1
|
Pollack M, Rapsomaniki E, Anzueto A, Rhodes K, Hawkins NM, Vogelmeier CF, Marshall J, Müllerová H. Effectiveness of Single Versus Multiple Inhaler Triple Therapy on Mortality and Cardiopulmonary Risk Reduction in COPD: The SKOPOS-MAZI Study. Am J Med 2025; 138:650-659.e10. [PMID: 39566703 DOI: 10.1016/j.amjmed.2024.11.007] [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: 06/20/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Patients with chronic obstructive pulmonary disease (COPD) have elevated cardiopulmonary and mortality risk, particularly following exacerbations. While single inhaler triple therapies (SITTs), such as budesonide/glycopyrrolate/formoterol fumarate (BGF), reduce cardiopulmonary risk versus dual bronchodilator therapy, there is limited evidence comparing outcomes with SITTs versus multiple inhaler triple therapies (MITTs). METHODS SKOPOS-MAZI was a retrospective comparative effectiveness study in patients with COPD aged ≥40 years using US administrative claims data from Optum's deidentified Clinformatics® Data Mart Database. The primary and secondary endpoints were time to all-cause mortality and time to first severe cardiopulmonary event following initiation of BGF or MITT (identification period: October 1, 2020-June 30, 2023; index date: first prescription fill). Relative hazards of outcomes were assessed until a censoring event using Cox proportional hazards models, with inverse propensity treatment weighting accounting for between-group imbalances (standardized mean difference >0.1) in baseline characteristics. RESULTS In the primary cohort, risk (hazard ratio [95% confidence intervals]) of all-cause mortality and a first severe cardiopulmonary event were 18% (0.82 [0.75, 0.91]) and 12% (0.88 [0.83, 0.93]) lower in patients initiating BGF versus MITT; results were consistent across censoring definitions, landmark periods, and sensitivity cohorts. CONCLUSION In this real-world comparative effectiveness study of patients with COPD initiating BGF or MITT, BGF was associated with lower all-cause mortality and severe cardiopulmonary event risk versus MITT after accounting for between-group differences in baseline sociodemographic and clinical characteristics. This study supports the benefits of BGF over MITT and the need to consider proactive use of SITTs in COPD management.
Collapse
Affiliation(s)
| | | | - Antonio Anzueto
- University of Texas Health, South Texas Veterans Health Care System, San Antonio, Tex
| | - Kirsty Rhodes
- BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| | | | - Claus F Vogelmeier
- University of Marburg, German Center for Lung Research (DZL), Marburg, Hessen, Germany
| | - Jonathan Marshall
- BioPharmaceuticals Respiratory and Immunology Medical, AstraZeneca, Cambridge, UK
| | - Hana Müllerová
- Respiratory Evidence Strategy, BioPharmaceuticals Medical, AstraZeneca, Cambridge, UK
| |
Collapse
|
2
|
Mismetti V, Bertoletti L. Acute exacerbation of chronic obstructive pulmonary disease: An underestimated cardiovascular risk factor. Eur J Intern Med 2025:S0953-6205(25)00093-7. [PMID: 40074637 DOI: 10.1016/j.ejim.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Affiliation(s)
- Valentine Mismetti
- Department of Pulmonology and Thoracic Oncology, University Hospital of Saint-Etienne, 42270, Saint-Etienne, France; INSERM, U1059, SAINBIOSE, Jean Monnet University, 42023, Saint-Etienne, France.
| | - Laurent Bertoletti
- INSERM, U1059, SAINBIOSE, Jean Monnet University, 42023, Saint-Etienne, France; Therapeutic and Vascular Medicine Department, University Hospital of Saint-Etienne, 42270, Saint-Etienne, France; INSERM, CIC 1408, 42023, Saint-Etienne, France
| |
Collapse
|
3
|
Cano A, Esteban C, Larrea N, Iriberri M, Sánchez R, Jiménez-Puente A, de-Miguel-Díez J, Orive M, Quintana JM. Factors associated with short- and medium-term mortality after hospitalization for COPD exacerbation. Respir Med 2025; 240:108010. [PMID: 39988227 DOI: 10.1016/j.rmed.2025.108010] [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] [Received: 11/18/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVE Hospitalization due to exacerbation is a critical event for patients with chronic obstructive pulmonary disease (COPD). This study aimed to identify predictive factors for mortality in patients post-hospitalization for COPD exacerbation and to determine differences in these predictors in the short and medium term. METHODS A prospective observational study involving 1635 patients hospitalized for COPD exacerbation, followed for one year. Sociodemographic and clinical data, comorbidities, treatments, and quality of life questionnaires were assessed. Cox regression analysis identified mortality predictors at 2 months and >2-12 months post-hospitalization. RESULTS Mean age was 72.4 years with 76.6 % males. Overall, one-year mortality was 14.1 % (30.3 % of the deaths occurred within 2 months of discharge and 69.7 % occurred >2-12 months post-discharge). Short-term mortality predictors included: age (HR [95 % CI] = 2.483 [1.501-4.107]), lower Barthel index (HR [95 % CI] = 1.274 [1.063-1.526]), pulmonary function (FEV1 < 30 %: HR [95 % CI] = 5.153 [1.511-17.577]), and length of stay ≥8 days (HR [95 % CI] = 6.974 [2.504-19.419]). Medium-term predictors included: heart failure (HR [95 % CI] = 2.493 [1.318-4.717]), age (HR [95 % CI] = 1.690 [1.224-2.334]), lower Barthel index (HR [95 % CI] = 1.300 [1.149-1.472]), and pulmonary function (FEV1 < 30 %: HR [95 % CI] = 3.000 [1.351-6.658] and FEV1 30-50 %: HR [95 % CI] = 2.010 [1.046-3.862]). CONCLUSIONS Mortality risk factors with exacerbated COPD after hospitalization vary over time. In the short term, length of hospital stay is prominent, while heart failure is more significant in the medium term. Age, dependency and pulmonary function were common predictors in both periods. Tailoring clinical interventions over time may improve health outcomes in this population.
Collapse
Affiliation(s)
- Arantza Cano
- Osakidetza Basque Health Service, Hospital Santa Marina, Respiratory Department, Bilbao, Bizkaia, Spain.
| | - Cristóbal Esteban
- Osakidetza Basque Health Service, Hospital Universitario de Galdakao, Respiratory Department, Galdakao, Bizkaia, Spain; BioBizkaia Health Research Institute, Barakaldo, Spain; Health Services Research on Chronic Patients Network (REDISSEC), Bizkaia, Spain
| | - Nere Larrea
- Research Unit, Hospital Galdakao, Galdakao, Bizkaia, Spain; Research Network on Chronicity, Primary Care, and Prevention and Health Promotion- RICAPPS, Galdakao 48960, Bizkaia, Spain; Biosistemak Institute for Health Systems Research, Baracaldo, Spain
| | - Milagros Iriberri
- BioBizkaia Health Research Institute, Barakaldo, Spain; Osakidetza Basque Health Service, Hospital Universitario de Cruces, Respiratory Department, Barakaldo, Bizkaia, Spain
| | - Raquel Sánchez
- Osakidetza Basque Health Service, Hospital Universitario de Basurto, Respiratory Department, Bilbao, Bizkaia, Spain
| | - Alberto Jiménez-Puente
- Health Services Research on Chronic Patients Network (REDISSEC), Bizkaia, Spain; Research Network on Chronicity, Primary Care, and Prevention and Health Promotion- RICAPPS, Galdakao 48960, Bizkaia, Spain; Evaluation Unit, Hospital Costa del Sol, Marbella, Málaga, Spain
| | - Javier de-Miguel-Díez
- Health Services Research on Chronic Patients Network (REDISSEC), Bizkaia, Spain; Research Network on Chronicity, Primary Care, and Prevention and Health Promotion- RICAPPS, Galdakao 48960, Bizkaia, Spain; Respiratory Department, Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Universidad Complutense de Madrid, Madrid, Spain
| | - Miren Orive
- Health Services Research on Chronic Patients Network (REDISSEC), Bizkaia, Spain; Research Network on Chronicity, Primary Care, and Prevention and Health Promotion- RICAPPS, Galdakao 48960, Bizkaia, Spain; Department of Social Psychology, Faculty of Pharmacy, UPV/EHU, Vitoria-Gasteiz, Spain
| | - Jose María Quintana
- Research Unit, Hospital Galdakao, Galdakao, Bizkaia, Spain; Research Network on Chronicity, Primary Care, and Prevention and Health Promotion- RICAPPS, Galdakao 48960, Bizkaia, Spain; Biosistemak Institute for Health Systems Research, Baracaldo, Spain
| |
Collapse
|
4
|
Pirera E, Di Raimondo D, D'Anna L, Tuttolomondo A. Risk trajectory of cardiovascular events after an exacerbation of chronic obstructive pulmonary disease: A systematic review and meta-analysis. Eur J Intern Med 2025:S0953-6205(25)00026-3. [PMID: 39884921 DOI: 10.1016/j.ejim.2025.01.016] [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] [Received: 10/24/2024] [Revised: 01/05/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Exacerbations of chronic obstructive pulmonary disease (COPD) are known to increase the risk of cardiovascular (CV) events and mortality. However, the temporal trend of this risk has not fully elucidated. This systematic review and meta-analysis aims to quantify the risk of CV events after COPD exacerbations over different time periods. OBJECTIVES To assess the temporal association between CV events, including acute coronary syndrome (ACS), heart failure (HF), acute cereberovascular events, arrhythmia and all-cause mortality after the onset of COPD exacerbations in the following timepoints: 1-30 and 31-180 days; 1-7, 8-14, 15-30, 31-180, 181-365 and >365 days. METHODS A comprehensive literature search was conducted in PubMed, Embase, Web of Science and Cochrane databases, identifying observational studies that reported CV outcomes following COPD exacerbations. Studies were included if they enrolled adults diagnosed with COPD and compared CV event rates during exacerbation and non-exacerbation periods (PROSPERO, CRD42024561490). RESULTS Sixteen studies with over 1.8 million participants were included. Our meta-analysis demonstrated a significantly increased risk of ACS, HF, cerebrovascular events and arrhythmia, with the highest magnitude of risk observed in the period 1-30 days following an exacerbation. This increased risk showed a decline in time points 31-180, 181-365 days and remained persistently higher for ACS even one year after an acute exacerbation. Notably, the risk of HF was found to be greater compared to the other CV outcomes. CONCLUSION COPD exacerbations significantly increase the risk of acute CV events, particularly within the first 30 days. Optimal strategies to reduce cardiopulmonary risk are needed.
Collapse
Affiliation(s)
- Edoardo Pirera
- Internal Medicine and Stroke Care ward, Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (Promise) G. D'Alessandro, University of Palermo, Palermo Italy.
| | - Domenico Di Raimondo
- Internal Medicine and Stroke Care ward, Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (Promise) G. D'Alessandro, University of Palermo, Palermo Italy
| | - Lucio D'Anna
- Department of Stroke and Neuroscience, Charing Cross Hospital, Imperial College London NHS Healthcare Trust, London, United Kingdom
| | - Antonino Tuttolomondo
- Internal Medicine and Stroke Care ward, Department of Promoting Health, Maternal-Infant. Excellence and Internal and Specialized Medicine (Promise) G. D'Alessandro, University of Palermo, Palermo Italy
| |
Collapse
|
5
|
Gillan JL, Jaeschke L, Kuebler WM, Grune J. Immune mediators in heart-lung communication. Pflugers Arch 2025; 477:17-30. [PMID: 39256247 PMCID: PMC11711577 DOI: 10.1007/s00424-024-03013-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
It is often the case that serious, end-stage manifestations of disease result from secondary complications in organs distinct from the initial site of injury or infection. This is particularly true of diseases of the heart-lung axis, given the tight anatomical connections of the two organs within a common cavity in which they collectively orchestrate the two major, intertwined circulatory pathways. Immune cells and the soluble mediators they secrete serve as effective, and targetable, messengers of signals between different regions of the body but can also contribute to the spread of pathology. In this review, we discuss the immunological basis of interorgan communication between the heart and lung in various common diseases, and in the context of organ crosstalk more generally. Gaining a greater understanding of how the heart and lung communicate in health and disease, and viewing disease progression generally from a more holistic, whole-body viewpoint have the potential to inform new diagnostic approaches and strategies for better prevention and treatment of comorbidities.
Collapse
Affiliation(s)
- Jonathan L Gillan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Lara Jaeschke
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany.
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
6
|
Santos S, Manito N, Sánchez-Covisa J, Hernández I, Corregidor C, Escudero L, Rhodes K, Nordon C. Risk of severe cardiovascular events following COPD exacerbations: results from the EXACOS-CV study in Spain. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2025; 78:138-150. [PMID: 38936468 DOI: 10.1016/j.rec.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION AND OBJECTIVES This real-world study-the first of its kind in a Spanish population-aimed to explore severe risk for cardiovascular events and all-cause death following exacerbations in a large cohort of patients with chronic obstructive pulmonary disease (COPD). METHODS We included individuals with a COPD diagnosis code between 2014 and 2018 from the BIG-PAC health care claims database. The primary outcome was a composite of a first severe cardiovascular event (acute coronary syndrome, heart failure decompensation, cerebral ischemia, arrhythmia) or all-cause death following inclusion in the cohort. Time-dependent Cox proportional hazards models estimated HRs for associations between exposed time periods (1-7, 8-14, 15-30, 31-180, 181-365, and >365 days) following an exacerbation of any severity, and following moderate or severe exacerbations separately (vs unexposed time before a first exacerbation following cohort inclusion). RESULTS During a median follow-up of 3.03 years, 18 901 of 24 393 patients (77.5%) experienced ≥ 1 moderate/severe exacerbation, and 8741 (35.8%) experienced the primary outcome. The risk of a severe cardiovascular event increased following moderate/severe COPD exacerbation onset vs the unexposed period, with rates being most increased during the first 1 to 7 days following exacerbation onset (HR, 10.10; 95%CI, 9.29-10.97) and remaining increased >365 days after exacerbation onset (HR, 1.65; 95%CI, 1.49-1.82). CONCLUSIONS The risk of severe cardiovascular events or death increased following moderate/severe exacerbation onset, illustrating the need for proactive multidisciplinary care of patients with COPD to prevent exacerbations and address other cardiovascular risk factors.
