201
|
Martinez-Garcia MÁ, Faner R, Oscullo G, la Rosa-Carrillo D, Soler-Cataluña JJ, Ballester M, Muriel A, Agusti A. Chronic bronchial infection and incident cardiovascular events in chronic obstructive pulmonary disease patients: A long-term observational study. Respirology 2021; 26:776-785. [PMID: 34002922 DOI: 10.1111/resp.14086] [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: 10/05/2020] [Revised: 03/27/2021] [Accepted: 04/26/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND OBJECTIVE Cardiovascular (CV) diseases are frequent in patients with chronic obstructive pulmonary disease (COPD). Likewise, chronic bronchial infection (CBI) is also frequent in COPD and it is associated with systemic inflammation, a well-known CV risk factor. The objective of this study was to investigate the relationship between CBI, systemic inflammation and incident CV events. METHODS A post hoc analysis of prospectively collected cohort of 201 COPD patients [Global Initiative for Chronic Obstructive Lung Disease (GOLD) II-IV] followed up every 3-6 months for 84 months was conducted. CBI was defined as ≥3 positive pathogenic microorganisms sputum cultures over 1 year, separated by ≥3 months. Systemic inflammation was assessed by circulating levels of C-reactive protein and fibrinogen. Fatal and non-fatal CV events, including coronary and cerebrovascular events as well as arrhythmia episodes, were prospectively recorded. For analysis, they were analysed separately and combined in a composite variable. RESULTS As hypothesized, CBI was associated with persistent systemic inflammation and a significantly higher incidence of CV events (HR: 3.88; 95% CI: 1.83-8.22), mainly of coronary origin independent of age, number and severity of exacerbations, comorbidities, other CV risk factors, lung function, BMI, smoking status and treatments. These associations were particularly significant in patients with CBI by Pseudomonas aeruginosa (PA). CONCLUSION CBI, particularly by PA, is associated with sustained and enhanced systemic inflammation and a higher incidence of CV events (especially coronary events). The possibility that treating CBI may decrease systemic inflammation and CV events in COPD deserves prospective, interventional studies.
Collapse
Affiliation(s)
- Miguel Ángel Martinez-Garcia
- Respiratory Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain.,Centro de Investigación Biomédica en Red (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Faner
- Centro de Investigación Biomédica en Red (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Institut d'investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Grace Oscullo
- Respiratory Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | | | - Marta Ballester
- Pneumology Unit, Hospital General de Requena, Valencia, Spain
| | - Alfonso Muriel
- Biostatistic Unit, Hospital Ramón y Cajal, IRYCIS, CIBERESP and Universidad de Alcalá, Madrid, Spain
| | - Alvar Agusti
- Centro de Investigación Biomédica en Red (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.,Institut d'investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Respiratory Institute, Hospital Clinic University, Barcelona, Spain
| |
Collapse
|
202
|
Abstract
IL (interleukin)-6 is a pivotal cytokine of innate immunity, which enacts a broad set of physiological functions traditionally associated with host defense, immune cell regulation, proliferation, and differentiation. Following recognition of innate immune pathways leading from the NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome to IL-1 to IL-6 and on to the hepatically derived clinical biomarker CRP (C-reactive protein), an expanding literature has led to understanding of the proatherogenic role for IL-6 in cardiovascular disease and thus the potential for IL-6 inhibition as a novel method for vascular protection. In this review, we provide an overview of the mechanisms by which IL-6 signaling occurs and how that impacts upon pharmacological inhibition; describe murine models of IL-6 and atherogenesis; summarize human epidemiological data outlining the utility of IL-6 as a biomarker of vascular risk; outline genetic data suggesting a causal role for IL-6 in systemic atherothrombosis and aneurysm formation; and then detail the potential role of IL-6 inhibition in stable coronary disease, acute coronary syndromes, heart failure, and the atherothrombotic complications associated with chronic kidney disease and end-stage renal failure. Finally, we review anti-inflammatory and antithrombotic findings for ziltivekimab, a novel IL-6 ligand inhibitor being developed specifically for use in atherosclerotic disease and poised to be tested formally in a large-scale cardiovascular outcomes trial focused on individuals with chronic kidney disease and elevated levels of CRP, a population at high residual atherothrombotic risk, high residual inflammatory risk, and considerable unmet clinical need.
Collapse
MESH Headings
- Aneurysm/etiology
- Animals
- Antibodies, Monoclonal, Humanized/therapeutic use
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- C-Reactive Protein/metabolism
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/therapy
- Cell Differentiation
- Cell Proliferation
- Disease Models, Animal
- Humans
- Immunity, Cellular
- Immunity, Innate
- Inflammasomes
- Inflammation/complications
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Interleukin-6/antagonists & inhibitors
- Interleukin-6/genetics
- Interleukin-6/immunology
- Interleukin-6/metabolism
- Mice
- Myocardial Ischemia/therapy
- NLR Family, Pyrin Domain-Containing 3 Protein
- Receptors, Interleukin-6/antagonists & inhibitors
- Receptors, Interleukin-6/metabolism
- Renal Dialysis
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/therapy
- Thrombosis/etiology
Collapse
Affiliation(s)
- Paul M Ridker
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Manas Rane
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
203
|
Nasir K, Cainzos-Achirica M. Role of coronary artery calcium score in the primary prevention of cardiovascular disease. BMJ 2021; 373:n776. [PMID: 33947652 DOI: 10.1136/bmj.n776] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
First developed in 1990, the Agatston coronary artery calcium (CAC) score is an international guideline-endorsed decision aid for further risk assessment and personalized management in the primary prevention of atherosclerotic cardiovascular disease. This review discusses key international studies that have informed this 30 year journey, from an initial coronary plaque screening paradigm to its current role informing personalized shared decision making. Special attention is paid to the prognostic value of a CAC score of zero (the so called "power of zero"), which, in a context of low estimated risk thresholds for the consideration of preventive therapy with statins in current guidelines, may be used to de-risk individuals and thereby inform the safe delay or avoidance of certain preventive therapies. We also evaluate current recommendations for CAC scoring in clinical practice guidelines around the world, and past and prevailing barriers for its use in routine patient care. Finally, we discuss emerging approaches in this field, with a focus on the potential role of CAC informing not only the personalized allocation of statins and aspirin in the general population, but also of other risk-reduction therapies in special populations, such as individuals with diabetes and people with severe hypercholesterolemia.
Collapse
Affiliation(s)
- Khurram Nasir
- Division of Cardiovascular Prevention and Wellness, Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
- Center for Outcomes Research, Houston Methodist, Houston, TX, USA
| | - Miguel Cainzos-Achirica
- Division of Cardiovascular Prevention and Wellness, Department of Cardiology, Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
- Center for Outcomes Research, Houston Methodist, Houston, TX, USA
| |
Collapse
|
204
|
Marcos-Jubilar M, Orbe J, Roncal C, Machado FJD, Rodriguez JA, Fernández-Montero A, Colina I, Rodil R, Pastrana JC, Páramo JA. Association of SDF1 and MMP12 with Atherosclerosis and Inflammation: Clinical and Experimental Study. Life (Basel) 2021; 11:life11050414. [PMID: 34062730 PMCID: PMC8147178 DOI: 10.3390/life11050414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Atherosclerosis is the main etiology of cardiovascular diseases (CVD), associated to systemic inflammation. Matrix metalloproteinases (MMPs) are related to atherosclerosis progression through the SDF1/CXCR4 axis promoting macrophages recruitment within the vascular wall. The goal was to assess new circulatory inflammatory markers in relation to atherosclerosis. METHODS Measurement of SDF1, MMP12 and CRP in blood samples of 298 prospective patients with cardiovascular risk. To explore atherosclerosis progression, CXCR4/SDF1 axis and MMP12 expression were determined by RT-qPCR and by immunohistochemistry in the aorta of accelerated and delayed atherosclerosis mice models (Apoe-/- and Apoe-/-Mmp10-/-). RESULTS SDF1, MMP12 and CRP were elevated in patients with clinical atherosclerosis, but after controlling by confounding factors, only SDF1 and CRP remained increased. Having high levels of both biomarkers showed 2.8-fold increased risk of presenting clinical atherosclerosis (p = 0.022). Patients with elevated SDF1, MMP12 and CRP showed increased risk of death in follow-up (HR = 3.2, 95%CI: 1.5-7.0, p = 0.004). Gene and protein expression of CXCR4 and MMP12 were increased in aortas from Apoe-/- mice. CONCLUSIONS The combination of high circulating SDF1, MMP12 and CRP identified patients with particular inflammatory cardiovascular risk and increased mortality. SDF1/CXCR4 axis and MMP12 involvement in atherosclerosis development suggests that they could be possible atherosclerotic targets.
Collapse
Affiliation(s)
- María Marcos-Jubilar
- Haematology Service, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Correspondence: (M.M.-J.); (J.A.P.)
| | - Josune Orbe
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (J.O.); (C.R.); (F.J.D.M.); (J.A.R.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Roncal
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (J.O.); (C.R.); (F.J.D.M.); (J.A.R.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Florencio J. D. Machado
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (J.O.); (C.R.); (F.J.D.M.); (J.A.R.)
| | - José Antonio Rodriguez
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (J.O.); (C.R.); (F.J.D.M.); (J.A.R.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Inmaculada Colina
- Internal Medicine Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.C.); (J.C.P.)
| | - Raquel Rodil
- Internal Medicine Department, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain;
| | - Juan C. Pastrana
- Internal Medicine Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (I.C.); (J.C.P.)
| | - José A. Páramo
- Haematology Service, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain; (J.O.); (C.R.); (F.J.D.M.); (J.A.R.)
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (M.M.-J.); (J.A.P.)
| |
Collapse
|
205
|
Wu X, He Z, Sun R, Xie X, Chen Q, Wang J, Bao J, Huang J, Jiang Y, Zhang Y, Wang J. Large HDL 2 combined with inflammatory factors as superior predictors for coronary artery disease than small HDL 3. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:672. [PMID: 33987370 PMCID: PMC8106016 DOI: 10.21037/atm-21-948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background This study investigated whether combinations of high-density lipoprotein (HDL) subfractions and inflammatory markers would add value to coronary artery disease (CAD) prediction. Methods Non-CAD subjects (n=245) were stratified into low/moderate/high-Framingham risk (L/M/H-FR) groups and 180 CAD patients were enrolled. Levels of HDL-C, HDL2, HDL3, monocyte chemoattractant protein-1 (MCP-1), and high-sensitivity C-reactive protein (hsCRP) were measured. Multivariable logistic models for CAD were estimated with a single parameter or all parameters together after adjustment for conventional risk factors (CRFs), and Z statistics, net reclassification improvement (NRI), and integrated discrimination improvement (IDI) were used to compare discrimination among different models. Results The results show that HDL-C, HDL2, and HDL3 gradually decreased, while MCP-1 and hsCRP gradually increased from L/M/H-FR to the CAD group. When applying a single factor in the CRFs-adjusted models, HDL-C (OR 0.011, 95% CI, 0.002–0.071, P<0.05) and HDL2 (OR 0.000072, 95% CI, 0.000001–0.004, P<0.05), but not HDL3, were significantly related to CAD risk. Only HDL2 (OR 0.000072, 95% CI, 0.000001–0.004, P<0.001) remained significant when applying all HDL parameters. In the model including all HDL and inflammatory parameters, HDL2 (OR 0.001, 95% CI, 0.000027–0.051), MCP-1 (OR 1.066, 95% CI, 1.039–1.094), and hsCRP (OR 1.130, 95% CI, 1.041–1.227) showed significant differences (all P<0.05). This combined model showed improved discrimination over the models with a single factor (P<0.05) or all HDL parameters (Z=3.299, NRI =0.179, IDI =0.081, P<0.001). Conclusions Large HDL2 is superior to small HDL3 in the inverse association with CAD. The combination of HDL2, MCP-1, and hsCRP with CRFs provides an optimal prediction for CAD.
Collapse
Affiliation(s)
- Xiaoying Wu
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhijian He
- Cardiovascular Medicine Department, The First Affiliated Hospital/School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Runlu Sun
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiangkun Xie
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingqun Chen
- Cardiovascular Medicine Department, Qingyuan People's Hospital, Qingyuan, China
| | - Junjie Wang
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinlan Bao
- Comprehensive Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingjing Huang
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan Jiang
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuling Zhang
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingfeng Wang
- Cardiovascular Medicine Department, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
206
|
Zegeye MM, Andersson JSO, Wennberg P, Repsilber D, Sirsjö A, Ljungberg LU. IL-6 as a Mediator of the Association Between Traditional Risk Factors and Future Myocardial Infarction: A Nested Case-Control Study. Arterioscler Thromb Vasc Biol 2021; 41:1570-1579. [PMID: 33657883 DOI: 10.1161/atvbaha.120.315793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mulugeta M Zegeye
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| | - Jonas S O Andersson
- Skellefteå Research Unit, Department of Public Health and Clinical Medicine (J.S.O.A.), Umeå University, Sweden
| | - Patrik Wennberg
- Department of Public Health and Clinical Medicine, Family Medicine (P.W.), Umeå University, Sweden
| | - Dirk Repsilber
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| | - Allan Sirsjö
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| | - Liza U Ljungberg
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| |
Collapse
|
207
|
Moorman AJ, Dean LS, Yang E, Drezner JA. Cardiovascular Risk Assessment in the Older Athlete. Sports Health 2021; 13:622-629. [PMID: 33733939 DOI: 10.1177/19417381211004877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
CONTEXT Limited data are available to guide cardiovascular screening in adult or masters athletes (≥35 years old). This review provides recommendations and the rationale for the cardiovascular risk assessment of older athletes. EVIDENCE ACQUISITION Review of available clinical guidelines, original investigations, and additional searches across PubMed for articles relevant to cardiovascular screening, risk assessment, and prevention in adult athletes (1990-2020). STUDY DESIGN Clinical review. LEVEL OF EVIDENCE Level 3. RESULTS Atherosclerotic coronary artery disease (CAD) is the leading cause of exercise-induced acute coronary syndromes, myocardial infarction, and sudden cardiac death in older athletes. Approximately 50% of adult patients who experience acute coronary syndromes and sudden cardiac arrest do not have prodromal symptoms of myocardial ischemia. The risk of atherosclerotic cardiovascular disease (ASCVD) can be estimated by using existing risk calculators. ASCVD 10-year risk is stratified into 3 categories: low-risk (≤10%), intermediate-risk (between 10% and 20%), and high-risk (≥20%). Coronary artery calcium (CAC) scoring with noncontrast computed tomography provides a noninvasive measure of subclinical CAD. Evidence supports a significant association between elevated CAC and the risk of future cardiovascular events, independent of traditional risk factors or symptoms. Statin therapy is recommended for primary prevention if 10-year ASCVD risk is ≥10% (intermediate- or high-risk patients) or if the Agatston score is >100 or >75th percentile for age and sex. Routine stress testing in asymptomatic, low-risk patients is not recommended. CONCLUSION We propose a comprehensive risk assessment for older athletes that combines conventional and novel risk factors for ASCVD, a 12-lead resting electrocardiogram, and a CAC score. Available risk calculators provide a 10-year estimate of ASCVD risk allowing for risk stratification and targeted management strategies. CAC scoring can refine risk estimates to improve the selection of patients for initiation or avoidance of pharmacological therapy.
