1
|
Chen YL, You J, Guo Y, Zhang Y, Yao BR, Wang JJ, Chen SD, Ge YJ, Yang L, Wu XR, Wu BS, Zhang YR, Dong Q, Feng JF, Tian M, Cheng W, Yu JT. Identifying proteins and pathways associated with multimorbidity in 53,026 adults. Metabolism 2024:156126. [PMID: 39740741 DOI: 10.1016/j.metabol.2024.156126] [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: 10/13/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND AND AIMS Multimorbidity, the coexistence of multiple chronic diseases, is a rapidly expanding global health challenge, carrying profound implications for patients, caregivers, healthcare systems, and society. Investigating the determinants and drivers underlying multiple chronic diseases is a priority for disease management and prevention. METHOD This prospective cohort study analyzed data from the 53,026 participants in the UK Biobank from baseline (2006 to 2010) across 13.3 years of follow-up. Using Cox proportional hazards regression model, we characterized shared and unique associations across 38 incident outcomes (31 chronic diseases, 6 system mortality and all-cause mortality). Furthermore, ordinal regression models were used to assess the association between protein levels and multimorbidity (0-1, 2, 3-4, or ≥ 5 chronic diseases). Functional and tissue enrichment analysis were employed for multimorbidity-associated proteins. The upstream regulators of above proteins were identified. RESULTS We demonstrated 972 (33.3 %) proteins were shared across at least two incident chronic diseases after Bonferroni correction (P < 3.42 × 10-7, 93.3 % of those had consistent effects directions), while 345 (11.8 %) proteins were uniquely linked to a single chronic disease. Remarkably, GDF15, PLAUR, WFDC2 and AREG were positively associated with 20-24 incident chronic diseases (hazards ratios: 1.21-3.77) and showed strong associations with multimorbidity (odds ratios: 1.33-1.89). We further identified that protein levels are explained by common risk factors, especially renal function, liver function, inflammation, and obesity, providing potential intervention targets. Pathway analysis has underscored the pivotal role of the immune response, with the top three transcription factors associated with proteomics being NFKB1, JUN and RELA. CONCLUSIONS Our results enhance the understanding of the biological basis underlying multimorbidity, offering biomarkers for disease identification and novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yi-Lin Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jia You
- Harvard Medical School, Boston, MA, USA
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bing-Ran Yao
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | | | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China
| | - Mei Tian
- Huashan Hospital & Human Phenome Institute, Fudan University, Shanghai, China; Department of Nuclear Medicine/PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-inspired Intelligence (ISTBI), Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Wen Y, Zhang X, Liu H, Ye H, Wang R, Ma C, Duo T, Wang J, Yang X, Yu M, Wang Y, Wu L, Zhao Y, Wang L. SGLT2 inhibitor downregulates ANGPTL4 to mitigate pathological aging of cardiomyocytes induced by type 2 diabetes. Cardiovasc Diabetol 2024; 23:430. [PMID: 39633372 PMCID: PMC11619200 DOI: 10.1186/s12933-024-02520-8] [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: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Senescence is recognized as a principal risk factor for cardiovascular diseases, with a significant association between the senescence of cardiomyocytes and inferior cardiac function. Furthermore, type 2 diabetes exacerbates this aging process. Sodium-glucose co-transporter 2 inhibitor (SGLT2i) has well-established cardiovascular benefits and, in recent years, has been posited to possess anti-aging properties. However, there are no reported data on their improvement of cardiomyocytes function through the alleviation of aging. Consequently, our study aims to investigate the mechanism by which SGLT2i exerts anti-aging and protective effects at the cardiac level through its action on the FOXO1-ANGPTL4 pathway. METHODS To elucidate the underlying functions and mechanisms, we established both in vivo and in vitro disease models, utilizing mice with diabetic cardiomyopathy (DCM) induced by type 2 diabetes mellitus (T2DM) through high-fat diet combined with streptozotocin (STZ) administration, and AC16 human cardiomyocyte cell subjected to stimulation with high glucose (HG) and palmitic acid (PA). These models were employed to assess the changes in the senescence phenotype of cardiomyocytes and cardiac function following treatment with SGLT2i. Concurrently, we identified ANGPTL4, a key factor contributing to senescence in DCM, using RNA sequencing (RNA-seq) technology and bioinformatics methods. We further clarified ANGPTL4 role in promoting pathological aging of cardiomyocytes induced by hyperglycemia and hyperlipidemia through knockdown and overexpression of the factor, as well as analyzed the impact of SGLT2i intervention on ANGPTL4 expression. Additionally, we utilized chromatin immunoprecipitation followed by quantitative real-time PCR (ChIP-qPCR) to confirm that FOXO1 is essential for the transcriptional activation of ANGPTL4. RESULTS The therapeutic intervention with SGLT2i alleviated the senescence phenotype in cardiomyocytes of the DCM mouse model constructed by high-fat feeding combined with STZ, as well as in the AC16 model stimulated by HG and PA, while also improving cardiac function in DCM mice. We observed that the knockdown of ANGPTL4, a key senescence-promoting factor in DCM identified through RNA-seq technology and bioinformatics, mitigated the senescence of cardiomyocytes, whereas overexpression of ANGPTL4 exacerbated it. Moreover, SGLT2i improved the senescence phenotype by suppressing the overexpression of ANGPTL4. In fact, we discovered that SGLT2i exert their effects by regulating the upstream transcription factor FOXO1 of ANGPTL4. Under conditions of hyperglycemia and hyperlipidemia, compared to the control group without FOXO1, the overexpression of FOXO1 in conjunction with SGLT2i intervention significantly reduced both ANGPTL4 mRNA and protein levels. This suggests that the FOXO1-ANGPTL4 axis may be a potential target for the cardioprotective effects of SGLT2i. CONCLUSIONS Collectively, our study demonstrates that SGLT2i ameliorate the pathological aging of cardiomyocytes induced by a high glucose and high fat metabolic milieu by regulating the interaction between FOXO1 and ANGPTL4, thereby suppressing the transcriptional synthesis of the latter, and consequently restoring cardiac function.
Collapse
Affiliation(s)
- Yun Wen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiaofang Zhang
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, China
| | - Han Liu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Haowen Ye
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ruxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Caixia Ma
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Tianqi Duo
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jiaxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xian Yang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Meixin Yu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Liangyan Wu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yongting Zhao
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lihong Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China.
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
3
|
Eilat-Tsanani S, Ernst P, Suissa S. Real-World Effectiveness of Single-Inhaler Triple Therapy for COPD: Impact of Diabetes Comorbidity. COPD 2024; 21:2327345. [PMID: 38509685 DOI: 10.1080/15412555.2024.2327345] [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: 02/05/2024] [Accepted: 03/01/2024] [Indexed: 03/22/2024]
Abstract
Type 2 diabetes is a frequent comorbidity in chronic obstructive pulmonary disease (COPD) patients, with the GOLD treatment recommendations asserting that the presence of diabetes be disregarded in the choice of treatment. In a cohort of COPD patients with frequent exacerbations, initiators of single-inhaler triple therapy or dual bronchodilators were compared on the incidence of COPD exacerbation and pneumonia over one year, adjusted by propensity score weighting and stratified by type 2 diabetes. The COPD cohort included 1,114 initiators of triple inhalers and 4,233 of dual bronchodilators (28% with type 2 diabetes). The adjusted hazard ratio (HR) of exacerbation with triple therapy was 1.04 (95% CI: 0.86-1.25) among COPD patients with type 2 diabetes and 0.74 (0.65-0.85) in those without. The incidence of severe pneumonia was elevated with triple therapy among patients with type 2 diabetes (HR 1.77; 1.14-2.75). Triple therapy in COPD is effective among those without, but not those with, type 2 diabetes. Future therapeutic trials in COPD should consider diabetes comorbidity.