Collapse
Affiliation(s)
- Salud Santos
- Servicio de Neumología, Hospital Universitario de Bellvitge, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Nicolás Manito
- Servicio de Cardiología, Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | - Kirsty Rhodes
- Real World Science & Analytics, BioPharmaceuticals Medical, AstraZeneca, Cambridge, United Kingdom
| | - Clementine Nordon
- Epidemiology Medical Evidence Strategy, BioPharmaceuticals Medical, AstraZeneca, Cambridge, United Kingdom
| |
Collapse
|
7
|
Zo S, Lee H, Jeong CY, Kim BG, Chung JE, Kim Y, Moon JY, Lim YH, Min KH, Yoo KH, Yoon HJ, Kim JS, Kim SH. COVID-19 and Long-term Risk of Ischemic Heart Disease in Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2025; 17:135-146. [PMID: 39895608 PMCID: PMC11791370 DOI: 10.4168/aair.2025.17.1.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/29/2024] [Accepted: 10/15/2024] [Indexed: 02/04/2025]
Abstract
Despite the importance of comorbid ischemic heart disease (IHD) in the prognosis of asthma, the long-term impact of coronavirus disease 2019 (COVID-19) on IHD in adults with asthma remains unclear. This study investigated the long-term effects of COVID-19 on the risk of IHD in individuals with asthma, particularly regarding COVID-19 severity. Using the Korean National Health Insurance Service claims database, we identified individuals with asthma who had recovered from COVID-19 between October 8, 2020, and December 31, 2021 (n = 8,011) and 1:1 propensity score-matched controls (n = 8,011). The incidence and risk of IHD were compared between the two groups. Overall, during a median follow-up of 95 days (interquartile range, 34-213 days; range, 1-448 days), which includes a median of 14 lag days, the COVID-19 cohort did not show a higher risk of IHD (hazard ratio [HR], 2.11; 95% confidence interval [CI], 0.99-4.48) compared to matched controls. However, when the severity of COVID-19 was considered, the severe COVID-19 cohort exhibited a higher risk of IHD (HR, 4.89; 95% CI, 1.86-12.84) than matched controls; in contrast, the non-severe COVID-19 cohort showed no significantly increased risk of IHD (HR, 1.64; 95% CI, 0.73-3.70). Severe COVID-19 is associated with an increased long-term risk of IHD in adults with asthma, emphasizing the importance of cardiovascular events monitoring to improve asthma treatment outcomes in the era of COVID-19.
Collapse
Affiliation(s)
- Sungmin Zo
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Hyun Lee
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Cho Yun Jeong
- Department of Medical Informatics, Jeonbuk National University Medical School, Jeonju, Korea
| | - Bo-Guen Kim
- Division of Pulmonary Medicine, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jee Eun Chung
- College of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Korea
| | - Youlim Kim
- Division of Pulmonary and Allergy, Department of Internal Medicine, Konkuk University Hospital, School of Medicine, Konkuk University, Seoul, Korea
| | - Ji-Yong Moon
- Division of Pulmonary and Allergy, Department of Internal Medicine, Konkuk University Hospital, School of Medicine, Konkuk University, Seoul, Korea
| | - Young-Hyo Lim
- Division of Cardiology, Department of Internal Medicine, Hanyang Medical Center, Hanyang University College of Medicine, Seoul, Korea
| | - Kyung Hoon Min
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Kwang-Ha Yoo
- Division of Pulmonary and Allergy, Department of Internal Medicine, Konkuk University Hospital, School of Medicine, Konkuk University, Seoul, Korea
| | - Ho Joo Yoon
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Jong Seung Kim
- Department of Medical Informatics, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Jeonbuk National University Medical School, Jeonju, Korea.
| | - Sang-Heon Kim
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea.
| |
Collapse
|
8
|
Hawkins NM, Kaplan A, Ko DT, Penz E, Bhutani M. Is 'Cardiopulmonary' the New 'Cardiometabolic'? Making a Case for Systems Change in COPD. Pulm Ther 2024; 10:363-376. [PMID: 39249675 PMCID: PMC11573969 DOI: 10.1007/s41030-024-00270-2] [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: 07/04/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and cardiovascular disease (CVD) have a syndemic relationship with shared risk factors and complex interplay between genetic, environmental, socioeconomic, and pathophysiological mechanisms. CVD is among the most common comorbidities in patients with COPD and vice versa. Patients with COPD, irrespective of their disease severity, are at increased risk of CVD morbidity and mortality, driven in part by COPD exacerbations. Despite these known interrelationships, CVD is underestimated and undertreated in patients with COPD. Similarly, COPD is an independent risk-enhancing factor for adverse cardiovascular (CV) events, yet it is not incorporated into current CV risk assessment tools, leading to under-recognition and undertreatment. There is a pressing need for systems change in COPD management to move beyond symptom control towards a comprehensive cardiopulmonary disease paradigm with proactive prevention of exacerbations and adverse cardiopulmonary outcomes and mortality. However, there is a dearth of evidence defining optimal cardiopulmonary care pathways. Fortunately, there is a precedent to support systems-level change in the field of diabetes, which evolved from glycemic control to comprehensive multi-organ risk assessment and management. Key elements included integrated multidisciplinary care, intensive risk factor management, coordination between primary and specialist care, care pathways and protocols, education and self management, and disease-modifying therapies. This commentary article draws parallels between the cardiometabolic and cardiopulmonary paradigms and makes a case for systems change towards multidisciplinary, integrated cardiopulmonary care, using the evolution in diabetes care as a potential framework.
Collapse
Affiliation(s)
- Nathaniel M Hawkins
- Centre for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, 2775 Laurel Street, 9th Floor Room 9123, Vancouver, BC, V5Z 1M9, Canada.
| | - Alan Kaplan
- Family Physician Airways Group of Canada, University of Toronto, Toronto, ON, Canada
| | - Dennis T Ko
- Division of Cardiology, Schulich Heart Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Erika Penz
- Division of Respirology, Critical Care and Sleep Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mohit Bhutani
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
9
|
Simons SO, Heptinstall AB, Marjenberg Z, Marshall J, Mullerova H, Rogliani P, Nordon C, Hawkins NM. Temporal Dynamics of Cardiovascular Risk in Patients with Chronic Obstructive Pulmonary Disease During Stable Disease and Exacerbations: Review of the Mechanisms and Implications. Int J Chron Obstruct Pulmon Dis 2024; 19:2259-2271. [PMID: 39411574 PMCID: PMC11474009 DOI: 10.2147/copd.s466280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Exacerbations of chronic obstructive pulmonary disease (COPD) are risk factors for severe cardiovascular (CV) events, with the risk remaining significantly elevated long after the symptomatic phase of the exacerbation. The pathophysiology underpinning the relationship between acute events of both COPD and CV diseases has been understudied. Our objectives were to review the mechanisms by which COPD exacerbations increase the risk of CV events and understand the temporality of this risk. Methods A pragmatic and targeted literature review was conducted with a focus on identifying recent, high-impact papers up to June 2023, guided by insights from subject matter experts including pulmonologists and cardiologists. Results A substantial number of inter-related mechanisms underpin the spiral of anatomical and functional deterioration of lung and heart affecting COPD patients during stable state. In turn, an exacerbation of COPD may trigger a CV event, during and beyond the symptomatic phase, due to ventilation/perfusion mismatch, oxygen supply-demand imbalance, oxidative stress, systemic inflammation, hypercoagulable state, dynamic hyperinflation, pulmonary hypertension, and sympathetic activation. However, no study was identified that explored the mechanisms by which an exacerbation confers a sustained risk of CV event. Conclusion While our review identified multiple dynamic and interacting pathophysiological mechanisms during and after an exacerbation of COPD that contribute to increasing the risk of a wide range of cardiac events, little is known regarding the precise long-term mechanisms after acute exacerbation to explain the persistent increased CV event risk beyond the symptomatic phase. The temporal changes in static and dynamic substrates need further characterization to better understand the different risk factors and risk periods for a CV event following the onset of an exacerbation. Moreover, guideline-directed cardiopulmonary therapies should be implemented at every opportunity; preventing exacerbations and intensively treating traditional CV risk factors should be a focus in COPD management.
Collapse
Affiliation(s)
- Sami O Simons
- Department of Respiratory Medicine, NUTRIM Institute for Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | | | - Jonathan Marshall
- BioPharmaceuticals Medical, Respiratory and Immunology, AstraZeneca, Cambridge, UK
| | - Hana Mullerova
- BioPharmaceuticals Medical, Respiratory and Immunology, AstraZeneca, Cambridge, UK
| | - Paola Rogliani
- Department of Experimental Medicine, Unit of Respiratory Medicine, University of Rome ‘Tor Vergata’, Rome, Italy
| | - Clementine Nordon
- BioPharmaceuticals Medical, Respiratory and Immunology, AstraZeneca, Cambridge, UK
| | | |
Collapse
|
10
|
Alexandrova Y, Yero A, Olivenstein R, Orlova M, Schurr E, Estaquier J, Costiniuk CT, Jenabian MA. Dynamics of pulmonary mucosal cytotoxic CD8 T-cells in people living with HIV under suppressive antiretroviral therapy. Respir Res 2024; 25:240. [PMID: 38867225 PMCID: PMC11170847 DOI: 10.1186/s12931-024-02859-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Despite the success of antiretroviral therapy (ART), people living with HIV (PLWH) suffer from a high burden of pulmonary diseases, even after accounting for their smoking status. Cytotoxic CD8 T-cells are likely implicated in this phenomenon and may act as a double-edged sword. While being essential in viral infection control, their hyperactivation can also contribute to lung mucosal tissue damage. The effects of HIV and smoking on pulmonary mucosal CD8 T-cell dynamics has been a neglected area of research, which we address herein. METHODS Bronchoalveolar lavage (BAL) fluid were obtained from ART-treated PLWH (median duration of supressed viral load: 9 years; smokers: n = 14; non-smokers: n = 21) and HIV-uninfected controls (smokers: n = 11; non-smokers: n = 20) without any respiratory symptoms or active infection. Lymphocytes were isolated and CD8 T-cell subsets and homing markers were characterized by multiparametric flow cytometry. RESULTS Both smoking and HIV infection were independently associated with a significant increase in frequencies of total pulmonary mucosal CD8 T-cell. BAL CD8 T-cells were primarily CD69 + expressing CD103 and/or CD49a, at least one of the two granzymes (GzmA/GzmB), and little Perforin. Higher expression levels of CD103, CD69, and GzmB were observed in smokers versus non-smokers. The ex vivo phenotype of GzmA + and GzmB + cells revealed increased expression of CD103 and CXCR6 in smokers, while PLWH displayed elevated levels of CX3CR1 compared to controls. CONCLUSION Smoking and HIV could promote cytotoxic CD8 T-cell retention in small airways through different mechanisms. Smoking likely increases recruitment and retention of GzmB + CD8 Trm via CXCR6 and CD103. Heightened CX3CR1 expression could be associated with CD8 non-Trm recruitment from the periphery in PLWH.
Collapse
Affiliation(s)
- Yulia Alexandrova
- Department of Biological Sciences, Université du Québec à Montréal (UQAM), 141, Avenue President Kennedy, Montreal, QC, H2X 1Y4, Canada
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Alexis Yero
- Department of Biological Sciences, Université du Québec à Montréal (UQAM), 141, Avenue President Kennedy, Montreal, QC, H2X 1Y4, Canada
| | - Ronald Olivenstein
- Division of Respirology, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Marianna Orlova
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada
| | - Erwin Schurr
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Departments of Human Genetics and Medicine, McGill University, Montreal, QC, Canada
| | - Jerome Estaquier
- Centre de recherche de CHU de Québec - Université Laval Research Center, Québec City, Québec, Canada
| | - Cecilia T Costiniuk
- Infectious Diseases and Immunity in Global Health Program, Research Institute of McGill University Health Centre, Montreal, QC, Canada
- Division of Infectious Diseases and Chronic Viral Illness Service, McGill University Health Centre, Montreal, QC, Canada
| | - Mohammad-Ali Jenabian
- Department of Biological Sciences, Université du Québec à Montréal (UQAM), 141, Avenue President Kennedy, Montreal, QC, H2X 1Y4, Canada.
| |
Collapse
|
11
|
Singh D, Han MK, Hawkins NM, Hurst JR, Kocks JWH, Skolnik N, Stolz D, El Khoury J, Gale CP. Implications of Cardiopulmonary Risk for the Management of COPD: A Narrative Review. Adv Ther 2024; 41:2151-2167. [PMID: 38664329 PMCID: PMC11133105 DOI: 10.1007/s12325-024-02855-4] [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: 02/23/2024] [Accepted: 03/22/2024] [Indexed: 05/29/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) constitutes a major global health burden and is the third leading cause of death worldwide. A high proportion of patients with COPD have cardiovascular disease, but there is also evidence that COPD is a risk factor for adverse outcomes in cardiovascular disease. Patients with COPD frequently die of respiratory and cardiovascular causes, yet the identification and management of cardiopulmonary risk remain suboptimal owing to limited awareness and clinical intervention. Acute exacerbations punctuate the progression of COPD in many patients, reducing lung function and increasing the risk of subsequent exacerbations and cardiovascular events that may lead to early death. This narrative review defines and summarises the principles of COPD-associated cardiopulmonary risk, and examines respiratory interventions currently available to modify this risk, as well as providing expert opinion on future approaches to addressing cardiopulmonary risk.
Collapse
Affiliation(s)
- Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Trust, University of Manchester, Manchester, M23 9QZ, UK.
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - John R Hurst
- UCL Respiratory, University College London, London, UK
| | - Janwillem W H Kocks
- General Practitioners Research Institute, Groningen, The Netherlands
- Observational and Pragmatic Research Institute, Singapore, Singapore
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Daiana Stolz
- Clinic of Respiratory Medicine, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | | | - Chris P Gale
- Department of Cardiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| |
Collapse
|
12
|
Hawkins NM, Nordon C, Rhodes K, Talukdar M, McMullen S, Ekwaru P, Pham T, Randhawa AK, Sin DD. Heightened long-term cardiovascular risks after exacerbation of chronic obstructive pulmonary disease. Heart 2024; 110:702-709. [PMID: 38182279 PMCID: PMC11103306 DOI: 10.1136/heartjnl-2023-323487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024] Open
Abstract
OBJECTIVE To examine the risk of adverse cardiovascular (CV) events following an exacerbation of chronic obstructive pulmonary disease (COPD). METHODS This retrospective cohort study identified patients with COPD using administrative data from Alberta, Canada from 2014 to 2019. Exposure periods were 12 months following moderate or severe exacerbations; the reference period was time preceding a first exacerbation. The primary outcome was the composite of all-cause death or a first hospitalisation for acute coronary syndrome, heart failure (HF), arrhythmia or cerebral ischaemia. Time-dependent Cox regression models estimated covariate-adjusted risks associated with six exposure subperiods following exacerbation. RESULTS Among 1 42 787 patients (mean age 68.1 years and 51.7% men) 61 981 (43.4%) experienced at least one exacerbation and 34 068 (23.9%) died during median follow-up of 64 months. The primary outcome occurred in 43 564 (30.5%) patients with an incidence rate prior to exacerbation of 5.43 (95% CI 5.36 to 5.50) per 100 person-years. This increased to 95.61 per 100 person-years in the 1-7 days postexacerbation (adjusted HR 15.86, 95% CI 15.17 to 16.58) and remained increased for up to 1 year. The risk of both the composite and individual CV events was increased following either a moderate or a severe exacerbation, though greater and more prolonged following severe exacerbation. The highest magnitude of increased risk was observed for HF decompensation (1-7 days, HR 72.34, 95% CI 64.43 to 81.22). CONCLUSION Moderate and severe COPD exacerbations are independent risk factors for adverse CV events, especially HF decompensation. The impact of optimising COPD management on CV outcomes should be evaluated.