Collapse
Affiliation(s)
- Alec J Moorman
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington
| | - Larry S Dean
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington
| | - Eugene Yang
- Department of Medicine, Division of Cardiology, University of Washington, Seattle, Washington
| | - Jonathan A Drezner
- Department of Family Medicine, Sports Medicine Section and UW Medicine Center for Sports Cardiology, University of Washington, Seattle, Washington
| |
Collapse
|
208
|
Watanabe K, Onoue A, Omori H, Kubota K, Yoshida M, Katoh T. Association Between Airflow Limitation and Carotid Intima-Media Thickness in the Japanese Population. Int J Chron Obstruct Pulmon Dis 2021; 16:715-726. [PMID: 33776430 PMCID: PMC7989542 DOI: 10.2147/copd.s291477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/22/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE This study aimed to reveal the association between airflow limitation (AL) and carotid intima-media thickness (IMT) according to smoking status in Japan. SUBJECTS AND METHODS This cross-sectional study was performed in 2809 subjects, who underwent a comprehensive health examination with pulmonary function tests and carotid ultrasonographic measurement. AL was defined as forced expiratory volume in 1 s/forced vital capacity of <0.7. The subjects were divided into the following four groups: never smokers without AL, never smokers with AL, former/current smokers without AL, and former/current smokers with AL. Mean IMT, the maximum measurable IMT value in the left and right common carotid arteries (IMT-C max), and mean IMT-C max were measured by carotid ultrasonography. The carotid wall thickness as defined as follows: IMT ≥ 1.1 mm (IMT1.1), IMT-C max ≥ 1.2 mm (IMTc1.2), and IMT-C max > 1.5 mm (IMTc1.5), based on each measured region. The association between AL and the carotid wall thickness according to smoking status was assessed by logistic regression analysis. RESULTS The mean carotid IMT and mean IMT-C max were significantly higher in never smokers with AL and former/current smokers with or without AL than in never smokers without AL. In logistic regression models adjusted for sex, age, body mass index, hypertension, dyslipidemia, hyperglycemia, physical activity, and alcohol consumption, the risk of carotid wall thickness (IMT1.1 [odds ratio {OR}: 1.55; 95% confidence interval {CI}: 1.07-2.24]; IMTc1.2 [OR: 1.52; 95% CI: 1.03-2.24]; IMTc1.5 [OR: 1.99; 95% CI: 1.15-3.46]) were significantly higher in former/current smokers with AL than in never smokers without AL. CONCLUSION The present results suggest that greater IMT and risk of carotid wall thickness were associated with AL and smoking experience.
Collapse
Affiliation(s)
- Kazuhiko Watanabe
- Department of Public Health, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ayumi Onoue
- Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisamitsu Omori
- Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenichi Kubota
- Japanese Red Cross Kumamoto Health Care Center, Kumamoto, Japan
| | - Minoru Yoshida
- Japanese Red Cross Kumamoto Health Care Center, Kumamoto, Japan
| | - Takahiko Katoh
- Department of Public Health, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
209
|
Rezaei-Tavirani M, Rostami Nejad M, Arjmand B, Rezaei Tavirani S, Razzaghi M, Mansouri V. Fibrinogen Dysregulation is a Prominent Process in Fatal Conditions of COVID-19 Infection; a Proteomic Analysis. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2021; 9:e26. [PMID: 34027421 PMCID: PMC8126351 DOI: 10.22037/aaem.v9i1.1128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Molecular pathophysiology of COVID-19 is not completely known. Expression changes in patients' plasma proteins have revealed new information about the disease. Introducing the key targeted plasma protein in fatal conditions of COVID-19 infection is the aim of this study. METHODS Significant differentially expressed proteins (DEPs) in the plasma of cases with a fatal condition of COVID-19 were extracted from an original article. These proteins were included in a network via STRING database along with 100 first neighbor proteins to determine central nodes of the network for analyzing. RESULTS Queried and added proteins were included in a scale free network. Three hub nodes were identified as critical target proteins. The top queried hub proteins were chains of fibrinogen; Fibrinogen Alpha chain (FGA), Fibrinogen gamma chain (FGG), and Fibrinogen beta chain (FGB), which are related to the coagulation process. CONCLUSIONS It seems that fibrinogen dysregulation has a deep impact on the fatality of COVID-19 infection.
Collapse
Affiliation(s)
- Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Razzaghi
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Mansouri
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
210
|
Best LG, Azure C, Martell K, Tsosie KS, Voels B. Unactivated leukocyte expression of C-reactive protein is minimal and not dependent on rs1205 genotype. Sci Rep 2021; 11:5691. [PMID: 33707594 PMCID: PMC7952394 DOI: 10.1038/s41598-021-85272-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/22/2021] [Indexed: 11/12/2022] Open
Abstract
C-reactive protein (CRP), a prominent component of the innate immune system, is implicated in the pathophysiology of many conditions. CRP production primarily occurs in the liver; but contributions from other tissues is unclear. The Genotype-Tissue Expression Portal shows essentially no expression in whole blood and reports in the literature are conflicting. Multiple genomic variants influence serum levels of CRP. We measured CRP mRNA expression in leukocytes and sought to determine if rs1205 genotype influences leukocyte expression. Leukocytes were obtained from 20 women differing by genotype. Quantitative, real-time PCR (RT-qPCR) detected CRP and reference gene (GAPDH) mRNA. Leukocyte expression was calculated by the 2ΔCT method, and against a standard curve. Digital drop PCR was also used to calculate expression ratios. Student's t test and linear regression methods examined possible differences between genotypes. During 32 runs (10 replicates each), the RT-qPCR mean (SD) CRP/GAPDH ratio was 3.39 × 10–4 (SD 1.73 × 10–4) and 3.15 × 10–4 (SD 1.64 × 10–4) for TT and CC genotypes respectively, p = 0.76; and digital drop PCR results were similar. Serum CRP was not significantly different between genotypes, nor correlated with leukocyte expression. CRP is minimally expressed in unactivated leukocytes and this expression is not likely influenced by rs1205 genotype.
Collapse
Affiliation(s)
- L G Best
- University of North Dakota, Grand Forks, ND, USA. .,Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA. .,, 1935 118th Ave NW, Watford City, ND, 58854, USA.
| | - C Azure
- Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA
| | - K Martell
- Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA
| | - K S Tsosie
- Natural Sciences, Turtle Mountain Community College, Belcourt, ND, USA
| | - B Voels
- Science, Cankdeska Cikana Community College, Fort Totten, ND, USA
| |
Collapse
|
211
|
Choi H, Dey AK, Priyamvara A, Aksentijevich M, Bandyopadhya D, Dey D, Dani S, Guha A, Nambiar P, Nasir K, Jneid H, Mehta NN, Lavie C, Amar S. Role of Periodontal Infection, Inflammation and Immunity in Atherosclerosis. Curr Probl Cardiol 2021; 46:100638. [PMID: 32646544 PMCID: PMC8761259 DOI: 10.1016/j.cpcardiol.2020.100638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Inflammation plays a major role in the development and progression of cardiovascular disease (CVD) morbidity and mortality. The well-established relationship between periodontal disease (PD) and CVD may be causal. Left untreated, PD can lead to high systemic inflammation, thus contributing to inflammatory CVD, such as atherosclerosis. Multiple mechanisms have been proposed to elucidate the causal relationship between PD and its contribution to CVD. OBJECTIVE This review article highlights the current evidence supporting the role of PD in the development and progression of atherosclerosis. METHODS After creating a list of relevant medical subject heading (MeSH) terms, a systematic search within PubMed in English for each MeSH term between 2000 and 2019 was used to generate evidence for this review article. CONCLUSION There is overwhelming evidence in the current literature that supports an association between PD and CVD that is independent of known CVD risk factors. However, the supporting evidence that PD directly causes CVD in humans continues to remain elusive. Multiple biologically plausible mechanisms have been proposed and investigated, yet most studies are limited to mouse models and in vitro cell cultures. Additional studies testing the various proposed mechanisms in longitudinal human studies are required to provide deeper insight into the mechanistic link between these 2 related diseases.
Collapse
Affiliation(s)
- Harry Choi
- National Heart Lung and Blood Institute, Bethesda, MD, USA
| | - Amit K. Dey
- National Heart Lung and Blood Institute, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | - Nehal N. Mehta
- National Heart Lung and Blood Institute, Bethesda, MD, USA
| | - Carl Lavie
- Ochsner Clinical School-UQ School of Medicine, New Orleans, LA, USA
| | | |
Collapse
|
212
|
Kahhan N, Hossain MJ, Lang J, Harrison C, Canas J, Wysocki T, Lochrie A, Balagopal PB. Durability of Changes in Biomarkers of Cardiometabolic Disease: 1-Year Family-Based Intervention in Children with Obesity. Metab Syndr Relat Disord 2021; 19:264-271. [PMID: 33650888 DOI: 10.1089/met.2020.0097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: The sustainability of health benefits in response to lifestyle-based interventions remains unclear in children with overweight and obesity, and cardiometabolic disease (CMD). We determined the changes in novel biomarkers of CMD in a 1-year family-based intervention (FBI) program, during 6-month active monitoring phase and at 12-month follow-up. Methods: Children with an age-adjusted body mass index (BMI) percentile ≥85 (N = 130; age 8-11 years) were recruited for a 1-year (6-month monitored and 6-month unmonitored) randomized controlled FBI program. Anthropometry and selected biomarkers of CMD were measured in 87 participants, randomly allocated to intervention (INT) and education-only (EDU) groups, at baseline, immediately after a 6-month active intervention or control period, and at 12-month unmonitored follow-up. Results: Samples from 87 participants (age 10.00 ± 0.11 years and Tanner stage ≤3) with obesity (BMI%ile = 97.45 ± 0.15) were available. Overall intervention effect (between groups), was observed for total (T) and high molecular weight (HMW) adiponectin, ratio of total to HMW adiponectin, fibrinogen, and interleukin (IL)-6 (P < 0.05 for all). However, between-group beneficial changes after adjusting for baseline levels were limited to BMI percentile, T and HMW adiponectin and their ratio, IL-6, and fibrinogen (P < 0.05 for all) mainly during the 6-month period of monitored intervention. Changes in traditional risk factors such as lipids and triglycerides were inconsistent. During the 6-month follow-up period, the changes in biomarkers leveled-off, except for T and HMW adiponectin, IL-6, and fibrinogen that continued to show benefits (P < 0.05) from the 6- to 12-month follow-up. Conclusions: The FBI program beneficially altered novel biomarkers of CMD during the monitored intervention phase in school-age children with obesity, but they mostly moved back toward baseline during the unmonitored follow-up phase. The changes in novel biomarkers of CMD appear to be more sensitive compared to the traditional risk factors. The study implies the need for refinements in lifestyle-based approaches in the preservation of cardiovascular health and calls for robust biomarkers to monitor the changes. The study was registered at ClinicalTrials.gov (NCT01146314).
Collapse
Affiliation(s)
- Nicole Kahhan
- Division of Psychology, Nemours Children's Specialty Care, Jacksonville, Florida, USA
| | - Md Jobayer Hossain
- Nemours Biostatistics Core, Alfred I duPont Hospital for Children, Wilmington, Delaware, USA
| | - Jason Lang
- Division of Allergy/Immunology and Pulmonary Medicine, Duke University, Durham, North Carolina, USA
| | - Charles Harrison
- Nemours Biomedical Research, Nemours Children's Specialty Care, Jacksonville, Florida, USA
| | - Jose Canas
- Division of Endocrinology, Johns Hopkins All Children's Hospital, St. Petersburg, Florida, USA
| | - Tim Wysocki
- Nemours Center for Health Care Delivery Science, Jacksonville, Florida, USA
| | - Amanda Lochrie
- Division of Psychology, Nemours Children's Specialty Care, Jacksonville, Florida, USA
| | - P Babu Balagopal
- Nemours Biomedical Research, Nemours Children's Specialty Care, Jacksonville, Florida, USA.,Division of Pediatrics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
213
|
Ekström I. Serum C-Reactive Protein Is Negatively Associated With Olfactory Identification Ability in Older Adults. Iperception 2021; 12:20416695211009928. [PMID: 33948158 PMCID: PMC8054135 DOI: 10.1177/20416695211009928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 03/25/2021] [Indexed: 01/08/2023] Open
Abstract
IMPORTANCE Olfactory deficits are common in aging and associated with several conditions linked to inflammation. A few studies suggest that increased concentration of pro-inflammatory biomarkers may be related to olfactory deficits, but these associations are understudied in population-based samples. OBJECTIVE To investigate the association between serum concentrations of C-reactive protein (CRP) and olfactory identification level as well as rate of change in aging. METHODS We included 1,721 participants (mean age 70.5 years; 61.9% female) with at least two olfactory assessments across the 12-year follow-up. Baseline level and change in odor identification were estimated with linear mixed models as a function of CRP levels, derived from blood plasma at baseline. RESULTS Results indicated a negative dose-response association between CRP level and odor identification scores at baseline, after adjustment for demographic, cognitive, health, and lifestyle factors. CRP levels ranging between 11 and 20 mg/L were significantly related to lower olfactory ability (β = -0.811, 95% confidence interval [CI] [-1.503 to -0.118]; p = .022). Likewise, CRP values above 20 mg/L were related to lower olfactory scores, an association that approached statistical significance (β = -0.996, 95% CI [-2.045 to 0.054]; p = .063). We found no associations between CRP and olfactory change (ps > .368). Sensitivity analyses showed that associations between CRP and olfaction were confined to younger participants (age ≤72 years) and men (ps < .034). CONCLUSIONS Our findings suggest a negative association between serum CRP levels and olfactory identification ability in aging that may be dependent on age and sex.
Collapse
Affiliation(s)
- Ingrid Ekström
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| |
Collapse
|
214
|
Yan Y, Li S, Liu Y, Bazzano L, He J, Mi J, Chen W. The impact of body weight trajectory from childhood on chronic inflammation in adulthood: The Bogalusa Heart Study. Pediatr Investig 2021; 5:21-27. [PMID: 33778423 PMCID: PMC7984006 DOI: 10.1002/ped4.12248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
IMPORTANCE The impact of long-term burden of excessive body weight, beginning in childhood, on inflammatory status in adulthood has been poorly described. OBJECTIVE To characterize the longitudinal body mass index (BMI) trajectory from childhood and examine its relationship with inflammatory status in adulthood. METHODS We included 1285 adults who had 4-15 repeat measurements of BMI from childhood to adulthood. The area under the curve (AUC) of growth curves was calculated to characterize long-term burden (total AUC) and trends (incremental AUC) of BMI. RESULTS After adjusting for covariates, higher values of BMI in terms of childhood and adulthood, as well as total and incremental AUC, were strongly associated with elevated levels of adult C-reactive protein (CRP) in the four race-sex groups. There were significant differences in linear and nonlinear curve parameters between the normal and high CRP groups for all race-sex groups (P < 0.01). Compared with participants who had consistently low BMI in both childhood and adulthood, participants with high BMI in adulthood had higher CRP levels (P < 0.001), irrespective of their childhood BMI status; participants with high BMI in childhood but low BMI in adulthood had similar adult CRP levels. INTERPRETATION The impact of excessive body weight on inflammation is cumulative and exacerbated over time. The influence of childhood overweight/obesity on inflammatory status in adulthood can be alleviated by reducing adiposity in adulthood.
