity can be gained by quantifying the biological aging process and understanding the roles of modifiable lifestyle and environmental factors, and chronic disease conditions. METHODS We developed a biological age (BioAge) index by applying multiple state-of-art machine learning models based on easily accessible blood test data from the Canadian Longitudinal Study of Aging (CLSA). The BioAge gap, which is the difference between BioAge index and chronological age, was used to quantify the differential aging, i.e., the difference between biological and chronological age, of the CLSA participants. We further investigated the associations between the BioAge gap and lifestyle, environmental factors, and current and future health conditions. RESULTS BioAge gap had strong associations with existing adverse health conditions (e.g., cancers, cardiovascular diseases, diabetes, and kidney diseases) and future disease onset (e.g., Parkinson's disease, diabetes, and kidney diseases). We identified that frequent consumption of processed meat, pork, beef, and chicken, poor outcomes in nutritional risk screening, cigarette smoking, exposure to passive smoking are associated with positive BioAge gap ("older" BioAge than expected). We also identified several modifiable factors, including eating fruits, legumes, vegetables, related to negative BioAge gap ("younger" BioAge than expected). CONCLUSIONS Our study shows that a BioAge index based on easily accessible blood tests has the potential to quantify the differential biological aging process that can be associated with current and future adverse health events. The identified risk and protective factors for differential aging indicated by BioAge gap are informative for future research and guidelines to promote healthy longevity.
Collapse
Affiliation(s)
- Yipeng Song
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada,
| | - Yang S Liu
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Fernanda Talarico
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Yanbo Zhang
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Jake Hayward
- University of Alberta, Department of Emergency Medicine, Edmonton, Alberta, Canada
| | - Mengzhe Wang
- Ministry of Health (Alberta), Edmonton, Alberta, Canada
| | - Eleni Stroulia
- University of Alberta, Department of Computing Science, Edmonton, Alberta, Canada
| | - Roger A Dixon
- University of Alberta, Department of Psychology, Edmonton, Alberta, Canada
| | - Russell Greiner
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
- University of Alberta, Department of Computing Science, Edmonton, Alberta, Canada
| | - Xinmin Li
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Andrew Greenshaw
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
| | - Sui Jie
- University of Aberdeen, The School of Psychology, Aberdeen, UK
| | - Bo Cao
- University of Alberta, Department of Psychiatry, Edmonton, Alberta, Canada
- University of Alberta, Department of Computing Science, Edmonton, Alberta, Canada
| |
Collapse
|
28
|
Chung C, Lee KN, Han K, Shin DW, Lee SW. Effect of smoking on the development of chronic obstructive pulmonary disease in young individuals: a nationwide cohort study. Front Med (Lausanne) 2023; 10:1190885. [PMID: 37593403 PMCID: PMC10428618 DOI: 10.3389/fmed.2023.1190885] [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: 03/21/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
Background Cigarette smoking is an important risk factor for developing chronic obstructive pulmonary disease (COPD). However, the effect of smoking on the development of COPD in young individuals remains unclear. We aimed to evaluate the effect of smoking on COPD development in young individuals. Methods Using the Korean National Health Information Database, we screened individuals aged 20-39 years who participated in the national health check-up between 2009 and 2012. We defined physician-diagnosed COPD based on health insurance claims and searched the database until December 2019. We identified 6,307,576 eligible individuals, and 13,789 had newly developed COPD. We used multivariate Cox proportional hazards models to estimate the adjusted hazard ratio (aHR) of risk factors for COPD. Results The incidence rate for developing COPD was 0.26/1000 person-year. The risk of developing COPD was significantly higher in current smokers [aHR 1.46, 95% confidence interval (CI) 1.39-1.53] and former smokers (aHR 1.21, 95% CI 1.14-1.29) than in non-smokers. Furthermore, the risk increased with increasing smoking amounts (≥20 pack-years, aHR 2.24; 10-20 pack-years, aHR 1.55; <10 pack-years, aHR 1.27). Female participants had a higher relative risk of developing COPD due to smoking, compared with their male counterparts. Conclusion Cigarette smoking increased the risk of developing COPD in young individuals. Current and heavy smokers had higher risks of developing COPD than non-smokers. Female smokers were more likely to develop COPD than male smokers.
Collapse
Affiliation(s)
- Chiwook Chung
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Pulmonary, Allergy, and Critical Care Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Republic of Korea
| | - Kyu Na Lee
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Dong Wook Shin
- Supportive Care Center, Samsung Comprehensive Cancer Center, Department of Family Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
29
|
Sucre JM, Bock F, Negretti NM, Benjamin JT, Gulleman PM, Dong X, Ferguson KT, Jetter CS, Han W, Liu Y, Kook S, Gokey JJ, Guttentag SH, Kropski JA, Blackwell TS, Zent R, Plosa EJ. Alveolar repair following LPS-induced injury requires cell-ECM interactions. JCI Insight 2023; 8:e167211. [PMID: 37279065 PMCID: PMC10443799 DOI: 10.1172/jci.insight.167211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
During alveolar repair, alveolar type 2 (AT2) epithelial cell progenitors rapidly proliferate and differentiate into flat AT1 epithelial cells. Failure of normal alveolar repair mechanisms can lead to loss of alveolar structure (emphysema) or development of fibrosis, depending on the type and severity of injury. To test if β1-containing integrins are required during repair following acute injury, we administered E. coli lipopolysaccharide (LPS) by intratracheal injection to mice with a postdevelopmental deletion of β1 integrin in AT2 cells. While control mice recovered from LPS injury without structural abnormalities, β1-deficient mice had more severe inflammation and developed emphysema. In addition, recovering alveoli were repopulated with an abundance of rounded epithelial cells coexpressing AT2 epithelial, AT1 epithelial, and mixed intermediate cell state markers, with few mature type 1 cells. AT2 cells deficient in β1 showed persistently increased proliferation after injury, which was blocked by inhibiting NF-κB activation in these cells. Lineage tracing experiments revealed that β1-deficient AT2 cells failed to differentiate into mature AT1 epithelial cells. Together, these findings demonstrate that functional alveolar repair after injury with terminal alveolar epithelial differentiation requires β1-containing integrins.