Collapse
Affiliation(s)
- Nathaniel M Hawkins
- Centre for Cardiovascular Innovation, Division of Cardiology, The University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | - Paul Ekwaru
- Medlior Health Outcomes Research Ltd, Calgary, Alberta, Canada
| | - Tram Pham
- Medlior Health Outcomes Research Ltd, Calgary, Alberta, Canada
| | | | - Don D Sin
- UBC Centre for Heart Lung Innovation and Department of Medicine (Respirology), The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
13
|
Perdijk O, Azzoni R, Marsland BJ. The microbiome: an integral player in immune homeostasis and inflammation in the respiratory tract. Physiol Rev 2024; 104:835-879. [PMID: 38059886 DOI: 10.1152/physrev.00020.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/07/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
The last decade of microbiome research has highlighted its fundamental role in systemic immune and metabolic homeostasis. The microbiome plays a prominent role during gestation and into early life, when maternal lifestyle factors shape immune development of the newborn. Breast milk further shapes gut colonization, supporting the development of tolerance to commensal bacteria and harmless antigens while preventing outgrowth of pathogens. Environmental microbial and lifestyle factors that disrupt this process can dysregulate immune homeostasis, predisposing infants to atopic disease and childhood asthma. In health, the low-biomass lung microbiome, together with inhaled environmental microbial constituents, establishes the immunological set point that is necessary to maintain pulmonary immune defense. However, in disease perturbations to immunological and physiological processes allow the upper respiratory tract to act as a reservoir of pathogenic bacteria, which can colonize the diseased lung and cause severe inflammation. Studying these host-microbe interactions in respiratory diseases holds great promise to stratify patients for suitable treatment regimens and biomarker discovery to predict disease progression. Preclinical studies show that commensal gut microbes are in a constant flux of cell division and death, releasing microbial constituents, metabolic by-products, and vesicles that shape the immune system and can protect against respiratory diseases. The next major advances may come from testing and utilizing these microbial factors for clinical benefit and exploiting the predictive power of the microbiome by employing multiomics analysis approaches.
Collapse
Affiliation(s)
- Olaf Perdijk
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Rossana Azzoni
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Murillo Moreno MA, López Gutiérrez LV, Vinck EE, Roncancio Villamil G, Gallego Muñoz C, Saldarriaga Giraldo CI. Coronary heart disease and tuberculosis: an unnoticed syndemia. Review of literature and management proposal. ARCHIVOS PERUANOS DE CARDIOLOGIA Y CIRUGIA CARDIOVASCULAR 2024; 5:e375. [PMID: 39015190 PMCID: PMC11247974 DOI: 10.47487/apcyccv.v5i2.375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/03/2024] [Indexed: 07/18/2024]
Abstract
Tuberculosis is an increasing disease that affects about one-third of the global population. In line with the rise of tuberculosis, cardiovascular disease has shown a similar trend, with ischemic coronary heart disease becoming the leading cause of death worldwide. Based on the literature, a relationship can be drawn between tuberculosis and ischemic coronary heart disease through their shared multiple risk factors and a possible pathophysiological substrate linking them. The presentation of these two conditions reported so far is varied: it has been found as the onset of acute coronary syndrome in patients with active tuberculosis, the progressive development of coronary atherosclerosis in patients with latent tuberculosis, among others. Given this possible link and the progressive increase in their incidence rates, we can assert that we are facing an unnoticed syndemic, with their concurrent management posing a challenge due to significant pharmacological interactions. The purpose of this review is to clarify this possible link, propose an approach for diagnosis, and provide a treatment algorithm for the entire spectrum of coronary disease coexisting with tuberculosis according to the current available literature.
Collapse
Affiliation(s)
- Mauricio Andrés Murillo Moreno
- Departamento de Medicina Interna, Universidad CES, Medellín, Colombia.Universidad CESDepartamento de Medicina InternaUniversidad CESMedellínColombia
| | - Laura Valentina López Gutiérrez
- Departamento de Cardiología, Clínica Cardio VID, Medellín, Colombia.Departamento de CardiologíaClínica Cardio VIDMedellínColombia
| | - Eric Edward Vinck
- Departamento de Cirugía Cardiovascular, Clínica Cardio VID, Medellín, Colombia.Departamento de Cirugía CardiovascularClínica Cardio VIDMedellínColombia
| | - Gustavo Roncancio Villamil
- Departamento de Enfermedades Infecciosas, Clínica Cardio VID, Medellín, Colombia.Departamento de Enfermedades InfecciosasClínica Cardio VIDMedellínColombia
| | - Catalina Gallego Muñoz
- Departamento de Cardiología, Clínica Cardio VID, Medellín, Colombia.Departamento de CardiologíaClínica Cardio VIDMedellínColombia
| | - Clara Inés Saldarriaga Giraldo
- Departamento de Cardiología, Clínica Cardio VID, Medellín, Colombia.Departamento de CardiologíaClínica Cardio VIDMedellínColombia
| |
Collapse
|
15
|
Berber NK, Atlı S, Geçkil AA, Erdem M, Kıran TR, Otlu Ö, İn E. Diagnostic Value of Galectin-3 in Exacerbations of Chronic Obstructive Pulmonary Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:529. [PMID: 38674175 PMCID: PMC11052179 DOI: 10.3390/medicina60040529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
Background and Objectives: Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease characterized by acute exacerbations. Systemic inflammation and oxidative stress play an important role in the pathogenesis of COPD. Exacerbations in COPD reduce the quality of life and are associated with rapid disease progression. Galectin-3 is a beta-galactoside-binding lectin of approximately 30 kDa with pro-inflammatory and pro-fibrotic properties. This study aims to analyze the efficacy of serum galectin-3 in predicting exacerbations in COPD patients. Materials and Methods: Baseline demographic and clinical characteristics of all patients were recorded and blood samples were collected. A total of 58 consecutive COPD patients, including 28 patients (19 male and 9 female) with stable COPD and 30 patients (23 male and 7 female) with acute exacerbation of COPD (AECOPD), were included in the study. Results: Serum galectin-3 levels were significantly higher in the AECOPD group compared to the stable COPD group. A logistic regression analysis revealed that increased galectin-3 levels and disease duration were independent predictors of COPD exacerbation (OR = 5.322, 95% CI: 1.178-24.052, p = 0.03; and OR = 1.297, 95% CI: 1.028-1.635, p = 0.028; respectively). Conclusions: The results of our study demonstrated that Galectin-3 was a strong and independent predictor of exacerbations in COPD patients.
Collapse
Affiliation(s)
- Nurcan Kırıcı Berber
- Department of Chest Diseases, Malatya Turgut Özal University, Malatya 44210, Turkey;
| | - Siahmet Atlı
- Department of Chest Diseases, Van Training and Research Hospital, Van 65100, Turkey;
| | | | - Mehmet Erdem
- Department of Medical Biochemistry, Malatya Turgut Özal University, Malatya 44210, Turkey; (M.E.); (T.R.K.); (Ö.O.)
| | - Tuğba Raika Kıran
- Department of Medical Biochemistry, Malatya Turgut Özal University, Malatya 44210, Turkey; (M.E.); (T.R.K.); (Ö.O.)
| | - Önder Otlu
- Department of Medical Biochemistry, Malatya Turgut Özal University, Malatya 44210, Turkey; (M.E.); (T.R.K.); (Ö.O.)
| | - Erdal İn
- Department of Pulmonary Diseases, Faculty of Medicine, İzmir University of Economics, İzmir 35330, Turkey;
| |
Collapse
|
16
|
Vogelmeier CF, Friedrich FW, Timpel P, Kossack N, Diesing J, Pignot M, Abram M, Halbach M. Impact of COPD on mortality: An 8-year observational retrospective healthcare claims database cohort study. Respir Med 2024; 222:107506. [PMID: 38151176 DOI: 10.1016/j.rmed.2023.107506] [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: 10/13/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is one of the leading causes of morbidity and mortality. Here we present a large observational study on the association of COPD and exacerbations with mortality (AvoidEx Mortality). METHODS A real-world, observational cohort study with longitudinal analyses of German healthcare claims data in patients ≥40 years of age with a COPD diagnosis from 2011 to 2018 (n = 250,723) was conducted. Patients entered the cohort (index date) upon the first COPD diagnosis. To assess the impact of COPD on all-cause death, a propensity score-matched control group of non-COPD patients was constructed. The number and severity of exacerbations during a 12-month pre-index period were used to form subgroups. For each exacerbation subgroup the exacerbations during 12 months prior to death were analysed. RESULTS COPD increases the all-cause mortality risk by almost 60% (HR 1.57 (95% CI 1.55-1.59)) in comparison to matched non-COPD controls, when controlling for other baseline covariates. The cumulative risk of death after 8 years was highest in patients with a history of more than one moderate or severe exacerbation. Among all deceased COPD patients, 17.2% had experienced a severe, and 34.8% a moderate exacerbation, within 3 months preceding death. Despite increasing exacerbation rates towards death, more than the half of patients were not receiving any recommended pharmacological COPD therapy in the year before death. CONCLUSION Our study illustrates the impact of COPD on mortality risk and highlights the need for consequent COPD management comprising exacerbation assessment and treatment.
Collapse
Affiliation(s)
- Claus F Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Philipps-Universität Marburg, German Center for Lung Research (DZL), Baldingerstraße, 35033, Marburg, Hessen, Germany
| | | | - Patrick Timpel
- WIG2 GmbH Scientific Institute for Health Economics and Health System Research, Markt 8, 04109, Leipzig, Sachsen, Germany
| | - Nils Kossack
- WIG2 GmbH Scientific Institute for Health Economics and Health System Research, Markt 8, 04109, Leipzig, Sachsen, Germany
| | - Joanna Diesing
- WIG2 GmbH Scientific Institute for Health Economics and Health System Research, Markt 8, 04109, Leipzig, Sachsen, Germany
| | - Marc Pignot
- ZEG - Center for Epidemiology and Health Research Berlin GmbH, Invalidenstraße 115, 10115, Berlin, Germany
| | - Melanie Abram
- AstraZeneca GmbH, Friesenweg 26, 22763, Hamburg, Germany
| | - Marija Halbach
- AstraZeneca GmbH, Friesenweg 26, 22763, Hamburg, Germany.
| |
Collapse
|
17
|
Abdul-Rahman T, Roy P, Bliss ZSB, Mohammad A, Corriero AC, Patel NT, Wireko AA, Shaikh R, Faith OE, Arevalo-Rios ECE, Dupuis L, Ulusan S, Erbay MI, Cedeño MV, Sood A, Gupta R. The impact of air quality on cardiovascular health: A state of the art review. Curr Probl Cardiol 2024; 49:102174. [PMID: 37913932 DOI: 10.1016/j.cpcardiol.2023.102174] [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: 10/28/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023]
Abstract
Air pollution is a global health challenge, increasing the risk of cardiovascular diseases such as heart disease, stroke, and arrhythmias. Particulate matter (PM), particularly PM2.5 and ultrafine particles (UFP), is a key contributor to the adverse effects of air pollution on cardiovascular health. PM exposure can lead to oxidative stress, inflammation, atherosclerosis, vascular dysfunction, cardiac arrhythmias, and myocardial injury. Reactive oxygen species (ROS) play a key role in mediating these effects. PM exposure can also lead to hypertension, a significant risk factor for cardiovascular disease. The COVID-19 pandemic resulted in a significant reduction of air pollutants, leading to a decline in the incidence of heart attacks and premature deaths caused by cardiovascular diseases. This review highlights the relationship between environmental air quality and cardiovascular health, elucidating the pathways through which air pollutants affect the cardiovascular system. It also emphasizes the need for increased awareness, collective efforts to mitigate the adverse effects of air pollution, and strategic policies for long-term air quality improvement to prevent the devastating effects of air pollution on global cardiovascular health.
Collapse
Affiliation(s)
- Toufik Abdul-Rahman
- Medical Institute, Sumy State University, Sumy, Ukraine; Department of Research, Toufik's World Medical Association, Sumy, Ukraine
| | - Poulami Roy
- Department of Research, Toufik's World Medical Association, Sumy, Ukraine; Department of Medicine, North Bengal Medical College and Hospital, Siliguri, India
| | | | | | | | - Neal T Patel
- Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, FL, USA
| | - Andrew Awuah Wireko
- Medical Institute, Sumy State University, Sumy, Ukraine; Department of Research, Toufik's World Medical Association, Sumy, Ukraine
| | - Raheel Shaikh
- Broward Health Medical Center, Fort Lauderdale, FL, USA
| | | | | | - Léonie Dupuis
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sebahat Ulusan
- Medical School, Suleyman Demirel University, Isparta, Turkey
| | | | | | - Aayushi Sood
- Department of Medicine, The Wright Center for Graduate Medical Education, Scranton, PA, USA
| | - Rahul Gupta
- Department of Cardiology, Lehigh Valley Health Network, Allentown, PA, USA.
| |
Collapse
|
18
|
Ellis P, Bailey E, Choate R, Holm KE, Sandhaus RA, Turner AM, Newnham M. Prevalence of Cardiovascular Disease and Rate of Major Adverse Cardiovascular Events in Severe Alpha-1 Antitrypsin Deficiency COPD. Int J Chron Obstruct Pulmon Dis 2024; 19:149-159. [PMID: 38249829 PMCID: PMC10800105 DOI: 10.2147/copd.s419846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/05/2023] [Indexed: 01/23/2024] Open
Abstract
Aim Alpha-1 antitrypsin deficiency is an autosomal co-dominant condition that predisposes individuals to early-onset emphysema. As with COPD, AATD-COPD is associated with pulmonary exacerbations, which impacts on overall mortality and quality of life. Though there is evidence that COPD is associated with a higher prevalence of cardiovascular disease and major adverse cardiovascular events (MACE), it is unclear if this is true for patients with AATD-COPD. Methods Prevalence of cardiovascular disease was determined in two separate severe AATD cohorts: AlphaNet, USA and the Birmingham AATD registry, UK. All patients had preexisting lung disease. Cardiovascular disease was defined as presence of any of the following: heart failure, ischaemic heart disease, atrial fibrillation, stroke, and myocardial infarction. A Cox proportional hazards model was used to assess the impact of prior cardiovascular disease and frequent exacerbator phenotype on risk of future MACE. Results Out of 3493 patients with severe AATD, 14.7% had prior cardiovascular disease, including stroke (2.3%), myocardial infarction (2.2%), and heart failure (2.5%). Frequent exacerbators were more likely to have preexisting cardiovascular disease compared with those with one or no exacerbations in the preceding year (63% vs 44.8%, p = 0.001). There was increased risk of future MACE in frequent exacerbators (HR 1.85, 95% CI 1.24 to 2.75), former and current smokers (HR 1.80, 95% CI 1.07 to 3.02, p = 0.026, and HR 4.04, 95% CI 1.44 to 11.32, p = 0.008, respectively), and those with prior cardiovascular disease (HR 3.81, 95% CI 2.60 to 5.58, p < 0.001). Conclusion In severe AATD-COPD, MACE are associated with an increased exacerbation frequency, previous cardiovascular disease, and a history of smoking.