Collapse
Affiliation(s)
- Yinkun Yan
- Department of Non‐communicable Disease ManagementBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
- Department of EpidemiologyTulane University School of Public Health and Tropical MedicineNew OrleansLAUSA
| | - Shengxu Li
- Children’s Minnesota Research InstituteChildren’s Hospitals and Clinics of MinnesotaMinneapolisMNUSA
| | - Yang Liu
- Department of EpidemiologyTulane University School of Public Health and Tropical MedicineNew OrleansLAUSA
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lydia Bazzano
- Department of EpidemiologyTulane University School of Public Health and Tropical MedicineNew OrleansLAUSA
| | - Jiang He
- Department of EpidemiologyTulane University School of Public Health and Tropical MedicineNew OrleansLAUSA
| | - Jie Mi
- Department of Non‐communicable Disease ManagementBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Wei Chen
- Department of EpidemiologyTulane University School of Public Health and Tropical MedicineNew OrleansLAUSA
| |
Collapse
|
215
|
Fibrinogen and hemoglobin predict near future cardiovascular events in asymptomatic individuals. Sci Rep 2021; 11:4605. [PMID: 33633217 PMCID: PMC7907085 DOI: 10.1038/s41598-021-84046-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Abstract
To identify circulating proteins predictive of acute cardiovascular disease events in the general population, we performed a proteomic screen in plasma from asymptomatic individuals. A "Discovery cohort" of 25 individuals who subsequently incurred a cardiovascular event within 3 years (median age = 70 years, 80% male) was matched to 25 controls remaining event-free for > 5 years (median age = 72 years, 80% male). Plasma proteins were assessed by data independent acquisition mass spectrometry (DIA-MS). Associations with cardiovascular events were tested using Cox regression, adjusted for the New Zealand Cardiovascular Risk Score. Concentrations of leading protein candidates were subsequently measured with ELISAs in a larger (n = 151) independent subset. In the Discovery cohort, 76 plasma proteins were robustly quantified by DIA-MS, with 8 independently associated with cardiovascular events. These included (HR = hazard ratio [95% confidence interval] above vs below median): fibrinogen alpha chain (HR = 1.84 [1.19-2.84]); alpha-2-HS-glycoprotein (also called fetuin A) (HR = 1.86 [1.19-2.93]); clusterin isoform 2 (HR = 1.59 [1.06-2.38]); fibrinogen beta chain (HR = 1.55 [1.04-2.30]); hemoglobin subunit beta (HR = 1.49 [1.04-2.15]); complement component C9 (HR = 1.62 [1.01-2.59]), fibronectin isoform 3 (HR = 0.60 [0.37-0.99]); and lipopolysaccharide-binding protein (HR = 1.58 [1.00-2.49]). The proteins for which DIA-MS and ELISA data were correlated, fibrinogen and hemoglobin, were analyzed in an Extended cohort, with broader inclusion criteria and longer time to events, in which these two proteins were not associated with incident cardiovascular events. We have identified eight candidate proteins that may independently predict cardiovascular events occurring within three years in asymptomatic, low-to-moderate risk individuals, although these appear not to predict events beyond three years.
Collapse
|
216
|
Egorov AI, Converse RR, Griffin SM, Styles JN, Sams E, Hudgens E, Wade TJ. Latent Toxoplasma gondii infections are associated with elevated biomarkers of inflammation and vascular injury. BMC Infect Dis 2021; 21:188. [PMID: 33602170 PMCID: PMC7890825 DOI: 10.1186/s12879-021-05882-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 02/09/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Toxoplasma gondii is a protozoan parasite that infects cats as definitive hosts and other warm-blooded animals including humans as intermediate hosts. It forms infectious cysts in the brain, muscle and other tissues establishing life-long latent infection. Approximately 10% of the US population is infected. While latent infections are largely asymptomatic, they are associated with neurological deficits and elevated risks of neuropsychiatric diseases. METHODS This cross-sectional epidemiological study investigated associations of T. gondii infections with biomarkers of inflammation and vascular injury: soluble intercellular adhesion molecule 1 (ICAM-1), soluble vascular cell adhesion molecule 1 (VCAM-1), C-reactive protein (CRP), and serum amyloid A (SAA). Serum samples from 694 adults in the Raleigh-Durham-Chapel Hill, North Carolina metropolitan area were tested for IgG antibody response to T. gondii, and for the above biomarkers using commercially available assays. RESULTS T. gondii seroprevalence rate in this sample was 9.7%. Seropositivity was significantly associated with 11% (95% confidence limits 4, 20%) greater median levels of VCAM-1 (p = 0.003), and marginally significantly with 9% (1, 17%), and 36% (1, 83%) greater median levels of ICAM-1, and CRP, respectively (p = 0.04 for each) after adjusting for sociodemographic and behavioral covariates, while the 23% (- 7, 64%) adjusted effect on SAA was not statistically significant (p = 0.15). CONCLUSIONS Latent infections with T. gondii are associated with elevated biomarkers of chronic inflammation and vascular injury that are also known to be affected by ambient air pollution.
Collapse
Affiliation(s)
- Andrey I. Egorov
- United States Environmental Protection Agency, Office of Research and Development, EPA, MD 58-C, 109. T.W. Alexander Drive, Research Triangle Park, NC 27711 USA
| | - Reagan R. Converse
- United States Environmental Protection Agency, Office of Research and Development, EPA, MD 58-C, 109. T.W. Alexander Drive, Research Triangle Park, NC 27711 USA
| | - Shannon M. Griffin
- United States Environmental Protection Agency, Office of Research and Development, Cincinnati, OH USA
| | - Jennifer N. Styles
- United States Environmental Protection Agency, Office of Research and Development, EPA, MD 58-C, 109. T.W. Alexander Drive, Research Triangle Park, NC 27711 USA
- Gillings School of Global Public Health, Environmental Sciences and Engineering Department, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Elizabeth Sams
- United States Environmental Protection Agency, Office of Research and Development, EPA, MD 58-C, 109. T.W. Alexander Drive, Research Triangle Park, NC 27711 USA
| | - Edward Hudgens
- United States Environmental Protection Agency, Office of Research and Development, EPA, MD 58-C, 109. T.W. Alexander Drive, Research Triangle Park, NC 27711 USA
| | - Timothy J. Wade
- United States Environmental Protection Agency, Office of Research and Development, EPA, MD 58-C, 109. T.W. Alexander Drive, Research Triangle Park, NC 27711 USA
| |
Collapse
|
217
|
Testa C, DI Lorenzo A, Parlato A, D'Ambrosio G, Merolla A, Pacileo M, Iannuzzo G, Gentile M, Nugara C, Sarullo FM, DE Gregorio C, D'Andrea A, Vigorito C, Venturini E, Giallauria F. Exercise for slowing the progression of atherosclerotic process: effects on inflammatory markers. Panminerva Med 2021; 63:122-132. [PMID: 33565757 DOI: 10.23736/s0031-0808.21.04266-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is a dynamic process driven by all cardiovascular risk factors that can be briefly divided into an early and a late phase. Inflammation is one of the fundamental substrates that initiates the atherosclerotic process in the early stages and promotes and maintains it in the final stages. In the last decades, clinical and experimental data have shown that inflammation is supported by mediators that respond to physical activity. The present review aimed at investigating the effect of physical exercise on inflammatory mediators, both the positive ones that have a proinflammatory effect (interleukin 6, c-reactive protein and tumor necrosis factor α, interferon γ, high-mobility group box-1), and the negative ones which have an anti-inflammatory effect (interleukin 10). Pooled data support the evidence that physical exercise can directly modulate the activity of inflammatory cytokines slowing down or preventing the formation of the atherosclerotic stage.
Collapse
Affiliation(s)
- Crescenzo Testa
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Anna DI Lorenzo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandro Parlato
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giuseppe D'Ambrosio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Aurora Merolla
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Mario Pacileo
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Marco Gentile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Cinzia Nugara
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Filippo M Sarullo
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Cesare DE Gregorio
- Unit of Cardiology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy.,Post-graduate Residency School in Cardiovascular Diseases, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Antonello D'Andrea
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Carlo Vigorito
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Elio Venturini
- Cardiac Rehabilitation Unit, AUSL Toscana Nord-Ovest, Cecina Civil Hospital, Cecina, Livorno, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Federico II University, Naples, Italy - .,Faculty of Sciences and Technology, University of New England, Armidale, Australia
| |
Collapse
|
218
|
Zhang DP, Mao XF, Wu TT, Chen Y, Hou XG, Yang Y, Ma X, Zhang JY, Ma YT, Xie X, Zheng YY. The Fibrinogen-to-Albumin Ratio Is Associated With Outcomes in Patients With Coronary Artery Disease Who Underwent Percutaneous Coronary Intervention. Clin Appl Thromb Hemost 2021; 26:1076029620933008. [PMID: 32598182 PMCID: PMC7427009 DOI: 10.1177/1076029620933008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Coronary artery disease (CAD) is a systemic chronic inflammatory disease, and serum fibrinogen and albumin are 2 important factors in systemic inflammation. We aimed to investigate the relationship between the fibrinogen–albumin ratio (FAR) and outcomes in patients with CAD who underwent percutaneous coronary intervention (PCI). All patients were from the Clinical Outcomes and Risk Factors of Patients with Coronary Heart Disease after PCI (CORFCHD-PCI) study, which is a retrospective cohort study (Identifier: ChiCTR-ORC-16010153) that includes a total of 6050 patients with CAD after PCI from January 2008 to December 2016. A total of 5829 patients with CAD after PCI were recruited in the present study. They were divided into 2 groups according to the FAR cutoff value, which was calculated using a receiver operating characteristic curve, a low group (FAR < 0.095, n = 3811), and a high group (FAR ≥ 0.095, n = 2018). The average follow-up time was 35.9 ± 22.6 months. The multivariate Cox proportional hazards model showed that FAR was independently correlated with all-cause mortality (adjusted hazard ratio [HR] = 1.432 [1.134-1.808], P = .003), cardiac mortality (adjusted HR = 1.579 [1.218-2.047], P = .001), major adverse cardiac and cerebrovascular events (adjusted HR = 1.296 [1.125-1.494], P < .001), major adverse cardiac events (adjusted HR = 1.357 [1.170-1.572], P < .001), and heart failure (adjusted HR = 1.540 [1.135-2.091], P = .006). The present study indicated that the FAR was associated with adverse outcomes in patients with CAD who underwent PCI.
Collapse
Affiliation(s)
- Da-Peng Zhang
- Heart Center & Beijing Key Laboratory of Hypertension Disease, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiao-Feng Mao
- Department of Science and Technology, Xinjiang Medical University, Urumqi, China
| | - Ting-Ting Wu
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - You Chen
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xian-Geng Hou
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yi Yang
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiang Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jin-Ying Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Yi-Tong Ma
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiang Xie
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ying-Ying Zheng
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
219
|
Lin J, Hu S, Shi Y, Lu F, Luo W, Lin Y. Effects of continuous positive airway pressure on plasma fibrinogen levels in obstructive sleep apnea patients: a systemic review and meta-analysis. Biosci Rep 2021; 41:BSR20203856. [PMID: 33427285 PMCID: PMC7846965 DOI: 10.1042/bsr20203856] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/29/2020] [Accepted: 01/08/2021] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Fibrinogen has been implicated to play a role in the pathophysiology of obstructive sleep apnea (OSA). Many studies have evaluated the effect of continuous positive airway pressure (CPAP) on plasma fibrinogen levels in OSA patients. However, results from different reports were not consistent. To assess the effect of CPAP treatment on plasma fibrinogen levels of patients with OSA, a meta-analysis was performed. METHODS A systematic search of Pubmed, Embase, Cochrane, Wanfang Database and Chinese National Knowledge Infrastructure was performed. Data were extracted, and then weighted mean difference (WMD) and 95% confidence intervals (CIs) were calculated using a random-effects model. RESULTS Twenty-two studies involving 859 patients were included in this meta-analysis. Combined data showed that plasma fibrinogen concentrations decreased after CPAP therapy (WMD = -0.38 g/l, 95% CI [-0.54 to -0.22 g/l], P<0.001). In the subgroup analyses by therapy duration, plasma fibrinogen concentrations declined significantly in the long-term (≥1 month) CPAP therapy subgroup (WMD = -0.33 g/l, 95% CI [-0.49 to -0.16 g/l], P<0.001) but not in the short-term (<1 month) CPAP therapy subgroup (WMD = -0.84 g/l, 95% CI [-1.70 to 0.03 g/l], P=0.058). Moreover, in patients with long-term CPAP therapy duration, plasma fibrinogen levels decreased with good CPAP compliance (≥4 h/night) (WMD = -0.37 g/l, 95% CI [-0.55 to -0.19 g/l], P<0.001) but not with poor CPAP compliance (<4 h/night) (WMD = 0.12 g/l, 95% CI [-0.09 to 0.33 g/l], P=0.247). CONCLUSION Long-term CPAP treatment with good compliance can reduce the plasma fibrinogen levels in patients with OSA.
Collapse
Affiliation(s)
- Juan Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Suxian Hu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yonghong Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Fang Lu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Wen Luo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| | - Yihua Lin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Department of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
220
|
Jin Z, Wu Q, Chen S, Gao J, Li X, Zhang X, Zhou Y, He D, Cheng Z, Zhu Y, Wu S. The Associations of Two Novel Inflammation Indexes, SII and SIRI with the Risks for Cardiovascular Diseases and All-Cause Mortality: A Ten-Year Follow-Up Study in 85,154 Individuals. J Inflamm Res 2021; 14:131-140. [PMID: 33500649 PMCID: PMC7822090 DOI: 10.2147/jir.s283835] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/31/2020] [Indexed: 01/12/2023] Open
Abstract
Background SII and SIRI are two novel systemic inflammation indexes that were suggested in predicting poor outcomes in cancers. However, no studies have examined their effect on cardiovascular diseases (CVDs) and all-cause mortality. Thus, this study aims to investigate associations between SII, SIRI, and the risks for CVDs and all-cause mortality. Methods A total of 85,154 participants from the Kailuan cohort were included and followed up for incidents of CVDs (including MI, stroke) and all-cause death for 10 years. Multiple Cox regression was used to calculate the adjusted hazard ratios (HRs). Results During the follow-up period, 4262 stroke events, 1233 MI events, and 7225 all-cause deaths were identified, respectively. Compared with the lowest quantile (Q1) of SII or SIRI, after adjusted for most cardiovascular risk factors, both indexes showed positive associations with the risk for stroke (adjusted HRs in Q4 were 1.264 (95% CI: 1.157,1.382) for SII, 1.194 (95% CI: 1.087,1.313) for SIRI), and all-cause death (adjusted HRs in Q4 were 1.246 (95% CI: 1.165,1.331) for SII, 1.393 (95% CI: 1.296,1.498) for SIRI). Additionally, higher SII and SIRI are also associated with increased risk of hemorrhagic stroke and ischemic stroke. Higher SIRI but not SII exhibited a higher MI risk, the adjusted HR in Q4 was 1.204 (1.013,1.431). The significant association remained after additional adjustment for CRP. Subgroup analysis and sensitivity analysis displayed consistent results except for SIRI with MI, where the association did not arrive at significance in subjects aged ≥60. Conclusion Elevated SII and SIRI increased the risk of stroke, two stroke subtypes, and all-cause death. Higher SIRI, but not SII associated with increased MI incidence, and the association of SIRI was only significant in subjects aged <60.