Collapse
Affiliation(s)
- Jennifer M.S. Sucre
- Department of Pediatrics, Division of Neonatology
- Department of Cell and Developmental Biology
| | - Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | | | - Xinyu Dong
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | - Wei Han
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yang Liu
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jason J. Gokey
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jonathan A. Kropski
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Timothy S. Blackwell
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Nephrology and Hypertension; and
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
30
|
Bateman G, Guo-Parke H, Rodgers AM, Linden D, Bailey M, Weldon S, Kidney JC, Taggart CC. Airway Epithelium Senescence as a Driving Mechanism in COPD Pathogenesis. Biomedicines 2023; 11:2072. [PMID: 37509711 PMCID: PMC10377597 DOI: 10.3390/biomedicines11072072] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Cellular senescence is a state of permanent cell cycle arrest triggered by various intrinsic and extrinsic stressors. Cellular senescence results in impaired tissue repair and remodeling, loss of physiological integrity, organ dysfunction, and changes in the secretome. The systemic accumulation of senescence cells has been observed in many age-related diseases. Likewise, cellular senescence has been implicated as a risk factor and driving mechanism in chronic obstructive pulmonary disease (COPD) pathogenesis. Airway epithelium exhibits hallmark features of senescence in COPD including activation of the p53/p21WAF1/CIP1 and p16INK4A/RB pathways, leading to cell cycle arrest. Airway epithelial senescent cells secrete an array of inflammatory mediators, the so-called senescence-associated secretory phenotype (SASP), leading to a persistent low-grade chronic inflammation in COPD. SASP further promotes senescence in an autocrine and paracrine manner, potentially contributing to the onset and progression of COPD. In addition, cellular senescence in COPD airway epithelium is associated with telomere dysfunction, DNA damage, and oxidative stress. This review discusses the potential mechanisms of airway epithelial cell senescence in COPD, the impact of cellular senescence on the development and severity of the disease, and highlights potential targets for modulating cellular senescence in airway epithelium as a potential therapeutic approach in COPD.
Collapse
Affiliation(s)
- Georgia Bateman
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK
| | - Hong Guo-Parke
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK
| | - Aoife M Rodgers
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK
| | - Dermot Linden
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK
| | - Melanie Bailey
- Department of Respiratory Medicine, Mater Hospital Belfast, Belfast BT14 6AB, UK
| | - Sinéad Weldon
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK
| | - Joseph C Kidney
- Department of Respiratory Medicine, Mater Hospital Belfast, Belfast BT14 6AB, UK
| | - Clifford C Taggart
- Airway Innate Immunity Research Group, Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast BT9 7AE, UK
| |
Collapse
|
31
|
Jia M, Sayed K, Kapetanaki MG, Dion W, Rosas L, Irfan S, Valenzi E, Mora AL, Lafyatis RA, Rojas M, Zhu B, Benos PV. LEF1 isoforms regulate cellular senescence and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.20.549883. [PMID: 37502913 PMCID: PMC10370160 DOI: 10.1101/2023.07.20.549883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background The study of aging and its mechanisms, such as cellular senescence, has provided valuable insights into age-related pathologies, thus contributing to their prevention and treatment. The current abundance of high throughput data combined with the surge of robust analysis algorithms has facilitated novel ways of identifying underlying pathways that may drive these pathologies. Methods With the focus on identifying key regulators of lung aging, we performed comparative analyses of transcriptional profiles of aged versus young human subjects and mice, focusing on the common age-related changes in the transcriptional regulation in lung macrophages, T cells, and B immune cells. Importantly, we validated our findings in cell culture assays and human lung samples. Results We identified Lymphoid Enhancer Binding Factor 1 (LEF1) as an important age-associated regulator of gene expression in all three cell types across different tissues and species. Follow-up experiments showed that the differential expression of long and short LEF1 isoforms is a key regulatory mechanism of cellular senescence. Further examination of lung tissue from patients with Idiopathic Pulmonary Fibrosis (IPF), an age-related disease with strong ties to cellular senescence, we demonstrated a stark dysregulation of LEF1. Conclusions Collectively, our results suggest that the LEF1 is a key factor of aging, and its differential regulation is associated with human and murine cellular senescence.