Collapse
Affiliation(s)
- Paul Ellis
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Emily Bailey
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Radmila Choate
- University of Kentucky College of Public Health, Lexington, KY, USA
| | - Kristen E Holm
- Division of Neurology and Behavioural Health, National Jewish Health, Denver, CO, USA
- AlphaNet, Kissimmee, FL, USA
| | - Robert A Sandhaus
- AlphaNet, Kissimmee, FL, USA
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, USA
| | - Alice M Turner
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Michael Newnham
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
19
|
Yang Y, Zoulikha M, Xiao Q, Huang F, Jiang Q, Li X, Wu Z, He W. Pulmonary endothelium-targeted nanoassembly of indomethacin and superoxide dismutase relieves lung inflammation. Acta Pharm Sin B 2023; 13:4607-4620. [PMID: 37969734 PMCID: PMC10638505 DOI: 10.1016/j.apsb.2023.05.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 11/17/2023] Open
Abstract
Lung inflammation is an essential inducer of various diseases and is closely related to pulmonary-endothelium dysfunction. Herein, we propose a pulmonary endothelium-targeted codelivery system of anti-inflammatory indomethacin (IND) and antioxidant superoxide dismutase (SOD) by assembling the biopharmaceutical SOD onto the "vector" of rod-like pure IND crystals, followed by coating with anti-ICAM-1 antibody (Ab) for targeting endothelial cells. The codelivery system has a 237 nm diameter in length and extremely high drug loading of 39% IND and 2.3% SOD. Pharmacokinetics and biodistribution studies demonstrate the extended blood circulation and the strong pulmonary accumulation of the system after intravenous injection in the lipopolysaccharide (LPS)-induced inflammatory murine model. Particularly, the system allows a robust capacity to target pulmonary endothelium mostly due to the rod-shape and Ab coating effect. In vitro, the preparation shows the synergistic anti-inflammatory and antioxidant effects in LPS-activated endothelial cells. In vivo, the preparation exhibits superior pharmacodynamic efficacy revealed by significantly downregulating the inflammatory/oxidative stress markers, such as TNF-α, IL-6, COX-2, and reactive oxygen species (ROS), in the lungs. In conclusion, the codelivery system based on rod-like pure crystals could well target the pulmonary endothelium and effectively alleviate lung inflammation. The study offers a promising approach to combat pulmonary endothelium-associated diseases.
Collapse
Affiliation(s)
- Yi Yang
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Makhloufi Zoulikha
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
- Department of Traditional Chinese Medicine Processing and Preparation, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feifei Huang
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Qi Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| |
Collapse
|
20
|
Romankiewicz L, Schaare HL, Nestler S, Villringer A, Blöchl M. Mediation of the Association Between Vascular Risk Factors and Depressive Symptoms by C-Reactive Protein. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:642-650. [PMID: 37881535 PMCID: PMC10593949 DOI: 10.1016/j.bpsgos.2023.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 10/27/2023] Open
Abstract
Background This study examined whether C-reactive protein (CRP), a marker of low-grade systemic inflammation, mediates the association between vascular risk factor (VRF) burden and depressive symptoms. Methods We drew on the prospective design of the UK Biobank to include participants with longitudinal data on VRF burden, CRP, and depressive symptoms. Total, direct, and indirect effects were estimated using regression-based mediation models while controlling for confounding by sociodemographic factors, baseline CRP, and baseline depression. Sensitivity analyses probed the robustness of results to unmeasured confounding. Results We analyzed data from 10,470 participants from the UK Biobank (mean age = 56.75 years at baseline). Net of covariates, VRFs at baseline were associated with higher depressive symptoms at follow-up (total effect = 0.099; 95% CI, 0.002-0.163). CRP mediated this association (indirect effect = 0.010; 95% CI, 0.004-0.017), accounting for 10.0% (95% CI, 0.3%-30.0%) of the total effect of VRF burden on depressive symptoms. Exploratory analyses suggested that the total and indirect effects pertained to somatic depressive symptoms (tiredness and appetite). Conclusions These results suggest that inflammation-promoting effects of VRFs may contribute to depressive symptoms in mid- and later life. However, the mediating pathway via CRP explains only a small part of the association between VRFs and depression after accounting for important covariates and may pertain to specific depressive symptoms. Future studies leveraging similar longitudinal designs are needed to further disentangle the time-varying effects between VRFs, inflammation, and certain depressive symptoms while addressing important confounders.
Collapse
Affiliation(s)
| | - H. Lina Schaare
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour) Research Centre, Jülich, Germany
| | - Steffen Nestler
- Department of Psychology, University of Münster, Münster, Germany
| | - Arno Villringer
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Clinic for Cognitive Neurology, University Clinic Leipzig, Leipzig, Germany
- Center for Stroke Research Berlin, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Blöchl
- Department of Psychology, University of Münster, Münster, Germany
- International Max Planck Research School: Neuroscience of Communication: Structure, Function, and Plasticity, Leipzig, Germany
| |
Collapse
|
21
|
Jacobson PK, Lind L, Persson HL. Applying the Rome Proposal on Exacerbations of Chronic Obstructive Pulmonary Disease: Does Comorbid Chronic Heart Failure Matter? Int J Chron Obstruct Pulmon Dis 2023; 18:2055-2064. [PMID: 37744733 PMCID: PMC10517701 DOI: 10.2147/copd.s425592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Background Chronic heart failure (CHF) is a common comorbidity among patients with chronic obstructive pulmonary disease (COPD). Both exacerbations of COPD (ECOPDs) and exacerbations of CHF (ECHFs) display worsening of breathlessness at rest (BaR) and breathlessness at physical activity (BaPA). Comorbid CHF may have an impact on the vital signs assessed, when the Rome proposal (adopted by GOLD 2023) is applied on ECOPDs. Thus, the aim of the present study was to investigate the impact of comorbid CHF on ECOPDs severity, particularly focusing on the influence of comorbid CHF on BaR and BaPA. Methods We analysed data on COPD symptoms collected from the telehealth study The eHealth Diary. Patients with COPD (n = 43) and patients with CHF (n = 41) were asked to daily monitor BaR and BaPA, employing a digital pen and scales for BaR and BaPA (from 0 to 10). Twenty-eight patients of the COPD patients presented with comorbid CHF. Totally, 125 exacerbations were analysed. Results Exacerbations in the group with COPD patients and comorbid CHF were compared to the group with COPD patients without comorbid CHF and the group with CHF patients. Compared with GOLD 2022, the GOLD 2023 (the Rome proposal) significantly downgraded the ECOPD severity. Comorbid CHF did not interfere significantly on the observed difference. Comorbid CHF did not worsen BaR scores, assessed at inclusion and at the symptom peak of the exacerbations. Conclusion In the present study, we find no evidence that comorbid CHF would interfere significantly with the parameters included in the Rome proposal (GOLD 2023). We conclude that the Rome proposal can be safely applied even on COPD patients with very advanced comorbid CHF.
Collapse
Affiliation(s)
- Petra Kristina Jacobson
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Respiratory Medicine in Linköping, Linköping University, Linköping, Sweden
| | - Leili Lind
- Department of Biomedical Engineering/Health Informatics, Linköping University, Linköping, Sweden
- Digital Systems Division, Unit Digital Health, RISE Research Institutes of Sweden, Linköping, Sweden
| | - Hans L Persson
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Respiratory Medicine in Linköping, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
Bedford R, Smith G, Rothwell E, Martin S, Medhane R, Casentieri D, Daunt A, Freiberg G, Hollings M. A multi-organ, lung-derived inflammatory response following in vitro airway exposure to cigarette smoke and next-generation nicotine delivery products. Toxicol Lett 2023; 387:35-49. [PMID: 37774809 DOI: 10.1016/j.toxlet.2023.09.010] [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: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Despite increasing use of in vitro models that closely resemble in vivo human biology, their application in understanding downstream effects of airway toxicity, such as inflammation, are at an early stage. In this study, we used various assays to examine the inflammatory response induced in MucilAir™ tissues and A549 cells exposed to three products known to induce toxicity. Reduced barrier integrity was observed in tissues following exposure to each product, with reduced viability and increased cytotoxicity also shown. Similar changes in viability were also observed in A549 cells. Furthermore, whole cigarette smoke (CS) induced downstream phenotypic THP-1 changes and endothelial cell adhesion, an early marker of atherosclerosis. In contrast, exposure to next-generation delivery product (NGP) aerosol did not induce this response. Cytokine, histological and RNA analysis highlighted increased biomarkers linked to inflammatory pathways and immune cell differentiation following exposure to whole cigarette smoke, including GM-CSF, IL-1β, cleaved caspase-3 and cytochrome P450 enzymes. As a result of similar observations in human airway inflammation, we propose that our exposure platform could act as a representative model for studying such events in vitro. Furthermore, this model could be used to test the inflammatory or anti-inflammatory impact posed by inhaled compounds delivered to the lung.
Collapse
Affiliation(s)
- R Bedford
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| | - G Smith
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - E Rothwell
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - S Martin
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - R Medhane
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - D Casentieri
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - A Daunt
- Labcorp Early Development Laboratories Limited, Harrogate, UK
| | - G Freiberg
- Labcorp Early Development Laboratories Limited, Eye, UK
| | - M Hollings
- Labcorp Early Development Laboratories Limited, Harrogate, UK.
| |
Collapse
|
23
|
Templeton S, McVeigh CM, Nguyen C, Hunter R, Scieszka D, Herbert GW, Barr EB, Liu R, Gu H, Bleske BE, Campen MJ, Bolt AM. Acute inhalation of tungsten particles results in early signs of cardiac injury. Toxicol Lett 2023; 384:52-62. [PMID: 37442282 PMCID: PMC10528412 DOI: 10.1016/j.toxlet.2023.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023]
Abstract
Epidemiological studies have established that exposure to tungsten increases the risk of developing cardiovascular diseases. However, no studies have investigated how tungsten affects cardiac function or the development of cardiovascular disease. Inhalation of tungsten particulates is relevant in occupational settings, and inhalation of particulate matter has a known causative role in driving cardiovascular disease. This study examined if acute inhalation to tungsten particulates affects cardiac function and leads to heart tissue alterations. Female BALB/c mice were exposed to Filtered Air or 1.5 ± 0.23 mg/m3 tungsten particles, using a whole-body inhalation chamber, 4 times over the course of two weeks. Inhalation exposure resulted in mild pulmonary inflammation characterized by an increased percentage and number of macrophages and metabolomic changes in the lungs. Cardiac output was significantly decreased in the tungsten-exposed group. Additionally, A', an indicator of the amount of work required by the atria to fill the heart was elevated. Cardiac gene expression analysis revealed, tungsten exposure increased expression of pro-inflammatory cytokines, markers of remodeling and fibrosis, and oxidative stress genes. These data strongly suggest exposure to tungsten results in cardiac injury characterized by early signs of diastolic dysfunction. Functional findings are in parallel, demonstrating cardiac oxidative stress, inflammation, and early fibrotic changes. Tungsten accumulation data would suggest these cardiac changes are driven by systemic consequences of pulmonary damage.
Collapse
Affiliation(s)
- Sage Templeton
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Charlotte M McVeigh
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Colin Nguyen
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Russell Hunter
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - David Scieszka
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Guy W Herbert
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Edward B Barr
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Rui Liu
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Barry E Bleske
- The University of New Mexico College of Pharmacy, Department of Pharmacy Practice and Administrative Sciences, Albuquerque, NM 87131, USA
| | - Matthew J Campen
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Alicia M Bolt
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, USA.
| |
Collapse
|
24
|
Vlachopoulos CV, Solomou EG, Terentes Printzios DG, Pouli AG, Sioni A, Giannouli SE, Angelopoulou MK, Kafouris P, Metaxas MG, Chondropoulos SD, Stergiou IE, Marinakis TP, Koutagiar I, Miliou AA, Ioakeimidis N, Tsalamandris ST, Katsi V, Aggeli CI, Voulgarelis M, Tousoulis DM, Tsioufis C, Anagnostopoulos CD. Effects of Chemotherapy on Aortic 18-Fluorodeoxyglucose Uptake in Patients With Hodgkin and Non-Hodgkin Lymphoma. JACC. ADVANCES 2023; 2:100277. [PMID: 38938303 PMCID: PMC11198565 DOI: 10.1016/j.jacadv.2023.100277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 06/29/2024]
Abstract
Background Despite advances in the treatment of oncology patients, therapy-related side effects may lead to premature morbidity. Inflammatory activation that has been linked to cardiovascular disease is crucial for the pathogenesis of both Hodgkin (HL) and non-Hodgkin lymphoma (NHL). Objectives The purpose of this study was to assess the vascular effects of chemotherapy in patients with HL and NHL by positron emission tomography/computed tomography with 18-fluorodeoxyglucose (18-FDG PET/CT) and to investigate interactions with systemic inflammation as assessed by circulating inflammatory markers. Methods Between July 2015 and July 2019, 65 consecutive patients (mean age 56 ± 17.78 years) with confirmed diagnosis of either HL (n = 33) or NHL (n = 32) were prospectively studied. PET/CT imaging was performed at baseline, at an interim phase, and after first-line treatment. Aortic FDG uptake was assessed by measuring global aortic target-to-background ratio (GLA-TBR). Serum biomarkers interleukin (IL)-6 and IL-1b were measured at each phase. Results Patients with HL demonstrated significant reduction in aortic TBR after first-line treatment (median GLA-TBR baseline: 1.98, median GLA-TBR third scan: 1.75, median difference = -0.20, 95% CI: -0.07 to -0.33, P = 0.006), which remained significant after adjustment for confounders (adj. R2 of model = 0.53). In contrast, patients with NHL did not demonstrate a significant aortic inflammation response (P = 0.306). Furthermore, patients with HL demonstrated a significant reduction in IL-6 (P = 0.048) and IL-1b (P = 0.045), whereas patients with NHL did not demonstrate significant reduction in IL-6 (P = 0.085) and IL-1b levels (P = 0.476). Conclusions Aortic inflammation, as assessed by 18-FDG PET/CT, is reduced in HL patients after first-line treatment but not in NHL patients. These findings imply that different pathophysiological pathways and different therapies might affect the arterial bed in different ways for patients with lymphoma.