Collapse
Affiliation(s)
- Ziqi Jin
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Qiong Wu
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Shuohua Chen
- Health Care Center, Kailuan Group, Tangshan 063000, People's Republic of China
| | - Jingli Gao
- Department of Intensive Medicine, Kailuan General Hospital, Tangshan 063000, People's Republic of China
| | - Xiaolan Li
- Department of Intensive Medicine, Kailuan General Hospital, Tangshan 063000, People's Republic of China
| | - Xuhui Zhang
- Hangzhou Center for Disease Control and Prevention, Hangzhou 310051, Zhejiang, People's Republic of China
| | - Yaohan Zhou
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Di He
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Zongxue Cheng
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China.,Department of Respiratory Diseases, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310060, People's Republic of China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, 063000, People's Republic of China
| |
Collapse
|
221
|
Abstract
The risk of coronary events with non-steroidal anti-inflammatory drugs has been the subject of much debate since the original trial of rofecoxib raised the issue. Since then, over almost 20 years, such risks have been shown in clinical trials of long-term high-dose users, and in observational studies comparing users with non-users. The roles of cyclooxygenase (COX)-2/COX-1 selectivity and COX-2 inhibitory potency have been proposed to explain this increased risk of myocardial infarction (MI). Among NSAIDs, diclofenac appeared to be associated with a relatively higher risk of MI, similar to that of rofecoxib, compatible with the drug's high COX-2 inhibitory potency. Recent studies have resulted in further information being available. A study in the Danish healthcare system using active comparators found a slightly increased risk of MI in healthy persons. However, risk decreased with increasing baseline cardiovascular risk, to the point that in patients at high cardiovascular risk, there was no additional risk associated with diclofenac compared with paracetamol or other NSAIDs. The other major study, from the SOS project, studied several million persons in four countries in Europe, comparing the use of many NSAIDs with non-use. That study found a slightly increased risk with diclofenac compared with non-use, but this was not different from other NSAIDs. Comparing risks with selectivity or potency found no effect of either. These studies refute the main hypotheses to explain the coronary risk of NSAIDs. Finding risk in healthy low-risk patients only questions the reality of a link between the use of the drugs and the occurrence of MI in these conditions. Biases or confounding may be the major reason for small increases in cardiovascular risks in healthy users of NSAIDs in real life.
Collapse
Affiliation(s)
- Nicholas Moore
- University of Bordeaux, Bordeaux, France. .,Bordeaux PharmacoEpi INSERM CIC1401, 126 rue Leo Saignat, 33076, Bordeaux, France.
| |
Collapse
|
222
|
Cosentino F, Grant PJ, Aboyans V, Bailey CJ, Ceriello A, Delgado V, Federici M, Filippatos G, Grobbee DE, Hansen TB, Huikuri HV, Johansson I, Jüni P, Lettino M, Marx N, Mellbin LG, Östgren CJ, Rocca B, Roffi M, Sattar N, Seferović PM, Sousa-Uva M, Valensi P, Wheeler DC. 2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD. Eur Heart J 2021; 41:255-323. [PMID: 31497854 DOI: 10.1093/eurheartj/ehz486] [Citation(s) in RCA: 2597] [Impact Index Per Article: 649.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
223
|
Allen NE, Arnold M, Parish S, Hill M, Sheard S, Callen H, Fry D, Moffat S, Gordon M, Welsh S, Elliott P, Collins R. Approaches to minimising the epidemiological impact of sources of systematic and random variation that may affect biochemistry assay data in UK Biobank. Wellcome Open Res 2021; 5:222. [PMID: 33364437 PMCID: PMC7739095 DOI: 10.12688/wellcomeopenres.16171.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 02/04/2023] Open
Abstract
Background: UK Biobank is a large prospective study that recruited 500,000 participants aged 40 to 69 years, between 2006-2010.The study has collected (and continues to collect) extensive phenotypic and genomic data about its participants. In order to enhance further the value of the UK Biobank resource, a wide range of biochemistry markers were measured in all participants with an available biological sample. Here, we describe the approaches UK Biobank has taken to minimise error related to sample collection, processing, retrieval and assay measurement. Methods: During routine quality control checks, the laboratory team observed that some assay results were lower than expected for samples acquired during certain time periods. Analyses were undertaken to identify and correct for the unexpected dilution identified during sample processing, and for expected error caused by laboratory drift of assay results. Results: The vast majority (92%) of biochemistry serum assay results were assessed to be not materially affected by dilution, with an estimated difference in concentration of less than 1% (i.e. either lower or higher) than that expected if the sample were unaffected; 8.3% were estimated to be diluted by up to 10%; very few samples appeared to be diluted more than this. Biomarkers measured in urine (creatinine, microalbumin, sodium, potassium) and red blood cells (HbA1c) were not affected. In order to correct for laboratory variation over the assay period, all assay results were adjusted for date of assay, with the exception of those that had a high biological coefficient of variation or evident seasonal variability: vitamin D, lipoprotein (a), gamma glutamyltransferase, C-reactive protein and rheumatoid factor. Conclusions: Rigorous approaches related to sample collection, processing, retrieval, assay measurement and data analysis have been taken to mitigate the impact of both systematic and random variation in epidemiological analyses that use the biochemistry assay data in UK Biobank.
Collapse
Affiliation(s)
- Naomi E. Allen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Matthew Arnold
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
| | - Sarah Parish
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, UK
| | - Michael Hill
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, UK
| | | | - Howard Callen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Daniel Fry
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Stewart Moffat
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, UK
| | - Mark Gordon
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | | | - Paul Elliott
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Rory Collins
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| |
Collapse
|
224
|
Kataoka A, Katagiri S, Kawashima H, Nagura F, Nara Y, Hioki H, Nakashima M, Sasaki N, Hatasa M, Maekawa S, Ohsugi Y, Shiba T, Watanabe Y, Shimokawa T, Iwata T, Kozuma K. Association between periodontal bacteria and degenerative aortic stenosis: a pilot study. J Periodontal Implant Sci 2021; 51:226-238. [PMID: 34387043 PMCID: PMC8367646 DOI: 10.5051/jpis.2006040302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 12/15/2020] [Accepted: 03/05/2021] [Indexed: 11/10/2022] Open
Abstract
Purpose Although several reports have described the relationship between periodontal disease and cardiovascular disease, information about the association between periodontal disease and the progression of degenerative aortic stenosis (AS) is lacking. Therefore, we performed a retrospective, single-center, pilot study to provide insight into this potential association. Methods Data from 45 consecutive patients (19 men; median age, 83 years) with mild or moderate degenerative aortic stenosis were analyzed for a mean observation period of 3.3±1.9 years. The total amount of Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis and titers of serum immunoglobulin G (IgG) against periodontal bacteria and high-sensitivity C-reactive protein (hs-CRP) were evaluated. Aortic valve area (AVA), maximal velocity (Vmax), mean pressure gradient (mean PG), and the Doppler velocity index (DVI) were evaluated. The change in each parameter per year ([ParameterLATEST–ParameterBASELINE]/Follow-up Years) was calculated from the retrospective follow-up echocardiographic data (baseline vs. the most recently collected data [latest]). Results No correlation was found between the concentration of periodontopathic bacteria in the saliva and AS status/progression. The anti-P. gingivalis antibody titer in the serum showed a significant positive correlation with AVA and DVI. Additionally, there was a negative correlation between the anti-P. gingivalis IgG antibody titer and mean PG. The hs-CRP concentration showed positive correlations with Vmax and mean PG. Meanwhile, a negative correlation was observed between the anti-P. gingivalis IgG antibody titer and ΔAVA/year and Δmean PG/year. The hs-CRP concentration showed positive correlations with Vmax and mean PG, and it was significantly higher in patients with rapid aortic stenosis progression (ΔAVA/year <−0.1) than in their counterparts. Conclusions Our results suggest that periodontopathic bacteria such as A. actinomycetemcomitans and P. gingivalis are not directly related to the status/progression of degenerative AS. However, inflammation and a lower immune response may be associated with disease progression.
Collapse
Affiliation(s)
- Akihisa Kataoka
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan.
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Hideyuki Kawashima
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| | - Fukuko Nagura
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| | - Yugo Nara
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| | - Hirofumi Hioki
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| | - Makoto Nakashima
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| | - Naoki Sasaki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masahiro Hatasa
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shogo Maekawa
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiko Shiba
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Watanabe
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| | - Tomoki Shimokawa
- Department of Cardiovascular Surgery, Teikyo University, Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ken Kozuma
- Division of Cardiology, Department of Medicine, Teikyo University, Tokyo, Japan
| |
Collapse
|
225
|
Salzmann S, Euteneuer F, Laferton JAC, Shedden-Mora MC, Schedlowski M, Moosdorf R, Rief W. IL-8 and CRP moderate the effects of preoperative psychological interventions on postoperative long-term outcomes 6 months after CABG surgery - The randomized controlled PSY-HEART trial. Brain Behav Immun 2021; 91:202-211. [PMID: 33002633 DOI: 10.1016/j.bbi.2020.09.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/24/2020] [Accepted: 09/23/2020] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Inflammation has been related to several somatic and psychological disorders and may moderate effects of psychological interventions. In the PSY-HEART trial patients benefitted from preoperative psychological interventions before undergoing coronary artery bypass graft surgery (CABG) and, if necessary, concomitant valvular surgery, compared to standard medical care. In this study we examined whether patients' baseline inflammatory status moderated the intervention effects. MATERIAL AND METHODS In a prospective three-arm randomized clinical trial with 6-months follow-up, 124 patients scheduled for CABG surgery alone or concomitant with valvular surgery were randomized to (i) standard medical care only (SMC) or two preoperative psychological interventions: (ii) CBT-based optimizing expectations (EXPECT) and an (iii) an active control group focusing on emotional support (SUPPORT). Available baseline CRP- (n = 79), IL-6- (n = 78), IL-8- (n = 78) and TNF-alpha-(n = 80) parameters were considered as potential moderators (CRP as a categorical and continuous moderator). Linear mixed model analyses were calculated to test whether baseline inflammatory levels moderated intervention effects on disability, mental and physical quality of life at 6 months after surgery. RESULTS IL-8 moderated intervention effects on patients' disability and categorical CRP moderated intervention effects on mental quality of life. Follow-up tests indicated that EXPECT (and in part SUPPORT) led to lower postoperative disability and higher mental quality of life compared to SMC in patients with low baseline inflammatory markers. EXPECT indicated higher mental quality of life compared to SUPPORT in the high CRP subgroup. Patients in the SMC group had higher mental quality of life in the high CRP subgroup compared to the low CRP subgroup. CONCLUSION Especially for patients with a lower inflammatory baseline status preoperative psychological interventions might be helpful to optimize long-term CABG surgery outcomes.
Collapse
Affiliation(s)
- Stefan Salzmann
- Department of Clinical Psychology and Psychotherapy, Philipps University of Marburg, Marburg, Germany.
| | - Frank Euteneuer
- Department of Clinical Psychology and Psychotherapy, Philipps University of Marburg, Marburg, Germany; Clinical Psychology and Psychotherapy, Medical School Berlin, Berlin, Germany
| | - Johannes A C Laferton
- Department of Clinical Psychology and Psychotherapy, Philipps University of Marburg, Marburg, Germany; Department of Clinical Psychology and Psychotherapy, Psychologische Hochschule Berlin, Berlin, Germany
| | - Meike C Shedden-Mora
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Clinic Essen, Essen, Germany; Department of Clinical Neuroscience, Osher Center for Integrative Medicine, Karolinska Institute, 17177 Stockholm, Sweden
| | - Rainer Moosdorf
- Department for Cardiovascular Surgery, Heart Center, Philipps University of Marburg, Marburg, Germany
| | - Winfried Rief
- Department of Clinical Psychology and Psychotherapy, Philipps University of Marburg, Marburg, Germany
| |
Collapse
|
226
|
Fermont JM, Fisk M, Bolton CE, MacNee W, Cockcroft JR, Fuld J, Cheriyan J, Mohan D, Mäki-Petäjä KM, Al-Hadithi AB, Tal-Singer R, Müllerova H, Polkey MI, Wood AM, McEniery CM, Wilkinson IB. Cardiovascular risk prediction using physical performance measures in COPD: results from a multicentre observational study. BMJ Open 2020; 10:e038360. [PMID: 33372069 PMCID: PMC7772292 DOI: 10.1136/bmjopen-2020-038360] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 11/17/2020] [Accepted: 11/27/2020] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Although cardiovascular disease (CVD) is a common comorbidity associated with chronic obstructive pulmonary disease (COPD), it is unknown how to improve prediction of cardiovascular (CV) risk in individuals with COPD. Traditional CV risk scores have been tested in different populations but not uniquely in COPD. The potential of alternative markers to improve CV risk prediction in individuals with COPD is unknown. We aimed to determine the predictive value of conventional CVD risk factors in COPD and to determine if additional markers improve prediction beyond conventional factors. DESIGN Data from the Evaluation of the Role of Inflammation in Chronic Airways disease cohort, which enrolled 729 individuals with Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage II-IV COPD were used. Linked hospital episode statistics and survival data were prospectively collected for a median 4.6 years of follow-up. SETTING Five UK centres interested in COPD. PARTICIPANTS Population-based sample including 714 individuals with spirometry-defined COPD, smoked at least 10 pack years and who were clinically stable for >4 weeks. INTERVENTIONS Baseline measurements included aortic pulse wave velocity (aPWV), carotid intima-media thickness (CIMT), C reactive protein (CRP), fibrinogen, spirometry and Body mass index, airflow Obstruction, Dyspnoea and Exercise capacity (BODE) Index, 6 min walk test (6MWT) and 4 m gait speed (4MGS) test. PRIMARY AND SECONDARY OUTCOME MEASURES New occurrence (first event) of fatal or non-fatal hospitalised CVD, and all-cause and cause-specific mortality. RESULTS Out of 714 participants, 192 (27%) had CV hospitalisation and 6 died due to CVD. The overall CV risk model C-statistic was 0.689 (95% CI 0.688 to 0.691). aPWV and CIMT neither had an association with study outcome nor improved model prediction. CRP, fibrinogen, GOLD stage, BODE Index, 4MGS and 6MWT were associated with the outcome, independently of conventional risk factors (p<0.05 for all). However, only 6MWT improved model discrimination (C=0.727, 95% CI 0.726 to 0.728). CONCLUSION Poor physical performance defined by the 6MWT improves prediction of CV hospitalisation in individuals with COPD. TRIAL REGISTRATION NUMBER ID 11101.