Collapse
Affiliation(s)
- Sophia Eilat-Tsanani
- Department of Family Medicine, Clalit Health Services, North District, Israel
- Department of Family Medicine, Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
- Centre for Clinical Epidemiology, Lady Davis Institute-Jewish General Hospital, Montreal, Canada
| | - Pierre Ernst
- Centre for Clinical Epidemiology, Lady Davis Institute-Jewish General Hospital, Montreal, Canada
- Department of Epidemiology and Biostatistics, and of Medicine, McGill University, Montreal, Canada
| | - Samy Suissa
- Centre for Clinical Epidemiology, Lady Davis Institute-Jewish General Hospital, Montreal, Canada
- Department of Epidemiology and Biostatistics, and of Medicine, McGill University, Montreal, Canada
| |
Collapse
|
4
|
Golpe R, Figueira-Gonçalves JM, Arias-Zas L, Dacal-Rivas D, Blanco-Cid N, Castro-Añón O. Diabetes mellitus with poor glycemic control is a risk factor for pneumonia in COPD. Respir Med Res 2024; 86:101135. [PMID: 39191087 DOI: 10.1016/j.resmer.2024.101135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/29/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Pneumonias are events of great prognostic significance in COPD, so it is important to identify predictive factors. OBJECTIVE To determine whether poor glycemic control is related to an increased risk of pneumonia in COPD. METHOD A historical cohort study conducted in a COPD clinic. The first severe exacerbation after the first visit was analyzed. Exacerbations that presented with pulmonary infiltrates were identified. A Cox proportional hazards analysis was performed including the values of glycosylated hemoglobin (Hb1Ac) in patients with diabetes mellitus (DM) and variables that could plausibly be related to the risk of pneumonia. The best Hb1Ac value to predict pneumonia was assessed using receiver-operating characteristics analysis. RESULTS There were 1124 cases included in the study. A total of 411 patients were admitted to the hospital at least once and 87 were diagnosed with pneumonia. Variables associated with the risk of pneumonia were previous admissions due to COPD and Hb1Ac values (HR: 2.33, 95% CI: 1.06 - 5.08, p = 0.03). A higher body mass index (BMI) was associated with a lower risk of pneumonia. The optimal cutoff point for Hb1Ac to predict pneumonia risk was 7.8 %. The patients were classified into 3 groups: (1) no DM, (2) controlled DM (Hb1AC < 7.8 %), (3) uncontrolled DM (Hb1AC ≥ 7.8 %). The risk of pneumonia for group 2 was not different from group 1, while the risk for group 3 was significantly higher than for groups 1 and 2 (HR: 4.52, 95 % CI: 1.57 - 13.02). CONCLUSIONS Poor control of DM is a predictor of the risk of pneumonia in COPD. The cutoff point of 7.8 % for this variable seems to be the most useful to identify patients at risk.
Collapse
Affiliation(s)
- Rafael Golpe
- Servicio de Neumología. Hospital Universitario Lucus Augusti, Lugo, Spain.
| | - Juan-Marco Figueira-Gonçalves
- Servicio de Neumología y Cirugía Torácica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain; University Institute of Tropical Diseases and Public Health of the Canary Islands, University of La Laguna, Santa Cruz de Tenerife, Spain
| | - Laura Arias-Zas
- Servicio de Neumología. Hospital Universitario Lucus Augusti, Lugo, Spain
| | - David Dacal-Rivas
- Servicio de Neumología. Hospital Universitario Lucus Augusti, Lugo, Spain
| | - Nagore Blanco-Cid
- Servicio de Neumología. Hospital Universitario Lucus Augusti, Lugo, Spain
| | - Olalla Castro-Añón
- Servicio de Neumología. Hospital Universitario Lucus Augusti, Lugo, Spain
| |
Collapse
|
5
|
Krishnan JK, Martinez FJ, Altman P, Bilano VLF, Khokhlovich E, Przybysz R, Karcher H, Schoenberger M. Multimorbidities in COPD are Associated With Increased Exacerbations and Health Care Resource Utilization in Real-World Patients from a U.S. Database. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2024; 11:472-481. [PMID: 39133115 PMCID: PMC11548965 DOI: 10.15326/jcopdf.2024.0515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 08/13/2024]
Abstract
Background Patients with chronic obstructive pulmonary disease (COPD) often develop other morbidities, suggesting a systemic component to this disease. This retrospective noninterventional cohort study investigated relationships between multimorbidities in COPD and their impact on COPD exacerbations and COPD-related health care resource utilization (HCRU) using real-world evidence from Optum's de-identified Clinformatics® Data Mart Database. Methods Demographic and clinical characteristics were assessed. Overall comorbidity burden and proportion of individuals with gastroesophageal reflux disease (GERD), diabetes, or osteoporosis/osteopenia were compared in age-matched COPD versus non-COPD cohorts using descriptive statistics. COPD exacerbations and COPD-related HCRU (hospitalizations and emergency department visits) were compared between age-matched cohorts of COPD patients with and without specific common morbidities (GERD, diabetes, and osteoporosis/osteopenia). Additional weight-matching was performed for matched cohorts of COPD patients with and without diabetes, and with and without osteoporosis/osteopenia. The follow-up period was 5 years. Results Age-matched cohorts with and without COPD each comprised 158,106 patients. Morbidities were more common in the COPD cohort than the cohort without COPD (GERD: 44.9% versus 27.8%; diabetes: 40.8% versus 31.1%; osteoporosis/osteopenia: 18.8% versus 14.1%, respectively). Compared with matched cohorts with COPD only, cohorts of COPD patients with either GERD, diabetes, or osteoporosis/osteopenia experienced increased risk of severe exacerbations (odds ratio [OR]=1.819, OR=1.119, and OR=1.373, respectively), moderate exacerbations (OR=1.699, OR=1.102, and OR=1.322, respectively), or any exacerbations (OR=1.848, OR=1.099, and OR=1.384, respectively, p<0.001 for all comparisons) and increased risk of COPD-related HCRU (emergency department visits: OR=1.983, OR=1.098, and OR=1.343, respectively; hospitalization visits: OR=2.222, OR=1.26, and OR=1.368, respectively; p<0.001 for all comparisons). Conclusion These real-world data confirm that GERD, diabetes, and osteoporosis are common morbidities in patients with COPD and, moreover, that they affect frequency of exacerbation and HCRU. Determining and addressing the mechanisms behind the systemic effects of COPD may be beneficial for COPD patients and may also help reduce COPD exacerbations.
Collapse
Affiliation(s)
- Jamuna K. Krishnan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York City, New York, United States
| | - Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York City, New York, United States
| | - Pablo Altman
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, United States
- * at the time the study was completed
| | | | - Edward Khokhlovich
- Novartis Institutes for BioMedical Research, Inc, Cambridge, Massachusetts, United States
| | - Raymond Przybysz
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, United States
| | - Helene Karcher
- Novartis Pharma AG, Basel, Switzerland
- * at the time the study was completed
| | | |
Collapse
|
6
|
Liao WZ, Li JX, Feng WY, Xiao JQ, Wang ZX, Xie SJ, Hu YM, Mao JH, Huang ZM, Guo XG, Guan WJ. Association between coffee and caffeine intake and risk of COPD: Findings based on NHANES 2007-2012. Heart Lung 2024; 67:53-61. [PMID: 38701700 DOI: 10.1016/j.hrtlng.2024.04.015] [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: 12/17/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND The association between coffee and caffeine intake and the risk of COPD and lung function has not been thoroughly discussed in Americans, with subgroup and threshold effects remaining unclear. OBJECTIVES This study investigated the association between coffee and caffeine consumption and the risk of chronic obstructive pulmonary disease (COPD) as well as lung function utilizing data from the NHANES 2007-2012. METHODS We assessed the associations of coffee and caffeine consumption with the risk of COPD and lung function parameters, including FEV1 and FVC, adjusting for common demographic and disease characteristics in a cross-sectional analysis of NHANES data. RESULTS A total of 9763 participants were included in the study, and 592 were diagnosed with COPD. Multivariate regression models revealed positive associations between coffee and caffeine consumption and the risk of COPD and lung function. Subgroup analyses stratified by sex, DM, hypertension status, and smoking habits identified potential effect modifiers as well as inflection points from threshold effect examinations. CONCLUSIONS The results of this cross-sectional study indicated significant positive correlations between coffee and caffeine consumption and the risk of COPD. Additionally, positive correlations between exposure variables and FEV1 and FVC were detected. Among the stratification factors, smoking status exhibited the most potential for modifying effects. Future practices and research are needed to validate the results and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Wan-Zhe Liao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Jia-Xin Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wei-Yi Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jia-Qi Xiao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zi-Xun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China; Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shuo-Jia Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yi-Ming Hu
- Department of Electronic Information Engineering, Glasgow College, University of Electronic Science and Technology, Chengdu, 611731, China
| | - Jun-Hao Mao
- Department of Computer Science, Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Ze-Min Huang
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, King Med School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 510000, China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Wertman E. Essential New Complexity-Based Themes for Patient-Centered Diagnosis and Treatment of Dementia and Predementia in Older People: Multimorbidity and Multilevel Phenomenology. J Clin Med 2024; 13:4202. [PMID: 39064242 PMCID: PMC11277671 DOI: 10.3390/jcm13144202] [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: 06/10/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Dementia is a highly prevalent condition with devastating clinical and socioeconomic sequela. It is expected to triple in prevalence by 2050. No treatment is currently known to be effective. Symptomatic late-onset dementia and predementia (SLODP) affects 95% of patients with the syndrome. In contrast to trials of pharmacological prevention, no treatment is suggested to remediate or cure these symptomatic patients. SLODP but not young onset dementia is intensely associated with multimorbidity (MUM), including brain-perturbating conditions (BPCs). Recent studies showed that MUM/BPCs have a major role in the pathogenesis of SLODP. Fortunately, most MUM/BPCs are medically treatable, and thus, their treatment may modify and improve SLODP, relieving suffering and reducing its clinical and socioeconomic threats. Regrettably, the complex system features of SLODP impede the diagnosis and treatment of the potentially remediable conditions (PRCs) associated with them, mainly due to failure of pattern recognition and a flawed diagnostic workup. We suggest incorporating two SLODP-specific conceptual themes into the diagnostic workup: MUM/BPC and multilevel phenomenological themes. By doing so, we were able to improve the diagnostic accuracy of SLODP components and optimize detecting and favorably treating PRCs. These revolutionary concepts and their implications for remediability and other parameters are discussed in the paper.