Collapse
Affiliation(s)
- Minxue Jia
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Joint Carnegie Mellon University-University of Pittsburgh Ph.D. Program in Computational Biology, Pittsburgh, Pennsylvania, USA
| | - Khaled Sayed
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
| | - Maria G. Kapetanaki
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William Dion
- Aging Institute of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lorena Rosas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Saad Irfan
- Aging Institute of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eleanor Valenzi
- Department of Rheumatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ana L. Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Robert A. Lafyatis
- Department of Rheumatology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Bokai Zhu
- Aging Institute of UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pennsylvania, USA
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Joint Carnegie Mellon University-University of Pittsburgh Ph.D. Program in Computational Biology, Pittsburgh, Pennsylvania, USA
- Department of Epidemiology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
32
|
Zeng J, Liao S, Liang Z, Li C, Luo Y, Wang K, Zhang D, Lan L, Hu S, Li W, Lin R, Jie Z, Hu Y, Dai S, Zhang Z. Schisandrin A regulates the Nrf2 signaling pathway and inhibits NLRP3 inflammasome activation to interfere with pyroptosis in a mouse model of COPD. Eur J Med Res 2023; 28:217. [PMID: 37400851 PMCID: PMC10316617 DOI: 10.1186/s40001-023-01190-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/23/2023] [Indexed: 07/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a serious chronic lung disease. Schisandrin A (SchA) is one of the most important active ingredients in Schisandra chinensis and has been used to treat various lung diseases in several countries. Here, we studied the pharmacological effect of SchA on airway inflammation induced by cigarette smoke (CS) and explored the therapeutic mechanism of SchA in COPD model mice. Our results showed that SchA treatment significantly improved the lung function of CS-induced COPD model mice and reduced the recruitment of leukocytes and hypersecretion of interleukin-6 (IL-6), interleukin-1β (IL-1β) and tumor necrosis factor α (TNF-α) in bronchoalveolar lavage fluid (BALF). H&E staining showed that SchA treatment could effectively reduce emphysema, immune cell infiltration and airway wall destruction. In addition, we found that SchA treatment can stimulate the expression of heme oxygenase-1 (HO-1) through the nuclear factor-erythroid 2-related factor (Nrf2) pathway, significantly reduce oxidative stress, increase catalase (CAT) and superoxide dismutase (SOD) levels, and suppress the level of malondialdehyde (MDA) in COPD model mice. Moreover, SchA treatment suppressed the generation of the NLRP3/ASC/Caspase1 inflammasome complex to inhibit the inflammatory response caused by IL-1β and IL-18 and pyroptosis caused by GSDMD. In conclusion, our study shows that SchA treatment can inhibit the production of ROS and the activation of the NLRP3 inflammasome by upregulating Nrf-2, thereby producing anti-inflammatory effects and reducing lung injury in COPD model mice. More importantly, SchA exhibited similar anti-inflammatory effects to dexamethasone in COPD model mice, and we did not observe substantial side effects of SchA treatment. The high safety of SchA makes it a potential candidate drug for the treatment of COPD.
Collapse
Affiliation(s)
- Jiamin Zeng
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Sida Liao
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhu Liang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Caiping Li
- Guangzhou Medical University, Guangzhou, China
| | - Yuewen Luo
- Guangzhou Medical University, Guangzhou, China
| | - Kexin Wang
- Guangzhou Medical University, Guangzhou, China
| | - Dapeng Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lan Lan
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Suzhen Hu
- Huangdao District Chinese Medicine Hospital, Qingdao, China
| | - Wanyan Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ran Lin
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zichen Jie
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanlong Hu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Shiting Dai
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Zhimin Zhang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
33
|
Tanino R, Tsubata Y, Hotta T, Okimoto T, Amano Y, Takechi M, Tanaka T, Akita T, Nagase M, Yamashita C, Wada K, Isobe T. Characterization of a spontaneous mouse model of mild, accelerated aging via ECM degradation in emphysematous lungs. Sci Rep 2023; 13:10740. [PMID: 37400563 DOI: 10.1038/s41598-023-37638-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023] Open
Abstract
Emphysema limits airflow and causes irreversible progression of chronic obstructive pulmonary disease (COPD). Strain differences must be considered when selecting mouse models of COPD, owing to disease complexity. We previously reported that a novel C57BL/6JJcl substrain, the Mayumi-Emphysema (ME) mouse, exhibits spontaneous emphysema; however, the other characteristics remain unknown. We aimed to characterize the lungs of ME mice and determine their experimental availability as a model. ME mice had a lower body weight than the control C57BL/6JJcl mice, with a median survival time of ~80 weeks. ME mice developed diffused emphysema with respiratory dysfunction from 8 to 26 weeks of age, but did not develop bronchial wall thickening. Proteomic analyses revealed five extracellular matrix-related clusters in downregulated lung proteins in ME mice. Moreover, EFEMP2/fibulin-4, an essential extracellular matrix protein, was the most downregulated protein in the lungs of ME mice. Murine and human EFEMP2 were detected in the pulmonary artery. Furthermore, patients with mild COPD showed decreased EFEMP2 levels in the pulmonary artery when compared to those without COPD. The ME mouse is a model of mild, accelerated aging with low-inflammatory emphysema and respiratory dysfunction that progresses with age and pulmonary EFEMP2 decrease, similar to that observed in patients with mild COPD.
Collapse
Affiliation(s)
- Ryosuke Tanino
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Yukari Tsubata
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan.
| | - Takamasa Hotta
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Tamio Okimoto
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Yoshihiro Amano
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Mayumi Takechi
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| | - Tetsuya Tanaka
- Department of Human Nutrition, Faculty of Contemporary Life Science, Chugoku Gakuen University, Okayama, Japan
| | - Tomomi Akita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Mamiko Nagase
- Department of Organ Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Chikamasa Yamashita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Takeshi Isobe
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
34
|
Bao LK, Khoa ND, Chi LTK, Anh NT. Prevalence and Factors Affecting Appropriate Inhaler Use in Elderly Patients with Chronic Obstructive Pulmonary Disease: A Prospective Study. J Clin Med 2023; 12:4420. [PMID: 37445455 PMCID: PMC10342446 DOI: 10.3390/jcm12134420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) mainly affects individuals aged 60 and older. The proper use of inhalers is crucial for managing COPD. This study aimed to evaluate the prevalence and factors affecting the appropriate use of inhalers among elderly patients with COPD. METHODS We enrolled 91 elderly patients with COPD admitted to the Department of Respiratory, University Medical Center HCMC between October 2020 and May 2021. Patients who were capable of using the inhaler would have their inhaler usage recorded through video footage. Two respiratory experts carefully analyzed 133 video-recorded demonstrations for evaluation purposes. RESULTS 18.7% of the patients demonstrated the correct inhaler technique. Pressurized metered dose inhaler (pMDI) and Turbuhaler had the lowest documented correct usage rates (11.9% and 10.0%, respectively). Two critical steps, namely "holding breath for about five seconds or as long as comfortable" and "breathing out gently," were commonly performed incorrectly when using pMDI, Respimat, Breezhaler, or Turbuhaler. Multivariable logistic regression analysis showed that lower mMRC scores (AOR = 5.3, CI 1.1-25.5, p = 0.037) and receiving inhaler instruction within the past three months (AOR = 5.2, CI 1.3-20.1, p = 0.017) were associated with increased odds of using the inhaler correctly. CONCLUSIONS Our study found that less than 20% of elderly patients with COPD use inhalers correctly. Common errors include inadequate breath-holding and gentle exhalation. mMRC scores and recent inhaler instruction were predictors of proper use. These findings can aid clinicians in improving inhaler management for elderly patients with COPD.