Collapse
Affiliation(s)
- Jilles M Fermont
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Marie Fisk
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Charlotte E Bolton
- Division of Respiratory Medicine and NIHR Nottingham BRC respiratory theme, University of Nottingham, Nottingham, UK
| | - William MacNee
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - John R Cockcroft
- Department of Cardiology, Columbia University Medical Center, New York City, New York, USA
| | - Jonathan Fuld
- Department of Respiratory Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Divya Mohan
- Medical Innovation, Value Evidence Outcomes, GSK R&D, Philadelphia, Pennsylvania, USA
| | - Kaisa M Mäki-Petäjä
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ali B Al-Hadithi
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ruth Tal-Singer
- Medical Innovation, Value Evidence Outcomes, GSK R&D, Philadelphia, Pennsylvania, USA
| | | | - Michael I Polkey
- Department of Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Angela M Wood
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Carmel M McEniery
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ian B Wilkinson
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
227
|
Nielsen JB, Rom O, Surakka I, Graham SE, Zhou W, Roychowdhury T, Fritsche LG, Gagliano Taliun SA, Sidore C, Liu Y, Gabrielsen ME, Skogholt AH, Wolford B, Overton W, Zhao Y, Chen J, Zhang H, Hornsby WE, Acheampong A, Grooms A, Schaefer A, Zajac GJM, Villacorta L, Zhang J, Brumpton B, Løset M, Rai V, Lundegaard PR, Olesen MS, Taylor KD, Palmer ND, Chen YD, Choi SH, Lubitz SA, Ellinor PT, Barnes KC, Daya M, Rafaels N, Weiss ST, Lasky-Su J, Tracy RP, Vasan RS, Cupples LA, Mathias RA, Yanek LR, Becker LC, Peyser PA, Bielak LF, Smith JA, Aslibekyan S, Hidalgo BA, Arnett DK, Irvin MR, Wilson JG, Musani SK, Correa A, Rich SS, Guo X, Rotter JI, Konkle BA, Johnsen JM, Ashley-Koch AE, Telen MJ, Sheehan VA, Blangero J, Curran JE, Peralta JM, Montgomery C, Sheu WHH, Chung RH, Schwander K, Nouraie SM, Gordeuk VR, Zhang Y, Kooperberg C, Reiner AP, Jackson RD, Bleecker ER, Meyers DA, Li X, Das S, Yu K, LeFaive J, Smith A, Blackwell T, Taliun D, Zollner S, Forer L, Schoenherr S, Fuchsberger C, Pandit A, Zawistowski M, Kheterpal S, Brummett CM, Natarajan P, Schlessinger D, Lee S, Kang HM, Cucca F, Holmen OL, et alNielsen JB, Rom O, Surakka I, Graham SE, Zhou W, Roychowdhury T, Fritsche LG, Gagliano Taliun SA, Sidore C, Liu Y, Gabrielsen ME, Skogholt AH, Wolford B, Overton W, Zhao Y, Chen J, Zhang H, Hornsby WE, Acheampong A, Grooms A, Schaefer A, Zajac GJM, Villacorta L, Zhang J, Brumpton B, Løset M, Rai V, Lundegaard PR, Olesen MS, Taylor KD, Palmer ND, Chen YD, Choi SH, Lubitz SA, Ellinor PT, Barnes KC, Daya M, Rafaels N, Weiss ST, Lasky-Su J, Tracy RP, Vasan RS, Cupples LA, Mathias RA, Yanek LR, Becker LC, Peyser PA, Bielak LF, Smith JA, Aslibekyan S, Hidalgo BA, Arnett DK, Irvin MR, Wilson JG, Musani SK, Correa A, Rich SS, Guo X, Rotter JI, Konkle BA, Johnsen JM, Ashley-Koch AE, Telen MJ, Sheehan VA, Blangero J, Curran JE, Peralta JM, Montgomery C, Sheu WHH, Chung RH, Schwander K, Nouraie SM, Gordeuk VR, Zhang Y, Kooperberg C, Reiner AP, Jackson RD, Bleecker ER, Meyers DA, Li X, Das S, Yu K, LeFaive J, Smith A, Blackwell T, Taliun D, Zollner S, Forer L, Schoenherr S, Fuchsberger C, Pandit A, Zawistowski M, Kheterpal S, Brummett CM, Natarajan P, Schlessinger D, Lee S, Kang HM, Cucca F, Holmen OL, Åsvold BO, Boehnke M, Kathiresan S, Abecasis GR, Chen YE, Willer CJ, Hveem K. Loss-of-function genomic variants highlight potential therapeutic targets for cardiovascular disease. Nat Commun 2020; 11:6417. [PMID: 33339817 PMCID: PMC7749177 DOI: 10.1038/s41467-020-20086-3] [Show More Authors] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Pharmaceutical drugs targeting dyslipidemia and cardiovascular disease (CVD) may increase the risk of fatty liver disease and other metabolic disorders. To identify potential novel CVD drug targets without these adverse effects, we perform genome-wide analyses of participants in the HUNT Study in Norway (n = 69,479) to search for protein-altering variants with beneficial impact on quantitative blood traits related to cardiovascular disease, but without detrimental impact on liver function. We identify 76 (11 previously unreported) presumed causal protein-altering variants associated with one or more CVD- or liver-related blood traits. Nine of the variants are predicted to result in loss-of-function of the protein. This includes ZNF529:p.K405X, which is associated with decreased low-density-lipoprotein (LDL) cholesterol (P = 1.3 × 10-8) without being associated with liver enzymes or non-fasting blood glucose. Silencing of ZNF529 in human hepatoma cells results in upregulation of LDL receptor and increased LDL uptake in the cells. This suggests that inhibition of ZNF529 or its gene product should be prioritized as a novel candidate drug target for treating dyslipidemia and associated CVD.
Collapse
Affiliation(s)
- Jonas B Nielsen
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA.
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.
| | - Oren Rom
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Ida Surakka
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Sarah E Graham
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Wei Zhou
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Tanmoy Roychowdhury
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Lars G Fritsche
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sarah A Gagliano Taliun
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Carlo Sidore
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | - Yuhao Liu
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Brooke Wolford
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - William Overton
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Ying Zhao
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Jin Chen
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - He Zhang
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Whitney E Hornsby
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Akua Acheampong
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Austen Grooms
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Amanda Schaefer
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Gregory J M Zajac
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Luis Villacorta
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Ben Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Mari Løset
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
- Department of Dermatology, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Vivek Rai
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Pia R Lundegaard
- Laboratory for Molecular Cardiology, Department of Cardiology, Centre for Cardiac, Vascular, Pulmonary and Infectious Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten S Olesen
- Laboratory for Molecular Cardiology, Department of Cardiology, Centre for Cardiac, Vascular, Pulmonary and Infectious Diseases, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics and Los Angeles Biomedical Research Institute, Harbor-UCLA, Torrance, CA, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yii-Der Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics and Los Angeles Biomedical Research Institute, Harbor-UCLA, Torrance, CA, USA
| | - Seung H Choi
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Steven A Lubitz
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen C Barnes
- Colorado Center for Personalized Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Michelle Daya
- Colorado Center for Personalized Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Nicholas Rafaels
- Colorado Center for Personalized Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Department of Medicine Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Ramachandran S Vasan
- Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
- Framingham Heart Study, Framingham, MA, USA
| | - L Adrienne Cupples
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Rasika A Mathias
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lisa R Yanek
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lewis C Becker
- GeneSTAR Research Program, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Stella Aslibekyan
- The University of Alabama at Birmingham, Birmingham, AL, USA
- 23andMe, Inc., Sunnyvale, CA, USA
| | | | - Donna K Arnett
- Deans Office, College of Public Health, University of Kentucky, Lexington, KY, USA
| | | | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
- Jackson Heart Study, Jackson, MS, USA
| | - Solomon K Musani
- Jackson Heart Study, Jackson, MS, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Adolfo Correa
- Jackson Heart Study, Jackson, MS, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics and Los Angeles Biomedical Research Institute, Harbor-UCLA, Torrance, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics and Los Angeles Biomedical Research Institute, Harbor-UCLA, Torrance, CA, USA
| | - Barbara A Konkle
- BloodWorks Northwest, University of Washington, Seattle, WA, USA
| | - Jill M Johnsen
- BloodWorks Northwest, University of Washington, Seattle, WA, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Marilyn J Telen
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Vivien A Sheehan
- Department of Pediatrics, Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Joanne E Curran
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Juan M Peralta
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Courtney Montgomery
- Department of Genes and Human Disease, Oklahoma Medical Research Foundation, Oklahoma, OK, USA
| | - Wayne H-H Sheu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ren-Hua Chung
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Seyed M Nouraie
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Yingze Zhang
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Rebecca D Jackson
- Division of Endocrinology, Diabetes and Metabolism, Ohio State University, Columbus, OH, USA
| | | | - Deborah A Meyers
- Division of Pharmacogenomics University of Arizona, Tucson, AR, USA
| | - Xingnan Li
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AR, USA
| | - Sayantan Das
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Ketian Yu
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jonathon LeFaive
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Albert Smith
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Tom Blackwell
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Daniel Taliun
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sebastian Zollner
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Lukas Forer
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, AR, USA
| | - Sebastian Schoenherr
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Fuchsberger
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Anita Pandit
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Matthew Zawistowski
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sachin Kheterpal
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, US National Institutes of Health, Baltimore, MD, USA
| | - Seunggeun Lee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Hyun Min Kang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Oddgeir L Holmen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
| | - Bjørn O Åsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Michael Boehnke
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sekar Kathiresan
- Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MD, USA
| | - Goncalo R Abecasis
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Y Eugene Chen
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA.
| | - Cristen J Willer
- Department of Internal Medicine: Cardiology, University of Michigan, Ann Arbor, MI, USA.
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA.
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway.
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, Levanger, Norway.
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway.
| |
Collapse
|
228
|
Loss-of-function genomic variants highlight potential therapeutic targets for cardiovascular disease. Nat Commun 2020. [PMID: 33339817 DOI: 10.1038/s41467−020−17792−3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Pharmaceutical drugs targeting dyslipidemia and cardiovascular disease (CVD) may increase the risk of fatty liver disease and other metabolic disorders. To identify potential novel CVD drug targets without these adverse effects, we perform genome-wide analyses of participants in the HUNT Study in Norway (n = 69,479) to search for protein-altering variants with beneficial impact on quantitative blood traits related to cardiovascular disease, but without detrimental impact on liver function. We identify 76 (11 previously unreported) presumed causal protein-altering variants associated with one or more CVD- or liver-related blood traits. Nine of the variants are predicted to result in loss-of-function of the protein. This includes ZNF529:p.K405X, which is associated with decreased low-density-lipoprotein (LDL) cholesterol (P = 1.3 × 10-8) without being associated with liver enzymes or non-fasting blood glucose. Silencing of ZNF529 in human hepatoma cells results in upregulation of LDL receptor and increased LDL uptake in the cells. This suggests that inhibition of ZNF529 or its gene product should be prioritized as a novel candidate drug target for treating dyslipidemia and associated CVD.
Collapse
|
229
|
Pope JE, Choy EH. C-reactive protein and implications in rheumatoid arthritis and associated comorbidities. Semin Arthritis Rheum 2020; 51:219-229. [PMID: 33385862 DOI: 10.1016/j.semarthrit.2020.11.005] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/19/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
C-reactive protein (CRP) is routinely assessed as a marker of systemic inflammation in rheumatoid arthritis (RA). However, it is also an immune regulator that plays an important role in inflammatory pathways associated with RA and promotes atherogenic effects. Comorbidities linked to systemic inflammation are common in RA, and CRP has been associated with the risk for cardiovascular disease, diabetes, metabolic syndrome, pulmonary diseases, and depression. The relationship between systemic inflammation, CRP, and comorbidities in RA is complex, and it is challenging to determine how changing CRP levels may affect the risk or progression of these comorbidities. We review the biological role of CRP in RA and its implications for disease activity and treatment response. We also discuss the impact of treatment on CRP levels and whether reducing systemic inflammation and inhibiting CRP-mediated inflammatory pathways may have an impact on conditions commonly comorbid with RA.
Collapse
Affiliation(s)
- Janet E Pope
- Janet E. Pope: Schulich School of Medicine, University of Western Ontario, St. Joseph's Health Care, London, ON, Canada
| | - Ernest H Choy
- Ernest H. Choy: Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom.
| |
Collapse
|
230
|
Pieters M, Ferreira M, de Maat MPM, Ricci C. Biomarker association with cardiovascular disease and mortality - The role of fibrinogen. A report from the NHANES study. Thromb Res 2020; 198:182-189. [PMID: 33360152 DOI: 10.1016/j.thromres.2020.12.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/26/2020] [Accepted: 12/09/2020] [Indexed: 01/27/2023]
Abstract
BACKGROUND While fibrinogen is a known cardiovascular disease (CVD) risk marker, its quantitative input to mortality risk is a topic of debate. METHODS We investigated the contribution of fibrinogen, among that of other biomarkers, to prevalent CVD and incident CVD mortality in 4487 participants of the US National Health and Nutrition Examination Survey (NHANES). Participants were observed for a median period of 14 years, resulting in more than 58,000 person-years. RESULTS At baseline 551 participants had CVD and during follow up, 1339 all-cause deaths occurred, 321 (24%) of which were due to CVD. Hierarchical cluster analysis and principal component analysis (PCA) were performed to derive clusters of association between biomarkers. Next, structural equation modelling was performed to investigate the association of these clusters with baseline CVD and all-cause and CVD mortality during follow-up. Mediation analysis was used to determine which biomarkers played a mediatory role between prevalent CVD and future mortality. Fibrinogen clustered with C-reactive protein only and was associated with CVD at baseline (p < 0.0001) and with all-cause (p < 0.001) and CVD (p < 0.001) mortality at follow-up. Only fibrinogen (4.7%), followed by gamma-glutamyl transferase (GGT) (3.5%) and uric acid (2.3%) were identified as possible mediators between CVD status and all-cause mortality, with fibrinogen (3.2%) and GGT (3.1%) the only mediators between CVD status and CVD mortality. CONCLUSION This data shows that fibrinogen is not only cross-sectionally associated with CVD, but also contributes to all-cause and CVD mortality at follow-up. It furthermore appears to mediate the association between prevalent CVD and both all-cause and CVD mortality.