Collapse
Affiliation(s)
- Charalambos V. Vlachopoulos
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Eirini G. Solomou
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Dimitrios G. Terentes Printzios
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | | | - Anastasia Sioni
- Department of Hematology, “Aghios Savvas” Hospital, Athens, Greece
| | - Stavroula E. Giannouli
- 2nd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Pavlos Kafouris
- Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, Athens, Greece
| | - Marinos G. Metaxas
- Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Pathophysiology, School of Medicine, University of Athens, Athens, Greece
| | | | | | | | - Iosif Koutagiar
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Antigoni A. Miliou
- Biochemistry, Immunology and Molecular Biology Department, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Nikolaos Ioakeimidis
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Sotirios T. Tsalamandris
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Vasiliki Katsi
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Constantina I. Aggeli
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | | | - Dimitrios M. Tousoulis
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Constantinos Tsioufis
- 1st Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Constantinos D. Anagnostopoulos
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
25
|
Yu S, Lee SC, Hong JH, Han CH, Park SC, Jung JY. Cardiovascular and cerebrovascular mortality in patients with preceding asthma exacerbation. Respirology 2023; 28:281-286. [PMID: 36596607 DOI: 10.1111/resp.14444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/11/2022] [Indexed: 01/05/2023]
Affiliation(s)
- Soojoung Yu
- International Healthcare Center, Soonchunhyang University Seoul Hospital, Yongsan-gu, South Korea
| | - Sang Chul Lee
- Division of Pulmonology, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | - Jung Hwa Hong
- Department of Research and Analysis, National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | - Chang Hoon Han
- Division of Pulmonology, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | - Seon Cheol Park
- Division of Pulmonology, Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, South Korea
| | - Ji Ye Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seodaemun-gu, South Korea
| |
Collapse
|
26
|
Dodd KE, Blackley DJ, Mazurek JM. Cardiovascular Disease Among Adults With Work-Related Asthma, 2012-2017. Am J Prev Med 2023; 64:194-203. [PMID: 36371324 DOI: 10.1016/j.amepre.2022.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/23/2022] [Accepted: 09/12/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION Asthma is associated with an increased risk for cardiovascular disease, and adults with persistent, severe asthma have a significantly higher risk of cardiovascular disease than adults with intermittent or no asthma. METHODS The objective of this cross-sectional study was to assess the association between work-related asthma status and cardiovascular disease among ever-employed adults (aged 18-64 years) with current asthma using data from the 2012-2017 Behavioral Risk Factor Surveillance System Asthma Call-Back Survey from 37 states and the District of Columbia. Weighted prevalence ratios and 95% CIs, adjusted for age, sex, race/ethnicity, education, household income, smoking status, chronic obstructive pulmonary disease, diabetes, and BMI, were calculated. In addition, the associations of cardiovascular disease with adverse asthma outcomes and asthma control among adults with work-related asthma were examined. Analyses were conducted in 2021. RESULTS Among an estimated annualized 14.8 million ever-employed adults aged 18-64 years with current asthma, adults with work-related asthma (prevalence ratio=1.5; 95% CI=1.2, 1.8) and possible work-related asthma (prevalence ratio=1.2; 95% CI=1.0, 1.5) were significantly more likely to have cardiovascular disease than adults with non-work-related asthma. Among adults with work-related asthma, those with very poorly controlled asthma (prevalence ratio=1.8; 95% CI=1.3, 2.5) and an asthma-related emergency room visit (prevalence ratio=1.5; 95% CI=1.1, 2.0) were significantly more likely to have cardiovascular disease. CONCLUSIONS Adults with work-related asthma were more likely to have cardiovascular disease than those with non-work-related asthma. Primary prevention, early diagnosis, and implementation of optimal work-related asthma management are essential for workers' health. Cardiovascular disease should be considered where appropriate when diagnosing and recommending treatment and interventions for adults with work-related asthma.
Collapse
Affiliation(s)
- Katelynn E Dodd
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention (CDC), Morgantown, West Virginia.
| | - David J Blackley
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention (CDC), Morgantown, West Virginia
| | - Jacek M Mazurek
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention (CDC), Morgantown, West Virginia
| |
Collapse
|
27
|
Marcu DTM, Adam CA, Mitu F, Cumpat C, Aursulesei Onofrei V, Zabara ML, Burlacu A, Crisan Dabija R. Cardiovascular Involvement in Tuberculosis: From Pathophysiology to Diagnosis and Complications-A Narrative Review. Diagnostics (Basel) 2023; 13:432. [PMID: 36766543 PMCID: PMC9914020 DOI: 10.3390/diagnostics13030432] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Although primarily a lung disease, extra-pulmonary tuberculosis (TB) can affect any organ or system. Of these, cardiovascular complications associated with disease or drug toxicity significantly worsen the prognosis. Approximately 60% of patients with TB have a cardiovascular disease, the most common associated pathological entities being pericarditis, myocarditis, and coronary artery disease. We searched the electronic databases PubMed, MEDLINE, and EMBASE for studies that evaluated the impact of TB on the cardiovascular system, from pathophysiological mechanisms to clinical and paraclinical diagnosis of cardiovascular involvement as well as the management of cardiotoxicity associated with antituberculosis medication. The occurrence of pericarditis in all its forms and the possibility of developing constrictive pericarditis, the association of concomitant myocarditis with severe systolic dysfunction and complication with acute heart failure phenomena, and the long-term development of aortic aneurysms with risk of complications, as well as drug-induced toxicity, pose complex additional problems in the management of patients with TB. In the era of multidisciplinarity and polymedication, evidence-based medicine provides various tools that facilitate an integrative management that allows early diagnosis and treatment of cardiac pathologies associated with TB.
Collapse
Affiliation(s)
- Dragos Traian Marius Marcu
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
- Clinical Hospital of Pneumophthisiology Iași, Doctor Iosif Cihac Street no 30, 700115 Iasi, Romania
| | - Cristina Andreea Adam
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
- Clinical Rehabilitation Hospital, Cardiovascular Rehabilitation Clinic, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
| | - Florin Mitu
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
- Clinical Rehabilitation Hospital, Cardiovascular Rehabilitation Clinic, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
- Academy of Medical Sciences, Ion C. Brătianu Boulevard No 1, 030167 Bucharest, Romania
- Academy of Romanian Scientists, Dimitrie Mangeron Boulevard No. 433, 700050 Iasi, Romania
| | - Carmen Cumpat
- Clinical Rehabilitation Hospital, Cardiovascular Rehabilitation Clinic, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
- Department of Management, Alexandru Ioan Cuza University, Carol I Boulevard, 700506 Iasi, Romania
| | - Viviana Aursulesei Onofrei
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
- St. Spiridon Clinical Emergency Hospital, Independence Boulevard No. 1, 700111 Iasi, Romania
| | - Mihai Lucian Zabara
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
| | - Alexandru Burlacu
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
- Institute of Cardiovascular Diseases George I.M. Georgescu, 700503 Iasi, Romania
| | - Radu Crisan Dabija
- Department of Medical Specialties (I and III) and Surgical Specialties, Grigore T. Popa University of Medicine and Pharmacy, University Street No. 16, 700115 Iaşi, Romania
- Clinical Hospital of Pneumophthisiology Iași, Doctor Iosif Cihac Street no 30, 700115 Iasi, Romania
| |
Collapse
|
28
|
Wang K, Wang W, Lei L, Lan Y, Liu Q, Ren L, Wu S. Association between short-term exposure to ambient air pollution and biomarkers of coagulation: A systematic review and meta-analysis. ENVIRONMENTAL RESEARCH 2022; 215:114210. [PMID: 36030918 DOI: 10.1016/j.envres.2022.114210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
Ambient air pollution is one of the major global risk factors for cardiovascular health, and coagulation changes have been proposed to mediate this risk. Plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor (vWF), soluble P-selectin (sP-selectin) and tissue plasminogen activator (t-PA) are major coagulation biomarkers. However, there has been no systematic meta-analysis to summarize associations of ambient air pollution with these coagulation biomarkers. To assess the overall associations between ambient particulate matter (PM2.5, PM10), ozone (O3), nitrogen dioxide (NO2), carbon monoxide (CO) and major coagulation biomarkers including PAI-1, vWF, sP-selectin and t-PA based on the existing epidemiological research. We performed a systematic literature search of publications reporting the associations of ambient air pollutants (PM2.5, PM10, O3, NO2, and CO) with coagulation biomarkers (PAI-1, vWF, sP-selectin and t-PA) in PubMed, Web of Science, EMBASE, and Scopus databases as of April 5, 2022. Then, we performed a random-effect meta-analysis, which included 27 articles, and then identified the potential sources of heterogeneity. The pooled percent changes of coagulation biomarkers per 10 μg/m3 increase in short-term exposure to ambient PM2.5 were 2.43% (95% CI: 0.59%, 4.29%) in PAI-1, 1.08% (95% CI: 0.21%, 1.96%) in vWF and 1.14% (95% CI: 0.59%, 1.68%) in sP-selectin, respectively. We also found significant associations of short-term exposure to ambient O3 with PAI-1 (1.62%, 95% CI: 0.01%, 3.25%), sP-selectin (9.59%, 95% CI:2.78%, 16.86%) and t-PA (0.45%, 95% CI: 0.02%, 0.88%), respectively. Short-term exposures to ambient PM10, NO2 and CO were not significantly associated with changes in coagulation biomarkers. In conclusion, short-term exposures to PM2.5 and O3 are associated with significant increases in coagulation biomarkers, suggesting an activated coagulation state upon air pollution exposure.
Collapse
Affiliation(s)
- Kai Wang
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Wanzhou Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Lei Lei
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Yang Lan
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China
| | - Qisijing Liu
- Research Institute of Public Health, School of Medicine, Nankai University, Tianjin, China
| | - Lihua Ren
- School of Nursing, Peking University, Beijing, China
| | - Shaowei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China.
| |
Collapse
|
29
|
Uliński R, Kwiecień I, Domagała-Kulawik J. Lung Cancer in the Course of COPD-Emerging Problems Today. Cancers (Basel) 2022; 14:cancers14153819. [PMID: 35954482 PMCID: PMC9367492 DOI: 10.3390/cancers14153819] [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: 06/21/2022] [Revised: 07/26/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Tobacco smoking remains the main cause of tobacco-dependent diseases like lung cancer, chronic obstructive pulmonary disease (COPD), in addition to cardiovascular diseases and other cancers. Whilst the majority of smokers will not develop either COPD or lung cancer, they are closely related diseases, occurring as co-morbidities at a higher rate than if they were independently triggered by smoking. A patient with COPD has a four- to six-fold greater risk of developing lung cancer independent of smoking exposure, when compared to matched smokers with normal lung function. The 10 year risk is about 8.8% in the COPD group and only 2% in patients with normal lung function. COPD is not a uniform disorder: there are different phenotypes. One of them is manifested by the prevalence of emphysema and this is complicated by malignant processes most often. Here, we present and discuss the clinical problems of COPD in patients with lung cancer and against lung cancer in the course of COPD. There are common pathological pathways in both diseases. These are inflammation with participation of macrophages and neutrophils and proteases. It is known that anticancer immune regulation is distorted towards immunosuppression, while in COPD the elements of autoimmunity are described. Cytotoxic T cells, lymphocytes B and regulatory T cells with the important role of check point molecules are involved in both processes. A growing number of lung cancer patients are treated with immune check point inhibitors (ICIs), and it was found that COPD patients may have benefits from this treatment. Altogether, the data point to the necessity for deeper analysis and intensive research studies to limit the burden of these serious diseases by prevention and by elaboration of specific therapeutic options.
Collapse
Affiliation(s)
- Robert Uliński
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Iwona Kwiecień
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine, 04-141 Warsaw, Poland
| | - Joanna Domagała-Kulawik
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, 02-097 Warsaw, Poland
- Correspondence:
| |
Collapse
|
30
|
Rögnvaldsson KG, Bjarnason A, Kristinsson K, Bragason HT, Erlendsdóttir H, Þorgeirsson G, Gottfreðsson M. Acetylsalicylic acid use is associated with improved survival in bacteremic pneumococcal pneumonia: A long-term nationwide study. J Intern Med 2022; 292:321-332. [PMID: 35315156 PMCID: PMC9543431 DOI: 10.1111/joim.13485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Pneumonia is commonly caused by Streptococcus pneumoniae (pneumococcus) and associated with subsequent cardiovascular complications and increased mortality. Potential short-term survival benefits conferred by acetylsalicylic acid (ASA) use in pneumonia remain controversial, and long-term outcomes have not been studied. OBJECTIVES To evaluate the association between ASA use and survival for up to 1 year following bacteremic pneumococcal pneumonia. METHODS All bacteremic pneumococcal episodes in Iceland from 1975 to 2019 were reviewed. The study cohort consisted of individuals at least 18 years of age with symptoms and imaging results consistent with pneumonia. Differences in survival were assessed at 30 days, 90 days and 1 year using propensity score weighting (inverse probability weighting). Splitting and stratifying on survival at 7 days was done for the 30-day survival, because of nonproportionality. RESULTS In total, 815 bacteremic pneumococcal pneumonia episodes (median age 67 years, females 48%) were identified. Cox regression using propensity score weighting on the association of ASA with survival at 30 days showed an average hazard ratio (HR) of 0.60 (95% confidence interval [CI] 0.34-1.05). A significantly improved survival was observed within 7 days (HR = 0.42, 95% CI 0.19-0.92) but not during days 7-30 (HR = 1.08, 95% CI 0.46-2.55). ASA was associated with survival at 90 days (HR = 0.53, 95% CI 0.32-0.87) and 1 year (HR = 0.48, 95% CI 0.31-0.75). CONCLUSION Use of ASA upon admission for bacteremic pneumococcal pneumonia is associated with significantly reduced mortality for up to 1 year after diagnosis. ASA therapy in patients with pneumonia and other infectious syndromes warrants further study.