Collapse
Affiliation(s)
- Eli Wertman
- Department of Neurology, Hadassah University Hospital, The Hebrew University, Jerusalem 9190500, Israel;
- Section of Neuropsychology, Department of Psychology, The Hebrew University, Jerusalem 9190500, Israel
- Or’ad: Organization for Cognitive and Behavioral Changes in the Elderly, Jerusalem 9458118, Israel
- Merhav Neuropsychogeriatric Clinics, Nehalim 4995000, Israel
| |
Collapse
|
8
|
Zhang L, Nishi H, Kinoshita K. Multi-Omics Profiling Reveals Phenotypic and Functional Heterogeneity of Neutrophils in COVID-19. Int J Mol Sci 2024; 25:3841. [PMID: 38612651 PMCID: PMC11011481 DOI: 10.3390/ijms25073841] [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: 03/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Accumulating evidence has revealed unexpected phenotypic heterogeneity and diverse functions of neutrophils in several diseases. Coronavirus disease (COVID-19) can alter the leukocyte phenotype based on disease severity, including neutrophil activation in severe cases. However, the plasticity of neutrophil phenotypes and their relative impact on COVID-19 pathogenesis has not been well addressed. This study aimed to identify and validate the heterogeneity of neutrophils in COVID-19 and evaluate the functions of each subpopulation. We analyzed public single-cell RNA-seq, bulk RNA-seq, and proteome data from healthy donors and patients with COVID-19 to investigate neutrophil subpopulations and their response to disease pathogenesis. We identified eight neutrophil subtypes: pro-neutrophil, pre-neutrophil, immature neutrophil, and five mature neutrophil subpopulations. The subtypes exhibited distinct features, including diverse activation signatures and multiple enriched pathways. The pro-neutrophil subtype was associated with severe and fatal disease, while the pre-neutrophil subtype was particularly abundant in mild/moderate disease. One of the mature neutrophil subtypes showed consistently large fractions in patients with different disease severity. Bulk RNA-seq dataset analyses using a cellular deconvolution approach validated the relative abundances of neutrophil subtypes and the expansion of pro-neutrophils in severe COVID-19 patients. Cell-cell communication analysis revealed representative ligand-receptor interactions among the identified neutrophil subtypes. Further investigation into transcription factors and differential protein abundance revealed the regulatory network differences between healthy donors and patients with severe COVID-19. Overall, we demonstrated the complex interactions among heterogeneous neutrophil subtypes and other blood cell types during COVID-19 disease. Our work has great value in terms of both clinical and public health as it furthers our understanding of the phenotypic and functional heterogeneity of neutrophils and other cell populations in multiple diseases.
Collapse
Affiliation(s)
- Lin Zhang
- Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University, Sendai 980-8579, Miyagi, Japan
| | - Hafumi Nishi
- Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University, Sendai 980-8579, Miyagi, Japan
- Faculty of Core Research, Ochanomizu University, Tokyo 112-8610, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University, Sendai 980-8579, Miyagi, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Miyagi, Japan
- Department of In Silico Analyses, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai 980-8575, Miyagi, Japan
| |
Collapse
|
9
|
Fabbri LM, Celli BR, Agustí A, Criner GJ, Dransfield MT, Divo M, Krishnan JK, Lahousse L, Montes de Oca M, Salvi SS, Stolz D, Vanfleteren LEGW, Vogelmeier CF. COPD and multimorbidity: recognising and addressing a syndemic occurrence. THE LANCET. RESPIRATORY MEDICINE 2023; 11:1020-1034. [PMID: 37696283 DOI: 10.1016/s2213-2600(23)00261-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 09/13/2023]
Abstract
Most patients with chronic obstructive pulmonary disease (COPD) have at least one additional, clinically relevant chronic disease. Those with the most severe airflow obstruction will die from respiratory failure, but most patients with COPD die from non-respiratory disorders, particularly cardiovascular diseases and cancer. As many chronic diseases have shared risk factors (eg, ageing, smoking, pollution, inactivity, and poverty), we argue that a shift from the current paradigm in which COPD is considered as a single disease with comorbidities, to one in which COPD is considered as part of a multimorbid state-with co-occurring diseases potentially sharing pathobiological mechanisms-is needed to advance disease prevention, diagnosis, and management. The term syndemics is used to describe the co-occurrence of diseases with shared mechanisms and risk factors, a novel concept that we propose helps to explain the clustering of certain morbidities in patients diagnosed with COPD. A syndemics approach to understanding COPD could have important clinical implications, in which the complex disease presentations in these patients are addressed through proactive diagnosis, assessment of severity, and integrated management of the COPD multimorbid state, with a patient-centred rather than a single-disease approach.
Collapse
Affiliation(s)
- Leonardo M Fabbri
- Section of Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Bartolome R Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alvar Agustí
- Cátedra Salud Respiratoria, Universitat de Barcelona, Barcelona, Spain; Institut Respiratori, Clínic Barcelona, Barcelona, Spain; Institut d'Investigacions Biomédicas August Pi i Sunyer, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Respiratorias, Spain
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Mark T Dransfield
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Miguel Divo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jamuna K Krishnan
- Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, USA
| | - Lies Lahousse
- Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Maria Montes de Oca
- School of Medicine, Universidad Central de Venezuela, Caracas, Venezuela; Hospital Centro Medico de Caracas, Caracas, Venezuela
| | - Sundeep S Salvi
- Pulmocare Research and Education (PURE) Foundation, Pune, India; School of Health Sciences, Symbiosis International Deemed University, Pune, India
| | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland; Clinic of Respiratory Medicine and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lowie E G W Vanfleteren
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claus F Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Centre Giessen and Marburg, Philipps University of Marburg, Member of the German Centre for Lung Research, Marburg, Germany.
| |
Collapse
|
10
|
Correa-Gutiérrez CA, Ji Z, Aragón-Espinosa P, Rodrigues-Oliveira S, Zeng L, Meizoso-Pita O, Sevillano-Collantes C, Hernández-Vázquez J, Puente-Maestu L, de Miguel-Díez J. Influence of Diabetes Mellitus and Nutritional Parameters on Clinical and Functional Aspects and Quality of Life in Patients Hospitalized Due to Exacerbation of Chronic Obstructive Pulmonary Disease. J Clin Med 2023; 12:6874. [PMID: 37959339 PMCID: PMC10650144 DOI: 10.3390/jcm12216874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/28/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) may experience exacerbations. During severe exacerbations, nutritional and endocrinological comorbidities can play an important role in the clinical and functional aspects of these patients. The aim of this study was to analyse the influence of the presence of diabetes mellitus (DM) and nutritional parameters on the deterioration of symptoms and quality of life during a severe exacerbation in patients with COPD. An observational study was conducted on COPD patients admitted due to an exacerbation. The COPD Assessment Test (CAT) questionnaire was administered, and clinical and functional parameters were compared based on the presence of nutritional and endocrinological alterations. A total of 50 patients were included, of whom 30 (60%) were male. The mean age was 70.5 years (standard deviation (SD) 9.6). The median CAT score during exacerbation was 25 (interquartile range (IQR) 17.5-30), and the baseline score was 13.5 (IQR 7-19), which represented a statistically significant difference (p < 0.001). Patients with iron deficiencies had a lower total CAT score (p = 0.041), specifically for items related to daily activity (p = 0.009) and energy (p = 0.007). Diabetic patients exhibited a greater decline in pulmonary function during exacerbation (p = 0.016), while patients with high thyroid-stimulating hormone (TSH) levels had a shorter hospital stay (p = 0.016). For COPD patients admitted due to an exacerbation, the metabolic assessment is useful and relevant in the clinical set-up, as endocrinological comorbidities negatively affect clinical and functional aspects of these patients.
Collapse
Affiliation(s)
- Cristhian Alonso Correa-Gutiérrez
- Respiratory Department, Gregorio Marañón General University Hospital, 28007 Madrid, Spain; (C.A.C.-G.); (L.P.-M.); (J.d.M.-D.)
- Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.A.-E.); (S.R.-O.)
| | - Zichen Ji
- Respiratory Department, Gregorio Marañón General University Hospital, 28007 Madrid, Spain; (C.A.C.-G.); (L.P.-M.); (J.d.M.-D.)
- Gregorio Marañón Biomedical Research Institute, 28007 Madrid, Spain
| | - Patricia Aragón-Espinosa
- Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.A.-E.); (S.R.-O.)
| | - Sarah Rodrigues-Oliveira
- Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.A.-E.); (S.R.-O.)
| | - Luyi Zeng
- Endocrinology and Nutrition Department, Infanta Leonor University Hospital, 28031 Madrid, Spain; (L.Z.); (O.M.-P.); (C.S.-C.)
| | - Olalla Meizoso-Pita
- Endocrinology and Nutrition Department, Infanta Leonor University Hospital, 28031 Madrid, Spain; (L.Z.); (O.M.-P.); (C.S.-C.)
| | - Cristina Sevillano-Collantes
- Endocrinology and Nutrition Department, Infanta Leonor University Hospital, 28031 Madrid, Spain; (L.Z.); (O.M.-P.); (C.S.-C.)
| | | | - Luis Puente-Maestu
- Respiratory Department, Gregorio Marañón General University Hospital, 28007 Madrid, Spain; (C.A.C.-G.); (L.P.-M.); (J.d.M.-D.)
- Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.A.-E.); (S.R.-O.)
- Gregorio Marañón Biomedical Research Institute, 28007 Madrid, Spain
| | - Javier de Miguel-Díez
- Respiratory Department, Gregorio Marañón General University Hospital, 28007 Madrid, Spain; (C.A.C.-G.); (L.P.-M.); (J.d.M.-D.)
- Faculty of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (P.A.-E.); (S.R.-O.)
- Gregorio Marañón Biomedical Research Institute, 28007 Madrid, Spain
| |
Collapse
|
11
|
Ye B, Zhou Y, Chen M, Chen C, Tan J, Xu X. The association between depression during perimenopause and progression of chronic conditions and multimorbidity: results from a Chinese prospective cohort. Arch Womens Ment Health 2023; 26:697-705. [PMID: 37550508 DOI: 10.1007/s00737-023-01354-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/21/2023] [Indexed: 08/09/2023]
Abstract
PURPOSE The association between perimenopausal depression and many chronic conditions among women has been well-established. However, the role of depression during perimenopause in the progression of multiple chronic conditions (multimorbidity) remains poorly understood. MATERIAL AND METHODS A total of 1,216 community-dwelling women in their perimenopause period between 2010 and 2016 were enrolled in our analysis, and followed up for the progression of multimorbidity. Depression, as well as its severity, was evaluated by the Center for Epidemiologic Studies Depression 10-item scale (CES-D-10). Progression of multimorbidity was defined as the first report of two or more chronic conditions for participants without multimorbidity or the new report of one or more conditions for those with multimorbidity. Univariable and multivariable Cox proportional hazards model and the restricted cubic spline regression model were performed to assess the prospective association between perimenopausal depression and the progression of multimorbidity. RESULTS A total of 480 (39.5%) women reported depression during perimenopause, and 529 (43.5%) women progressed to multimorbidity. After adjusting for socio-demographic and lifestyle factors, perimenopausal depression was independently associated with the progression of multimorbidity (hazard ratio [HR]: 1.34; 95% confidence interval [CI]: 1.13 to 1.60). Moreover, the severity of depression was positively and linearly associated with the progression of multimorbidity (P < 0.05). CONCLUSIONS Our finding reveals a prospective association between perimenopausal depression and the progression of multimorbidity, indicating interventions targeting perimenopausal depression may reduce the burden of chronic diseases and multimorbidity in women's post-menopausal life.
Collapse
Affiliation(s)
- Bingqi Ye
- School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China
| | - Yaguan Zhou
- School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China
| | - Mengsha Chen
- School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China
| | - Chen Chen
- School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China
| | - Jie Tan
- School of Public Health, Wuhan University, Wuhan, Hubei, China
- Global Health Research Center, Duke Kunshan University, Kunshan, Jiangsu, China
| | - Xiaolin Xu
- School of Public Health, The Second Affiliated Hospital, Zhejiang University School of Medicine, Yuhangtang Road 866, Hangzhou, 310058, Zhejiang, China.
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
- School of Public Health, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
12
|
Baliunas D, Voci S, Selby P, de Oliveira C, Kurdyak P, Rosella L, Zawertailo L, Fu L, Sutradhar R. Incidence of chronic disease following smoking cessation treatment: A matched cohort study using linked administrative healthcare data in Ontario, Canada. PLoS One 2023; 18:e0288759. [PMID: 37494345 PMCID: PMC10370896 DOI: 10.1371/journal.pone.0288759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/04/2023] [Indexed: 07/28/2023] Open
Abstract
Scarce evidence is available on the impact of real-world smoking cessation treatment on subsequent health outcomes, such as incidence of chronic disease. This study compared two cohorts of people that smoke-those that enrolled in a smoking cessation program, and a matched control that had not accessed the program-to assess the incidence of cancer, chronic obstructive pulmonary disease, diabetes, hypertension, and major cardiovascular events over a 5-year follow-up period. We selected five sub-cohorts with matched treatment-control pairs in which both individuals were at risk of the five chronic diseases. Incident chronic disease from index date until December 31, 2017, was determined through linkage with routinely collected healthcare data. The cumulative incidence of each chronic disease was estimated using the cumulative incidence function with death as a competing risk. Gray's test was used to test for a difference between matched treatment and control groups in the chronic disease-specific cumulative incidence function over follow-up. Analyses were stratified by sex. Among females, cumulative incidence of diabetes was higher over follow-up for the treatment group (5-year cumulative incidence 5.8% vs 4.2%, p = 0.004), but did not differ for the four other chronic diseases. Among males, cumulative incidence of chronic obstructive pulmonary disease (12.2% vs 9.1%, p < 0.001) and diabetes (6.7% vs 4.8%, p < 0.001) both had higher 5-year cumulative incidence for the treated versus control groups but did not differ for the other three chronic diseases. We conclude that accessing primary-care based smoking cessation treatment is associated with increased incidence of diabetes for both sexes, and chronic obstructive pulmonary disease for males (possibly due to under diagnosis prior to treatment), within 5 years of treatment. The associations detected require further research to understand causal relationships.
Collapse
Affiliation(s)
- Dolly Baliunas
- School of Health and Medical Sciences, University of Southern Queensland, Ipswich, Queensland, Australia
- Clinical Research - Addictions, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- School of Public Health, University of Queensland, Herston, Queensland, Australia
| | - Sabrina Voci
- Nicotine Dependence Service, INTREPID Lab, Addictions Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Peter Selby
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Nicotine Dependence Service, INTREPID Lab, Addictions Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Claire de Oliveira
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Centre for Health Economics and Hull York Medical School, University of York, Heslington, York, United Kingdom
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| | - Paul Kurdyak
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Laura Rosella
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| | - Laurie Zawertailo
- Nicotine Dependence Service, INTREPID Lab, Addictions Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute for Mental Health Policy Research, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | | | - Rinku Sutradhar
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Jung I, Han KD, Joon Moon S, Kwon H, Eun Park S, Rhee EJ, Lee WY. Association of Physical Activity with Total and Cause-specific Mortality in Patients with Diabetes: A Nationwide Population-based Cohort Study. Diabetes Res Clin Pract 2023:110819. [PMID: 37422165 DOI: 10.1016/j.diabres.2023.110819] [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: 04/20/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/10/2023]
Abstract
AIMS Physical inactivity is a modifiable risk factor for cardiovascular disease (CVD) in patients with type 2 diabetes mellitus (T2DM); however, little is known about its association with mortality due to other causes. Herein, we investigated the association between physical activity (PA) and cause-specific mortality in patients with T2DM. METHODS We analyzed data from the Korean National Health Insurance Service and claims database of adults with T2DM aged >20 years at baseline (n=2,651,214). Each participant's PA volume was measured as the metabolic equivalent of tasks (METs)-min per week, and hazard ratios of all-cause and cause-specific mortality relative to PA levels were estimated. RESULTS During the 7.8 years of follow-up, all-cause, CVD, respiratory, cancer, and other causes of mortality were lowest in patients engaged in vigorous PA. MET-min/week was inversely associated with mortality after adjusting for covariates. The reduction in total and cause-specific mortality was greater in patients aged ≥65 years than in those aged <65 years. CONCLUSIONS Increasing PA may facilitate a reduction in mortality from various causes, especially among older patients with T2DM. Clinicians should encourage such patients to increase their daily PA levels to reduce their risk of mortality.
Collapse
Affiliation(s)
- Inha Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Kyung-Do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Sun Joon Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyemi Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se Eun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Eun-Jung Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Won-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Fan H, Xiong Y, Huang Y, Xu C, Feng X, Li W, Yang Y, Hua R, Wang Z, Yuan Z, Zhou J. Lung function indices do not affect the incidence of coronary heart disease in patients with sleep-disordered breathing. Sleep Med 2023; 108:22-28. [PMID: 37307697 DOI: 10.1016/j.sleep.2023.06.005] [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: 04/11/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Currently, it's unclear whether sleep-disordered breathing (SDB) and reduced lung function could synergistically increase the incidence of coronary heart disease (CHD). Furthermore, the predictive value of different lung function indices for the incidence of CHD remains unknown. METHODS We enrolled 3749 participants from the Sleep Heart Health Study (SHHS) to conduct a retrospective study. The individuals were divided into the SDB and non-SDB subgroups according to Apnoea-Hypopnoea Index (AHI). Cox regression models were used to evaluate the association between lung function and CHD. We also conducted a ROC analysis to assess the predictive value of different lung function indices. RESULTS 512 cases of CHD were identified during an average of 10.40 years of follow-up in participants without CVD at baseline. We observed that lung function was a better predictor of CHD in non-SDB participants compared with SDB participants. Reduced lung function was associated with a higher risk of CHD in participants without SDB, while the inverse association became non-significant in participants with SDB. Furthermore, the incremental contribution of lung function to CHD diminished with increasing severity of SDB. CONCLUSION We need to focus more on the lung function of individuals without SDB rather than those with SDB to reduce the risk of CHD.