Collapse
Affiliation(s)
- Marlien Pieters
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa; Medical Research Council Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa.
| | - Maylene Ferreira
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa
| | - Moniek P M de Maat
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Cristian Ricci
- Centre of Excellence for Nutrition, North-West University, Potchefstroom, South Africa; Pediatric Epidemiology, Department of Pediatrics, University Medicine Leipzig, Leipzig, Germany
| |
Collapse
|
231
|
Tang H, Cheng Z, Li N, Mao S, Ma R, He H, Niu Z, Chen X, Xiang H. The short- and long-term associations of particulate matter with inflammation and blood coagulation markers: A meta-analysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115630. [PMID: 33254709 PMCID: PMC7687019 DOI: 10.1016/j.envpol.2020.115630] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 05/16/2023]
Abstract
Inflammation and the coagulation cascade are considered to be the potential mechanisms of ambient particulate matter (PM) exposure-induced adverse cardiovascular events. Tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-8 (IL-8), and fibrinogen are arguably the four most commonly assayed markers to reflect the relationships of PM with inflammation and blood coagulation. This review summarized and quantitatively analyzed the existing studies reporting short- and long-term associations of PM2.5(PM with an aerodynamic diameter ≤2.5 μm)/PM10 (PM with an aerodynamic diameter≤10 μm) with important inflammation and blood coagulation markers (TNF-α, IL-6, IL-8, fibrinogen). We reviewed relevant studies published up to July 2020, using three English databases (PubMed, Web of Science, Embase) and two Chinese databases (Wang-Fang, China National Knowledge Infrastructure). The OHAT tool, with some modification, was applied to evaluate risk of bias. Meta-analyses were conducted with random-effects models for calculating the pooled estimate of markers. To assess the potential effect modifiers and the source of heterogeneity, we conducted subgroup analyses and meta-regression analyses where appropriate. The assessment and correction of publication bias were based on Begg's and Egger's test and "trim-and-fill" analysis. We identified 44 eligible studies. For short-term PM exposure, the percent change of a 10 μg/m3 PM2.5 increase on TNF-α and fibrinogen was 3.51% (95% confidence interval (CI): 1.21%, 5.81%) and 0.54% (95% confidence interval (CI): 0.21%, 0.86%) respectively. We also found a significant short-term association between PM10 and fibrinogen (percent change = 0.17%, 95% CI: 0.04%, 0.29%). Overall analysis showed that long-term associations of fibrinogen with PM2.5 and PM10 were not significant. Subgroup analysis showed that long-term associations of fibrinogen with PM2.5 and PM10 were significant only found in studies conducted in Asia. Our findings support significant short-term associations of PM with TNF-α and fibrinogen. Future epidemiological studies should address the role long-term PM exposure plays in inflammation and blood coagulation markers level change.
Collapse
Affiliation(s)
- Hong Tang
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Zilu Cheng
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, 122# Luoshi Road, Wuhan, China
| | - Na Li
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Shuyuan Mao
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Runxue Ma
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Haijun He
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Zhiping Niu
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Xiaolu Chen
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China
| | - Hao Xiang
- Department of Global Health, School of Health Sciences, Wuhan University, 115# Donghu Road, Wuhan, China; Global Health Institute, Wuhan University, 115# Donghu Road, Wuhan, China.
| |
Collapse
|
232
|
Zhu C, Li W, Wang X, Xue J, Zhao L, Song Y, Zhou T, Zhang M. Thiopental sodium loaded solid lipid nano-particles attenuates obesity-induced cardiac dysfunction and cardiac hypertrophy via inactivation of inflammatory pathway. Drug Deliv 2020; 27:1188-1200. [PMID: 32762480 PMCID: PMC7470049 DOI: 10.1080/10717544.2020.1803449] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/27/2020] [Indexed: 11/30/2022] Open
Abstract
This work evaluates solid lipid nanoparticles of thiopental sodium against obesity-induced cardiac dysfunction and hypertrophy and explores the possible mechanism of action. TS loaded SLNs were formulated by hot-homogenization and solvent diffusion method. TS-SLNs were scrutinized for entrapment efficiency, drug loading capacity, gastric stability, particle size, in vitro drug release. Mice were feed with the normal chow or high-fat diet for 08 weeks to induce obesity and primary cardiomyocytes. The therapeutic effects of thiopental sodium in the high fat diet (HFD) induced cardiac hypertrophy. Systolic blood pressure (SBP) was estimated at a regular time interval. At the end of the experimental study, systolic pressure left ventricular, LV end-diastolic pressure and rate of increase of LV pressure and antioxidant, apoptosis, cytokines and inflammatory scrutinized. HFD induced group mice exhibited a reduction in the body weight and enhancement of cardiac hypertrophy marker and dose-dependent treatment of thiopental sodium up-regulation the body weight and down-regulated the cardiac hypertrophy. Thiopental sodium significantly (p < .001) dose-dependently altered the antioxidant, biochemical, cardiac parameters and remodeling. Thiopental sodium significantly (p < .001) dose-dependently reduced the SBP. Thiopental sodium altered the apoptosis marker, pro-inflammatory cytokines, inflammatory parameters along with reduced the p38-MAPK level. The cardiac protective effect of thiopental sodium shed light on future therapeutic interventions in obesity and related cardiovascular complications via inflammatory pathway.
Collapse
Affiliation(s)
- Canzhan Zhu
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Wanjing Li
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Xinhong Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Jiahong Xue
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Ling Zhao
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Yafan Song
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Tian Zhou
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| | - Mingjuan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi’an, Shaanxi, 710004, China
| |
Collapse
|
233
|
Kott KA, Vernon ST, Hansen T, de Dreu M, Das SK, Powell J, Fazekas de St Groth B, Di Bartolo BA, McGuire HM, Figtree GA. Single-Cell Immune Profiling in Coronary Artery Disease: The Role of State-of-the-Art Immunophenotyping With Mass Cytometry in the Diagnosis of Atherosclerosis. J Am Heart Assoc 2020; 9:e017759. [PMID: 33251927 PMCID: PMC7955359 DOI: 10.1161/jaha.120.017759] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Coronary artery disease remains the leading cause of death globally and is a major burden to every health system in the world. There have been significant improvements in risk modification, treatments, and mortality; however, our ability to detect asymptomatic disease for early intervention remains limited. Recent discoveries regarding the inflammatory nature of atherosclerosis have prompted investigation into new methods of diagnosis and treatment of coronary artery disease. This article reviews some of the highlights of the important developments in cardioimmunology and summarizes the clinical evidence linking the immune system and atherosclerosis. It provides an overview of the major serological biomarkers that have been associated with atherosclerosis, noting the limitations of these markers attributable to low specificity, and then contrasts these serological markers with the circulating immune cell subtypes that have been found to be altered in coronary artery disease. This review then outlines the technique of mass cytometry and its ability to provide high-dimensional single-cell data and explores how this high-resolution quantification of specific immune cell subpopulations may assist in the diagnosis of early atherosclerosis in combination with other complimentary techniques such as single-cell RNA sequencing. We propose that this improved specificity has the potential to transform the detection of coronary artery disease in its early phases, facilitating targeted preventative approaches in the precision medicine era.
Collapse
Affiliation(s)
- Katharine A Kott
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia
| | - Stephen T Vernon
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia
| | - Thomas Hansen
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia
| | - Macha de Dreu
- School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Ramaciotti Facility for Human Systems Biology Charles Perkins Centre University of Sydney Sydney Australia
| | - Souvik K Das
- Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia
| | - Joseph Powell
- Garvan-Weizmann Centre for Cellular Genomics Garvan Institute Sydney Australia.,UNSW Cellular Genomics Futures Institute University of New South Wales Sydney Australia
| | - Barbara Fazekas de St Groth
- School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Ramaciotti Facility for Human Systems Biology Charles Perkins Centre University of Sydney Sydney Australia.,Charles Perkins Centre University of Sydney Sydney Australia
| | - Belinda A Di Bartolo
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia
| | - Helen M McGuire
- School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Ramaciotti Facility for Human Systems Biology Charles Perkins Centre University of Sydney Sydney Australia.,Charles Perkins Centre University of Sydney Sydney Australia
| | - Gemma A Figtree
- Cardiothoracic and Vascular Health Kolling Institute of Medical Research Sydney Australia.,Department of Cardiology Royal North Shore Hospital Northern Sydney Local Health District Sydney Australia.,School of Medical Sciences Faculty of Medicine and Health University of Sydney Sydney Australia.,Charles Perkins Centre University of Sydney Sydney Australia
| |
Collapse
|
234
|
Alfaddagh A, Martin SS, Leucker TM, Michos ED, Blaha MJ, Lowenstein CJ, Jones SR, Toth PP. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am J Prev Cardiol 2020; 4:100130. [PMID: 34327481 PMCID: PMC8315628 DOI: 10.1016/j.ajpc.2020.100130] [Citation(s) in RCA: 206] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation constitutes a complex, highly conserved cascade of molecular and cellular events. Inflammation has been labeled as “the fire within,” is highly regulated, and is critical to host defense and tissue repair. In general, inflammation is beneficial and has evolved to promote survival. However, inflammation can also be maladaptive when chronically activated and sustained, leading to progressive tissue injury and reduced survival. Examples of a maladaptive response include rheumatologic disease and atherosclerosis. Despite evidence gathered by Virchow over 100 years ago showing that inflammatory white cells play a role in atherogenesis, atherosclerosis was until recently viewed as a disease of passive cholesterol accumulation in the subendothelial space. This view has been supplanted by considerable basic scientific and clinical evidence demonstrating that every step of atherogenesis, from the development of endothelial cell dysfunction to foam cell formation, plaque formation and progression, and ultimately plaque rupture stemming from architectural instability, is driven by the cytokines, interleukins, and cellular constituents of the inflammatory response. Herein we provide an overview of the role of inflammation in atherosclerotic cardiovascular disease, discuss the predictive value of various biomarkers involved in inflammation, and summarize recent clinical trials that evaluated the capacity of various pharmacologic interventions to attenuate the intensity of inflammation and impact risk for acute cardiovascular events.
Collapse
Affiliation(s)
- Abdulhamied Alfaddagh
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth S Martin
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten M Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erin D Michos
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael J Blaha
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles J Lowenstein
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven R Jones
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter P Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
235
|
Paoli A, Gorini S, Caprio M. The dark side of the spoon - glucose, ketones and COVID-19: a possible role for ketogenic diet? J Transl Med 2020; 18:441. [PMID: 33218357 PMCID: PMC7677746 DOI: 10.1186/s12967-020-02600-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/30/2020] [Indexed: 02/06/2023] Open
Abstract
The novel coronavirus disease (COVID-19) is posing a serious challenge to the health-care systems worldwide, with an enormous impact on health conditions and loss of lives. Notably, obesity and its related comorbidities are strictly related with worse clinical outcomes of COVID-19 disease. Recently, there is a growing interest in the clinical use of ketogenic diets (KDs), particularly in the context of severe obesity with related metabolic complications. KDs have been proven effective for a rapid reduction of fat mass, preserving lean mass and providing an adequate nutritional status. In particular, the physiological increase in plasma levels of ketone bodies exerts important anti-inflammatory and immunomodulating effects, which may reveal as precious tools to prevent infection and potential adverse outcomes of COVID-19 disease. We discuss here the importance of KDs for a rapid reduction of several critical risk factors for COVID-19, such as obesity, type 2 diabetes and hypertension, based on the known effects of ketone bodies on inflammation, immunity, metabolic profile and cardiovascular function. We do believe that a rapid reduction of all modifiable risk factors, especially obesity with its metabolic complications, should be a pillar of public health policies and interventions, in view of future waves of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta, 247, 00166, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta, 247, 00166, Rome, Italy. .,Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta, 247, 00166, Rome, Italy.
| |
Collapse
|
236
|
The Effects of Exposure Time on Systemic Inflammation in Subjects With Exposure to Zinc- and Copper-Containing Brazing Fumes. J Occup Environ Med 2020; 61:806-811. [PMID: 31348430 DOI: 10.1097/jom.0000000000001676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Inhalation of copper and zinc containing brazing fumes (2.5 mg/m for 6 hours) is able to induce asymptomatic systemic inflammation which is supposed to be connected with an increased risk for cardiovascular disease. In this study it was investigated if inflammation can be prevented by reducing the exposure time. METHODS A total of 15 healthy male subjects were exposed to such brazing fumes in a crossover design for 3, 4, and 5 hours in randomized order. Before and 24 hours after exposure, blood samples were taken and c-reactive protein (CRP) as marker for an acute phase reaction was measured. RESULTS Five-hour exposure induced an increase of CRP, whereas the shorter exposure times did not result in a significant inflammatory reaction. CONCLUSIONS Reducing daily exposure times below 5 hours is able to prevent systemic inflammatory reactions.
Collapse
|
237
|
Sesti F, Pofi R, Pozza C, Minnetti M, Gianfrilli D, Kanakis GA. Cardiovascular Complications in Patients with Klinefelter's Syndrome. Curr Pharm Des 2020; 26:5556-5563. [PMID: 33138758 DOI: 10.2174/1381612826666201102105408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 10/01/2020] [Indexed: 12/26/2022]
Abstract
More than 70 years have passed since the first description of Klinefelter Syndrome (KS), the most frequent chromosome disorder causing male infertility and hypogonadism. KS is associated with increased cardiovascular (CV) mortality due to several comorbidities, including hypogonadism, as well as metabolic syndrome and type 2 diabetes, which are highly prevalent in these patients. Aside from metabolic disturbances, patients with KS suffer from both acquired and congenital CV abnormalities, cerebrovascular thromboembolic disease, subclinical atherosclerosis and endothelial dysfunction, which may all contribute to increased CV mortality. The mechanisms involved in this increased risk of CV morbidity and mortality are not entirely understood. More research is needed to better characterise the CV manifestations, elucidate the pathophysiological mechanisms and define the contribution of testosterone replacement to restoring CV health in KS patients. This review explores the complex association between KS, metabolic syndrome and CV risk in order to plan future studies and improve strategies to reduce mortality in this high-risk population.
Collapse
Affiliation(s)
- Franz Sesti
- Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, 00161, Rome, Italy
| | - Riccardo Pofi
- Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, 00161, Rome, Italy
| | - Carlotta Pozza
- Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, 00161, Rome, Italy
| | - Marianna Minnetti
- Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, 00161, Rome, Italy
| | - Daniele Gianfrilli
- Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, 00161, Rome, Italy
| | - George A Kanakis
- Department of Endocrinology, Athens Naval & VA Hospital, 11525, Athens, Greece
| |
Collapse
|
238
|
Cardiovascular events prediction by left ventricular longitudinal strain and serum high-sensitivity troponin I in patients with axial spondyloarthritis. Clin Rheumatol 2020; 39:3373-3382. [DOI: 10.1007/s10067-020-05112-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/08/2020] [Accepted: 04/16/2020] [Indexed: 10/24/2022]
|
239
|
Joint effect of blood pressure and C-reactive protein and the risk of sudden cardiac death: A prospective cohort study. Int J Cardiol 2020; 326:184-188. [PMID: 33130259 DOI: 10.1016/j.ijcard.2020.10.071] [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: 05/23/2020] [Revised: 09/26/2020] [Accepted: 10/23/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Both blood pressure and C-reactive protein (CRP) are each independently related to mortality risk. However, the combined effect of systolic blood pressure (SBP) and CRP on sudden cardiac death (SCD) risk has not been studied. PATIENTS AND METHODS We studied the joint impact of SBP and CRP and the risk of SCD in the Kuopio Ischemic Heart Disease prospective cohort study of 1953 men aged 42-61 years with no history of ischemic heart disease. Baseline investigations were conducted between March 1984 and December 1989. SBP and CRP were measured. SBP was divided based on median values to low and high (median cutoffs 132 mmHg) and CRP as low and high (median cut-off 1.30 mg/L). Hazard ratios (HRs) with confidence intervals (CIs) were calculated after multivariate adjustment. RESULTS Subjects were followed-up for 23.2 years, and 137 SCDs occurred. In this study, elevated SBP (>132 mmHg) combined with elevated (CRP >1.30 mg/L) were associated with SCD risk. Adjustment for age, examination year, alcohol consumption, BMI, energy expenditure during exercise, total cholesterol, HDL-cholesterol, type 2 diabetes, smoking, antihypertension medication and aspirin use, the risk of SCD remained statistically significant (HR, 2,73, 95% CI, 1.62-4.60, p < .001). Further adjustment for socio-economic status, years of education and history of cardiovascular disease in a family the results were only slightly changed (HR, 2.65, 95% CI, 1.57-4.49, p < .001). CONCLUSIONS In our male cohort study, the joint effect of high SBP together with increased CRP levels is a risk predictor of SCD compared with low SBP and CRP.