Collapse
Affiliation(s)
- Le Khac Bao
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, 217 Hong Bang, Ward 11, District 5, Ho Chi Minh City 700000, Vietnam; (L.K.B.); (N.D.K.); (L.T.K.C.)
| | - Nguyen Dang Khoa
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, 217 Hong Bang, Ward 11, District 5, Ho Chi Minh City 700000, Vietnam; (L.K.B.); (N.D.K.); (L.T.K.C.)
| | - Le Thi Kim Chi
- Department of Internal Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, 217 Hong Bang, Ward 11, District 5, Ho Chi Minh City 700000, Vietnam; (L.K.B.); (N.D.K.); (L.T.K.C.)
| | - Nguyen Tuan Anh
- Department of Respiratory, University Medical Center Ho Chi Minh City, 215 Hong Bang, Ward 11, District 5, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
35
|
Liu X, Zhang X, Yao C, Liang J, Noble PW, Jiang D. A transcriptional cell atlas identifies the decline in the AT2 niche in aged human lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545378. [PMID: 37398304 PMCID: PMC10312782 DOI: 10.1101/2023.06.16.545378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Aging poses a global public health challenge, associated with molecular and physiological changes in the lungs. It increases susceptibility to acute and chronic lung diseases, yet the underlying molecular and cellular drivers in aged populations are not fully appreciated. To systematically profile the genetic changes associated with age, we present a single-cell transcriptional atlas comprising nearly half a million cells from the healthy lungs of human subjects spanning various ages, sexes, and smoking statuses. Most annotated cell lineages in aged lungs exhibit dysregulated genetic programs. Specifically, the aged alveolar epithelial cells, including both alveolar type II (AT2) and type I (AT1) cells, demonstrate loss of epithelial identities, heightened inflammaging characterized by increased expression of AP-1 transcription factor and chemokine genes, and significantly increased cellular senescence. Furthermore, the aged mesenchymal cells display a remarkable decrease in Collagen and Elastin transcription. The decline of the AT2 niche is further exacerbated by a weakened endothelial cell phenotype and a dysregulated genetic program in macrophages. These findings highlight the dysregulation observed in both AT2 stem cells and their supportive niche cells, potentially contributing to the increased susceptibility of aged populations to lung diseases.
Collapse
|
36
|
Laiman V, Hsiao TC, Fang YT, Chen YY, Lo YC, Lee KY, Chen TT, Chen KY, Ho SC, Wu SM, Chen JK, Heriyanto DS, Chung KF, Ho KF, Chuang KJ, Chang JH, Chuang HC. Hippo signaling pathway contributes to air pollution exposure-induced emphysema in ageing rats. JOURNAL OF HAZARDOUS MATERIALS 2023; 452:131188. [PMID: 36963197 DOI: 10.1016/j.jhazmat.2023.131188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/07/2023] [Accepted: 03/08/2023] [Indexed: 05/03/2023]
Affiliation(s)
- Vincent Laiman
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Ting Fang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - You-Yin Chen
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Industrial Ph.D. Program of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chun Lo
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; TMU Research Center of Thoracic Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Didik Setyo Heriyanto
- Department of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kin-Fai Ho
- School of Public Health and Primary Care, the Chinese University of Hong Kong, Hong Kong, China
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan; Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Departments of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; National Heart and Lung Institute, Imperial College London, London, UK; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
37
|
Pellegrino D, Casas-Recasens S, Faner R, Palange P, Agusti A. When GETomics meets aging and exercise in COPD. Respir Med 2023:107294. [PMID: 37295536 DOI: 10.1016/j.rmed.2023.107294] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
The term GETomics has been recently proposed to illustrate that human health and disease are actually the final outcome of many dynamic, interacting and cumulative gene (G) - environment (E) interactions that occur through the lifetime (T) of the individual. According to this new paradigm, the final outcome of any GxE interactions depends on both the age of the individual at which such GxE interaction occurs as well as on the previous, cumulative history of previous GxE interactions through the induction of epigenetic changes and immune memory (both lasting overtime). Following this conceptual approach, our understanding of the pathogenesis of chronic obstructive pulmonary disease (COPD) has changed dramatically. Traditionally believed to be a self-inflicted disease induced by tobacco smoking occurring in older men and characterized by an accelerated decline of lung function with age, now we understand that there are many other risk factors associated with COPD, that it occurs also in females and young individuals, that there are different lung function trajectories through life, and that COPD is not always characterized by accelerated lung function decline. In this paper we discuss how a GETomics approach to COPD may open new perspectives to better understand its relationship with exercise limitation and the ageing process.