Collapse
Affiliation(s)
- Heze Fan
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Ying Xiong
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Yuzhi Huang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Chenbo Xu
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Xueying Feng
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Wenyuan Li
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Yuxuan Yang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Rui Hua
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Zihao Wang
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China
| | - Zuyi Yuan
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China.
| | - Juan Zhou
- Cardiovascular Department, First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, 710061, China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, 710061, China.
| |
Collapse
|
15
|
Marunaka Y. Molecular Mechanisms of Obesity-Induced Development of Insulin Resistance and Promotion of Amyloid-β Accumulation: Dietary Therapy Using Weak Organic Acids via Improvement of Lowered Interstitial Fluid pH. Biomolecules 2023; 13:biom13050779. [PMID: 37238649 DOI: 10.3390/biom13050779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/31/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Insulin resistance is one of the etiologies of type 2 diabetes mellitus (T2DM) and has been suggested to contribute to the development of Alzheimer's disease by promoting amyloid-β accumulation. Various causes of insulin resistance have been suggested; however, mechanisms of insulin resistance development remain to be elucidated in many respects. Elucidating the mechanisms underlying the development of insulin resistance is one of the key factors in developing methods to prevent the onset of T2DM and Alzheimer's disease. It has been suggested that the body pH environment plays an important role in the control of cellular functions by regulating the action of hormones including insulin and the activity of enzymes and neurons, thereby maintaining homeostatic conditions of the body. This review introduces: (1) Mitochondrial dysfunction through oxidative stress caused by obesity-induced inflammation. (2) Decreased pH of interstitial fluid due to mitochondrial dysfunction. (3) Development of insulin resistance due to diminution of insulin affinity to its receptor caused by the lowered interstitial fluid pH. (4) Accelerated accumulation of amyloid-β due to elevated activities of β- and γ-secretases caused by the lowered interstitial fluid pH. (5) Diet therapies for improving insulin resistance with weak organic acids that act as bases in the body to raise the pH of lowered interstitial fluid and food factors that promote absorption of weak organic acids in the gut.
Collapse
Affiliation(s)
- Yoshinori Marunaka
- Medical Research Institute, Kyoto Industrial Health Association, Kyoto 604-8472, Japan
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu 525-8577, Japan
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
16
|
Lipoxin and glycation in SREBP signaling: Insight into diabetic cardiomyopathy and associated lipotoxicity. Prostaglandins Other Lipid Mediat 2023; 164:106698. [PMID: 36379414 DOI: 10.1016/j.prostaglandins.2022.106698] [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: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
Abstract
Diabetes and cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Diabetes increases cardiovascular risk through hyperglycemia and atherosclerosis. Chronic hyperglycemia accelerates glycation reaction, which forms advanced glycation end products (AGEs). Additionally, hyperglycemia with enhanced levels of cholesterol, native and oxidized low-density lipoproteins, free fatty acids, and oxidative stress induces lipotoxicity. Accelerated glycation and disturbed lipid metabolism are characteristic features of diabetic heart failure. SREBP signaling plays a significant role in lipid and glucose homeostasis. AGEs increase lipotoxicity in diabetic cardiomyopathy by inhibiting SREBP signaling. While anti-inflammatory lipid mediators, lipoxins resolve inflammation caused by lipotoxicity by upregulating the PPARγ expression and regulating CD36. PPARγ connects the bridge between glycation and lipoxin in SREBP signaling. A summary of treatment modalities against diabetic cardiomyopathy is given in brief. This review indicates the novel therapeutic approach in the crosstalk between glycation and lipoxin in SREBP signaling.
Collapse
|
17
|
Serum Amyloid A in Stable Patients with Chronic Obstructive Pulmonary Disease Does Not Reflect the Clinical Course of the Disease. Int J Mol Sci 2023; 24:ijms24032478. [PMID: 36768801 PMCID: PMC9916457 DOI: 10.3390/ijms24032478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/09/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
Serum amyloid A (SAA) is a good systemic marker of the exacerbations of chronic obstructive pulmonary disease (COPD), but the significance of SAA in stable patients with COPD has not been widely investigated. We aimed to evaluate the SAA level in peripheral blood from stable patients with COPD and to search for correlations between SAA and other inflammatory markers and clinical characteristics of the disease. Serum SAA, IL-6, IL-8, TNF-alpha, basic blood investigations, pulmonary function testing and a 6-min walk test were performed. The correlations between SAA and other inflammatory markers, functional performance and the number of disease exacerbations were evaluated. A total of 100 consecutive patients with COPD were analyzed. No correlations between SAA and inflammatory markers as well as pulmonary function were found. Hierarchical clustering identified two clusters incorporating SAA: one comprised SAA, PaO2 and FEV1 and the second was formed of SAA and nine other disease markers. The SAA level was higher in patients with blood eosinophils < 2% when compared to those with blood eosinophils ≥ 2% (41.8 (19.5-69.7) ng/mL vs. 18.9 (1.0-54.5) ng/mL, respectively, p = 0.04). We conclude that, in combination with other important disease features, SAA may be useful for patient evaluation in stable COPD.
Collapse
|
18
|
Balk-Møller E, Hebsgaard MMB, Lilleør NB, Møller CH, Gøtze JP, Kissow H. Glucagon-like peptide-1 stimulates acute secretion of pro-atrial natriuretic peptide from the isolated, perfused pig lung exposed to warm ischemia. FRONTIERS IN TRANSPLANTATION 2022; 1:1082634. [PMID: 38994393 PMCID: PMC11235333 DOI: 10.3389/frtra.2022.1082634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/16/2022] [Indexed: 07/13/2024]
Abstract
Glucagon-like peptide-1 (GLP-1) has proven to be protective in animal models of lung disease but the underlying mechanisms are unclear. Atrial natriuretic peptide (ANP) is mainly produced in the heart. As ANP possesses potent vaso- and bronchodilatory effects in pulmonary disease, we hypothesised that the protective functions of GLP-1 could involve potentiation of local ANP secretion from the lung. We examined whether the GLP-1 receptor agonist liraglutide was able to improve oxygenation in lungs exposed to 2 h of warm ischemia and if liraglutide stimulated ANP secretion from the lungs in the porcine ex vivo lung perfusion (EVLP) model. Pigs were given a bolus of 40 µg/kg liraglutide or saline 1 h prior to sacrifice. The lungs were then left in vivo for 2 h, removed en bloc and placed in the EVLP machinery. Lungs from the liraglutide treated group were further exposed to liraglutide in the perfusion buffer (1.125 mg). Main endpoints were oxygenation capacity, and plasma and perfusate concentrations of proANP and inflammatory markers. Lung oxygenation capacity, plasma concentrations of proANP or concentrations of inflammatory markers were not different between groups. ProANP secretion from the isolated perfused lungs were markedly higher in the liraglutide treated group (area under curve for the first 30 min in the liraglutide group: 635 ± 237 vs. 38 ± 38 pmol/L x min in the saline group) (p < 0.05). From these results, we concluded that liraglutide potentiated local ANP secretion from the lungs.
Collapse
Affiliation(s)
- Emilie Balk-Møller
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mathilde M. B. Hebsgaard
- Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nikolaj B. Lilleør
- Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian H. Møller
- Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Jens P. Gøtze
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hannelouise Kissow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Yang X, Gao C, Liu Y, Zhu L, Yang K. Simplified Cell Magnetic Isolation Assisted SC 2 Chip to Realize "Sample in and Chemotaxis Out": Validated by Healthy and T2DM Patients' Neutrophils. MICROMACHINES 2022; 13:1820. [PMID: 36363840 PMCID: PMC9692824 DOI: 10.3390/mi13111820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Neutrophil migration in tissues critically regulates the human immune response and can either play a protective role in host defense or cause health problems. Microfluidic chips are increasingly applied to study neutrophil migration, attributing to their advantages of low reagent consumption, stable chemical gradients, visualized cell chemotaxis monitoring, and quantification. Most chemotaxis chips suffered from low throughput and fussy cell separation operations. We here reported a novel and simple "sample in and chemotaxis out" method for rapid neutrophils isolation from a small amount of whole blood based on a simplified magnetic method, followed by a chemotaxis assay on a microfluidic chip (SC2 chip) consisting of six cell migration units and six-cell arrangement areas. The advantages of the "sample in and chemotaxis out" method included: less reagent consumption (10 μL of blood + 1 μL of magnetic beads + 1 μL of lysis buffer); less time (5 min of cell isolation + 15 min of chemotaxis testing); no ultracentrifugation; more convenient; higher efficiency; high throughput. We have successfully validated the approach by measuring neutrophil chemotaxis to frequently-used chemoattractant (i.e., fMLP). The effects of D-glucose and mannitol on neutrophil chemotaxis were also analyzed. In addition, we demonstrated the effectiveness of this approach for testing clinical samples from diabetes mellitus type 2 (T2DM) patients. We found neutrophils' migration speed was higher in the "well-control" T2DM than in the "poor-control" group. Pearson coefficient analysis further showed that the migration speed of T2DM was negatively correlated with physiological indicators, such as HbA1c (-0.44), triglyceride (-0.36), C-reactive protein (-0.28), and total cholesterol (-0.28). We are very confident that the developed "sample in and chemotaxis out" method was hoped to be an attractive model for analyzing the chemotaxis of healthy and disease-associated neutrophils.