Collapse
|
240
|
Morris G, Puri BK, Olive L, Carvalho A, Berk M, Walder K, Gustad LT, Maes M. Endothelial dysfunction in neuroprogressive disorders-causes and suggested treatments. BMC Med 2020; 18:305. [PMID: 33070778 PMCID: PMC7570030 DOI: 10.1186/s12916-020-01749-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Potential routes whereby systemic inflammation, oxidative stress and mitochondrial dysfunction may drive the development of endothelial dysfunction and atherosclerosis, even in an environment of low cholesterol, are examined. MAIN TEXT Key molecular players involved in the regulation of endothelial cell function are described, including PECAM-1, VE-cadherin, VEGFRs, SFK, Rho GEF TRIO, RAC-1, ITAM, SHP-2, MAPK/ERK, STAT-3, NF-κB, PI3K/AKT, eNOS, nitric oxide, miRNAs, KLF-4 and KLF-2. The key roles of platelet activation, xanthene oxidase and myeloperoxidase in the genesis of endothelial cell dysfunction and activation are detailed. The following roles of circulating reactive oxygen species (ROS), reactive nitrogen species and pro-inflammatory cytokines in the development of endothelial cell dysfunction are then described: paracrine signalling by circulating hydrogen peroxide, inhibition of eNOS and increased levels of mitochondrial ROS, including compromised mitochondrial dynamics, loss of calcium ion homeostasis and inactivation of SIRT-1-mediated signalling pathways. Next, loss of cellular redox homeostasis is considered, including further aspects of the roles of hydrogen peroxide signalling, the pathological consequences of elevated NF-κB, compromised S-nitrosylation and the development of hypernitrosylation and increased transcription of atherogenic miRNAs. These molecular aspects are then applied to neuroprogressive disorders by considering the following potential generators of endothelial dysfunction and activation in major depressive disorder, bipolar disorder and schizophrenia: NF-κB; platelet activation; atherogenic miRs; myeloperoxidase; xanthene oxidase and uric acid; and inflammation, oxidative stress, nitrosative stress and mitochondrial dysfunction. CONCLUSIONS Finally, on the basis of the above molecular mechanisms, details are given of potential treatment options for mitigating endothelial cell dysfunction and activation in neuroprogressive disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | | | - Lisa Olive
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- School of Psychology, Faculty of Health, Deakin University, Geelong, Australia
| | - Andre Carvalho
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Berk
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia.
- Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia.
| | - Ken Walder
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Lise Tuset Gustad
- Department of Circulation and medical imaging, Norwegian University of Technology and Science (NTNU), Trondheim, Norway
- Nord-Trøndelag Hospital Trust, Levanger Hospital, Levanger, Norway
| | - Michael Maes
- IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
241
|
van Zyl S, van Rooyen FC, Joubert G, Kruger WH, Walsh CM. A Comparison of the Socio-Behavioral-Metabolic Risk Profiles and Associated Factors for Chronic Diseases of Lifestyle in Urban and Rural Communities in Central South Africa. Front Public Health 2020; 8:570676. [PMID: 33178661 PMCID: PMC7596254 DOI: 10.3389/fpubh.2020.570676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/15/2020] [Indexed: 11/30/2022] Open
Abstract
Background: The global escalating prevalence of lifestyle-related non-communicable diseases places a significant burden on health systems. Chronic diseases of lifestyle (CDL) are a group of diseases that share similar modifiable risk factors that can result in long-term disease processes. Considering the socio-behavioral-metabolic risk profiles of communities and risk factors predictive of the presence of CDL can assist in the development of focused and effective community-based prevention, intervention, and treatment programs for CDL. Aim: To determine the socio-behavioral-metabolic risk profiles and identify associated factors for the following CDL: obesity, cardiovascular disease, hypertension, and type 2 diabetes mellitus in rural and urban communities in central South Africa. Methodology: This cross-sectional study included adults aged 25–65 years in the rural Southern Free State and urban Mangaung. Social determinants, behavioral and metabolic risk factors, and inflammatory biomarkers for CDL were determined. Results: In total, 575 rural (mean age: 42 years; 71% female) and 429 urban (mean age: 44 years; 76% female) participants were included in the study. More than 20% of participants in both communities reported being previously diagnosed with cardiovascular diseases; with reported hypertension and diabetes mellitus more prevalent among rural participants. Insufficient intake of fruit and vegetables, alcohol use, and high blood pressure were among the top five risk factors in both communities. Physical inactivity ranked among the top two risk factors in the urban community; while alcohol and tobacco use was significantly higher in the rural community. Fibrinogen was the most prevalent inflammatory marker in both communities (32.9 rural vs. 48.3% urban). High sensitivity C-reactive protein (Hs-CRP), only available for rural participants, was high with increased levels in more than 80% of participants. In both communities, being female, having high blood pressure and increased fibrinogen levels were associated with obesity. Conclusion: This study illustrated the high prevalence of socio-behavioral-metabolic risk factors for CDL, and identified similarities and distinct differences in the risk profiles of rural and urban communities. Considering the CDL risk profiles of communities can assist in prioritizing health needs and contribute to the development of tailor-made community-based primary health care prevention, intervention, and health promotion programs.
Collapse
Affiliation(s)
- Sanet van Zyl
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Francios C van Rooyen
- Department of Biostatistics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Gina Joubert
- Department of Biostatistics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Willem H Kruger
- Department of Community Health, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Corinna M Walsh
- Department of Nutrition and Dietetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
242
|
Reduction in hsCRP levels is associated with decreased incidence of cardiovascular events in Japanese hypertensive women but not in men. Sci Rep 2020; 10:17040. [PMID: 33046765 PMCID: PMC7550334 DOI: 10.1038/s41598-020-73905-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/23/2020] [Indexed: 12/03/2022] Open
Abstract
To test our hypothesis that the magnitude of reduction in hsCRP achieved by antihypertensive medications may predict the benefit for cardiovascular outcomes in hypertensive individuals, we performed subanalysis of the ATTEMPT-CVD study. The hypertensive participants enrolled in the ATTMEPT-CVD study were categorized into two groups according to whether achieved reduction in hsCRP levels at 6 months after initiation of antihypertensive medications from baseline was equal to or greater than 40% (responder group) or less than 40% (non-responder group). Baseline characteristics and blood pressure during follow-up period were similar between the groups. For women, the incidence of cardiovascular events was significantly less in responder group than non-responder group (P < 0.0221). However, for men, there was no significant difference between the groups regarding incident cardiovascular events (P = 0.2434). There was a significant interaction (P = 0.0187) between sexes for incident cardiovascular events. Our results provide the evidence suggesting that substantial reduction (40% or greater reduction) in hsCRP on antihypertensive medication predicts the benefit for cardiovascular outcomes in hypertensive women but it does not in hypertensive men. The magnitude of achieved reduction in hsCRP by antihypertensive medications seems to be a useful indicator of successful treatment in Japanese hypertensive women. This trial was registered with ClinicalTrials.gov, number NCT01075698.
Collapse
|
243
|
Combating Inflammation in Cardiovascular Disease. Heart Lung Circ 2020; 30:197-206. [PMID: 33039279 DOI: 10.1016/j.hlc.2020.09.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 08/10/2020] [Accepted: 09/03/2020] [Indexed: 12/24/2022]
Abstract
The role of inflammation in promoting atherosclerosis and subsequent cardiovascular disease is increasingly recognised, particularly after the publication of Anti-inflammatory Therapy with Canakinumab for Atherosclerotic Disease (CANTOS) and Colchicine Cardiovascular Outcomes (COLCOT) trials. It appears that specifically targeting the Nod-like receptor protein 3 (NLRP3) inflammasome-interleukin 1/interleukin 18-interleukin 6 pathway appears to be most beneficial in cardiovascular risk reduction. High sensitivity C-reactive protein (CRP) is a downstream biomarker of inflammation that can be used to monitor treatment. This article will discuss the role of inflammation in cardiovascular disease, the utility of high sensitivity C-reactive protein and treatments that target this inflammation. While further research is needed into the cost effectiveness and safety of newer agents, it remains an evolving approach to manage cardiovascular risk.
Collapse
|
244
|
Welsh P, Welsh C, Celis-Morales CA, Brown R, Ho FK, Ferguson LD, Mark PB, Lewsey J, Gray SR, Lyall DM, Gill JMR, Pell JP, de Lemos JA, Willeit P, Sattar N. Lipoprotein(a) and cardiovascular disease: prediction, attributable risk fraction, and estimating benefits from novel interventions. Eur J Prev Cardiol 2020; 28:1991-2000. [PMID: 33624048 DOI: 10.1093/eurjpc/zwaa063] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022]
Abstract
AIMS To investigate the population attributable fraction due to elevated lipoprotein (a) (Lp(a)) and the utility of measuring Lp(a) in cardiovascular disease (CVD) risk prediction. METHODS AND RESULTS In 413 734 participants from UK Biobank, associations of serum Lp(a) with composite fatal/non-fatal CVD (n = 10 066 events), fatal CVD (n = 3247), coronary heart disease (CHD; n = 18 292), peripheral vascular disease (PVD; n = 2716), and aortic stenosis (n = 901) were compared using Cox models. Median Lp(a) was 19.7 nmol/L (interquartile interval 7.6-75.3 nmol/L). About 20.8% had Lp(a) values >100 nmol/L; 9.2% had values >175 nmol/L. After adjustment for classical risk factors, 1 SD increment in log Lp(a) was associated with a hazard ratio for fatal/non-fatal CVD of 1.12 [95% confidence interval (CI) 1.10-1.15]. Similar associations were observed with fatal CVD, CHD, PVD, and aortic stenosis. Adding Lp(a) to a prediction model containing traditional CVD risk factors in a primary prevention group improved the C-index by +0.0017 (95% CI 0.0008-0.0026). In the whole cohort, Lp(a) above 100 nmol/L was associated with a population attributable fraction (PAF) of 5.8% (95% CI 4.9-6.7%), and for Lp(a) above 175 nmol/L the PAF was 3.0% (2.4-3.6%). Assuming causality and an achieved Lp(a) reduction of 80%, an ongoing trial to lower Lp(a) in patients with CVD and Lp(a) above 175 nmol/L may reduce CVD risk by 20.0% and CHD by 24.4%. Similar benefits were also modelled in the whole cohort, regardless of baseline CVD. CONCLUSION Population screening for elevated Lp(a) may help to predict CVD and target Lp(a) lowering drugs, if such drugs prove efficacious, to those with markedly elevated levels.
Collapse
Affiliation(s)
- Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Claire Welsh
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Carlos A Celis-Morales
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK.,Institute of Health and Wellbeing, University of Glasgow, 1 Lilybank Gardens, Glasgow G12 8RZ, UK
| | - Rosemary Brown
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Frederick K Ho
- Institute of Health and Wellbeing, University of Glasgow, 1 Lilybank Gardens, Glasgow G12 8RZ, UK
| | - Lyn D Ferguson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - James Lewsey
- Institute of Health and Wellbeing, University of Glasgow, 1 Lilybank Gardens, Glasgow G12 8RZ, UK
| | - Stuart R Gray
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, 1 Lilybank Gardens, Glasgow G12 8RZ, UK
| | - Jason M R Gill
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, 1 Lilybank Gardens, Glasgow G12 8RZ, UK
| | - James A de Lemos
- Division of Cardiology, Department of Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Peter Willeit
- Department of Neurology, Medical University of Innsbruck, Anichstraße 35, Innsbruck 6020, Austria.,Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
245
|
Ozdemir B. Correlation of C-Reactive Protein and Serum Iron Levels with Syntax Score. ARCHIVES OF RAZI INSTITUTE 2020; 75:413-418. [PMID: 33025782 PMCID: PMC8418810 DOI: 10.22092/ari.2020.128122.1404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/12/2020] [Indexed: 09/30/2022]
Abstract
Cardiovascular disease is one of the most common causes of morbidity and mortality in the world. Atherosclerosis is an inflammatory process, and serum C-reactive protein (CRP) is an acute-phase protein rising in response to inflammation. Serum iron (Fe) is one of the essential metals for the human body. Inflammation and infection are characterized by changes in Fe metabolism. Since atherosclerosis is an inflammatory process, changes in CRP and serum Fe levels are expected. However, the distribution of the disease in the coronary arteries is important for mortality and morbidity. The distribution of the disease can be determined by the syntax score. This study included 407 patients with a mean age of 56.4±10.7 years. The majority of the patients were male (51.4%). In this study, 53 and 354 patients had critical and no critical lesions, respectively. According to the baseline coronary angiograms, the syntax score was calculated in all patients. The laboratory variables, including hemoglobin levels, blood glucose, creatinine, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, triglycerides, Fe, and CRP were also evaluated in this study. Regarding the laboratory parameters of all groups, the mean CRP levels, Fe levels, and syntax score were estimated at 0.75±1.8 mg/dl, 80.4±27.5 mg/dl, and 1.5±4.8, respectively. Furthermore, a high syntax score correlated with Fe and CRP levels. Based on the findings of the present study, elevated serum Fe and CRP concentrations were associated with increased syntax score and atherosclerosis severity.
Collapse
Affiliation(s)
- B Ozdemir
- Department of Cardiology Faculty of Medicine, Nigde Omer Halisdemir University, Campus, 51240, Nigde, Turkey
- Department of Cardiology Faculty of Medicine, Nigde Omer Halisdemir University, Campus, 51240, Nigde, Turkey
| |
Collapse
|
246
|
Liu J, Zhang Y, Lavie CJ, Tabung FK, Xu J, Hu Q, He L, Zhang Y. Associations of C-reactive protein and fibrinogen with mortality from all-causes, cardiovascular disease and cancer among U.S. adults. Prev Med 2020; 139:106044. [PMID: 32097752 DOI: 10.1016/j.ypmed.2020.106044] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/22/2020] [Accepted: 02/22/2020] [Indexed: 01/05/2023]
Abstract
C-reactive protein (CRP) and fibrinogen are associated with an increased risk of death with suggested differences by gender, diet quality and race/ethnicity. However, the current evidence is limited. We used data including 8646 men and 9880 women from the National Health and Nutrition Examination Survey (NHANES) Linked Morality cohort (1999-2011) to investigate the associations of CRP and fibrinogen with mortality overall and by gender, race/ethnicity and diet quality. Cox-proportional hazard model was used to quantify the associations. With a median follow-up of 6 years, a strong dose-response relationship was observed between CRP levels and mortality risk in men after multivariable adjustment. For subjects who survived the first two years, the adjusted hazard ratios (HRs) for total mortality across quartiles (from lower to higher) of CRP were 1.97 (95% CI: 0.62-6.33), 1.89 (0.59, 6.06) and 6.34 (2.28-17.7) (P for trend <0.001). For cardiovascular disease (CVD) mortality, its association with CRP varied by diet quality. For cancer mortality, its association differed by history of cancer, and positive associations were observed only among subjects with history of cancer. In contrast, no such association of CRP with mortality was found in women. For fibrinogen, we observed its positive association with total mortality and the HRs across quartiles of fibrinogen (from lower to higher) were 1.21 (0.88, 1.67), 1.49 (1.22, 1.82) and 1.99 (1.56, 2.55). The association with CVD mortality differed by diet quality whereas no association was found with cancer mortality. Our findings suggest that higher levels of CRP and fibrinogen were associated with lower survival from total and CVD; the associations of CRP with mortality were more pronounced in men than women. Diet quality is an effect modifier for the association of CRP and fibrinogen with CVD mortality.