Collapse
Affiliation(s)
| | - Agnar Bjarnason
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Karl Kristinsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Akutklinikken, Rikshospitalet, Oslo, Norway
| | - Hörður T Bragason
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Helga Erlendsdóttir
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Guðmundur Þorgeirsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - Magnús Gottfreðsson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| |
Collapse
|
31
|
Label-free detection and quantification of ultrafine particulate matter in lung and heart of mouse and evaluation of tissue injury. Part Fibre Toxicol 2022; 19:51. [PMID: 35883088 PMCID: PMC9316794 DOI: 10.1186/s12989-022-00493-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022] Open
Abstract
While it is known that air borne ultrafine particulate matter (PM) may pass through the pulmonary circulation of blood at the alveolar level between lung and heart and cross the air-blood barrier, the mechanism and effects are not completely clear. In this study the imaging method fluorescence lifetime imaging microscopy is adopted for visualization with high spatial resolution and quantification of ultrafine PM particles in mouse lung and heart tissues. The results showed that the median numbers of particles in lung of mice exposed to ultrafine particulate matter of diameter less than 2.5 µm was about 2.0 times more than that in the filtered air (FA)-treated mice, and about 1.3 times more in heart of ultrafine PM-treated mice than in FA-treated mice. Interestingly, ultrafine PM particles were more abundant in heart than lung, likely due to how ultrafine PM particles are cleared by phagocytosis and transport via circulation from lungs. Moreover, heart tissues showed inflammation and amyloid deposition. The component analysis of concentrated airborne ultrafine PM particles suggested traffic exhausts and industrial emissions as predominant sources. Our results suggest association of ultrafine PM exposure to chronic lung and heart tissue injuries. The current study supports the contention that industrial air pollution is one of the causative factors for rising levels of chronic pulmonary and cardiac diseases.
Collapse
|
32
|
Memaran N, Onnen M, Müller C, Schwerk N, Carlens J, Borchert-Mörlins B, Bauer E, Blöte R, Sugianto RI, Zürn K, Wühl E, Warnecke G, Tudorache I, Hansen G, Gjertson DW, Schmidt BMW, Melk A. Cardiovascular Burden Is High in Pediatric Lung Transplant Recipients. Transplantation 2022; 106:1465-1472. [PMID: 34982755 DOI: 10.1097/tp.0000000000004025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cardiovascular morbidity is common in adults after lung transplantation (LTx) but has not been described for pediatric LTx recipients. Early subclinical cardiovascular damage is reflected by increases in pulse wave velocity (PWV; indicating arteriosclerosis), intima-media thickness (IMT; indicating atherosclerosis), and left ventricular mass index (LVMI; indicating left ventricular hypertrophy). METHODS We annually assessed 47 pediatric LTx recipients in a prospective longitudinal study (144 observations, mean 3.1 visits/patient, range of 1-4 visits, mean follow-up 2.2 y). RESULTS At inclusion, increased PWV and IMT were detected in 13% and 30%, respectively, and elevated LVMI was detected in 33%. Higher PWV was associated with male sex, longer time since LTx, higher diastolic blood pressure, and lower glomerular filtration rate. Male sex and lower hemoglobin levels were associated with higher IMT, and the presence of diabetes was associated with higher LVMI. CONCLUSIONS Pediatric LTx recipients suffer from a high and sustained burden of subclinical cardiovascular damage. In light of improving long-term outcomes, cardiovascular morbidity needs to be addressed. Our analysis identified classical and nonclassical risk factors to be associated with the measures for cardiovascular damage, which could serve as targets for intervention.
Collapse
Affiliation(s)
- Nima Memaran
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Mareike Onnen
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Carsten Müller
- Clinic for Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Nicolaus Schwerk
- Clinic for Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Julia Carlens
- Clinic for Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
| | - Bianca Borchert-Mörlins
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Elena Bauer
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Ricarda Blöte
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Rizky I Sugianto
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Katharina Zürn
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Elke Wühl
- Division of Pediatric Nephrology, University Children's Hospital, University of Heidelberg, Heidelberg, Germany
| | - Gregor Warnecke
- Department of Cardiothoracic Surgery, Hannover Medical School, Hannover, Germany
| | - Igor Tudorache
- Department of Cardiothoracic Surgery, Hannover Medical School, Hannover, Germany
| | - Gesine Hansen
- Clinic for Pediatric Pneumology, Allergology, and Neonatology, Hannover Medical School, Hannover, Germany
| | - David W Gjertson
- Division of Biostatistics, University of California, Los Angeles School of Public Health, Los Angeles, CA
| | | | - Anette Melk
- Department of Pediatric Kidney, Liver, and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Curtis K, Clarke D, Hanegan M, Stapley B, Wendt R, Beckett N, Litchfield C, Campbell K, Reynolds P, Arroyo J. Lung Inflammation Is Associated with Preeclampsia Development in the Rat. Cells 2022; 11:1884. [PMID: 35741013 PMCID: PMC9220878 DOI: 10.3390/cells11121884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/01/2023] Open
Abstract
Preeclampsia (PE) is an obstetric complication associated with significant health implications for the fetus and mother. Studies have shown a correlation between lung disease development and PE. Gas6 protein is expressed in the lung and placenta, and binds to the AXL Tyrosine kinase receptor. Recently, our laboratory utilized Gas6 to induce preeclamptic-like conditions in rats. Our objective was to determine the role of Gas6/AXL signaling in the maternal lung during PE development. Briefly, pregnant rats were divided into control, Gas6, or Gas6 + R428 (an AXL inhibitor). Immunofluorescence was performed to determine AXL expression. Bronchoalveolar lavage fluid (BALF) was procured for the assessment of inflammatory cell secretion. Western blot was performed to detect signaling molecules and ELISA determined inflammatory cytokines. We observed increased proteinuria and increased blood pressure in Gas6-treated animals. AXL was increased in the lungs of the treated animals and BALF fluid revealed elevated total protein abundance in Gas6 animals. Extracellular-signal regulated kinase (ERK) and protein kinase B (AKT) signaling in the lung appeared to be mediated by Gas6 as well as the secretion of inflammatory cytokines. We conclude that Gas6 signaling is capable of inducing PE and that this is associated with increased lung inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Juan Arroyo
- Lung and Placenta Laboratory, Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA; (K.C.); (D.C.); (M.H.); (B.S.); (R.W.); (N.B.); (C.L.); (K.C.); (P.R.)
| |
Collapse
|
34
|
Kenney AD, Aron SL, Gilbert C, Kumar N, Chen P, Eddy A, Zhang L, Zani A, Vargas-Maldonado N, Speaks S, Kawahara J, Denz PJ, Dorn L, Accornero F, Ma J, Zhu H, Rajaram MVS, Cai C, Langlois RA, Yount JS. Influenza virus replication in cardiomyocytes drives heart dysfunction and fibrosis. SCIENCE ADVANCES 2022; 8:eabm5371. [PMID: 35544568 PMCID: PMC9094651 DOI: 10.1126/sciadv.abm5371] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 03/24/2022] [Indexed: 05/04/2023]
Abstract
Cardiac dysfunction is a common complication of severe influenza virus infection, but whether this occurs due to direct infection of cardiac tissue or indirectly through systemic lung inflammation remains unclear. To test the etiology of this aspect of influenza disease, we generated a novel recombinant heart-attenuated influenza virus via genome incorporation of target sequences for miRNAs expressed in cardiomyocytes. Compared with control virus, mice infected with miR-targeted virus had significantly reduced heart viral titers, confirming cardiac attenuation of viral replication. However, this virus was fully replicative in the lungs and induced similar systemic inflammation and weight loss compared to control virus. The miR-targeted virus induced fewer cardiac conduction irregularities and significantly less fibrosis in mice lacking interferon-induced transmembrane protein 3 (IFITM3), which serve as a model for influenza-associated cardiac pathology. We conclude that robust virus replication in the heart is required for pathology, even when lung inflammation is severe.
Collapse
Affiliation(s)
- Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| | - Stephanie L. Aron
- Department of Microbiology and Immunology, The University of Minnesota, Minneapolis, MN, USA
| | - Clara Gilbert
- Department of Microbiology and Immunology, The University of Minnesota, Minneapolis, MN, USA
| | - Naresh Kumar
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Peng Chen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Adrian Eddy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| | - Lizhi Zhang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| | - Ashley Zani
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| | - Nahara Vargas-Maldonado
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Samuel Speaks
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Jeffrey Kawahara
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| | - Parker J. Denz
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| | - Lisa Dorn
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Federica Accornero
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Jianjie Ma
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Hua Zhu
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
| | - Chuanxi Cai
- Department of Surgery, The Ohio State University, Columbus, OH, USA
| | - Ryan A. Langlois
- Department of Microbiology and Immunology, The University of Minnesota, Minneapolis, MN, USA
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA
- Infectious Diseases Institute, Viruses and Emerging Pathogens Program, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
35
|
Wang H, Wang T, Rui W, Xie J, Xie Y, Zhang X, Guan L, Li G, Lei Z, Schiffelers RM, Sluijter JPG, Xiao J. Extracellular vesicles enclosed-miR-421 suppresses air pollution (PM 2.5 )-induced cardiac dysfunction via ACE2 signalling. J Extracell Vesicles 2022; 11:e12222. [PMID: 35536587 PMCID: PMC9089227 DOI: 10.1002/jev2.12222] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/03/2022] [Accepted: 04/15/2022] [Indexed: 01/05/2023] Open
Abstract
Air pollution, via ambient PM2.5, is a big threat to public health since it associates with increased hospitalisation, incidence rate and mortality of cardiopulmonary injury. However, the potential mediators of pulmonary injury in PM2.5‐induced cardiovascular disorder are not fully understood. To investigate a potential cross talk between lung and heart upon PM2.5 exposure, intratracheal instillation in vivo, organ culture ex vivo and human bronchial epithelial cells (Beas‐2B) culture in vitro experiments were performed respectively. The exposed supernatants of Beas‐2B were collected to treat primary neonatal rat cardiomyocytes (NRCMs). Upon intratracheal instillation, subacute PM2.5 exposure caused cardiac dysfunction, which was time‐dependent secondary to lung injury in mice, thereby demonstrating a cross‐talk between lungs and heart potentially mediated via small extracellular vesicles (sEV). We isolated sEV from PM2.5‐exposed mice serum and Beas‐2B supernatants to analyse the change of sEV subpopulations in response to PM2.5. Single particle interferometric reflectance imaging sensing analysis (SP‐IRIS) demonstrated that PM2.5 increased CD63/CD81/CD9 positive particles. Our results indicated that respiratory system‐derived sEV containing miR‐421 contributed to cardiac dysfunction post‐PM2.5 exposure. Inhibition of miR‐421 by AAV9‐miR421‐sponge could significantly reverse PM2.5‐induced cardiac dysfunction in mice. We identified that cardiac angiotensin converting enzyme 2 (ACE2) was a downstream target of sEV‐miR421, and induced myocardial cell apoptosis and cardiac dysfunction. In addition, we observed that GW4869 (an inhibitor of sEV release) or diminazene aceturate (DIZE, an activator of ACE2) treatment could attenuate PM2.5‐induced cardiac dysfunction in vivo. Taken together, our results suggest that PM2.5 exposure promotes sEV‐linked miR421 release after lung injury and hereby contributes to PM2.5‐induced cardiac dysfunction via suppressing ACE2.
Collapse
Affiliation(s)
- Hongyun Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Tianhui Wang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Wei Rui
- Institute for Immunology, Tsinghua University, Beijing, China
| | - Jinxin Xie
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Yuling Xie
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xiao Zhang
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Longfei Guan
- China-America Institute Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhiyong Lei
- CDL Research, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | | | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht, The Netherlands
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| |
Collapse
|
36
|
Guo J, Zhang Y, Liu T, Levy BD, Libby P, Shi GP. Allergic asthma is a risk factor for human cardiovascular diseases. NATURE CARDIOVASCULAR RESEARCH 2022; 1:417-430. [PMID: 39195946 DOI: 10.1038/s44161-022-00067-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 04/08/2022] [Indexed: 08/29/2024]
Abstract
Asthma is an allergic airway disease in which type 2-mediated inflammation has a pathogenic role. Cardiovascular diseases (CVDs) are type 1-dominant inflammatory diseases in which type 2 cytokines often have a protective role. However, clinical studies demonstrate that allergic asthma and associated allergies are essential risk factors for CVD, including coronary heart diseases, aortic diseases, peripheral arterial diseases, pulmonary embolism, right ventricular dysfunction, atrial fibrillation, cardiac hypertrophy and even hypertension. Mast cells, eosinophils, inflammatory cytokines and immunoglobulin (Ig)E accumulate in asthmatic lungs and in the injured heart and vasculature of patients with CVD. Clinical studies show that many anti-asthmatic therapies affect the risk of CVD. As such, allergic asthma and CVD may share common pathogenic mechanisms. Preclinical investigations indicate that anti-asthmatic drugs have therapeutic potential in certain CVDs. In this Review, we discuss how asthma and allied allergic conditions may contribute to the prevalence, incidence and progression of CVD and vice versa.