Collapse
Affiliation(s)
- Xiao Yang
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Chaoru Gao
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- School of Biomedical Engineering, Anhui Medical University, Hefei 230032, China
| | - Yong Liu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Ling Zhu
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Ke Yang
- Anhui Institute of Optics and Fine Mechanics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
20
|
Kingston A, Wittenberg R, Hu B, Jagger C. Projections of dependency and associated social care expenditure for the older population in England to 2038: effect of varying disability progression. Age Ageing 2022; 51:6649132. [PMID: 35871421 PMCID: PMC9308990 DOI: 10.1093/ageing/afac158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives to assess the effect of recent stalling of life expectancy and various scenarios for disability progression on projections of social care expenditure between 2018 and 2038, and the likelihood of reaching the Ageing Society Grand Challenge mission of five extra healthy, independent years at birth. Design two linked projections models: the Population Ageing and Care Simulation (PACSim) model and the Care Policy and Evaluation Centre long-term care projections model, updated to include 2018-based population projections. Population PACSim: about 303,589 individuals aged 35 years and over (a 1% random sample of the England population in 2014) created from three nationally representative longitudinal ageing studies. Main outcome measures Total social care expenditure (public and private) for older people, and men and women’s independent life expectancy at age 65 (IndLE65) under five scenarios of changing disability progression and recovery with and without lower life expectancy. Results between 2018 and 2038, total care expenditure was projected to increase by 94.1%–1.25% of GDP; men’s IndLE65 increasing by 14.7% (range 11.3–16.5%), exceeding the 8% equivalent of the increase in five healthy, independent years at birth, although women’s IndLE65 increased by only 4.7% (range 3.2–5.8%). A 10% reduction in disability progression and increase in recovery resulted in the lowest increase in total care expenditure and increases in both men’s and women’s IndLE65 exceeding 8%. Conclusions interventions that slow down disability progression, and improve recovery, could significantly reduce social care expenditure and meet government targets for increases in healthy, independent years.
Collapse
Affiliation(s)
- Andrew Kingston
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Raphael Wittenberg
- Care Policy and Evaluation Centre (CPEC), London School of Economics and Political Science, London, UK
| | - Bo Hu
- Care Policy and Evaluation Centre (CPEC), London School of Economics and Political Science, London, UK
| | - Carol Jagger
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
21
|
Allen CNS, Santerre M, Arjona SP, Ghaleb LJ, Herzi M, Llewellyn MD, Shcherbik N, Sawaya BE. SARS-CoV-2 Causes Lung Inflammation through Metabolic Reprogramming and RAGE. Viruses 2022; 14:983. [PMID: 35632725 PMCID: PMC9143006 DOI: 10.3390/v14050983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 12/26/2022] Open
Abstract
Clinical studies indicate that patients infected with SARS-CoV-2 develop hyperinflammation, which correlates with increased mortality. The SARS-CoV-2/COVID-19-dependent inflammation is thought to occur via increased cytokine production and hyperactivity of RAGE in several cell types, a phenomenon observed for other disorders and diseases. Metabolic reprogramming has been shown to contribute to inflammation and is considered a hallmark of cancer, neurodegenerative diseases, and viral infections. Malfunctioning glycolysis, which normally aims to convert glucose into pyruvate, leads to the accumulation of advanced glycation end products (AGEs). Being aberrantly generated, AGEs then bind to their receptor, RAGE, and activate several pro-inflammatory genes, such as IL-1b and IL-6, thus, increasing hypoxia and inducing senescence. Using the lung epithelial cell (BEAS-2B) line, we demonstrated that SARS-CoV-2 proteins reprogram the cellular metabolism and increase pyruvate kinase muscle isoform 2 (PKM2). This deregulation promotes the accumulation of AGEs and senescence induction. We showed the ability of the PKM2 stabilizer, Tepp-46, to reverse the observed glycolysis changes/alterations and restore this essential metabolic process.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Lea J. Ghaleb
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Muna Herzi
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Megan D. Llewellyn
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Natalia Shcherbik
- Department for Cell Biology and Neuroscience, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA;
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
- Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
22
|
Petchakup C, Yang H, Gong L, He L, Tay HM, Dalan R, Chung AJ, Li KHH, Hou HW. Microfluidic Impedance-Deformability Cytometry for Label-Free Single Neutrophil Mechanophenotyping. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104822. [PMID: 35253966 DOI: 10.1002/smll.202104822] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/03/2022] [Indexed: 06/14/2023]
Abstract
The intrinsic biophysical states of neutrophils are associated with immune dysfunctions in diseases. While advanced image-based biophysical flow cytometers can probe cell deformability at high throughput, it is nontrivial to couple different sensing modalities (e.g., electrical) to measure other critical cell attributes including cell viability and membrane integrity. Herein, an "optics-free" impedance-deformability cytometer for multiparametric single cell mechanophenotyping is reported. The microfluidic platform integrates hydrodynamic cell pinching, and multifrequency impedance quantification of cell size, deformability, and membrane impedance (indicative of cell viability and activation). A newly-defined "electrical deformability index" is validated by numerical simulations, and shows strong correlations with the optical cell deformability index of HL-60 experimentally. Human neutrophils treated with various biochemical stimul are further profiled, and distinct differences in multimodal impedance signatures and UMAP analysis are observed. Overall, the integrated cytometer enables label-free cell profiling at throughput of >1000 cells min-1 without any antibodies labeling to facilitate clinical diagnostics.
Collapse
Affiliation(s)
- Chayakorn Petchakup
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Haoning Yang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lingyan Gong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Linwei He
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Hui Min Tay
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Rinkoo Dalan
- Endocrinology Department, Tan Tock Seng Hospital, 11 Jln Tan Tock Seng Road, Singapore, 308433, Singapore
| | - Aram J Chung
- School of Biomedical Engineering, Korea University, Seoul, 02841, Republic of Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul, 02841, Republic of Korea
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Clinical Sciences Building Level 11, Singapore, 308232, Singapore
| |
Collapse
|
23
|
Pournaras N, Andersson A, Kovach MA, Padra M, Che KF, Brundin B, Yoshihara S, Bozinovski S, Lindén SK, Jansson PA, Sköld MC, Qvarfordt I, Lindén A. Glucose Homeostasis in Relation to Neutrophil Mobilization in Smokers with COPD. Int J Chron Obstruct Pulmon Dis 2022; 17:1179-1194. [PMID: 35620349 PMCID: PMC9129100 DOI: 10.2147/copd.s353753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/03/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Nikolaos Pournaras
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
- Correspondence: Nikolaos Pournaras, Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden and Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden, Email
| | - Anders Andersson
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
- COPD Center, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Melissa A Kovach
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Médea Padra
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karlhans F Che
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Bettina Brundin
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shigemi Yoshihara
- Pediatric Allergology and Respiratory Medicine, Department of Pediatrics, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Sara K Lindén
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per-Anders Jansson
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus C Sköld
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
- Division for Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ingemar Qvarfordt
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Lindén
- Division for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
24
|
Jurevičienė E, Burneikaitė G, Dambrauskas L, Kasiulevičius V, Kazėnaitė E, Navickas R, Puronaitė R, Smailytė G, Visockienė Ž, Danila E. Epidemiology of Chronic Obstructive Pulmonary Disease (COPD) Comorbidities in Lithuanian National Database: A Cluster Analysis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:970. [PMID: 35055792 PMCID: PMC8775709 DOI: 10.3390/ijerph19020970] [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] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 02/04/2023]
Abstract
Various comorbidities and multimorbidity frequently occur in chronic obstructive pulmonary disease (COPD), leading to the overload of health care systems and increased mortality. We aimed to assess the impact of COPD on the probability and clustering of comorbidities. The cross-sectional analysis of the nationwide Lithuanian database was performed based on the entries of the codes of chronic diseases. COPD was defined on the code J44.8 entry and six-month consumption of bronchodilators. Descriptive statistics and odds ratios (ORs) for associations and agglomerative hierarchical clustering were carried out. 321,297 patients aged 40-79 years were included; 4834 of them had COPD. A significantly higher prevalence of cardiovascular diseases (CVD), lung cancer, kidney diseases, and the association of COPD with six-fold higher odds of lung cancer (OR 6.66; p < 0.0001), a two-fold of heart failure (OR 2.61; p < 0.0001), and CVD (OR 1.83; p < 0.0001) was found. Six clusters in COPD males and five in females were pointed out, in patients without COPD-five and four clusters accordingly. The most prevalent cardiovascular cluster had no significant difference according to sex or COPD presence, but a different linkage of dyslipidemia was found. The study raises the need to elaborate adjusted multimorbidity case management and screening tools enabling better outcomes.