Collapse
Affiliation(s)
- Junxiu Liu
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
| | - Yanan Zhang
- Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-the University of Queensland School of Medicine, New Orleans, LA 70121, USA
| | - Fred K Tabung
- Division of Medical Oncology, The Ohio State University College of Medicine and Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, OH 43221, USA
| | - Jiting Xu
- Department of Computer Science and Engineering, University of South Carolina, Columbia, SC 29201, USA
| | - Qingwei Hu
- Department of Public Health Sciences, University of Clemson, Clemson, SC 29631, USA
| | - Lixia He
- Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Yunxiang Zhang
- Department of Pathology, Weifang People's Hospital, Weifang, Shandong 261053, China.
| |
Collapse
|
247
|
Matsushita K, Jassal SK, Sang Y, Ballew SH, Grams ME, Surapaneni A, Arnlov J, Bansal N, Bozic M, Brenner H, Brunskill NJ, Chang AR, Chinnadurai R, Cirillo M, Correa A, Ebert N, Eckardt KU, Gansevoort RT, Gutierrez O, Hadaegh F, He J, Hwang SJ, Jafar TH, Kayama T, Kovesdy CP, Landman GW, Levey AS, Lloyd-Jones DM, Major RW, Miura K, Muntner P, Nadkarni GN, Naimark DMJ, Nowak C, Ohkubo T, Pena MJ, Polkinghorne KR, Sabanayagam C, Sairenchi T, Schneider MP, Shalev V, Shlipak M, Solbu MD, Stempniewicz N, Tollitt J, Valdivielso JM, van der Leeuw J, Wang AYM, Wen CP, Woodward M, Yamagishi K, Yatsuya H, Zhang L, Schaeffner E, Coresh J. Incorporating kidney disease measures into cardiovascular risk prediction: Development and validation in 9 million adults from 72 datasets. EClinicalMedicine 2020; 27:100552. [PMID: 33150324 PMCID: PMC7599294 DOI: 10.1016/j.eclinm.2020.100552] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) measures (estimated glomerular filtration rate [eGFR] and albuminuria) are frequently assessed in clinical practice and improve the prediction of incident cardiovascular disease (CVD), yet most major clinical guidelines do not have a standardized approach for incorporating these measures into CVD risk prediction. "CKD Patch" is a validated method to calibrate and improve the predicted risk from established equations according to CKD measures. METHODS Utilizing data from 4,143,535 adults from 35 datasets, we developed several "CKD Patches" incorporating eGFR and albuminuria, to enhance prediction of risk of atherosclerotic CVD (ASCVD) by the Pooled Cohort Equation (PCE) and CVD mortality by Systematic COronary Risk Evaluation (SCORE). The risk enhancement by CKD Patch was determined by the deviation between individual CKD measures and the values expected from their traditional CVD risk factors and the hazard ratios for eGFR and albuminuria. We then validated this approach among 4,932,824 adults from 37 independent datasets, comparing the original PCE and SCORE equations (recalibrated in each dataset) to those with addition of CKD Patch. FINDINGS We confirmed the prediction improvement with the CKD Patch for CVD mortality beyond SCORE and ASCVD beyond PCE in validation datasets (Δc-statistic 0.027 [95% CI 0.018-0.036] and 0.010 [0.007-0.013] and categorical net reclassification improvement 0.080 [0.032-0.127] and 0.056 [0.044-0.067], respectively). The median (IQI) of the ratio of predicted risk for CVD mortality with CKD Patch vs. the original prediction with SCORE was 2.64 (1.89-3.40) in very high-risk CKD (e.g., eGFR 30-44 ml/min/1.73m2 with albuminuria ≥30 mg/g), 1.86 (1.48-2.44) in high-risk CKD (e.g., eGFR 45-59 ml/min/1.73m2 with albuminuria 30-299 mg/g), and 1.37 (1.14-1.69) in moderate risk CKD (e.g., eGFR 60-89 ml/min/1.73m2 with albuminuria 30-299 mg/g), indicating considerable risk underestimation in CKD with SCORE. The corresponding estimates for ASCVD with PCE were 1.55 (1.37-1.81), 1.24 (1.10-1.54), and 1.21 (0.98-1.46). INTERPRETATION The "CKD Patch" can be used to quantitatively enhance ASCVD and CVD mortality risk prediction equations recommended in major US and European guidelines according to CKD measures, when available. FUNDING US National Kidney Foundation and the NIDDK.
Collapse
Affiliation(s)
- Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Simerjot K Jassal
- Division of General Internal Medicine, University of California, San Diego and VA San Diego Healthcare, San Diego, California
| | - Yingying Sang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Shoshana H Ballew
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Corresponding author.
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Aditya Surapaneni
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Johan Arnlov
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Nisha Bansal
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Milica Bozic
- Vascular & Renal Translational Research Group, IRBLleida, Spain and Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ) and Network Aging Research, University of Heidelberg, Heidelberg, Germany
| | - Nigel J Brunskill
- John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Alex R Chang
- Department of Nephrology and Kidney Health Research Institute, Geisinger Medical Center, Danville, Pennsylvania
| | - Rajkumar Chinnadurai
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
| | - Massimo Cirillo
- Department of Public Health, University of Naples “Federico II”, Italy
| | - Adolfo Correa
- University of Mississippi Medical Center, Jackson, United States
| | - Natalie Ebert
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Orlando Gutierrez
- Departments of Epidemiology and Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute, Framingham, MA, United States
| | - Tazeen H Jafar
- Program in Health Services and Systems Research, Duke-National University of Singapore Medical School, Singapore
- Duke Global Health Institute, Durham, Duke University, NC, United States
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Takamasa Kayama
- Global Center of Excellence, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Csaba P Kovesdy
- Medicine-Nephrology, Memphis Veterans Affairs Medical Center and University of Tennessee Health Science Center, Memphis, TN, United States
| | | | - Andrew S Levey
- Division of Nephrology, Tufts Medical Center, Boston, MA, United States
| | - Donald M Lloyd-Jones
- Department of Preventive Medicine, Northwestern University, Chicago, Illinois, United States
| | - Rupert W. Major
- John Walls Renal Unit, Leicester General Hospital, University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Katsuyuki Miura
- Department of Public Health, Center for Epidemiologic Research in Asia (CERA) Shiga University of Medical Science (SUMS) Seta-Tsukinowa-cho, Shiga, Japan
| | - Paul Muntner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Girish N Nadkarni
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Takayoshi Ohkubo
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Michelle J Pena
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Kevan R Polkinghorne
- Department of Nephrology, Monash Medical Centre, Monashhealth, Melbourne, Australia and Department of Medicine, and Department of Epidemiology & Preventive Medicine, Monash University, Melbourne, Australia
| | | | - Toshimi Sairenchi
- Department of Public Health, Dokkyo Medical University School of Medicine, Mibu, Japan
| | - Markus P Schneider
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Varda Shalev
- Institute for Health and Research and Innovation, Maccabi Healthcare Services and Tel Aviv University, Tel Aviv, Israel
| | - Michael Shlipak
- Department of Epidemiology and Biostatistics, University of California, San Francisco, and San Francisco VA Medical Center, San Francisco, United States
| | - Marit D Solbu
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway and UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikita Stempniewicz
- AMGA (American Medical Group Association), Alexandria, Virginia and OptumLabs Visiting Fellow, United States
| | - James Tollitt
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - José M Valdivielso
- Vascular & Renal Translational Research Group, IRBLleida, Spain and Spanish Research Network for Renal Diseases (RedInRen. ISCIII), Lleida, Spain
| | - Joep van der Leeuw
- Department of Vascular Medicine and Department of Nephrology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Angela Yee-Moon Wang
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Chi-Pang Wen
- China Medical University Hospital, Taichung, Taiwan
| | - Mark Woodward
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- George Institute for Global Health, Australia, and George Institute for Global Health, Imperial College, London, United Kingdom
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Japan
| | - Hiroshi Yatsuya
- Department of Public Health, Fujita Health University School of Medicine, Aichi, Japan
- Department of Public Health and Health Systems, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Luxia Zhang
- Peking University First Hospital and Peking University, Beijing, China
| | - Elke Schaeffner
- Institute of Public Health, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
248
|
Allen NE, Arnold M, Parish S, Hill M, Sheard S, Callen H, Fry D, Moffat S, Gordon M, Welsh S, Elliott P, Collins R. Approaches to minimising the epidemiological impact of sources of systematic and random variation that may affect biochemistry assay data in UK Biobank. Wellcome Open Res 2020; 5:222. [PMID: 33364437 PMCID: PMC7739095 DOI: 10.12688/wellcomeopenres.16171.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2020] [Indexed: 01/09/2023] Open
Abstract
Background: UK Biobank is a large prospective study that recruited 500,000 participants aged 40 to 69 years, between 2006-2010.The study has collected (and continues to collect) extensive phenotypic and genomic data about its participants. In order to enhance further the value of the UK Biobank resource, a wide range of biochemistry markers were measured in all participants with an available biological sample. Here, we describe the approaches UK Biobank has taken to minimise error related to sample collection, processing, retrieval and assay measurement. Methods: During routine quality control checks, the laboratory team observed that some assay results were lower than expected for samples acquired during certain time periods. Analyses were undertaken to identify and correct for the unexpected dilution identified during sample processing, and for expected error caused by laboratory drift of assay results. Results: The vast majority (92%) of biochemistry serum assay results were assessed to be not materially affected by dilution, with an estimated difference in concentration of less than 1% (i.e. either lower or higher) than that expected if the sample were unaffected; 8.3% were estimated to be diluted by up to 10%; very few samples appeared to be diluted more than this. Biomarkers measured in urine (creatinine, microalbumin, sodium, potassium) and red blood cells (HbA1c) were not affected. In order to correct for laboratory variation over the assay period, all assay results were adjusted for date of assay, with the exception of those that had a high biological coefficient of variation or evident seasonal variability: vitamin D, lipoprotein (a), gamma glutamyltransferase, C-reactive protein and rheumatoid factor. Conclusions: Rigorous approaches related to sample collection, processing, retrieval, assay measurement and data analysis have been taken to mitigate the impact of both systematic and random variation in epidemiological analyses that use the biochemistry assay data in UK Biobank.
Collapse
Affiliation(s)
- Naomi E. Allen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Matthew Arnold
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
| | - Sarah Parish
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, UK
| | - Michael Hill
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, UK
| | | | - Howard Callen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Daniel Fry
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | - Stewart Moffat
- MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, UK
| | - Mark Gordon
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| | | | - Paul Elliott
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Rory Collins
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, OXFORD, Oxon, OX3 7LF, UK
- UK Biobank, Stockport, Cheshire, SK3 0SA, UK
| |
Collapse
|
249
|
Fu Y, Zhang Y, Khoo BL. Liquid biopsy technologies for hematological diseases. Med Res Rev 2020; 41:246-274. [PMID: 32929726 DOI: 10.1002/med.21731] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/10/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022]
Abstract
Since the discovery of circulating tumor cells in 1869, technological advances in studying circulating biomarkers from patients' blood have made the diagnosis of nonhematologic cancers less invasive. Technological advances in the detection and analysis of biomarkers provide new opportunities for the characterization of other disease types. When compared with traditional biopsies, liquid biopsy markers, such as exfoliated bladder cancer cells, circulating cell-free DNA (cfDNA), and extracellular vesicles (EV), are considered more convenient than conventional biopsies. Liquid biopsy markers undoubtedly have the potential to influence disease management and treatment dynamics. Our main focuses of this review will be the cell-based, gene-based, and protein-based key liquid biopsy markers (including EV and cfDNA) in disease detection, and discuss the research progress of these biomarkers used in conjunction with liquid biopsy. First, we highlighted the key technologies that have been broadly adopted used in hematological diseases. Second, we introduced the latest technological developments for the specific detection of cardiovascular disease, leukemia, and coronavirus disease. Finally, we concluded with perspectives on these research areas, focusing on the role of microfluidic technology and artificial intelligence in point-of-care medical applications. We believe that the noninvasive capabilities of these technologies have great potential in the development of diagnostics and can influence treatment options, thereby advancing precision disease management.
Collapse
Affiliation(s)
- Yatian Fu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Yiyuan Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong, China
| |
Collapse
|
250
|
Zou B, Miao C, Chen J. Depression and Perceived Stress, but Not Anxiety, are Associated with Elevated Inflammation in an Obese Adult Population. Risk Manag Healthc Policy 2020; 13:1489-1497. [PMID: 32982507 PMCID: PMC7490106 DOI: 10.2147/rmhp.s270359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 08/13/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Anxiety, depression and perceived stress are risk factors for adverse health problems. Inflammation participates in the development of chronic diseases such as psychiatric disorders. This study explored the relationships between inflammatory biomarkers and depression, anxiety and perceived stress in an obese adult population. METHODS The relationships between psychological scores and inflammatory markers were analyzed. RESULTS A higher BMI was not correlated with a higher anxiety score (P=0.152); however, BMI was positively associated with a higher depression score (P<0.001) and a higher perceived stress score (P<0.001). Multivariate linear regression analysis revealed that in participants with BMI≥30 and 25≤BMI<30, depression and perceived stress were significantly and independently associated with ICAM-1, E-selectin and CRP, but these associations were not observed in participants with BMI<25. The anxiety score was not associated with any inflammatory marker in any group of subjects, as determined by multivariate analysis. CONCLUSION Depression and perceived stress were strongly associated with increased serum levels of pro-inflammatory markers, including ICAM-1, E-selectin and CRP, among a general obese population from the United States. These results further suggest that depression and perceived stress might also be chronic systemic inflammatory diseases.
Collapse
Affiliation(s)
- Bin Zou
- Department of Orthopaedics, Affiliated Mindong Hospital of Fujian Medical University, Fuan City, Fu Jian355000, People’s Republic of China
| | - Chenfang Miao
- Department of Anesthesiology, Affiliated Mindong Hospital of Fujian Medical University, Fuan City, Fu Jian355000, People’s Republic of China
| | - Jiliang Chen
- Department of Orthopaedics, Affiliated Mindong Hospital of Fujian Medical University, Fuan City, Fu Jian355000, People’s Republic of China
| |
Collapse
|