Collapse
Affiliation(s)
- D Pellegrino
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy; Respiratory and Critical Care Unit, Policlinico Umberto I Hospital of Rome, Italy
| | - S Casas-Recasens
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain
| | - R Faner
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain; Cathedra Salut Respiratoria, University of Barcelona, Spain
| | - P Palange
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Italy; Respiratory and Critical Care Unit, Policlinico Umberto I Hospital of Rome, Italy
| | - A Agusti
- Institut d'investigacions biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Spain; Cathedra Salut Respiratoria, University of Barcelona, Spain; Respiratory Institute, Clinic Barcelona, Spain.
| |
Collapse
|
38
|
Shaikh SB, Goracci C, Tjitropranoto A, Rahman I. Impact of aging on immune function in the pathogenesis of pulmonary diseases: potential for therapeutic targets. Expert Rev Respir Med 2023; 17:351-364. [PMID: 37078192 PMCID: PMC10330361 DOI: 10.1080/17476348.2023.2205127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION Several immunological alterations that occur during pulmonary diseases often mimic alterations observed in the aged lung. From the molecular perspective, pulmonary diseases and aging partake in familiar mechanisms associated with significant dysregulation of the immune systems. Here, we summarized the findings of how aging alters immunity to respiratory conditions to identify age-impacted pathways and mechanisms that contribute to the development of pulmonary diseases. AREAS COVERED The current review examines the impact of age-related molecular alterations in the aged immune system during various lung diseases, such as COPD, IPF, Asthma, and alongside many others that could possibly improve on current therapeutic interventions. Moreover, our increased understanding of this phenomenon may play a primary role in shaping immunomodulatory strategies to boost outcomes in the elderly. Here, the authors present new insights into the context of lung-related diseases and describe the alterations in the functioning of immune cells during various pulmonary conditions altered with age. EXPERT OPINION The expert opinion provided the concepts on how aging alters immunity during pulmonary conditions, and suggests the associated mechanisms during the development of lung diseases. As a result, it becomes important to comprehend the complex mechanism of aging in the immune lung system.
Collapse
Affiliation(s)
- Sadiya Bi Shaikh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Chiara Goracci
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Ariel Tjitropranoto
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
39
|
Kheradmand F, Zhang Y, Corry DB. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol Rev 2023; 103:1059-1093. [PMID: 36201635 PMCID: PMC9886356 DOI: 10.1152/physrev.00036.2021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/01/2023] Open
Abstract
The pathophysiology of chronic obstructive pulmonary disease (COPD) and the undisputed role of innate immune cells in this condition have dominated the field in the basic research arena for many years. Recently, however, compelling data suggesting that adaptive immune cells may also contribute to the progressive nature of lung destruction associated with COPD in smokers have gained considerable attention. The histopathological changes in the lungs of smokers can be limited to the large or small airways, but alveolar loss leading to emphysema, which occurs in some individuals, remains its most significant and irreversible outcome. Critically, however, the question of why emphysema progresses in a subset of former smokers remained a mystery for many years. The recognition of activated and organized tertiary T- and B-lymphoid aggregates in emphysematous lungs provided the first clue that adaptive immune cells may play a crucial role in COPD pathophysiology. Based on these findings from human translational studies, experimental animal models of emphysema were used to determine the mechanisms through which smoke exposure initiates and orchestrates adaptive autoreactive inflammation in the lungs. These models have revealed that T helper (Th)1 and Th17 subsets promote a positive feedback loop that activates innate immune cells, confirming their role in emphysema pathogenesis. Results from genetic studies and immune-based discoveries have further provided strong evidence for autoimmunity induction in smokers with emphysema. These new findings offer a novel opportunity to explore the mechanisms underlying the inflammatory landscape in the COPD lung and offer insights for development of precision-based treatment to halt lung destruction.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| | - Yun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
40
|
Li S, Huang Q, He B. SIRT1 as a Potential Therapeutic Target for Chronic Obstructive Pulmonary Disease. Lung 2023; 201:201-215. [PMID: 36790647 DOI: 10.1007/s00408-023-00607-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/04/2023] [Indexed: 02/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common, preventable, and treatable disease characterized by irreversible airflow obstruction and lung function decline. It is well established that COPD represents a major cause of morbidity and mortality globally. Due to the substantial economic and social burdens associated with COPD, it is necessary to discover new targets and develop novel beneficial therapies. Although the pathogenesis of COPD is complex and remains to be robustly elucidated, numerous studies have shown that oxidative stress, inflammatory responses, cell apoptosis, autophagy, and aging are involved in the pathogenesis of COPD. Sirtuin 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase belonging to the silent information regulator 2 (Sir2) family. Multiple studies have indicated that SIRT1 plays an important role in oxidative stress, apoptosis, inflammation, autophagy, and cellular senescence, which contributes to the pathogenesis and development of COPD. This review aimed to discuss the functions and mechanisms of SIRT1 in the progression of COPD and concluded that SIRT1 activation might be a potential therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Siqi Li
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Baimei He
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China. .,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
41
|
Role of a small GTPase Cdc42 in aging and age-related diseases. Biogerontology 2023; 24:27-46. [PMID: 36598630 DOI: 10.1007/s10522-022-10008-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/13/2022] [Indexed: 01/05/2023]
Abstract
A small GTPase, Cdc42 is evolutionarily one of the most ancient members of the Rho family, which is ubiquitously expressed and involved in a wide range of fundamental cellular functions. The crucial role of Cdc42 includes regulation of the actin cytoskeleton, cell polarity, morphology and migration, endocytosis and exocytosis, cell cycle, and proliferation in many different cell types. Many studies have provided compelling yet contradicting evidence that Cdc42 dysregulation plays an important role in cellular and tissue aging. Furthermore, Cdc42 is a critical factor in the development and progression of aging-related pathologies, such as neurodegenerative and cardiovascular disorders, diabetes type 2, and aging-related disorders of the joints and bones, and the inhibition of the Cdc42 demonstrates potentially significant therapeutic and anti-aging effects in animal models of aging and disease. However, regulation of Cdc42 expression and activity is very complex and depends on many factors, such as the origin and complexity of the tissues, hormonal status, etc. Therefore, this review is focused on current advances in understanding the underlying cellular and molecular mechanisms associated with Cdc42 activity and regulation of senescence in different cell types since they may provide a foundation for novel therapeutic strategies and targeted drugs to reverse the aging process and treat aging-associated disorders.