Collapse
Affiliation(s)
- Elena Jurevičienė
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| | - Greta Burneikaitė
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| | - Laimis Dambrauskas
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| | - Vytautas Kasiulevičius
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
| | - Edita Kazėnaitė
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| | - Rokas Navickas
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| | - Roma Puronaitė
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
- Faculty of Mathematics and Informatics, Institute of Data Science and Digital Technologies, Vilnius University, Naugarduko g. 24, LT-03225 Vilnius, Lithuania
| | - Giedrė Smailytė
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
| | - Žydrūnė Visockienė
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| | - Edvardas Danila
- Faculty of Medicine, Vilnius University, Čiurlionio Str. 21, LT-03101 Vilnius, Lithuania; (G.B.); (L.D.); (V.K.); (E.K.); (R.N.); (R.P.); (G.S.); (Ž.V.); (E.D.)
- Vilnius University Hospital, Santaros Klinikos, Santariškių Str. 2, LT-08661 Vilnius, Lithuania
| |
Collapse
|
25
|
Henson SM, Aksentijevic D. Senescence and Type 2 Diabetic Cardiomyopathy: How Young Can You Die of Old Age? Front Pharmacol 2021; 12:716517. [PMID: 34690759 PMCID: PMC8529062 DOI: 10.3389/fphar.2021.716517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Inflammation is well understood to be a physiological process of ageing however it also underlies many chronic diseases, including conditions without an obvious pathogenic inflammatory element. Recent findings have unequivocally identified type 2 diabetes (T2D) as a chronic inflammatory disease characterized by inflammation and immune senescence. Immunosenescence is a hallmark of the prolonged low-grade systemic inflammation, in particular associated with metabolic syndrome and can be a cause as well as a consequence of T2D. Diabetes is a risk factor for cardiovascular mortality and remodelling and with particular changes to myocardial structure, function, metabolism and energetics collectively resulting in diabetic cardiomyopathy. Both cardiomyocytes and immune cells undergo metabolic remodelling in T2D and as a result become trapped in a vicious cycle of lost metabolic flexibility, thus losing their key adaptive mechanisms to dynamic changes in O2 and nutrient availability. Immunosenescence driven by metabolic stress may be both the cause and key contributing factor to cardiac dysfunction in diabetic cardiomyopathy by inducing metabolic perturbations that can lead to impaired energetics, a strong predictor of cardiac mortality. Here we review our current understanding of the cross-talk between inflammaging and cardiomyocytes in T2D cardiomyopathy. We discuss potential mechanisms of metabolic convergence between cell types which, we hypothesize, might tip the balance between resolution of the inflammation versus adverse cardiac metabolic remodelling in T2D cardiomyopathy. A better understanding of the multiple biological paradigms leading to T2D cardiomyopathy including the immunosenescence associated with inflammaging will provide a powerful target for successful therapeutic interventions.
Collapse
Affiliation(s)
- Sian M Henson
- Centre for Translational Medicine and Therapeutics, London, United Kingdom
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
26
|
Zykov MV, Barbarash OL. Inflammation and Comorbidity. Are There any Chances to Improve the Prognosis in Patients with Extremely High Cardiovascular Risk? RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2021. [DOI: 10.20996/1819-6446-2021-08-06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The review contains actual data on possible approaches aimed at improving the prognosis in a special category of patients with extremely high cardiovascular risk, as well as in patients with recent acute coronary syndrome combined with comorbidity, including multifocal atherosclerosis. Currently, there are no class I recommendations for such patients aimed at reducing the risk of adverse cardiovascular events. It is suggested that suppression of inflammation may be a new therapeutic goal in this category of patients. Given the importance of inflammation in the development and course of atherosclerosis, in recent years there have been repeated attempts to influence the various components of the pro-inflammatory cascade involved in atherogenesis, but not all of them have been successful. Special attention is given to the anti-inflammatory effects of colchicine, a drug that can improve cardiovascular outcomes in patients with proven atherosclerosis. The review provides numerous pathogenetic and clinical evidence for the effectiveness of colchicine in patients with various manifestations of atherosclerosis. It is concluded that colchicine is the most promising anti-inflammatory drug that can improve the outcome of cardiovascular diseases. Thus, there is a need to initiate new clinical trial protocols aimed at studying the anti-inflammatory potential of this drug in patients with extreme cardiovascular risk.
Collapse
Affiliation(s)
- M. V. Zykov
- Research Institute for Complex Issues of Cardiovascular Diseases
| | - O. L. Barbarash
- Research Institute for Complex Issues of Cardiovascular Diseases
| |
Collapse
|
27
|
A Novel Microfluidic Device for the Neutrophil Functional Phenotype Analysis: Effects of Glucose and Its Derivatives AGEs. MICROMACHINES 2021; 12:mi12080944. [PMID: 34442566 PMCID: PMC8399494 DOI: 10.3390/mi12080944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 11/17/2022]
Abstract
Neutrophil dysfunction is closely related to the pathophysiology of patients with diabetes mellitus, but existing immunoassays are difficult to implement in clinical applications, and neutrophil’s chemotaxis as a functional biomarker for diabetes mellitus prognostic remains largely unexplored. Herein, a novel microfluidic device consisted of four independent test units with four cell docking structures was developed to study the neutrophil chemotaxis, which allowed multiple cell migration observations under a single field of view (FOV) and guaranteed more reliable results. In vitro studies, the chemotaxis of healthy neutrophils to N-Formyl-Met-Leu-Phe (fMLP) gradient (0, 10, 100, and 1000 nM) was concentration-dependent. The distinct promotion or suppression in the chemotaxis of metformin or pravastatin pretreated cells were observed after exposure to 100 nM fMLP gradient, indicating the feasibility and efficiency of this novel microfluidic device for clinically relevant evaluation of neutrophil functional phenotype. Further, the chemotaxis of neutrophils pretreated with 25, 50, or 70 mM of glucose was quantitatively lower than that of the control groups (i.e., 5 mM normal serum level). Neutrophils exposed to highly concentrated advanced glycation end products (AGEs) (0.2, 0.5, or 1.0 μM; 0.13 μM normal serum AGEs level), a product of prolonged hyperglycemia, showed that the higher the AGEs concentration was, the weaker the migration speed became. Specifically, neutrophils exposed to high concentrations of glucose or AGEs also showed a stronger drifting along with the flow, further demonstrating the change of neutrophil chemotaxis. Interestingly, adding the N-benzyl-4-chloro-N-cyclohexylbenzamide (FPS-ZM1) (i.e., high-affinity RAGE inhibitor) into the migration medium with AGEs could hinder the binding between AGEs and AGE receptor (RAGE) located on the neutrophil, thereby keeping the normal chemotaxis of neutrophils than the ones incubated with AGEs alone. These results revealed the negative effects of high concentrations of glucose and AGEs on the neutrophil chemotaxis, suggesting that patients with diabetes should manage serum AGEs and also pay attention to blood glucose indexes. Overall, this novel microfluidic device could significantly characterize the chemotaxis of neutrophils and have the potential to be further improved into a tool for risk stratification of diabetes mellitus.
Collapse
|
28
|
Kotlyarov S, Kotlyarova A. The Role of ABC Transporters in Lipid Metabolism and the Comorbid Course of Chronic Obstructive Pulmonary Disease and Atherosclerosis. Int J Mol Sci 2021; 22:6711. [PMID: 34201488 PMCID: PMC8269124 DOI: 10.3390/ijms22136711] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/12/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) ranks among the leading causes of morbidity and mortality worldwide. COPD rarely occurs in isolation and is often combined with various diseases. It is considered that systemic inflammation underlies the comorbid course of COPD. The data obtained in recent years have shown the importance of violations of the cross-links of lipid metabolism and the immune response, which are links in the pathogenesis of both COPD and atherosclerosis. The role of lipid metabolism disorders in the pathogenesis of the comorbid course of COPD and atherosclerosis and the participation of ATP-binding cassette (ABC) transporters in these processes is discussed in this article. It is known that about 20 representatives of a large family of ABC transporters provide lipid homeostasis of cells by moving lipids inside the cell and in its plasma membrane, as well as removing lipids from the cell. It was shown that some representatives of the ABC-transporter family are involved in various links of the pathogenesis of COPD and atherosclerosis, which can determine their comorbid course.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
29
|
The dynamicity of acute ozone-induced systemic leukocyte trafficking and adrenal-derived stress hormones. Toxicology 2021; 458:152823. [PMID: 34051339 DOI: 10.1016/j.tox.2021.152823] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/06/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022]
Abstract
Ozone exposure induces neuroendocrine stress response, which causes lymphopenia. It was hypothesized that ozone-induced increases in stress hormones will temporally follow changes in circulating granulocytes, monocytes- and lymphocyte subpopulations. The goal of this study was to chronicle the changes in circulating stress hormones, cytokines, and leukocyte trafficking during 4 h exposure to ozone. Male Wistar Kyoto rats were exposed to air or ozone (0.4 or 0.8 ppm) for 0.5, 1, 2, or 4 h. After each time point, circulating stress hormones, cytokines, and lung gene expression were assessed along with live and apoptotic granulocytes, monocytes (classical and non-classical), and lymphocytes (B, Th, and Tc) in blood, thymus, and spleen using flow cytometry. Circulating stress hormones began to increase at 1 h of ozone exposure. Lung expression of inflammatory cytokines (Cxcl2, Il6, and Hmox1) and glucocorticoid-responsive genes (Nr3c1, Fkbp5 and Tsc22d3) increased in both a time- and ozone concentration-dependent manner. Circulating granulocytes increased at 0.5 h of ozone exposure but tended to decrease at 2 and 4 h, suggesting a rapid egress and then margination to the lung. Classical monocytes decreased over 4 h of exposure periods (∼80 % at 0.8 ppm). B and Tc lymphocytes significantly decreased after ozone exposure at 2 and 4 h. Despite dynamic shifts in circulating immune cell populations, few differences were measured in serum cytokines. Ozone neither increased apoptotic cells nor altered thymus and spleen lymphocytes. The data show that ozone-induced increases in adrenal-derived stress hormones precede the dynamic migration of circulating immune cells, likely to the lung to mediate inflammation.