Collapse
Affiliation(s)
- Junli Guo
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province & Key Laboratory of Emergency and Trauma of Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yuanyuan Zhang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province & Key Laboratory of Emergency and Trauma of Ministry of Education, the First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Tianxiao Liu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bruce D Levy
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Puuvuori E, Liggieri F, Velikyan I, Chiodaroli E, Sigfridsson J, Romelin H, Ingvast S, Korsgren O, Hulsart-Billström G, Perchiazzi G, Eriksson O. PET-CT imaging of pulmonary inflammation using [ 68Ga]Ga-DOTA-TATE. EJNMMI Res 2022; 12:19. [PMID: 35394238 PMCID: PMC8994000 DOI: 10.1186/s13550-022-00892-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/28/2022] [Indexed: 12/21/2022] Open
Abstract
PURPOSE In the characterization of severe lung diseases, early detection of specific inflammatory cells could help to monitor patients' response to therapy and increase chances of survival. Macrophages contribute to regulating the resolution and termination of inflammation and have increasingly been of interest for targeted therapies. [68Ga]Ga-DOTA-TATE is an established clinical radiopharmaceutical targeting somatostatin receptor subtype 2 (SSTR 2). Since activated macrophages (M1) overexpress SSTR 2, the aim of this study was to investigate the applicability of [68Ga]Ga-DOTA-TATE for positron emission tomography (PET) imaging of M1 macrophages in pulmonary inflammation. METHODS Inflammation in the pig lungs was induced by warm saline lavage followed by injurious ventilation in farm pigs (n = 7). Healthy pigs (n = 3) were used as control. A 60-min dynamic PET scan over the lungs was performed after [68Ga]Ga-DOTA-TATE injection and [18F]FDG scan was executed afterward for comparison. The uptake of both tracers was assessed as mean standardized uptake values (SUVmean) 30-60-min post-injection. The PET scans were followed by computed tomography (CT) scans, and the Hounsfield units (HU) were quantified of the coronal segments. Basal and apical segments of the lungs were harvested for histology staining. A rat lung inflammation model was also studied for tracer specificity using lipopolysaccharides (LPS) by oropharyngeal aspiration. Organ biodistribution, ex vivo autoradiography (ARG) and histology samples were conducted on LPS treated, octreotide induced blocking and control healthy rats. RESULTS The accumulation of [68Ga]Ga-DOTA-TATE on pig lavage model was prominent in the more severely injured dorsal segments of the lungs (SUVmean = 0.91 ± 0.56), compared with control animals (SUVmean = 0.27 ± 0.16, p < 0.05). The tracer uptake corresponded to the damaged areas assessed by CT and histology and were in line with HU quantification. The [68Ga]Ga-DOTA-TATE uptake in LPS treated rat lungs could be blocked and was significantly higher compared with control group. CONCLUSION The feasibility of the noninvasive assessment of tissue macrophages using [68Ga]Ga-DOTA-TATE/PET was demonstrated in both porcine and rat lung inflammation models. [68Ga]Ga-DOTA-TATE has a great potential to be used to study the role and presence of macrophages in humans in fight against severe lung diseases.
Collapse
Affiliation(s)
- Emmi Puuvuori
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Dag Hammarskjölds väg 14C, 3tr, 751 83, Uppsala, Sweden
| | - Francesco Liggieri
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Irina Velikyan
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Dag Hammarskjölds väg 14C, 3tr, 751 83, Uppsala, Sweden
| | - Elena Chiodaroli
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jonathan Sigfridsson
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Hampus Romelin
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Sofie Ingvast
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Gry Hulsart-Billström
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Dag Hammarskjölds väg 14C, 3tr, 751 83, Uppsala, Sweden
| | - Gaetano Perchiazzi
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, Dag Hammarskjölds väg 14C, 3tr, 751 83, Uppsala, Sweden.
| |
Collapse
|
38
|
Botello-Manilla AE, López-Sánchez GN, Chávez-Tapia NC, Uribe M, Nuño-Lámbarri N. Hepatic steatosis and respiratory diseases: a new panorama. Ann Hepatol 2022; 24:100320. [PMID: 33549735 DOI: 10.1016/j.aohep.2021.100320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/04/2023]
Abstract
Non-alcoholic fatty liver disease is defined as hepatic fat accumulation in more than 5% of hepatocytes, without other liver steatosis causes. It comprises a broad spectrum that can range from benign steatosis and progress to non-alcoholic steatohepatitis, fibrosis, and ultimately hepatocellular carcinoma. Non-alcoholic fatty liver is considered a multisystemic disease since it is related to multiple disorders, such as type 2 diabetes mellitus, polycystic ovary syndrome, chronic kidney disease, psoriasis, osteoporosis, hypothyroidism, cardiovascular diseases, and obstructive sleep apnea syndrome; it is becoming increasingly clear that it is also a risk factor for developing certain respiratory diseases. This article aims to understand the liver and chronic obstructive pulmonary disease mechanisms, obstructive sleep apnea syndrome, asthma, and lung cancer. Given that non-alcoholic fatty liver disease has a considerable impact on the patient's well-being and life quality, as well as on the costs they generate for the country's health services, it is essential to continue research, especially in areas such as the respiratory tract, as there is much misinformation about it.
Collapse
Affiliation(s)
- Alan Eduardo Botello-Manilla
- Traslational Research Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Toriello Guerra Tlalpan, C.P. 14050, Mexico City, Mexico
| | - Guillermo Nahúm López-Sánchez
- Traslational Research Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Toriello Guerra Tlalpan, C.P. 14050, Mexico City, Mexico
| | - Norberto Carlos Chávez-Tapia
- Traslational Research Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Toriello Guerra Tlalpan, C.P. 14050, Mexico City, Mexico; Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Toriello Guerra Tlalpan, C.P. 14050, Mexico City, Mexico
| | - Misael Uribe
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Toriello Guerra Tlalpan, C.P. 14050, Mexico City, Mexico
| | - Natalia Nuño-Lámbarri
- Traslational Research Unit, Medica Sur Clinic & Foundation, Puente de Piedra 150, Toriello Guerra Tlalpan, C.P. 14050, Mexico City, Mexico.
| |
Collapse
|
39
|
Gilbert RF, Cichowitz C, Bibangambah P, Kim JH, Hemphill LC, Yang IT, Sentongo RN, Kakuhikire B, Christiani DC, Tsai AC, Okello S, Siedner MJ, North CM. Lung function and atherosclerosis: a cross-sectional study of multimorbidity in rural Uganda. BMC Pulm Med 2022; 22:12. [PMID: 34983492 PMCID: PMC8728924 DOI: 10.1186/s12890-021-01792-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a leading cause of global mortality. In high-income settings, the presence of cardiovascular disease among people with COPD increases mortality and complicates longitudinal disease management. An estimated 26 million people are living with COPD in sub-Saharan Africa, where risk factors for co-occurring pulmonary and cardiovascular disease may differ from high-income settings but remain uncharacterized. As non-communicable diseases have become the leading cause of death in sub-Saharan Africa, defining multimorbidity in this setting is critical to inform the required scale-up of existing healthcare infrastructure. METHODS We measured lung function and carotid intima media thickness (cIMT) among participants in the UGANDAC Study. Study participants were over 40 years old and equally divided into people living with HIV (PLWH) and an age- and sex-similar, HIV-uninfected control population. We fit multivariable linear regression models to characterize the relationship between lung function (forced expiratory volume in one second, FEV1) and pre-clinical atherosclerosis (cIMT), and evaluated for effect modification by age, sex, smoking history, HIV, and socioeconomic status. RESULTS Of 265 participants, median age was 52 years, 125 (47%) were women, and 140 (53%) were PLWH. Most participants who met criteria for COPD were PLWH (13/17, 76%). Median cIMT was 0.67 mm (IQR: 0.60 to 0.74), which did not differ by HIV serostatus. In models adjusted for age, sex, socioeconomic status, smoking, and HIV, lower FEV1 was associated with increased cIMT (β = 0.006 per 200 mL FEV1 decrease; 95% CI 0.002 to 0.011, p = 0.01). There was no evidence that age, sex, HIV serostatus, smoking, or socioeconomic status modified the relationship between FEV1 and cIMT. CONCLUSIONS Impaired lung function was associated with increased cIMT, a measure of pre-clinical atherosclerosis, among adults with and without HIV in rural Uganda. Future work should explore how co-occurring lung and cardiovascular disease might share risk factors and contribute to health outcomes in sub-Saharan Africa.
Collapse
Affiliation(s)
- Rebecca F Gilbert
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA
| | - Cody Cichowitz
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA
| | | | - June-Ho Kim
- Brigham and Women's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Linda C Hemphill
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Ruth N Sentongo
- Mbarara University of Science and Technology, Mbarara, Uganda
| | | | - David C Christiani
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA.,Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Alexander C Tsai
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA.,Mbarara University of Science and Technology, Mbarara, Uganda.,Harvard Medical School, Boston, MA, USA
| | - Samson Okello
- Mbarara University of Science and Technology, Mbarara, Uganda
| | - Mark J Siedner
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA.,Mbarara University of Science and Technology, Mbarara, Uganda.,Harvard Medical School, Boston, MA, USA
| | - Crystal M North
- Massachusetts General Hospital, 55 Fruit Street, BUL-148, Boston, MA, 02114, USA. .,Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Cho SN, Choi JA, Lee J, Son SH, Lee SA, Nguyen TD, Choi SY, Song CH. Ang II-Induced Hypertension Exacerbates the Pathogenesis of Tuberculosis. Cells 2021; 10:cells10092478. [PMID: 34572127 PMCID: PMC8465031 DOI: 10.3390/cells10092478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/23/2022] Open
Abstract
It has been known that infection plays a role in the development of hypertension. However, the role of hypertension in the progression of infectious diseases remain unknown. Many countries with high rates of hypertension show geographical overlaps with those showing high incidence rates of tuberculosis (TB). To explore the role of hypertension in tuberculosis, we compared the effects of hypertension during mycobacterial infection, we infected both hypertensive Angiotensin II (Ang II) and control mice with Mycobacterium tuberculosis (Mtb) strain H37Ra by intratracheal injection. Ang II-induced hypertension promotes cell death through both apoptosis and necrosis in Mtb H37Ra infected mouse lungs. Interestingly, we found that lipid accumulation in pulmonary tissues was significantly increased in the hypertension group compared to the normal controls. Ang II-induced hypertension increases the formation of foamy macrophages during Mtb infection and it leads to cell death. Moreover, the hypertension group showed more severe granuloma formation and fibrotic lesions in comparison with the control group. Finally, we observed that the total number of mycobacteria was increased in the lungs in the hypertension group compared to the normal controls. Taken together, these results suggest that hypertension increases intracellular survival of Mtb through formation of foamy macrophages, resulting in severe pathogenesis of TB.
Collapse
Affiliation(s)
- Soo-Na Cho
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Ji-Ae Choi
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Junghwan Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Sang-Hun Son
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Seong-Ahn Lee
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Tam-Doan Nguyen
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Song-Yi Choi
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon 35015, Korea;
- Translational Immunology Institute, Chungnam National University, Daejeon 34134, Korea
| | - Chang-Hwa Song
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Korea; (S.-N.C.); (J.-A.C.); (J.L.); (S.-H.S.); (S.-A.L.); (T.-D.N.)
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Korea
- Translational Immunology Institute, Chungnam National University, Daejeon 34134, Korea
- Correspondence: ; Tel.: +82-42-580-8245; Fax: +82-42-585-3686
| |
Collapse
|
41
|
Pham HH, Le TT, Tran DQ, Ngo DT, Nguyen HQ. Interpreting chest X-rays via CNNs that exploit hierarchical disease dependencies and uncertainty labels. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2020.03.127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
42
|
Gündel S, Setio AAA, Ghesu FC, Grbic S, Georgescu B, Maier A, Comaniciu D. Robust classification from noisy labels: Integrating additional knowledge for chest radiography abnormality assessment. Med Image Anal 2021; 72:102087. [PMID: 34015595 DOI: 10.1016/j.media.2021.102087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/24/2021] [Accepted: 04/16/2021] [Indexed: 12/29/2022]
Abstract
Chest radiography is the most common radiographic examination performed in daily clinical practice for the detection of various heart and lung abnormalities. The large amount of data to be read and reported, with more than 100 studies per day for a single radiologist, poses a challenge in consistently maintaining high interpretation accuracy. The introduction of large-scale public datasets has led to a series of novel systems for automated abnormality classification. However, the labels of these datasets were obtained using natural language processed medical reports, yielding a large degree of label noise that can impact the performance. In this study, we propose novel training strategies that handle label noise from such suboptimal data. Prior label probabilities were measured on a subset of training data re-read by 4 board-certified radiologists and were used during training to increase the robustness of the training model to the label noise. Furthermore, we exploit the high comorbidity of abnormalities observed in chest radiography and incorporate this information to further reduce the impact of label noise. Additionally, anatomical knowledge is incorporated by training the system to predict lung and heart segmentation, as well as spatial knowledge labels. To deal with multiple datasets and images derived from various scanners that apply different post-processing techniques, we introduce a novel image normalization strategy. Experiments were performed on an extensive collection of 297,541 chest radiographs from 86,876 patients, leading to a state-of-the-art performance level for 17 abnormalities from 2 datasets. With an average AUC score of 0.880 across all abnormalities, our proposed training strategies can be used to significantly improve performance scores.
Collapse
Affiliation(s)
- Sebastian Gündel
- Digital Technology and Inovation, Siemens Healthineers, Erlangen 91052, Germany; Pattern Recognition Lab, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91058, Germany.
| | - Arnaud A A Setio
- Digital Technology and Inovation, Siemens Healthineers, Erlangen 91052, Germany
| | - Florin C Ghesu
- Digital Technology and Inovation, Siemens Healthineers, Princeton, NJ 08540, USA
| | - Sasa Grbic
- Digital Technology and Inovation, Siemens Healthineers, Princeton, NJ 08540, USA
| | - Bogdan Georgescu
- Digital Technology and Inovation, Siemens Healthineers, Princeton, NJ 08540, USA
| | - Andreas Maier
- Pattern Recognition Lab, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Dorin Comaniciu
- Digital Technology and Inovation, Siemens Healthineers, Princeton, NJ 08540, USA
| |
Collapse
|
43
|
Poor Cognitive Function Is Associated with Obstructive Lung Diseases in Taiwanese Adults. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18052344. [PMID: 33673619 PMCID: PMC7957805 DOI: 10.3390/ijerph18052344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/21/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
Previous studies have reported an association between the impairment of cognitive performance and lung diseases. However, whether obstructive or restrictive lung diseases have an impact on cognitive function is still inconclusive. We aimed to investigate the association between cognitive function and obstructive or restrictive lung diseases in Taiwanese adults using the Mini-Mental State Examination (MMSE). In this study, we used data from the Taiwan Biobank. Cognitive function was evaluated using the MMSE. Spirometry measurements of forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) were obtained to assess lung function. Participants were classified into three groups according to lung function, namely, normal, restrictive, and obstructive lung function. In total, 683 patients enrolled, of whom 357 participants had normal lung function (52.3%), 95 had restrictive lung function (13.9%), and 231 had obstructive lung function (33.8%). Compared to the normal lung function group, the obstructive lung function group was associated with a higher percentage of cognitive impairment (MMSE < 24). In multivariable analysis, a low MMSE score was significantly associated with low FVC, low FEV1, and low FEV1/FVC. Furthermore, a low MMSE score was significantly associated with low FEV1 in the participants with FEV1/FVC < 70%, whereas MMSE was not significantly associated with FVC in the participants with FEV1/FVC ≥ 70%. Our results showed that a low MMSE score was associated with low FEV1, low FVC and low FEV1/FVC. Furthermore, a low MMSE score was associated with obstructive lung diseases but not with restrictive lung diseases.