Collapse
|
42
|
Adhikari TB, Paudel K, Paudel R, Bhusal S, Rijal A, Högman M, Neupane D, Sigsgaard T, Kallestrup P. Burden and risk factors of chronic respiratory diseases in Nepal, 1990-2019: An analysis of the global burden of diseases study. Health Sci Rep 2023; 6:e1091. [PMID: 36741854 PMCID: PMC9887632 DOI: 10.1002/hsr2.1091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
Background and Aims Chronic respiratory diseases (CRDs) substantially contribute to morbidity and mortality globally and in Nepal. However, there is a paucity of evidence on the trend and the burden of CRDs in Nepal. This study reports the trend of the burden and contribution of major risk factors to CRDs in Nepal from 1990 to 2019. Methods This study is an observational study using publicly available data from Global Burden of Disease 2019 estimations for Nepal. The age-standardized and age-specific prevalence, incidence, mortality, disability-adjusted life years (DALYs), and risk factors for CRDs in Nepal were extracted to measure the burden and its trend. The data are presented as percentages or as rates per 100,000 population. Results The age-standardized incidence rate of CRDs in Nepal in 2019 was 913.6 per 100,000 (95% uncertainty interval [UI]: 828.7-1000.1), which was an increase of 7.7% from 848.6 per 100,000 (95% UI: 780.2-918.2) in 1990. However, the age-standardized prevalence rate [4453/100,000 (4234.2-4671.8) in 1990; 4457.1/100,000 (4255.2-4666.8) in 2019] was almost stagnant. Most CRDs attributed to deaths and DALYs were due to chronic obstructive pulmonary disease. Conclusions Air pollution and smoking are the main risk factors for DALYs due to CRDs in Nepal. This surging burden of the incidence rate of CRDs in Nepal calls for more effective actions to curb the risk factors and diseases.
Collapse
Affiliation(s)
- Tara Ballav Adhikari
- Nepal Health FrontiersTokha‐5KathmanduNepal
- COBIN ProjectNepal Development SocietyChitwanNepal
- Department of Public Health, Section for Environment, Occupation & HealthAarhus UniversityAarhusDenmark
| | | | | | | | - Anupa Rijal
- Nepal Health FrontiersTokha‐5KathmanduNepal
- COBIN ProjectNepal Development SocietyChitwanNepal
- Department of Regional Health Research, The Faculty of Health SciencesUniversity of Southern DenmarkOdenseDenmark
| | - Marieann Högman
- Department of Medical Sciences, Respiratory, Allergy and Sleep ResearchUppsala UniversityUppsalaSweden
| | - Dinesh Neupane
- COBIN ProjectNepal Development SocietyChitwanNepal
- Department of International Health, Johns Hopkins Bloomberg School of Public HealthJohns Hopkins UniversityMDBaltimoreUSA
| | - Torben Sigsgaard
- Department of Public Health, Section for Environment, Occupation & HealthAarhus UniversityAarhusDenmark
| | - Per Kallestrup
- Department of Public Health, Section for Global HealthAarhus UniversityAarhusDenmark
| |
Collapse
|
43
|
Lee KY, Ho SC, Sun WL, Feng PH, Lin CW, Chen KY, Chuang HC, Tseng CH, Chen TT, Wu SM. Lnc-IL7R alleviates PM 2.5-mediated cellular senescence and apoptosis through EZH2 recruitment in chronic obstructive pulmonary disease. Cell Biol Toxicol 2022; 38:1097-1120. [PMID: 35303175 DOI: 10.1007/s10565-022-09709-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/11/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Long-term exposure to PM2.5 (particulate matter with an aerodynamic diameter of ≤ 2.5 μm) is associated with pulmonary injury and emphysema in patients with chronic obstructive pulmonary disease (COPD). We investigated mechanisms through which the long noncoding RNA lnc-IL7R contributes to cellular damage by inducing oxidative stress in COPD patients exposed to PM2.5. METHODS Associations of serum lnc-IL7R levels with lung function, emphysema, and previous PM2.5 exposure in COPD patients were analyzed. Reactive oxygen species and lnc-IL7R levels were measured in PM2.5-treated cells. The levels of lnc-IL7R and cellular senescence-associated genes, namely p16INK4a and p21CIP1/WAF1, were determined through lung tissue section staining. The effects of p16INK4a or p21CIP1/WAF1 regulation were examined by performing lnc-IL7R overexpression and knockdown assays. The functions of lnc-IL7R-mediated cell proliferation, cell cycle, senescence, colony formation, and apoptosis were examined in cells treated with PM2.5. Chromatin immunoprecipitation assays were conducted to investigate the epigenetic regulation of p21CIP1/WAF1. RESULTS Lnc-IL7R levels decreased in COPD patients and were negatively correlated with emphysema or PM2.5 exposure. Lnc-IL7R levels were upregulated in normal lung epithelial cells but not in COPD cells exposed to PM2.5. Lower lnc-IL7R expression in PM2.5-treated cells induced p16INK4a and p21CIP1/WAF1 expression by increasing oxidative stress. Higher lnc-IL7R expression protected against cellular senescence and apoptosis, whereas lower lnc-IL7R expression augmented injury in PM2.5-treated cells. Lnc-IL7R and the enhancer of zeste homolog 2 (EZH2) synergistically suppressed p21CIP1/WAF1 expression through epigenetic modulation. CONCLUSION Lnc-IL7R attenuates PM2.5-mediated p21CIP1/WAF1 expression through EZH2 recruitment, and its dysfunction may augment cellular injury in COPD.