Collapse
|
30
|
Gum Arabic modifies anti-inflammatory cytokine in mice fed with high fat diet induced obesity. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.bcdf.2020.100258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
31
|
Press VG, Myers LC, Feemster LC. Preventing COPD Readmissions Under the Hospital Readmissions Reduction Program: How Far Have We Come? Chest 2021; 159:996-1006. [PMID: 33065106 PMCID: PMC8501005 DOI: 10.1016/j.chest.2020.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/10/2020] [Accepted: 10/01/2020] [Indexed: 01/06/2023] Open
Abstract
The Hospital Readmissions Reduction Program (HRRP) was developed and implemented by the Centers for Medicare & Medicaid Services to curb the rate of 30-day hospital readmissions for certain common, high-impact conditions. In October 2014, COPD became a target condition for which hospitals were penalized for excess readmissions. The appropriateness, utility, and potential unintended consequences of the metric have been a topic of debate since it was first enacted. Nevertheless, there is evidence that hospital policies broadly implemented in response to the HRRP may have been responsible for reducing the rate of readmissions following COPD hospitalizations even before it was added as a target condition. Since the addition of the COPD condition to the HRRP, several predictive models have been developed to predict COPD survival and readmissions, with the intention of identifying modifiable risk factors. A number of interventions have also been studied, with mixed results. Bundled care interventions using the electronic health record and patient education interventions for inhaler education have been shown to reduce readmissions, whereas pulmonary rehabilitation, follow-up visits, and self-management programs have not been consistently shown to do the same. Through this program, COPD has become recognized as a public health priority. However, 5 years after COPD became a target condition for HRRP, there continues to be no single intervention that reliably prevents readmissions in this patient population. Further research is needed to understand the long-term effects of the policy, the role of competing risks in measuring quality, the optimal postdischarge care for patients with COPD, and the integrated use of predictive modeling and advanced technologies to prevent COPD readmissions.
Collapse
Affiliation(s)
- Valerie G Press
- Section of General Internal Medicine University of Chicago Medicine.
| | - Laura C Myers
- Divisions of Research and Pulmonary/Critical Care Medicine, Kaiser Permanente Northern California
| | - Laura C Feemster
- Division of Pulmonary, Critical Care, and Sleep Medicine, VA Puget Sound Health Care System
| |
Collapse
|
32
|
Bayani A, Dunster JL, Crofts JJ, Nelson MR. Spatial considerations in the resolution of inflammation: Elucidating leukocyte interactions via an experimentally-calibrated agent-based model. PLoS Comput Biol 2020; 16:e1008413. [PMID: 33137107 PMCID: PMC7660912 DOI: 10.1371/journal.pcbi.1008413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 11/12/2020] [Accepted: 10/01/2020] [Indexed: 01/13/2023] Open
Abstract
Many common medical conditions (such as cancer, arthritis, chronic obstructive pulmonary disease (COPD), and others) are associated with inflammation, and even more so when combined with the effects of ageing and multimorbidity. While the inflammatory response varies in different tissue types, under disease and in response to therapeutic interventions, it has common interactions that occur between immune cells and inflammatory mediators. Understanding these underlying inflammatory mechanisms is key in progressing treatments and therapies for numerous inflammatory conditions. It is now considered that constituent mechanisms of the inflammatory response can be actively manipulated in order to drive resolution of inflammatory damage; particularly, those mechanisms related to the pro-inflammatory role of neutrophils and the anti-inflammatory role of macrophages. In this article, we describe the assembly of a hybrid mathematical model in which the spatial spread of inflammatory mediators is described through partial differential equations, and immune cells (neutrophils and macrophages) are described individually via an agent-based modelling approach. We pay close attention to how immune cells chemotax toward pro-inflammatory mediators, presenting a model for cell chemotaxis that is calibrated against experimentally observed cell trajectories in healthy and COPD-affected scenarios. We illustrate how variations in key model parameters can drive the switch from resolution of inflammation to chronic outcomes, and show that aberrant neutrophil chemotaxis can move an otherwise healthy outcome to one of chronicity. Finally, we reflect on our results in the context of the on-going hunt for new therapeutic interventions. Inflammation is the body’s primary defence to harmful stimuli such as infections, toxins and tissue strain but also underlies a much broader range of conditions, including asthma, arthritis and cancer. The inflammatory response is key in resolving injury to facilitate recovery, and involves a range of interactions between immune cells (leukocytes, neutrophils and macrophages in particular) and inflammatory mediators. Immune cells are recruited from the blood stream in response to injury. Once in tissue, neutrophils release toxins to kill invading agents and resolve damage; however, if not carefully managed by other immune cells (mainly macrophages), their responses can increase inflammation instead of helping to resolve it. We model these interactions in response to damage using a spatial model, examining how a healthy response can prevent localised inflammation from spreading. We pay close attention to how cells migrate toward the damaged area, as many inflammatory conditions are associated with impairment of this process. We calibrate our model against experimentally-observed cell trajectories from healthy patients and patients with chronic obstructive pulmonary disease. We illustrate that a healthy outcome depends strongly upon efficient cell migration and a delicate balance between the pro- and anti-inflammatory effects of neutrophils and macrophages.
Collapse
Affiliation(s)
- Anahita Bayani
- Department of Physics & Mathematics, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, United Kingdom
| | - Joanne L. Dunster
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, RG6 6AS, United Kingdom
| | - Jonathan J. Crofts
- Department of Physics & Mathematics, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, United Kingdom
| | - Martin R. Nelson
- Department of Physics & Mathematics, Nottingham Trent University, Clifton Campus, Nottingham, NG11 8NS, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Isidori AM, Giannetta E, Pofi R, Venneri MA, Gianfrilli D, Campolo F, Mastroianni CM, Lenzi A, d'Ettorre G. Targeting the NO-cGMP-PDE5 pathway in COVID-19 infection. The DEDALO project. Andrology 2020; 9:33-38. [PMID: 32526061 PMCID: PMC7307129 DOI: 10.1111/andr.12837] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/08/2023]
Abstract
Background A pandemic outbreak of COVID‐19 has been sweeping the world since December. It begins as a respiratory infection that, mainly in men with diabetes or renal impairment, evolves into a systemic disease, with SARDS, progressive endothelial cell damage, abnormal clotting and impaired cardiovascular and liver function. Some clinical trials are testing biological drugs to limit the immune system dysregulation, “cytokines storm,” that causes the systemic complications of COVID‐19. The contraindications of these drugs and their cost raise concerns over the implications of their widespread availability. Objectives Numerous clinical and experimental studies have revealed a role for the nitric oxide (NO)‐cyclic GMP‐phosphodiesterase type 5 (PDE5) pathway in modulating low‐grade inflammation in patients with metabolic diseases, offering cardiovascular protection. PDE5 inhibition favors an anti‐inflammatory response by modulating activated T cells, reducing cytokine release, lowering fibrosis, increasing oxygen diffusion, stimulating vascular repair. PDE5 is highly expressed in the lungs, where its inhibition improves pulmonary fibrosis, a complication of severe COVID‐19 disease. Materials and methods We performed a systematic review of all evidence documenting any involvement of the NO‐cGMP‐PDE5 axis in the pathophysiology of COVID‐19, presenting the ongoing clinical trials aimed at modulating this axis, including our own “silDEnafil administration in DiAbetic and dysmetaboLic patients with COVID‐19 (DEDALO trial).” Results The reviewed evidence suggests that PDE5 inhibitors could offer a new strategy in managing COVID‐19 by (i) counteracting the Ang‐II‐mediated downregulation of AT‐1 receptor; (ii) acting on monocyte switching, thus reducing pro‐inflammatory cytokines, interstitial infiltration and the vessel damage responsible for alveolar hemorrhage‐necrosis; (iii) inhibiting the transition of endothelial and smooth muscle cells to mesenchymal cells in the pulmonary artery, preventing clotting and thrombotic complications. Discussion and Conclusion If the ongoing trials presented herein should provide positive findings, the low cost, wide availability and temperature stability of PDE5 inhibitors could make them a major resource to combat COVID‐19 in developing countries.
Collapse
Affiliation(s)
- Andrea M Isidori
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Mary A Venneri
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Claudio M Mastroianni
- Department of Public Health and Infectious Diseases, "Sapienza" University of Rome, Policlinico Umberto I of Rome, Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza" University of Rome, Rome, Italy
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, "Sapienza" University of Rome, Policlinico Umberto I of Rome, Rome, Italy
| |
Collapse
|