Collapse
|
44
|
Digital Health for Enhanced Understanding and Management of Chronic Conditions: COPD as a Use Case. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11690-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
45
|
Crosstalk Between Lung and Extrapulmonary Organs in Infection and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:333-350. [PMID: 33788201 DOI: 10.1007/978-3-030-63046-1_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute and chronic lung inflammation is a risk factor for various diseases involving lungs and extrapulmonary organs. Intercellular and interorgan networks, including crosstalk between lung and brain, intestine, heart, liver, and kidney, coordinate host immunity against infection, protect tissue, and maintain homeostasis. However, this interaction may be counterproductive and cause acute or chronic comorbidities due to dysregulated inflammation in the lung. In this chapter, we review the relationship of the lung with other key organs during normal cell processes and disease development. We focus on how pneumonia may lead to a systemic pathophysiological response to acute lung injury and chronic lung disease through organ interactions, which can facilitate the development of undesirable and even deleterious extrapulmonary sequelae.
Collapse
|
46
|
İn E, Kuluöztürk M, Turgut T, Altıntop Geçkil A, İlhan N. Endocan as a potential biomarker of disease severity and exacerbations in COPD. CLINICAL RESPIRATORY JOURNAL 2020; 15:445-453. [PMID: 33319462 DOI: 10.1111/crj.13320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/10/2020] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Endocan is a proteoglycan that is regarded as a novel marker of endothelial dysfunction. Endothelial dysfunction in pulmonary vascular bed is known to play an important role for the pathogenesis of COPD. OBJECTIVE This study aimed to determine serum endocan levels in patients with stable COPD and acute exacerbation of COPD (AECOPD) and to test the relationship between serum endocan levels and exacerbations. METHODS This study enrolled a total of 55 COPD patients, 24 of which had AECOPD and 31 had stable COPD. All patients' basic demographic and clinical data were recorded and blood samples were collected. RESULTS Serum endocan levels were significantly higher in the AECOPD group compared to the stable COPD and control groups (for both p < 0.001) and stable COPD group had higher levels than the control group (p < 0.005). Additionally, serum endocan levels were negatively correlated with FVC, FEV1, partial oxygen pressure and oxygen saturation (r = -0.30, p = 0.03; r = -0.34, p = 0.01; r = -0.34, p = 0.01 and r = -0.36, p = 0.007 respectively), and positively correlated with disease duration and systolic pulmonary artery pressure (r = 0.47, p < 0.001; r = 0.31, p = 0.02 respectively). A cut-off value of 434.29 pg/ml for endocan predicted exacerbation with a sensitivity of 79% and a specificity of 84% (AUC: 0.778, 95% Cl 0.648-0.909; p < 0.001). Logistic regression analysis revealed that increased endocan levels was independent predictor of COPD exacerbation (OR = 9.32, 95%CI, 1.64-52.95; p = 0.01). CONCLUSION Endocan may be a novel biomarker for detection of endothelial dysfunction and prediction of exacerbations in patients with COPD.
Collapse
Affiliation(s)
- Erdal İn
- Department of Pulmonary Medicine, School of Medicine, Firat University, Elazig, Turkey
| | - Mutlu Kuluöztürk
- Department of Pulmonary Medicine, School of Medicine, Firat University, Elazig, Turkey
| | - Teyfik Turgut
- Department of Pulmonary Medicine, School of Medicine, Firat University, Elazig, Turkey
| | | | - Nevin İlhan
- Department of Medical Biochemistry, School of Medicine, Firat University, Elazig, Turkey
| |
Collapse
|
47
|
Vlachopoulos CV, Koutagiar IP, Georgakopoulos AT, Pouli AG, Sioni AΚ, Giannouli SΕ, Chondropoulos SD, Stergiou IΕ, Solomou EG, Terentes-Printzios DG, Karakitsios IG, Kafouris PP, Gaitanis A, Pianou NK, Petrocheilou A, Aggeli CI, Stroumpouli E, Marinakis TP, Voulgarelis M, Tousoulis DM, Anagnostopoulos CD. Lymphoma Severity and Type Are Associated With Aortic FDG Uptake by 18F-FDG PET/CT Imaging. JACC: CARDIOONCOLOGY 2020; 2:758-770. [PMID: 34396292 PMCID: PMC8352324 DOI: 10.1016/j.jaccao.2020.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022]
Abstract
Background There is evidence that metabolic disease burden in lymphoma influences patient outcome. However, the impact of disease severity on the cardiovascular system is unknown. Objectives The aim of this study was to examine whether lymphoma is associated with arterial inflammation by investigating the relationship between disease metabolic burden and arterial fluorodeoxyglucose (FDG) uptake. Methods Sixty-two chemotherapy-naïve patients with active Hodgkin’s or non-Hodgkin’s lymphoma were matched (2:1) to individual control groups of lymphoma patients previously treated and free of active disease. All groups underwent 18F-FDG position emission tomography–computed tomography imaging. Disease severity was quantified by metabolic tumor volume (MTV) and total lesion glycolysis corresponding to standardized uptake values (SUVs) ≥41% or ≥2.5 of the maximum SUV within lymphoma regions, and aortic FDG uptake was quantified through the target-to-background ratio (TBR). Inflammatory and disease severity biomarkers were also measured. Results MTV and total lesion glycolysis measurements were significantly correlated with inflammatory and disease biomarkers. Aortic TBR was higher in patients with active non-Hodgkin’s lymphoma compared with control subjects (median difference 0.51; 95% confidence interval [CI]: 0.28 to 0.78; p < 0.001). Similarly, patients with active Hodgkin’s lymphoma had higher values of aortic TBR compared with control subjects (median difference 0.31; 95% CI: 0.15 to 0.49; p < 0.001). In addition, aortic TBR was modestly increased in patients with stage III to IV disease compared with those with stage I to II disease (median aortic TBR: 2.23 [interquartile range: 2.01 to 2.54] vs. 2.06 [interquartile range: 1.83 to 2.27; p = 0.050). In multivariable analysis, aortic FDG uptake and MTV≥2.5 values were independently associated (β = 0.425; 95% CI: 0.189 to 0.662; p = 0.001; R2 = 0.208), as were aortic FDG uptake and MTV≥41% (β = 0.407; 95% CI: 0.167 to 0.649, p = 0.001; R2 = 0.191). Conclusions Aortic wall FDG uptake is related with disease severity indicative of a possible vascular effect of lymphoma. This work highlights a new potential role of molecular imaging in cardio-oncology for evaluating disease severity and its consequences on the vasculature.
Collapse
Key Words
- 18F-FDG, 18F-fluorodeoxyglucose
- BMI, body mass index
- CI, confidence interval
- CT, computed tomography
- CVD, cardiovascular disease
- LDH, lactate dehydrogenase
- MTV, metabolic tumor burden
- PET, positron emission tomography
- SUV, standardized uptake value
- SUVmax, maximum standardized uptake value
- SUVmean, mean standardized uptake value
- TBR, target-to-background ratio
- TLG, total lesion glycolysis
- WBC, white blood cell count
- arterial inflammation
- hsCRP, high-sensitivity C-reactive protein
- lymphoma
- metabolic burden
- positron emission tomography
Collapse
Affiliation(s)
- Charalambos V Vlachopoulos
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Iosif P Koutagiar
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros T Georgakopoulos
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | | | - Stavroula Ε Giannouli
- 2nd Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Ioanna Ε Stergiou
- Department of Pathophysiology, School of Medicine, University of Athens, Athens, Greece
| | - Eirini G Solomou
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios G Terentes-Printzios
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis G Karakitsios
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Pavlos P Kafouris
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.,Department of Informatics and Telecommunications, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Gaitanis
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Nikoletta K Pianou
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Aikaterini Petrocheilou
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantina I Aggeli
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Euaggelia Stroumpouli
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Michael Voulgarelis
- Department of Pathophysiology, School of Medicine, University of Athens, Athens, Greece
| | - Dimitrios M Tousoulis
- Hypertension and Cardiometabolic Syndrome Unit, 1st Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos D Anagnostopoulos
- Center for Experimental Surgery, Clinical and Translational Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| |
Collapse
|
48
|
Alfaddagh A, Martin SS, Leucker TM, Michos ED, Blaha MJ, Lowenstein CJ, Jones SR, Toth PP. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am J Prev Cardiol 2020; 4:100130. [PMID: 34327481 PMCID: PMC8315628 DOI: 10.1016/j.ajpc.2020.100130] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation constitutes a complex, highly conserved cascade of molecular and cellular events. Inflammation has been labeled as “the fire within,” is highly regulated, and is critical to host defense and tissue repair. In general, inflammation is beneficial and has evolved to promote survival. However, inflammation can also be maladaptive when chronically activated and sustained, leading to progressive tissue injury and reduced survival. Examples of a maladaptive response include rheumatologic disease and atherosclerosis. Despite evidence gathered by Virchow over 100 years ago showing that inflammatory white cells play a role in atherogenesis, atherosclerosis was until recently viewed as a disease of passive cholesterol accumulation in the subendothelial space. This view has been supplanted by considerable basic scientific and clinical evidence demonstrating that every step of atherogenesis, from the development of endothelial cell dysfunction to foam cell formation, plaque formation and progression, and ultimately plaque rupture stemming from architectural instability, is driven by the cytokines, interleukins, and cellular constituents of the inflammatory response. Herein we provide an overview of the role of inflammation in atherosclerotic cardiovascular disease, discuss the predictive value of various biomarkers involved in inflammation, and summarize recent clinical trials that evaluated the capacity of various pharmacologic interventions to attenuate the intensity of inflammation and impact risk for acute cardiovascular events.
Collapse
Affiliation(s)
- Abdulhamied Alfaddagh
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth S Martin
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten M Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erin D Michos
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles J Lowenstein
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven R Jones
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter P Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
49
|
Sohrabi F, Dianat M, Badavi M, Radan M, Mard SA. Does gallic acid improve cardiac function by attenuation of oxidative stress and inflammation in an elastase-induced lung injury? IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 23:1130-1138. [PMID: 32963734 PMCID: PMC7491503 DOI: 10.22038/ijbms.2020.46427.10721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Objective(s): Cardiovascular disease has an important role in mortality caused by lung injury. Emphysema is associated with impaired pulmonary gas exchange efficiency and airflow limitation associated with small airway inflammation. The aim was to evaluate the interactions between lung injury, inflammation, and cardiovascular disease. Since gallic acid has antioxidant and anti-inflammatory effects, we hypothesized that gallic acid protects the lung and the related heart dysfunction in elastase-induced lung injury. Materials and Methods: Forty-eight Sprague-Dawley male rats were randomly divided into six groups: Control, Porcine pancreatic elastase (PPE) , PPE+GA, and 3 groups for different doses of gallic acid (GA 7.5, GA 15, GA 30 mg/kg). PPE was injected intra-tracheally on days 1 and 10 of the test. In each group, electrocardiography, hemodynamic parameters, oxidative stress, and bronchoalveolar lavage fluid were examined. Results: PPE administration showed a decrease in HR and QRS voltage of electrocardiogram parameters, as well as in hemodynamic parameters (P<0.05, P<0.01, and P<0.001) and superoxide dismutase (SOD) (P<0.05). Tumor Necrosis Factor α (TNF-α) (P<0.001), interleukin 6 (IL-6) (P<0.001), interleukin 6 (MDA) (P<0.001), and the total number of white blood cells (P<0.001) showed an increase in PPE groups. Gallic acid preserved the values of hemodynamic properties, oxidative stress, inflammation, and electrocardiogram parameters in comparison to the PPE group. Conclusion: Briefly, this study showed the valuable effect of gallic acid in cardiac dysfunction related to elastase-induced lung injury. These findings suggested that gallic acid, as a natural antioxidant, has a potential therapeutic effect on preventing oxidative stress, inflammation, and subsequent cardiovascular disease.
Collapse
Affiliation(s)
- Farzaneh Sohrabi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahin Dianat
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Badavi
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Radan
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyyed Ali Mard
- Department of Physiology, Physiology Research Center, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
50
|
Niu M, Shen F, Zhou F, Zhu T, Zheng Y, Yang Y, Sun Y, Li X, Wu Y, Fu P, Tao S. Indoor air filtration could lead to increased airborne endotoxin levels. ENVIRONMENT INTERNATIONAL 2020; 142:105878. [PMID: 32580116 DOI: 10.1016/j.envint.2020.105878] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/21/2020] [Accepted: 06/04/2020] [Indexed: 05/13/2023]
Abstract
Stand-alone portable air purifiers (APs) have become an increasingly popular method of controlling individual inhalation exposure. Exposure to bacterial endotoxins has a causative role in respiratory inhalation health. Here, we studied the changes in endotoxin levels in indoor air before and after purification by a portable AP equipped with HEPA (high-efficiency particulate air) filters. An increase in endotoxins was observed when a previously used AP was turned on to clean the air. Replacing the HEPA filters in the AP helped to mitigate the increase in endotoxins of larger sizes but not endotoxins of smaller sizes. Consequently, the use of APs could lead to increased endotoxin deposition in airways, especially in the alveolar region. The endotoxin concentrations on the HEPA filters were well correlated with the free DNA concentrations on the HEPA filters. This correlation indicates that the disrupted bacteria, which released free DNA, could also release endotoxins, thus making HEPA filters a source of indoor airborne endotoxins. Our results illustrate a potential endotoxin inhalation risk associated with HEPA-APs as an air cleaning strategy and highlight the importance of composition-specific air cleaning while reducing the particle number/mass.
Collapse
Affiliation(s)
- Mutong Niu
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China
| | - Fangxia Shen
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China.
| | - Feng Zhou
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China
| | - Tianle Zhu
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China
| | - Yunhao Zheng
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yi Yang
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China
| | - Ye Sun
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China
| | - Xinghua Li
- School of Space and Environment, Beijing Key Laboratory of Bio-Inspired Energy Materials and Devices, Beihang University, Beijing 100191, China
| | - Yan Wu
- School of Environmental Science and Engineering, Shandong University, Jinan 250100, China
| | - Pingqing Fu
- Institute of Surface-Earth System Science, Tianjin University, Tianjin 300072, China; State Key Laboratory of Atmospheric Boundary Layer Physics and Atmospheric Chemistry, Institute of Atmospheric Physics, Chinese Academy of Sciences, Beijing 100029, China
| | - Shu Tao
- College of Urban and Environmental Sciences, Laboratory for Earth Surface Processes, Sino-French Institute for Earth System Science, Peking University, Beijing 100871, China
| |
Collapse
|