Collapse
Affiliation(s)
- Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Chuan Ho
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Cheng-Wei Lin
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chien-Hua Tseng
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
44
|
Kaleem Ullah M, Parthasarathi A, Biligere Siddaiah J, Vishwanath P, Upadhyay S, Ganguly K, Anand Mahesh P. Impact of Acute Exacerbation and Its Phenotypes on the Clinical Outcomes of Chronic Obstructive Pulmonary Disease in Hospitalized Patients: A Cross-Sectional Study. TOXICS 2022; 10:toxics10110667. [PMID: 36355958 PMCID: PMC9695923 DOI: 10.3390/toxics10110667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 06/07/2023]
Abstract
Acute exacerbations of COPD (AECOPD) are clinically significant events having therapeutic and prognostic consequences. However, there is a lot of variation in its clinical manifestations described by phenotypes. The phenotypes of AECOPD were categorized in this study based on pathology and exposure. In our cross-sectional study, conducted between 1 January 2016 to 31 December 2020, the patients were categorized into six groups based on pathology: non-bacterial and non-eosinophilic; bacterial; eosinophilic; bacterial infection with eosinophilia; pneumonia; and bronchiectasis. Further, four groups were classified based on exposure to tobacco smoke (TS), biomass smoke (BMS), both, or no exposure. Cox proportional-hazards regression analyses were performed to assess hazard ratios, and Kaplan-Meier analysis was performed to assess survival, which was then compared using the log-rank test. The odds ratio (OR) and independent predictors of ward admission type and length of hospital stay were assessed using binomial logistic regression analyses. Of the 2236 subjects, 2194 were selected. The median age of the cohort was 67.0 (60.0 to 74.0) and 75.2% were males. Mortality rates were higher in females than in males (6.2% vs. 2.3%). AECOPD-B (bacterial infection) subjects [HR 95% CI 6.42 (3.06-13.46)], followed by AECOPD-P (pneumonia) subjects [HR (95% CI: 4.33 (2.01-9.30)], were at higher mortality risk and had a more extended hospital stay (6.0 (4.0 to 9.5) days; 6.0 (4.0 to 10.0). Subjects with TS and BMS-AECOPD [HR 95% CI 7.24 (1.53-34.29)], followed by BMS-AECOPD [HR 95% CI 5.28 (2.46-11.35)], had higher mortality risk. Different phenotypes have different impacts on AECOPD clinical outcomes. A better understanding of AECOPD phenotypes could contribute to developing an algorithm for the precise management of different phenotypes.
Collapse
Affiliation(s)
- Mohammed Kaleem Ullah
- Centre for Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSSAHER, Mysore 570015, Karnataka, India
- Global Infectious Diseases Fellow, Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Ashwaghosha Parthasarathi
- Allergy, Asthma, and Chest Centre, Krishnamurthypuram, Mysore 570004, Karnataka, India
- RUTGERS Centre for Pharmacoepidemiology and Treatment Science, New Brunswick, NJ 08901-1293, USA
| | | | - Prashant Vishwanath
- Centre for Excellence in Molecular Biology and Regenerative Medicine, Department of Biochemistry, JSS Medical College, JSSAHER, Mysore 570015, Karnataka, India
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine (IMM), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine (IMM), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Padukudru Anand Mahesh
- Department of Respiratory Medicine, JSS Medical College, JSSAHER, Mysore 570015, Karnataka, India
| |
Collapse
|
45
|
Chen S, Zhan Y, Chen J, Wu J, Gu Y, Huang Q, Deng Z, Wu X, Lv Y, Xie J. Identification and validation of genetic signature associated with aging in chronic obstructive pulmonary disease. Aging (Albany NY) 2022; 14:8568-8580. [DOI: 10.18632/aging.204358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/12/2022] [Indexed: 11/25/2022]
Affiliation(s)
- Shanshan Chen
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yuan Zhan
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jinkun Chen
- Department of Science, Western University, London, Ontario N6A 3K7, Canada
| | - Jixing Wu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Yiya Gu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Qian Huang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Zhesong Deng
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiaojie Wu
- Department of Respiratory and Critical Care Medicine, Wuhan No.1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan 430022, China
| | - Yongman Lv
- Health Management Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| |
Collapse
|
46
|
Ma JH, Zhang YT, Wang LP, Sun QY, Zhang H, Li JJ, Han NN, Zhu YY, Xie XY, Li X. K63 Ubiquitination of P21 Can Facilitate Pellino-1 in the Context of Chronic Obstructive Pulmonary Disease and Lung Cellular Senescence. Cells 2022; 11:cells11193115. [PMID: 36231077 PMCID: PMC9563803 DOI: 10.3390/cells11193115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic obstructive pulmonary diseases (COPD) is a kind of age-related, airflow-obstruction disease mostly caused by cigarette smoke. However, the relationship between COPD and lung cellular senescence is still not fully understood. Here, we found silencing Pellino-1 could inhibit the protein level of P21. Then, through constructing cell lines expressed ubiquitin-HA, we found that the E3 ubiquitin ligase Pellino-1 could bind to senescence marker p21 and modify p21 by K63-site ubiquitination by co-IP assays. Furthermore, we found that p21-mediated lung cellular senescence could be inhibited by silencing Pellino-1 in a D-galactose senescence mice model. Moreover, by constructing a COPD mouse model with shPellino-1 adenovirus, we found that silencing Pellino-1 could inhibit COPD and inflammation via reduction of SASPs regulated by p21. Taken together, our study findings elucidated that silencing E3 ligase Pellino-1 exhibits therapeutic potential for treatment to attenuate the progression of lung cellular senescence and COPD.
Collapse
Affiliation(s)
- Jia-Hui Ma
- Marine College, Shandong University, Weihai 264200, China
| | - Yi-Ting Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Lu-Ping Wang
- College of Biomedical Engineering and Instrumentation Science, Zhejiang University, Hangzhou 310000, China
| | - Qing-Yu Sun
- Marine College, Shandong University, Weihai 264200, China
| | - Hao Zhang
- Marine College, Shandong University, Weihai 264200, China
| | - Jian-Jiang Li
- Marine College, Shandong University, Weihai 264200, China
| | - Ning-Ning Han
- Marine College, Shandong University, Weihai 264200, China
| | - Yao-Yao Zhu
- Marine College, Shandong University, Weihai 264200, China
| | - Xiao-Yu Xie
- Marine College, Shandong University, Weihai 264200, China
| | - Xia Li
- Marine College, Shandong University, Weihai 264200, China
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
- Correspondence: ; Tel.: +86-531-88382612
| |
Collapse
|
47
|
Durso DF, Silveira-Nunes G, Coelho MM, Camatta GC, Ventura LH, Nascimento LS, Caixeta F, Cunha EHM, Castelo-Branco A, Fonseca DM, Maioli TU, Teixeira-Carvalho A, Sala C, Bacalini MJ, Garagnani P, Nardini C, Franceschi C, Faria AMC. Living in endemic area for infectious diseases accelerates epigenetic age. Mech Ageing Dev 2022; 207:111713. [PMID: 35931241 DOI: 10.1016/j.mad.2022.111713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022]
Abstract
Inflammaging is a low-grade inflammatory state generated by the aging process that can contribute to frailty and age-related diseases in the elderly. However, it can have distinct effects in the elderly living in endemic areas for infectious diseases. An increased inflammatory response may confer protection against infectious agents in these areas, although this advantage can cause accelerating epigenetic aging. In this study, we evaluated the inflammatory profile and the epigenetic age of infected and noninfected individuals from an endemic area in Brazil. The profile of cytokines, chemokines and growth factors analyzed in the sera of the two groups of individuals showed similarities, although infected individuals had a higher concentration of these mediators. A significant increase in IL-1ra, CXCL8, CCL2, CCL3 and CCL4 production was associated with leprosy infection. Notably, elderly individuals displayed distinct immune responses associated with their infection status when compared to adults suggesting an adaptive remodelling of their immune responses. Epigenetic analysis also showed that there was no difference in epigenetic age between the two groups of individuals. However, individuals from the endemic area had a significant accelerated aging when compared to individuals from São Paulo, a non-endemic area in Brazil. Moreover, the latter cohort was also epigenetically aged in relation to an Italian cohort. Our data shows that living in endemic areas for chronic infectious diseases results in remodelling of inflammaging and acceleration of epigenetic aging in individuals regardless of their infectious status. It also highlights that geographical, genetic and environmental factors influence aging and immunosenescence in their pace and profile.
Collapse
Affiliation(s)
- D F Durso
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - G Silveira-Nunes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Medicina, Universidade Federal de Juiz de Fora, Governador Valadares, Brazil
| | - M M Coelho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - G C Camatta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - L H Ventura
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - L S Nascimento
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - F Caixeta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - E H M Cunha
- Universidade Vale do Rio Doce, Governador Valadares, Brazil
| | - A Castelo-Branco
- Centro de Referência em Doenças Endêmicas e Programas Especiais, Governador Valadares, Brazil
| | - D M Fonseca
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - T U Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - A Teixeira-Carvalho
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - C Sala
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - M J Bacalini
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - P Garagnani
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - C Nardini
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - C Franceschi
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy; Research Laboratory of System Medicine for Healthy Ageing, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - A M C Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
48
|
Rivas M, Gupta G, Costanzo L, Ahmed H, Wyman AE, Geraghty P. Senescence: Pathogenic Driver in Chronic Obstructive Pulmonary Disease. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:817. [PMID: 35744080 PMCID: PMC9228143 DOI: 10.3390/medicina58060817] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 01/10/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is recognized as a disease of accelerated lung aging. Over the past two decades, mounting evidence suggests an accumulation of senescent cells within the lungs of patients with COPD that contributes to dysregulated tissue repair and the secretion of multiple inflammatory proteins, termed the senescence-associated secretory phenotype (SASP). Cellular senescence in COPD is linked to telomere dysfunction, DNA damage, and oxidative stress. This review gives an overview of the mechanistic contributions and pathologic consequences of cellular senescence in COPD and discusses potential therapeutic approaches targeting senescence-associated signaling in COPD.
Collapse
Affiliation(s)
- Melissa Rivas
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Gayatri Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Louis Costanzo
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Huma Ahmed
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Anne E. Wyman
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY 11203, USA; (M.R.); (L.C.); (H.A.); (A.E.W.)
| |
Collapse
|
49
|
Kaminsky DA, Irvin CG. The Physiology of Asthma-Chronic Obstructive Pulmonary Disease Overlap. Immunol Allergy Clin North Am 2022; 42:575-589. [DOI: 10.1016/j.iac.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
50
|
Giri A, Wang Q, Rahman I, Sundar IK. Circadian molecular clock disruption in chronic pulmonary diseases. Trends Mol Med 2022; 28:513-527. [DOI: 10.1016/j.molmed.2022.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/31/2022]
|